51
|
Zhang Y, Arrington L, Boardman D, Davis J, Xu Y, DiFelice K, Stirdivant S, Wang W, Budzik B, Bawiec J, Deng J, Beutner G, Seifried D, Stanton M, Gindy M, Leone A. The development of an in vitro assay to screen lipid based nanoparticles for siRNA delivery. J Control Release 2013; 174:7-14. [PMID: 24240015 DOI: 10.1016/j.jconrel.2013.11.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/13/2013] [Accepted: 11/04/2013] [Indexed: 12/12/2022]
Abstract
In order to rapidly screen and select lead candidates for in vivo evaluation of lipid nanoparticles (LNPs) for systemic small interfering RNA (siRNA) delivery, an in vitro assay amenable to high-throughput screening (HTS) is developed. The strategy is to mimic the in vivo experience of LNPs after systemic administration, such as interactions with serum components, exposure to endosomal pH environments, and interactions with endosomal membrane lipids. It is postulated that the amount of siRNA released from LNPs after going through these treatments can be used as a screening tool to rank order the effectiveness of siRNA delivery by lipid nanoparticles in vivo. LNPs were incubated with 50% serum from different species (i.e. mouse, rat, or rhesus) at 37°C. The resulting samples were then reacted with anionic, endosomal-mimicking lipids at different pHs. The amount of siRNA released from LNPs was determined using spectrophotometry employing the fluorescent indicator SYBR Gold. Our results indicated that the amount of siRNA liberated was highly dependent upon the species of serum used and the pH to which it was exposed. LNPs treated with mouse serum showed higher levels of siRNA release, as did those subjected to endosomal pH (6.0), compared to physiological pH. Most interestingly, a good correlation between the amount of siRNA released and the in vivo efficacy was observed. In conclusion, an in vitro siRNA release assay was developed to screen and rank order LNPs for in vivo evaluation.
Collapse
Affiliation(s)
- Ye Zhang
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA.
| | - Leticia Arrington
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - David Boardman
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Jared Davis
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Yan Xu
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Katie DiFelice
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Steve Stirdivant
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Weimin Wang
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Brian Budzik
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Jack Bawiec
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - James Deng
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Greg Beutner
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Darla Seifried
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Matthew Stanton
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Marian Gindy
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Anthony Leone
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| |
Collapse
|
52
|
Teo BM, van der Westen R, Hosta-Rigau L, Städler B. Cell response to PEGylated poly(dopamine) coated liposomes considering shear stress. Biochim Biophys Acta Gen Subj 2013; 1830:4838-47. [DOI: 10.1016/j.bbagen.2013.06.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 06/06/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
|
53
|
Loew M, Forsythe JC, McCarley RL. Lipid nature and their influence on opening of redox-active liposomes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:6615-23. [PMID: 23698020 PMCID: PMC3778659 DOI: 10.1021/la304340e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The pathway for content release from reduction-sensitive liposomes based on a quinone-dioleoylphosphatidylethanolamine lipid conjugate (Q-DOPE) is outlined using results from fluorescent dye content release assays as well as single- and multiple-angle light scattering. Experimental observations are consistent with a shape/size change of the reduced liposomes prior to their aggregation, with subsequent near-quantitative content release achieved only when the lipid membrane experiences conditions favorable to a lamellar to an inverted hexagonal phase transition. Addition of poly(ethyleneglycol)-modified DOPE (PEG-DOPE) to the Q-DOPE liposomal formulation results in stabilization of the lipid bilayer, whereas incorporation of DOPE yields faster content release. At high DOPE concentrations, DOPE/PEG-DOPE/Q-DOPE liposomes exhibit larger content release, indicating a change in pathway for content release. The outcomes here provide a better understanding of the underlying principles of triggered liposomal content release and the potential utility of specific lipid properties for the rational design of drug delivery systems based on the novel Q-DOPE lipid.
Collapse
Affiliation(s)
| | | | - Robin L. McCarley
- CORRESPONDING AUTHOR: Telephone: (225) 578-3239. Facsimile: (225) 578-3458.
| |
Collapse
|
54
|
Polysaccharide-based micelles for drug delivery. Pharmaceutics 2013; 5:329-52. [PMID: 24300453 PMCID: PMC3834947 DOI: 10.3390/pharmaceutics5020329] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 05/09/2013] [Accepted: 05/16/2013] [Indexed: 11/23/2022] Open
Abstract
Delivery of hydrophobic molecules and proteins has been an issue due to poor bioavailability following administration. Thus, micelle carrier systems are being investigated to improve drug solubility and stability. Due to problems with toxicity and immunogenicity, natural polysaccharides are being explored as substitutes for synthetic polymers in the development of new micelle systems. By grafting hydrophobic moieties to the polysaccharide backbone, self-assembled micelles can be readily formed in aqueous solution. Many polysaccharides also possess inherent bioactivity that can facilitate mucoadhesion, enhanced targeting of specific tissues, and a reduction in the inflammatory response. Furthermore, the hydrophilic nature of some polysaccharides can be exploited to enhance circulatory stability. This review will highlight the advantages of polysaccharide use in the development of drug delivery systems and will provide an overview of the polysaccharide-based micelles that have been developed to date.
Collapse
|
55
|
Sakurai Y, Hatakeyama H, Sato Y, Hyodo M, Akita H, Harashima H. Gene silencing via RNAi and siRNA quantification in tumor tissue using MEND, a liposomal siRNA delivery system. Mol Ther 2013; 21:1195-203. [PMID: 23568259 DOI: 10.1038/mt.2013.57] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Small interfering RNA (siRNA) would be predicted to function as a cancer drug, but an efficient siRNA delivery system is required for clinical development. To address this issue, we developed a liposomal siRNA carrier, a multifunctional envelope-type nanodevice (MEND). We previously reported that a MEND composed of a pH-sensitive cationic lipid, YSK05, showed significant knockdown in both in vitro and in tumor tissue by intratumoral injection. Here, we report on the development of an in vivo siRNA delivery system that is delivered by systemic injection and an analysis of the pharmacokinetics of an intravenously administered siRNA molecule in tumor tissue. Tumor delivery of siRNA was quantified by means of stem-loop primer quantitative reverse transcriptase PCR (qRT-PCR) method. PEGylation of the YSK-MEND results in the increase in the accumulation of siRNA in tumor tissue from 0.0079% ID/g tumor to 1.9% ID/g tumor. The Administration of the MEND (3 mg siRNA/kg body weight) showed about a 50% reduction in the target gene mRNA and protein. Moreover, we verified the induction of RNA interference by 5' RACE-PCR method. The collective results reported here indicate that an siRNA carrier was developed that can deliver siRNA to a target cell in tumor tissue through an improved siRNA bioavailability.
Collapse
Affiliation(s)
- Yu Sakurai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
56
|
Wan Y, Han J, Fan G, Zhang Z, Gong T, Sun X. Enzyme-responsive liposomes modified adenoviral vectors for enhanced tumor cell transduction and reduced immunogenicity. Biomaterials 2013; 34:3020-30. [PMID: 23360783 DOI: 10.1016/j.biomaterials.2012.12.051] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 12/14/2012] [Indexed: 10/27/2022]
Abstract
Limitations of adenoviral (Ad) vectors for cancer gene therapy could be overcome by their combination with pharmaceutical technologies. Here we show that an enzyme-responsive liposomal formulation could significantly enhance the tumor cell transduction abilities and reduce the immunogenicity of Ad vectors. In the current research, the enzymatically cleavable PEG-lipids composed of a PEG/matrix metalloproteinase (MMP)-substrate peptide/cholesterol (PPC) were synthesized and characterized by (1)H NMR and TOF MS ES(+). The obtained MMP-cleavable lipids were inserted into the anionic liposomal Ad vectors (AL-Ad) by the post-insertion method. The results of in vitro infection assays indicated that the enzymatically cleavable formulation (PPC-AL-Ad) displayed a much higher gene expression than naked Ad5 and the non-cleavable PEG-lipid modified Ad vectors in tumor cells. More importantly, PPC-AL-Ad induces a lower production of neutralizing antibody and lower innate immune response, as well as significantly reduced liver toxicity in vivo. These findings suggest that PPC-AL-Ad is a promising system for gene delivery in tumor therapy.
Collapse
Affiliation(s)
- Yu Wan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, PR China
| | | | | | | | | | | |
Collapse
|
57
|
Xu H, Deng YH, Wang KQ, Chen DW. Preparation and characterization of stable pH-sensitive vesicles composed of α-tocopherol hemisuccinate. AAPS PharmSciTech 2012; 13:1377-85. [PMID: 23054989 DOI: 10.1208/s12249-012-9863-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/21/2012] [Indexed: 11/30/2022] Open
Abstract
The current study aims to develop a stable pH-sensitive drug delivery system. First, cleavable polyethylene glycol-α-tocopherol hemisuccinate (PEG-THS) was synthesized. Conventional pH-sensitive vesicles composed of the Tris salt of α-tocopherol hemisuccinate (THST) were then prepared using the detergent removal technique. The vesicles had a mean particle size of (163.8 ± 5.5) nm and a zeta potential of -74.5 ± 6.4 mV. The THST vesicles were then modified using PEG-THS or uncleavable PEG-cholesterol (PEG-CHOL) (THST/PEG-lipids, 100:6 molar ratio). The mean vesicle particle size and absolute zeta potential decreased with increasing PEG-THS proportion. When the pH was decreased, the vesicle particle size and calcein release rate increased. The THST vesicles were initially Ca(2+)-unstable but exhibited significantly improved stability after modification with PEG-THS, especially at PEG-lipid ratios above 6%. Incubation in an acid serum increased the calcein release rate of conventional THST vesicles to 45 ± 1.98% at 10 min. However, the release rate of the PEG-CHOL vesicles remained low. The calcein release rate of PEG-THS vesicles was between those of conventional and PEG-CHOL-V. Therefore, PEG-THS can protect vesicles in serum and reconstitute their pH sensitivity in acidic conditions. Cleavable PEG-THS can be used in stable pH-sensitive preparations without loss of pH sensitivity. Free calcein and conventional vesicles eliminated from the plasma soon after injection, as well as the half-life (t(1/2)) and area under the curve of PEG-THS-V encapsulating calcein, were dramatically increased. This phenomenon indicates that the use of PEG-lipid derivatives has gained a favorably long circulation effect in mice.
Collapse
|
58
|
Plaunt AJ, Courbanou MB, Cuison KD, Harmatys KM, Smith BD. Selective non-covalent triggered release from liposomes. Chem Commun (Camb) 2012; 48:8123-5. [PMID: 22772732 PMCID: PMC3417318 DOI: 10.1039/c2cc32962j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A zinc(II)-dipicolylamine coordination complex selectively associates with anionic liposomes, including sterically protected PEGylated liposomes, and causes rapid leakage of encapsulated contents.
Collapse
Affiliation(s)
| | | | | | | | - Bradley D. Smith
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
59
|
Nguyen J, Walsh CL, Motion JPM, Perttu EK, Szoka F. Controlled nucleation of lipid nanoparticles. Pharm Res 2012; 29:2236-48. [PMID: 22544683 DOI: 10.1007/s11095-012-0752-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 04/02/2012] [Indexed: 12/17/2022]
Abstract
PURPOSE We describe a nucleation-based method which allows for the generation of monodisperse lipid nanoparticles over a range of diameters. Using a set of novel zwitterionic lipids and inverse phosphocholine lipids with pKas ranging from 2 to 5, we showed how the hydrodynamic diameter of lipid nanoparticles can be systematically manipulated over a 60 nm to 500 nm size range. METHOD Lipid nanoparticles were prepared by adding an anti-solvent, such as water, to the organic phase containing the lipid components. This led to super-saturation and the spontaneous formation of particles. RESULTS The growth and final particle size was controlled by the ratio of the components in the ternary system: lipid, organic solvent and aqueous phase. Particles with diameter below 125 nm were formed under conditions where the super-saturation coefficient was between 2.3 and 20. PEG-lipid served as an efficient growth inhibitor except at very high and low lipid concentrations. Encapsulation efficiency of siRNA into lipid nanoparticles was shown to be pH-dependent and requires the protonation of the anionic carboxylate groups of the zwitterionic lipids, emphasizing the importance of electrostatic forces. CONCLUSION This process enables high encapsulation efficiency of nucleic acids and allows the size of lipid nanoparticles to be controlled.
Collapse
Affiliation(s)
- Juliane Nguyen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
60
|
Chan CL, Majzoub RN, Shirazi RS, Ewert KK, Chen YJ, Liang KS, Safinya CR. Endosomal escape and transfection efficiency of PEGylated cationic liposome-DNA complexes prepared with an acid-labile PEG-lipid. Biomaterials 2012; 33:4928-35. [PMID: 22469293 DOI: 10.1016/j.biomaterials.2012.03.038] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 03/10/2012] [Indexed: 01/27/2023]
Abstract
Cationic liposome-DNA (CL-DNA) complexes are being pursued as nonviral gene delivery systems for use in applications that include clinic trials. However, to compete with viral vectors for systemic delivery in vivo, their efficiencies and pharmacokinetics need to be improved. The addition of poly (ethylene glycol)-lipids (PEGylation) prolongs circulation lifetimes of liposomes, but inhibits cellular uptake and endosomal escape of CL-DNA complexes. We show that this limits their transfection efficiency (TE) in a manner dependent on the amount of PEG-lipid, the lipid/DNA charge ratio, and the lipid membrane charge density. To improve endosomal escape of PEGylated CL-DNA complexes, we prepared an acid-labile PEG-lipid (HPEG2K-lipid, PEG MW 2000) which is designed to lose its PEG chains at the pH of late endosomes. The HPEG2K-lipid and a similar but acid-stable PEG-lipid were used to prepare PEGylated CL-DNA complexes. TLC and dynamic light scattering showed that HPEG2K-CL-DNA complexes are stable at pH 7.4 for more than 24 h, but the PEG chains are cleaved at pH 5 within 1 h, leading to complex aggregation. The acid-labile HPEG2K-CL-DNA complexes showed enhanced TE over complexes stabilized with the acid-stable PEG-lipid. Live-cell imaging showed that both types of complexes were internalized to quantitatively similar particle distributions within the first 2 h of incubation with cells. Thus, we attribute the increased TE of the HPEG2K-CL-DNA complexes to efficient endosomal escape, enabled by the acid-labile HPEG2K-lipid which sheds its PEG chains in the low pH environment of late endosomes, effectively switching on the electrostatic interactions that promote fusion of the membranes of complex and endosome.
Collapse
Affiliation(s)
- Chia-Ling Chan
- Department of Materials, University of California, Santa Barbara, CA 93106, USA
| | | | | | | | | | | | | |
Collapse
|
61
|
Magarkar A, Karakas E, Stepniewski M, Róg T, Bunker A. Molecular Dynamics Simulation of PEGylated Bilayer Interacting with Salt Ions: A Model of the Liposome Surface in the Bloodstream. J Phys Chem B 2012; 116:4212-9. [DOI: 10.1021/jp300184z] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Aniket Magarkar
- Centre for Drug Research,
Faculty
of Pharmacy, University of Helsinki, Helsinki,
Finland
| | - Esra Karakas
- Centre for Drug Research,
Faculty
of Pharmacy, University of Helsinki, Helsinki,
Finland
| | - Michał Stepniewski
- Centre for Drug Research,
Faculty
of Pharmacy, University of Helsinki, Helsinki,
Finland
| | - Tomasz Róg
- Department of Physics, Tampere University of Technology, Tampere, Finland
| | - Alex Bunker
- Centre for Drug Research,
Faculty
of Pharmacy, University of Helsinki, Helsinki,
Finland
- Department of Chemistry, Aalto University, Espoo, Finland
| |
Collapse
|
62
|
Abstract
RNA interference (RNAi) technology represents a fundamentally new category of treatments for human disease by addressing targets that are traditionally considered undruggable with existing medicines. The major challenge for RNAi-based therapy is the delivery system that meets human therapeutic needs. Therefore, engineering vectors with good delivery efficiency and safety profile is an intense area of research. Lipid-based nanoparticles for RNAi have yielded successful advances in vivo and to an extent in clinical trials. In this review, we discuss the barriers in developing lipid-based nanoparticles for in vivo RNAi and different strategies to overcome them. Rational designs that address safety concerns and ensure effective delivery will aid the translation of engineered lipid-based nanoparticles toward the clinic in the foreseeable future.
Collapse
Affiliation(s)
- Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599-7571, USA.
| | | |
Collapse
|
63
|
Sawant RR, Torchilin VP. Challenges in development of targeted liposomal therapeutics. AAPS JOURNAL 2012; 14:303-15. [PMID: 22415612 DOI: 10.1208/s12248-012-9330-0] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/03/2012] [Indexed: 01/09/2023]
Abstract
Liposomes, phospholipid vesicles with a bilayered membrane structure, have been widely used as pharmaceutical carriers for drugs and genes, in particular for treatment of cancer. To enhance the efficacy of the liposomal drugs, drug-loaded liposomes are targeted to the tumors by means of passive (enhanced permeability and retention mediated) targeting, based on the longevity of liposomes in blood and its accumulation in pathological sites with compromised vasculature, and active targeting, based on the attachment of specific ligands to the liposomal surface to bind certain antigens on the target cells. Antibody-targeted liposomes loaded with anticancer drugs demonstrate high potential for clinical applications. This review highlights evolution of liposomes for both passive and active targeting and challenges in development of targeted liposomal therapeutics specifically antibody-targeted liposomes.
Collapse
Affiliation(s)
- Rupa R Sawant
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Mugar Building, Room 312, 360 Huntington Avenue, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
64
|
El-Sayed A, Masuda T, Akita H, Harashima H. Stearylated INF7 Peptide Enhances Endosomal Escape and Gene Expression of PEGylated Nanoparticles both In Vitro and In Vivo. J Pharm Sci 2012; 101:879-82. [DOI: 10.1002/jps.22807] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/26/2011] [Accepted: 10/14/2011] [Indexed: 11/06/2022]
|
65
|
Gindy ME, Leone AM, Cunningham JJ. Challenges in the pharmaceutical development of lipid-based short interfering ribonucleic acid therapeutics. Expert Opin Drug Deliv 2012; 9:171-82. [DOI: 10.1517/17425247.2012.642363] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
66
|
Mendonça L, Pedroso de Lima M, Simões S. Targeted lipid-based systems for siRNA delivery. J Drug Deliv Sci Technol 2012. [DOI: 10.1016/s1773-2247(12)50006-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
67
|
Bunker A. Poly(Ethylene Glycol) in Drug Delivery, Why Does it Work, and Can We do Better? All Atom Molecular Dynamics Simulation Provides Some Answers. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.phpro.2012.05.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
68
|
Velluto D, Gasbarri C, Angelini G, Fontana A. Use of simple kinetic and reaction-order measurements for the evaluation of the mechanism of surfactant-liposome interactions. J Phys Chem B 2011; 115:8130-7. [PMID: 21604798 DOI: 10.1021/jp2026187] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Surfactant-liposome interactions have been previously studied through different methods and techniques. We present here a classical physical chemistry study on liposome solutions added to destabilizing agents at concentrations well above the solubilization concentration, which enable us to draw useful and interesting conclusions about the mechanism of surfactant-induced liposomal breakdown by simply exploiting the kinetics and the reaction order of the liposomal content release. In such excess of surfactant, the mechanism of surfactant-induced rupture of the liposomes has been demonstrated to be different from that proposed for low surfactant concentrations. Thus, depending on the surfactant concentration, two prevailing processes have been evidenced: (i) a cooperative mechanism that implies the assembly of a critical number of surfactant molecules to trigger the formation of a channel and therefore the release of the liposomal content and (ii) a mechanism driven by direct interaction of the surfactant molecules with the lipids that causes the complete solubilization of the liposomes. The former mechanism occurs at low surfactant concentrations, whereas the latter occurs at higher concentrations and above the CMC of the surfactants. The effect of different guests embedded into the liposomal bilayer on the mechanism of surfactant-induced liposomal breakdown has been compared by using the second-order rate constants measured for the liposome breakdown process.
Collapse
Affiliation(s)
- Diana Velluto
- Institute for Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | | | | |
Collapse
|
69
|
Hatakeyama H, Akita H, Harashima H. A multifunctional envelope type nano device (MEND) for gene delivery to tumours based on the EPR effect: a strategy for overcoming the PEG dilemma. Adv Drug Deliv Rev 2011; 63:152-60. [PMID: 20840859 DOI: 10.1016/j.addr.2010.09.001] [Citation(s) in RCA: 495] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 08/12/2010] [Accepted: 09/03/2010] [Indexed: 10/19/2022]
Abstract
Gene and nucleic acid therapy are expected to play a major role in the next generation of medicine. We recently developed a multifunctional envelope-type nano device (MEND) for use as a novel non-viral gene delivery system. Poly(ethylene glycol) (PEG)ylation is a useful method for achieving a longer circulation time for delivery of the MEND to a tumour via the enhanced permeability and retention (EPR) effect. However, PEGylation strongly inhibits cellular uptake and endosomal escape, which results in significant loss of activity for the delivery system. For successful gene delivery for cancer treatment, the crucial issue associated with the use of PEG, the 'PEG dilemma' must be addressed. In this review, we describe the development and applications of MEND, and discuss strategies for overcoming the PEG dilemma, based on the manipulation of intracellular trafficking of cellular uptake and endosomal release using functional devices such as specific ligands, cleavable PEG systems and endosomal fusogenic/disruptic peptides.
Collapse
|
70
|
Reduction-triggered delivery using nucleoside-lipid based carriers possessing a cleavable PEG coating. J Control Release 2011; 151:123-30. [PMID: 21354443 DOI: 10.1016/j.jconrel.2011.02.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 01/28/2011] [Accepted: 02/05/2011] [Indexed: 11/22/2022]
Abstract
A new non-ionic nucleoside based lipid (DOU-SS-PEG(2000), 5'-PEG(2000)-2',3'-dioleoyluridine) featuring uridine (U) as nucleoside and 2',3'-dioleyl (DO), as lipid moieties and a poly(ethylene glycol) (PEG) thiolytic cleavable spacer for in vitro delivery of drugs is described. The PEG detachable nucleotide lipid (DOU-SS-PEG(2000)) was prepared via a convergent synthesis starting from HS-PEG-OMe and uridine. The reduction-triggered delivery using the PEG detachable nucleoside lipid DOU-SS-PEG(2000) was evaluated on both liposomal and micellar objects. The liposomes were prepared from of a mixture of DOTAU (N-[5'-(2',3'-dioleoyl)uridine]-N',N',N'-trimethylammonium tosylate), the PEG detachable nucleoside lipid DOU-SS-PEG(2000) and DOPE-rhodamine (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(lissamine rhodamine B sulfonyl ammonium salt) (60/40/0.1), whereas a mixture of 99.9% of DOU-SS-PEG(2000) and 0.1% of DOPE-rhodamine was used to prepare micelles. In addition, the supramolecular systems underwent a reduction-induced morphology transition from a micellar to vesicular states, which was characterized by DLS, zeta potential and TEM. The disulfide bond of the PEG chain was cleaved, by adding a reducing agent such as dithiothréitol (DTT), to expose the cationic surface of the liposome. The internalization of the resulting liposomes was facilitated as shown by the enhanced fluorescence signal observed in ovarian cancer cells (SKOV3) compared to the pegylated liposome. Likewise, when DTT was added to the mixture of cells incubated in the presence of DOU-SS-PEG(2000)/DOPE-rhodamine micelle, the fluorescence was observed in almost 100% of the SKOV3 cells.
Collapse
|
71
|
Yang ST, Zaitseva E, Chernomordik LV, Melikov K. Cell-penetrating peptide induces leaky fusion of liposomes containing late endosome-specific anionic lipid. Biophys J 2011; 99:2525-33. [PMID: 20959093 DOI: 10.1016/j.bpj.2010.08.029] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 07/22/2010] [Accepted: 08/02/2010] [Indexed: 01/14/2023] Open
Abstract
Cationic cell-penetrating peptides (CPPs) are a promising vehicle for the delivery of macromolecular drugs. Although many studies have indicated that CPPs enter cells by endocytosis, the mechanisms by which they cross endosomal membranes remain elusive. On the basis of experiments with liposomes, we propose that CPP escape into the cytosol is based on leaky fusion (i.e., fusion associated with the permeabilization of membranes) of the bis(monoacylglycero)phosphate (BMP)-enriched membranes of late endosomes. In our experiments, prototypic CPP HIV-1 TAT peptide did not interact with liposomes mimicking the outer leaflet of the plasma membrane, but it did induce lipid mixing and membrane leakage as it translocated into liposomes mimicking the lipid composition of late endosome. Both membrane leakage and lipid mixing depended on the BMP content and were promoted at acidic pH, which is characteristic of late endosomes. Substitution of BMP with its structural isomer, phosphatidylglycerol (PG), significantly reduced both leakage of the aqueous probe from liposomes and lipid mixing between liposomes. Although affinity of binding to TAT was similar for BMP and PG, BMP exhibited a higher tendency to support the inverted hexagonal phase than PG. Finally, membrane leakage and peptide translocation were both inhibited by inhibitors of lipid mixing, further substantiating the hypothesis that cationic peptides cross BMP-enriched membranes by inducing leaky fusion between them.
Collapse
Affiliation(s)
- Sung-Tae Yang
- Section on Membrane Biology, Laboratory of Cellular and Molecular Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
72
|
Tomatsu I, Marsden HR, Rabe M, Versluis F, Zheng T, Zope H, Kros A. Influence of pegylation on peptide-mediated liposome fusion. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm11722j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
73
|
Guo S, Huang L. Nanoparticles Escaping RES and Endosome: Challenges for siRNA Delivery for Cancer Therapy. JOURNAL OF NANOMATERIALS 2011. [PMID: 0 DOI: 10.1155/2011/742895] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Small interfering RNAs (siRNAs) technology has emerged as a promising potential treatment for viral, genetic diseases and cancers. Despite the powerful therapeutic potential of siRNA, there are challenges for developing efficient and specific delivery systems for systemic administration. There are extracellular and intracellular barriers for nanoparticle-mediated delivery. First, nanoparticles are rapidly cleared from the circulation by the reticuloendothelial system (RES). Second, following their cellular uptake, nanoparticles are trapped in endosomes/lysosomes, where siRNA would be degraded by enzymes. In this review, we describe strategies for grafting a polyethylene glycol (PEG) brush to the nanoparticles for evading RES, such that they may effectively accumulate in the tumor by the enhanced permeability and retention (EPR) effect. PEG has to shed from the nanoparticles to allow close interaction with the tumor cells. Current strategies for facilitating endosome escape, such as ion pair formation, “proton sponge effect”, destabilizing endosome membrane, and hydrophobic modification of the vector, are discussed.
Collapse
Affiliation(s)
- Shutao Guo
- Division of Molecular Pharmaceutics, School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Molecular Pharmaceutics, School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
74
|
Liu D, Hu H, Zhang J, Zhao X, Tang X, Chen D. Drug pH-Sensitive Release in Vitro and Targeting Ability of Polyamidoamine Dendrimer Complexes for Tumor Cells. Chem Pharm Bull (Tokyo) 2011; 59:63-71. [DOI: 10.1248/cpb.59.63] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Dan Liu
- School of Pharmacy, Shenyang Pharmaceutical University
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University
| | - Jie Zhang
- School of Pharmacy, Shenyang Pharmaceutical University
| | - Xiuli Zhao
- School of Pharmacy, Shenyang Pharmaceutical University
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University
| |
Collapse
|
75
|
Gasbarri C, Guernelli S, Boncompagni S, Angelini G, Siani G, De Maria P, Fontana A. Fine-tuning of POPC liposomal leakage by the use of beta-cyclodextrin and several hydrophobic guests. J Liposome Res 2010; 20:202-10. [PMID: 19848554 DOI: 10.3109/08982100903244526] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The effect of entrapped beta-cyclodextrin (beta-CD) on the stability of multilamellar vesicles (MLVs) of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), prepared by the dehydration-rehydration method, was studied by monitoring the release of 5(6)-carboxyfluorescein encapsulated into the liposomes. Different hydrophobic guests, such as Fullerene C(60), have been incorporated into the POPC bilayer in order to modify the membrane composition. The kinetic results as well as ESI-MS measurements evidenced that the destabilizing activity of beta-CD is due to the formation of beta-CD inclusion complexes and the consequent removal of selected bilayer constituents from the liposomal membrane. Hence, when beta-CD was added to the liposomes in the form of a strong, water-soluble 2:1 beta-CD/C(60) inclusion complex, such a destabilizing effect was not observed. However, the same beta-CD/C(60) inclusion complex does not form as a result of C(60) extraction from the bilayer. This may be attributed either to the overwhelming concentration of POPC with respect to C(60) and/or to the fact that C(60) is largely aggregated in the bilayer. Turbidimetric and fluorimetric determinations of lamellarity and entrapped volume of the studied MLVs provided further evidence of the alteration of the liposomal bilayer as a consequence of the addition of beta-CD and/or the presence of the studied guests.
Collapse
Affiliation(s)
- Carla Gasbarri
- Dipartimento di Scienze del Farmaco, Università G. d'Annunzio, Chieti, Italy
| | | | | | | | | | | | | |
Collapse
|
76
|
Interaction of bacterial surface layer proteins with lipid membranes: Synergysm between surface charge density and chain packing. Colloids Surf B Biointerfaces 2010; 79:191-7. [DOI: 10.1016/j.colsurfb.2010.03.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 03/29/2010] [Accepted: 03/31/2010] [Indexed: 11/20/2022]
|
77
|
Sigot V, Arndt-Jovin DJ, Jovin TM. Targeted Cellular Delivery of Quantum Dots Loaded on and in Biotinylated Liposomes. Bioconjug Chem 2010; 21:1465-72. [DOI: 10.1021/bc100054c] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Valeria Sigot
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Donna J. Arndt-Jovin
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Thomas M. Jovin
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| |
Collapse
|
78
|
Oruch R, Lund A, Pryme IF, Holmsen H. An intercalation mechanism as a mode of action exerted by psychotropic drugs: results of altered phospholipid substrate availabilities in membranes? J Chem Biol 2010; 3:67-88. [PMID: 21270935 PMCID: PMC2852515 DOI: 10.1007/s12154-009-0034-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 10/29/2009] [Accepted: 11/03/2009] [Indexed: 12/27/2022] Open
Abstract
Patients respond differently to psychotropic drugs, and this is currently a controversial theme among psychiatrists. The effects of 16 psychotropics on cell membrane parameters have been reported. These drugs belong to three major groups used in therapeutic psychiatry: antipsychotics, antidepressants, and anxiolytic/hypnotics. Human platelets, lacking dopamine (D(2)) receptors (proposed targets of most psychotropics), have been used as a cell model. Here we discuss the effects of these drugs on three metabolic phenomena and also results from Langmuir experiments. Diazepam, in contrast to the remaining drugs, had negligible effects on metabolic phenomena and had no effects in Langmuir experiments. Psychotropic drugs may work through intercalation in membrane phospholipids. It is possible that the fluidity of membranes, rich in essential fatty acids, the content being influenced by diet, could be a contributing factor to the action of psychotropics. This might in turn explain the observed major differences in therapeutic response among patients.
Collapse
Affiliation(s)
- Ramadhan Oruch
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Anders Lund
- MoodNet, Haukeland University Hospital, University of Bergen, Bergen, Norway
| | - Ian F. Pryme
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Holm Holmsen
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| |
Collapse
|
79
|
Abstract
A major limiting factor for the wide application of pH-sensitive liposomes is their recognition and sequestration by the phagocytes of the reticulo-endothelial system, which conditions a very short circulation half-life. Typically prolonged circulation of liposomes is achieved by grafting their membranes with pegylated phospholipids (PEG-lipids), which have been shown, however, to deteriorate membrane integrity on one hand and to hamper the pH-responsiveness on the other. Hence, the need for novel alternative surface modifying agents to ensure effective half-life prolongation of pH-sensitive liposomes is a subject of intensive research. A series of copolymers having short blocks of lipid-mimetic units has been shown to sterically stabilize conventional liposomes based on different phospholipids. This has prompted us to broaden their utilization to pH-sensitive liposomes, too. The present contribution gives thorough account on the chemical synthesis of these copolymers their incorporation in DOPE:CHEMs pH-sensitive liposomes and detailed explanation on the battery of techniques for the biopharmaceutical characterization of the prepared formulations in terms of pH-responsiveness, cellular internalization, in vivo pharmacokinetics and biodistribution.
Collapse
Affiliation(s)
- Denitsa Momekova
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical University-Sofia, Sofia, Bulgaria
| | | | | |
Collapse
|
80
|
Subcellular neuropharmacology: the importance of intracellular targeting. Trends Pharmacol Sci 2009; 30:203-11. [DOI: 10.1016/j.tips.2009.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 01/09/2009] [Accepted: 01/21/2009] [Indexed: 01/03/2023]
|
81
|
Boomer JA, Qualls MM, Inerowicz HD, Haynes RH, Patri VS, Kim JM, Thompson DH. Cytoplasmic delivery of liposomal contents mediated by an acid-labile cholesterol-vinyl ether-PEG conjugate. Bioconjug Chem 2009; 20:47-59. [PMID: 19072698 DOI: 10.1021/bc800239b] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An acid-cleavable PEG lipid, 1'-(4'-cholesteryloxy-3'-butenyl)-omega-methoxy-polyethylene[112] glycolate (CVEP), has been developed that produces stable liposomes when dispersed as a minor component (0.5-5 mol %) in 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE). Cleavage of CVEP at mildly acidic pHs results in dePEGylation of the latently fusogenic DOPE liposomes, thereby triggering the onset of content release. This paper describes the synthesis of CVEP via a six-step sequence starting from the readily available precursors 1,4-butanediol, cholesterol, and mPEG acid. The hydrolysis rates and release kinetics from CVEP/DOPE liposome dispersions as a function of CVEP loading, as well as the cryogenic transmission electron microscopy and pH-dependent monolayer properties of 9:91 CVEP/DOPE mixtures, also are reported. When folate receptor-positive KB cells were exposed to calcein-loaded 5:95 CVEP/DOPE liposomes containing 0.1 mol % folate-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-polyethylene[76] glycolamide (folate-PEG-DSPE), delivery of the calcein cargo to the cytoplasm of the cells was observed as determined by fluorescence microscopy and flow cytometry. Fluorescence resonance energy transfer analysis of lipid mixing in these cells was consistent with membrane-membrane fusion between the liposome and endosomal membranes.
Collapse
Affiliation(s)
- Jeremy A Boomer
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907-1393, USA
| | | | | | | | | | | | | |
Collapse
|
82
|
Mustapa MFM, Grosse SM, Kudsiova L, Elbs M, Raiber EA, Wong JB, Brain APR, Armer HEJ, Warley A, Keppler M, Ng T, Lawrence MJ, Hart SL, Hailes HC, Tabor AB. Stabilized Integrin-Targeting Ternary LPD (Lipopolyplex) Vectors for Gene Delivery Designed To Disassemble Within the Target Cell. Bioconjug Chem 2009; 20:518-32. [DOI: 10.1021/bc800450r] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- M. Firouz Mohd Mustapa
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Stephanie M. Grosse
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Laila Kudsiova
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Martin Elbs
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Eun-Ang Raiber
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - John B. Wong
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Anthony P. R. Brain
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Hannah E. J. Armer
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Alice Warley
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Melanie Keppler
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Tony Ng
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - M. Jayne Lawrence
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Stephen L. Hart
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Helen C. Hailes
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| | - Alethea B. Tabor
- Department of Chemistry, University College London, Christopher Ingold Laboratories, 20 Gordon Street, London WC1H 0AJ, Wolfson Centre for Gene Therapy of Childhood Disease, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, School of Biomedical and Health Sciences, Pharmaceutical Science Research Division, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, Centre for Ultrastructure Imaging, King’s College London, New Hunt’s House,
| |
Collapse
|
83
|
El-Sayed A, Futaki S, Harashima H. Delivery of macromolecules using arginine-rich cell-penetrating peptides: ways to overcome endosomal entrapment. AAPS JOURNAL 2009; 11:13-22. [PMID: 19125334 DOI: 10.1208/s12248-008-9071-2] [Citation(s) in RCA: 374] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Accepted: 12/07/2008] [Indexed: 11/30/2022]
Abstract
Arginine-rich cell-penetrating peptides (AR-CPPs) are very promising tools for the delivery of therapeutic macromolecules such as peptides, proteins, and nucleic acids. These peptides allow efficient internalization of the linked cargos intracellularly through the endocytic pathway. However, when linked to bulky cargos, entrapment in the endocytic vesicles is a major limitation to the application of these peptides in cytosolic delivery. Attachment of a compatible endosomal escape device is, therefore, necessary to allow cytosolic delivery of the peptide-attached cargo. This review presents different endosomal escape devices currently in application in combination with AR-CPPs. Applications of fusogenic lipids, membrane-disruptive peptides, membrane-disruptive polymers, lysosomotropic agents, and photochemical internalization to enhance the cytosolic delivery of AR-CPPs-attached cargos are presented. The properties of each system and its mechanism of action for the enhancement of endosomal escape are discussed, together with its applications for the delivery of different macromolecules in vitro and, if applicable, in vivo.
Collapse
Affiliation(s)
- Ayman El-Sayed
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-Ku, Sapporo, Hokkaido, 060-0812, Japan
| | | | | |
Collapse
|
84
|
Puri A, Loomis K, Smith B, Lee JH, Yavlovich A, Heldman E, Blumenthal R. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to clinic. Crit Rev Ther Drug Carrier Syst 2009; 26:523-80. [PMID: 20402623 PMCID: PMC2885142 DOI: 10.1615/critrevtherdrugcarriersyst.v26.i6.10] [Citation(s) in RCA: 553] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In recent years, various nanotechnology platforms in the area of medical biology, including both diagnostics and therapy, have gained remarkable attention. Moreover, research and development of engineered multifunctional nanoparticles as pharmaceutical drug carriers have spurred exponential growth in applications to medicine in the last decade. Design principles of these nanoparticles, including nanoemulsions, dendrimers, nano-gold, liposomes, drug-carrier conjugates, antibody-drug complexes, and magnetic nanoparticles, are primarily based on unique assemblies of synthetic, natural, or biological components, including but not limited to synthetic polymers, metal ions, oils, and lipids as their building blocks. However, the potential success of these particles in the clinic relies on consideration of important parameters such as nanoparticle fabrication strategies, their physical properties, drug loading efficiencies, drug release potential, and, most importantly, minimum toxicity of the carrier itself. Among these, lipid-based nanoparticles bear the advantage of being the least toxic for in vivo applications, and significant progress has been made in the area of DNA/RNA and drug delivery using lipid-based nanoassemblies. In this review, we will primarily focus on the recent advances and updates on lipid-based nanoparticles for their projected applications in drug delivery. We begin with a review of current activities in the field of liposomes (the so-called honorary nanoparticles), and challenging issues of targeting and triggering will be discussed in detail. We will further describe nanoparticles derived from a novel class of amphipathic lipids called bolaamphiphiles with unique lipid assembly features that have been recently examined as drug/DNA delivery vehicles. Finally, an overview of an emerging novel class of particles (based on lipid components other than phospholipids), solid lipid nanoparticles and nanostructured lipid carriers will be presented. We conclude with a few examples of clinically successful formulations of currently available lipid-based nanoparticles.
Collapse
Affiliation(s)
- Anu Puri
- Center for Cancer Research Nanobiology Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702-1201, USA.
| | | | | | | | | | | | | |
Collapse
|
85
|
Mozafari MR, Reed CJ, Rostron C, Hasirci V. A Review of Scanning Probe Microscopy Investigations of Liposome-DNA Complexes. J Liposome Res 2008; 15:93-107. [PMID: 16194929 DOI: 10.1081/lpr-64965] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Liposome-DNA complexes are one of the most promising systems for the protection and delivery of nucleic acids to combat neoplastic, viral, and genetic diseases. In addition, they are being used as models in the elucidation of many biological phenomena such as viral infection and transduction. In order to understand these phenomena and to realize the mechanism of nucleic acid transfer by liposome-DNA complexes, studies at the molecular level are required. To this end, scanning probe microscopy (SPM) is increasingly being used in the characterization of lipid layers, lipid aggregates, liposomes, and their complexes with nucleic acid molecules. The most attractive attributes of SPM are the potential to image samples with subnanometer spatial resolution under physiological conditions and provide information on their physical and mechanical properties. This review describes the application of scanning tunneling microscopy and atomic force microscopy, the two most commonly applied SPM techniques, in the characterisation of liposome-DNA complexes.
Collapse
Affiliation(s)
- M R Mozafari
- School of Pharmacy and Chemistry, Liverpool John Moores University, England, UK.
| | | | | | | |
Collapse
|
86
|
Xu H, Deng Y, Chen D, Qin J, Liu J, Shi L, Lu Y. Preparation and Characterization of pH-Sensitive Vesicles Made of Cholesteryl Hemisuccinate. Drug Dev Ind Pharm 2008; 34:134-41. [DOI: 10.1080/03639040701410903] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
87
|
Bally MB, Lim H, Cullis PR, Mayer LD. Controlling the Drug Delivery Attributes of Lipid-Based Drug Formulations. J Liposome Res 2008. [DOI: 10.3109/08982109809035537] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
88
|
Zareýe MH, Mozafarý MR, Hasirci V, Pýkýn E. Scanning Tunnelling Microscopy Investigation of Liposome-DNA-Ca2+ Complexes. J Liposome Res 2008. [DOI: 10.3109/08982109709035518] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
89
|
Mori A, Chonn A, Choi LS, Israels A, Monck MA, Cullis PR. Stabilization and Regulated Fusion of Liposomes Containing a Cationic Lipid Using Amphipathic Polyethyleneglycol Derivatives. J Liposome Res 2008. [DOI: 10.3109/08982109809035526] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
90
|
Litzinger DC. Limitations of Cationic Liposomes for Antisense Oligonucleotide Delivery in Vivo. J Liposome Res 2008. [DOI: 10.3109/08982109709035481] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
91
|
Whateley TL. Literature Alerts. Drug Deliv 2008. [DOI: 10.3109/10717549609029464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
92
|
Holland JW, Deeth HC, Alewood PF. Analysis of disulphide linkages in bovine κ-casein oligomers using two-dimensional electrophoresis. Electrophoresis 2008; 29:2402-10. [DOI: 10.1002/elps.200700840] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
93
|
MacLachlan I. Lipid-Mediated in vivo Delivery of Small Interfering RNAs. THERAPEUTIC OLIGONUCLEOTIDES 2008. [DOI: 10.1039/9781847558275-00241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Ian MacLachlan
- Protiva Biotherapeutics Inc. 100-3480 Gilmore Way Burnaby BC Canada V5G 4Y1
| |
Collapse
|
94
|
Long-circulating, pH-sensitive liposomes sterically stabilized by copolymers bearing short blocks of lipid-mimetic units. Eur J Pharm Sci 2007; 32:308-17. [PMID: 17928206 DOI: 10.1016/j.ejps.2007.08.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 06/28/2007] [Accepted: 08/27/2007] [Indexed: 11/24/2022]
Abstract
A major hurdle towards in vivo utilization of pH-sensitive liposomes is their prompt sequestration by reticuloendothelial system and hence short circulation time. Prolonged circulation of liposomes is usually achieved by incorporation of pegylated lipids, which have been frequently reported to deteriorate the acid-triggered release. In this study we evaluate the ability of four novel nonionic copolymers, bearing short blocks of lipid-mimetic units to provide steric stabilization of DOPE:CHEMs liposomes. The vesicles were prepared using the lipid film hydration method and extrusion, yielding liposomes of 120-160 nm in size. Their pH-sensitivity was monitored via the release of encapsulated calcein. The incorporation of the block copolymers at concentration up to 10 mol% did not deteriorate the pH-sensitivity of the liposomes. A selected formulation was tested for stability in presence of 25% human plasma and proved to significantly outclass the plain DOPE:CHEMs liposomes. The ability of calcein-loaded liposomes to deliver their cargo inside EJ cells was investigated using fluorescent microscopy and the results show that the surface-modified vesicles are as effective to ensure intracellular delivery as plain liposomes. The pharmacokinetics and organ distribution of a selected formulation, containing a copolymer bearing four lipid anchors was investigated in comparison to plain liposomes and PEG (2000)-DSPE stabilized liposomes. The juxtaposition of the blood clearance curves and the calculated pharmacokinetic parameters show that the block copolymer confers superior longevity in vivo. The block copolymers utilized in this study can be consider as promising sterically stabilizing agents for pH-sensitive liposomes.
Collapse
|
95
|
Cummings JE, Vanderlick TK. Aggregation and hemi-fusion of anionic vesicles induced by the antimicrobial peptide cryptdin-4. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:1796-804. [PMID: 17531950 DOI: 10.1016/j.bbamem.2007.04.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 04/20/2007] [Accepted: 04/20/2007] [Indexed: 10/23/2022]
Abstract
We show that cryptdin-4 (Crp4), an antimicrobial peptide found in mice, induces the aggregation and hemi-fusion of charged phospholipid vesicles constructed of the anionic lipid POPG and the zwitterionic lipid POPC. Hemi-fusion is confirmed with positive total lipid-mixing assay results, negative inner monolayer lipid-mixing assay results, and negative results from contents-mixing assays. Aggregation, as quantified by absorbance and dynamic light scattering, is self-limiting, creating finite-sized vesicle assemblies. The rate limiting step in the formation process is the mixing of juxtaposed membrane leaflets, which is regulated by bound peptide concentration as well as vesicle radius (with larger vesicles less prone to hemi-fusion). Bound peptide concentration is readily controlled by total peptide concentration and the fraction of anionic lipid in the vesicles. As little as 1% PEGylated lipid significantly reduces aggregate size by providing a steric barrier for membrane apposition. Finally, as stable hemi-fusion is a rare occurrence, we compare properties of Crp4 to those of many peptides known to induce complete fusion and lend insight into conditions necessary for this unusual type of membrane merger.
Collapse
Affiliation(s)
- Jason E Cummings
- Department of Chemical Engineering, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
96
|
Romberg B, Hennink WE, Storm G. Sheddable coatings for long-circulating nanoparticles. Pharm Res 2007; 25:55-71. [PMID: 17551809 PMCID: PMC2190344 DOI: 10.1007/s11095-007-9348-7] [Citation(s) in RCA: 393] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Accepted: 05/14/2007] [Indexed: 11/26/2022]
Abstract
Nanoparticles, such as liposomes, polymeric micelles, lipoplexes and polyplexes are frequently studied as targeted drug carrier systems. The ability of these particles to circulate in the bloodstream for a prolonged period of time is often a prerequisite for successful targeted delivery. To achieve this, hydrophilic ‘stealth’ polymers, such as poly(ethylene glycol) (PEG), are used as coating materials. Such polymers shield the particle surface and thereby reduce opsonization by blood proteins and uptake by macrophages of the mononuclear phagocyte system. Yet, after localizing in the pathological site, nanoparticles should deliver their contents in an efficient manner to achieve a sufficient therapeutic response. The polymer coating, however, may hinder drug release and target cell interaction and can therefore be an obstacle in the realization of the therapeutic response. Attempts have been made to enhance the therapeutic efficacy of sterically stabilized nanoparticles by means of shedding, i.e. a loss of the coating after arrival at the target site. Such an ‘unmasking’ process may facilitate drug release and/or target cell interaction processes. This review presents an overview of the literature regarding different shedding strategies that have been investigated for the preparation of sterically stabilized nanoparticulates. Detach mechanisms and stimuli that have been used are described.
Collapse
Affiliation(s)
- Birgit Romberg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Sorbonnelaan 16, 3584 CA Utrecht, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Sorbonnelaan 16, 3584 CA Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Sorbonnelaan 16, 3584 CA Utrecht, The Netherlands
| |
Collapse
|
97
|
Xianghua Y, Zirong X. The use of immunoliposome for nutrient target regulation (a review). Crit Rev Food Sci Nutr 2006; 46:629-38. [PMID: 17092828 DOI: 10.1080/10408390500507167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Although research on the role of genetically engineered antibodies and liposomes in the immunology or the nutrition field is extensive, there is no case for immunoliposome to nutrient target regulation. It is known that liposomes are spherical particles that encapsulate a fraction of the solvent, in which they freely diffuse (float) into their interior. Therefore, identification of immunoliposomes in hypothalamic site or intestinal epithelial cells that are differentially regulated by liposomes encapsulating nutrients or drugs will be an important step toward understanding the role of immunoliposomes in nutrition regulation progression and ingredient stability. Consequently, a useful model (immunoliposomal nutrient delivery system, ILNDS) of nutrient target regulation via immunoliposomes is designed to regulate the endocrine system effectively. This review focuses on antibody libraries' construction, display and selection, a brief introduction of immunoliposome, and how to use ILNDS for nutrient target regulation.
Collapse
Affiliation(s)
- Yan Xianghua
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, P.R. China.
| | | |
Collapse
|
98
|
Wasungu L, Hoekstra D. Cationic lipids, lipoplexes and intracellular delivery of genes. J Control Release 2006; 116:255-64. [PMID: 16914222 DOI: 10.1016/j.jconrel.2006.06.024] [Citation(s) in RCA: 406] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Accepted: 06/21/2006] [Indexed: 11/30/2022]
Abstract
As a consequence of several setbacks encountered by viral technology in achieving efficient and safe gene therapy in clinical trials, non-viral gene delivery vectors are considered to date as a valuable alternative and to hold promise for future therapeutic applications. Nevertheless, the transfection efficiency mediated by these non-viral gene delivery vectors has to be improved, especially in vivo, to benefit fully from their advantages. Cationic lipid/nucleic acid complexes or lipoplexes have been the subject of intensive investigations in recent years to understand the parameters governing the efficiency of transfection. Specifically, the comprehension of such mechanisms, from the formation of the complexes to their intracellular delivery, will lead to the design of better adapted non-viral vectors for gene therapy applications. Here, we will discuss some recent developments in the field on the structure/function relationship of cationic lipids in the mechanism of transfection, and where appropriate, we will make a comparison with mechanisms of viral and polyplex-mediated gene delivery. Cationic lipids are often used in combination with helper lipids such as DOPE or cholesterol. The effect of DOPE on lipoplex assembly and the relevance of the structural properties of the lipoplexes in destabilizing endosomal membranes and mediating endosomal escape of DNA will be discussed.
Collapse
Affiliation(s)
- Luc Wasungu
- Department of Cell Biology, Section Membrane Cell Biology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | |
Collapse
|
99
|
Thorén PEG, Persson D, Lincoln P, Nordén B. Membrane destabilizing properties of cell-penetrating peptides. Biophys Chem 2005; 114:169-79. [PMID: 15829350 DOI: 10.1016/j.bpc.2004.11.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2004] [Revised: 11/25/2004] [Accepted: 11/26/2004] [Indexed: 11/28/2022]
Abstract
Although cell-penetrating peptides (CPPs), also denoted protein transduction domains (PTDs), have been widely used for intracellular delivery of large and hydrophilic molecules, the mechanism of uptake is still poorly understood. In a recent live cell study of the uptake of penetratin and tryptophan-containing analogues of Tat(48-60) and oligoarginine, denoted TatP59W, TatLysP59W and R(7)W, respectively, it was found that both endocytotic and non-endocytotic uptake pathways are involved [Thoren et al., Biochem. Biophys. Res. Commun. 307 (2003) 100-107]. Non-endocytotic uptake was only observed for the arginine-rich peptides TatP59W and R(7)W. In this paper, the interactions of penetratin, R(7)W, TatP59W and TatLysP59W with phospholipid vesicles are compared in the search for an understanding of the mechanisms for cellular uptake. While R(7)W, TatP59W and TatLysP59W are found to promote vesicle fusion, indicated by mixing of membrane components, penetratin merely induces vesicle aggregation. Studies of the leakage from dye-loaded vesicles indicate that none of the peptides forms membrane pores and that vesicle fusion is not accompanied by leakage of the aqueous contents of the vesicles. These observations are important for a proper interpretation of future experiments on the interactions of these peptides with model membranes. We suggest that the discovered variations in propensity to destabilize phospholipid bilayers between the peptides investigated, in some cases sufficient to induce fusion, may be related to their different cellular uptake properties.
Collapse
Affiliation(s)
- Per E G Thorén
- Department of Chemistry and Bioscience, Chalmers University of Technology, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
100
|
Ambegia E, Ansell S, Cullis P, Heyes J, Palmer L, MacLachlan I. Stabilized plasmid-lipid particles containing PEG-diacylglycerols exhibit extended circulation lifetimes and tumor selective gene expression. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2005; 1669:155-63. [PMID: 15893518 DOI: 10.1016/j.bbamem.2005.02.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 02/01/2005] [Accepted: 02/02/2005] [Indexed: 11/19/2022]
Abstract
Stabilized plasmid lipid particles (SPLP) consist of a single copy of DNA surrounded by a lipid bilayer. The particles are small ( approximately 100 nm), stable, monodisperse and have a low surface charge. A diffusible polyethylene glycol (PEG) coating attached to a lipid anchor is critical to the SPLP's functionality. The PEG-lipid exchanges out of the bilayer at a rate determined by the size of the lipid anchor. Here we show that SPLP can be prepared using a series of PEG-diacylglycerol lipids (PEG-S-DAGs). SPLP were prepared incorporating PEG-dimyristoylglycerol (C14), PEG-dipalmitoylglycerol (C16) or PEG-distearoylglycerol (C18) and the rate of PEG-lipid diffusion from the bi-layer determined using a FRET assay. SPLP pharmacokinetics confirm a correlation between the stability of the PEG-lipid component and circulation lifetime. PEG-S-DAGs with longer lipid anchors yield more stable SPLP particles with longer circulation half-lives yielding an increase in tumor delivery and gene expression. PEG-distearoylglycerol (C18) containing SPLP bypass so-called 'first pass' organs, including the lung, and elicit levels of gene expression in distal tumor tissue 100- to 1000-fold greater than that observed in any other tissue. The incorporation of PEG-S-DAG in SPLP confirms that small size, low surface charge and extended circulation lifetimes are prerequisite to the accumulation and tumor selective expression of plasmid DNA following systemic administration.
Collapse
Affiliation(s)
- E Ambegia
- Protiva Biotherapeutics Inc., 100-3480 Gilmore Way, Burnaby, BC, Canada V5G 4Y1
| | | | | | | | | | | |
Collapse
|