Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-16T20:33:42.569Z Has data issue: false hasContentIssue false

Lipid Membranes: Biological Inspiration for Micro and Nano Encapsulation Technologies, Especially Drug Delivery

Published online by Cambridge University Press:  15 February 2011

David Needham*
Affiliation:
Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USAd.needham@duke.edu
Get access

Abstract

Our approach to biologically inspired materials and materials systems recognizes biology (at all scale levels) as a series of products that fulfill particular functions. It then links material composition and structure to function through properties and therefore attempts to bring mechanism to processes and functions of biology. As an example of this approach we have focused on the lipid bilayer membranes of blood cells, like erythrocytes and neutrophils, as a bioinspired material system for drug delivery leading to the creation of waxy, nano capsules called liposomes that can be triggered to release their drug by hyperthermia. Thus, while Nature's encapsulation technology provides the inspiration, the mechanism of drug release is non-natural. The necessary design parameters for the required functions of drug encapsulation, i.e. drug retention, circulation half life, and eventual thermally-triggered drug release, were obtained through extensive experimentation and modeling of artificial lipid vesicles by us and others, with much of the mechanical and thermomechanical properties, molecular exchange, and in vitro performance investigated by a direct micropipet manipulation technique. With respect to cancer chemotherapy, the unmet need for primary solid tumors is to deliver more drug to the tumor tissue thereby reducing the tumor size (debulking) while at the same time reducing toxic side effects. It is with these criteria in mind that we developed the temperature-triggered liposome for the treatment of solid tumors. This paper then, describes this liposome development and its performance in vivo, where, in some cases, the temperature-triggered release of drug directly in the blood stream and tumor resulted in complete tumor regression. What this example also shows is that through material property measurement and modeling, new insights into Nature's functions and designs can be discovered in a reverse engineering process from which new products can then be forward engineered to solve engineering and product problems in health, technology, and the environment.

Type
Research Article
Copyright
Copyright © Materials Research Society 2003

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

[1] Binnig, G. Quate, C. and Gerber, C.Atomic force microscope,” Phys. Rev. Lett., vol. 56, pp. 930933, 1986.Google Scholar
[2] Drake, B. Prater, C. B. Weisenhorn, A. L. Gould, S. A. C., Albrecht, T. R. Quate, C. F. Cannell, D. S. Hansma, H. G. and Hansma, P. K.Imaging Crystals, Polymers, and Processes in Water with the Atomic Force Microscope,” Science, vol. 243, pp. 15861589, 1989.Google Scholar
[3] Ashkin, A. Schutze, K. Dziedzic, J. Euteneuer, U. and Schliwa, M.Force generation of organelle transport measured in vivo by an infrared laser trap,” Nature, vol. 348, pp. 346348, 1990.Google Scholar
[4] Ashkin, A.Forces of a single-beam gradient laser trap on a dielectric sphere in the ray optics regime,” Biophysical Journal, vol. 61, pp. 569582, 1992.Google Scholar
[5] Evans, E. Ritchie, K. and Merkel, R.Sensitive Force Technique to Probe Molecular Adhesion and Structural Linkages at Biological Interfaces,” Biophysical Journal, vol. 68, pp. 25802587, 1995.Google Scholar
[6] Evans, E.Energy landscapes of biomolecular adhesion and receptor anchoring at interfaces explored with dynamic force spectroscopy,” Faraday Discussions, vol. 111, pp. 116, 1998.Google Scholar
[7] Evans, E.Looking inside molecular bonds at biological interfaces with dynamic force spectroscopy,” Biophysical Chemistry, vol. 82, pp. 8397, 1999.Google Scholar
[8] Evans, E. and Kwok, R.Mechanical calorimetry of large dimyristoylphosphatidylcholine vesicles in the phase transition region,” Biochemistry, vol. 21, pp. 48744879, 1982.Google Scholar
[9] Needham, D. and Evans, E.Structure and mechanical properties of giant lipid (DMPC) vesicle bilayers from 20 degrees C below to 10 degrees C above the liquid crystalcrystalline phase transition at 24 degrees C,” Biochemistry, vol. 27, pp. 82618269, 1988.Google Scholar
[10] Needham, D. and Zhelev, D. V. “The Mechanochemistry of Lipid Vesicles Examined by Micropipet Manipulation Techniques,” in Vesicles, Rosoff, M. Ed. New York: Marcel Dekker, Inc., 1996, pp. 373444.Google Scholar
[11] Needham, D. and Zhelev, D. V. “Use of micropipet manipulation techniques to measure the properties of giant lipid vesicles,” in Giant Vesicles, Luisi, P. L. and Walde, P. Eds.: John Wiley & Sons, 2000, pp. 103147.Google Scholar
[12] Kwok, R. and Evans, E.Thermoelasticity of large lecithin bilayer vesicles,” Biophys. J., vol. 35, pp. 637652, 1981.Google Scholar
[13] Harrigan, P. R. Hope, M. J. Redelmeier, T. E. and Cullis, P. R.Determination of transmembrane pH gradients and membrane potentials in liposomes,” Biophys J, vol. 63, pp. 1336–45, 1992.Google Scholar
[14] Hope, M. J. Bally, M. B. Webb, G. and Cullis, P. R.Production of large unilamellar vesicles by a rapid extrusion procedure - characterization of size distribution, trapped volume and ability to maintain a membrane-potential,” Biochimica Et Biophysica Acta, vol. 812, pp. 5565, 1985.Google Scholar
[15] Mayer, L. D. Tai, L. C. Ko, D. S. Masin, D. Ginsberg, R. S. Cullis, P. R. and Bally, M. B.Influence of vesicle size, lipid composition, and drug-to-lipid ratio on the biological activity of liposomal doxorubicin in mice,” Cancer Res, vol. 49, pp. 5922–30, 1989.Google Scholar
[16] Mayer, L. D. Tai, L. C. Bally, M. B. Mitilenes, G. N. Ginsberg, R. S. and Cullis, P. R.Characterization of liposomal systems containing doxorubicin entrapped in response to pH gradients,” Biochim Biophys Acta, vol. 1025, pp. 143–51, 1990.Google Scholar
[17] Needham, D.Materials engineering of lipid bilayers for drug carrier performance,” MRS Bulletin, vol. 24, pp. 3240, 1999.Google Scholar
[18] Duwe, H. P. Engelhardt, H. Zilker, A. and Sackmann, E.Curvature elasticity of smectic A lipid bilayers and cell plasma membranes,” Mol. Cryst. Liq. Cryst., vol. 91, pp. 17, 1987.Google Scholar
[19] Faucon, J. F. Mitov, M. D. Meleard, P. Bivas, I. and Bothorel, P.Bending elasticity and thermal fluctuations of lipid membranes. Theoretical and experimental requirements,” J. Phys. France., vol. 50, pp. 23892414, 1989.Google Scholar
[20] Milner, S. T. and Safran, S. A.Dynamical fluctuations of droplet microemulsions and vesicles,” Phys. Rev. A, vol. 36, pp. 43714379, 1987.Google Scholar
[21] Schneider, M. B. Jenkins, J. T. and Webb, W. W.Thermal fluctuations of large cylindrical phospholipid vesicles,” Biophys. J., vol. 45, pp. 891899, 1984.Google Scholar
[22] Servuss, R. M. Harbich, W. and Helfrich, W.Measurement of the curvature elastic modulus of egg lecithin bilayers,” Biochim. Biophys. Acta, vol. 436, pp. 900, 1976.Google Scholar
[23] Bennett, V.The membrane skeleton of human erythrocytes and its implication for more complex cells,” Annu. Rev. Biochem., vol. 54, pp. 273304, 1985.Google Scholar
[24] Zhelev, D. V. Alteraifi, A. M. and Hochmuth, R. M.F-actin network formation in tethers and in pseudopods stimulated by chemoattractant,” Cell Motil Cytoskeleton, vol. 35, pp. 331–44, 1996.Google Scholar
[25] Zhelev, D. V. and Alteraifi, A.Signaling in the motility responses of the human neutrophil,” Ann Biomed Eng, vol. 30, pp. 356–70, 2002.Google Scholar
[26] Chodniewicz, D. and Zhelev, D. V.Chemoattractant receptor-stimulated F-actin polymerization in the human neutrophil is signaled by 2 distinct pathways,” Blood, vol. 101, pp. 1181–4, 2003.Google Scholar
[27] Rand, R. P. and Burton, A. C.Mechanical properties of the red cell membrane: II. Viscoelastic breakdown of the membrane,” Biophys. J., vol. 4, pp. 115, 1964.Google Scholar
[28] Evans, E. and Hochmuth, R. M.A solid-liquid composite model of the red blood cell membrane,” J. Membr. Biol., vol. 30, pp. 351362, 1977.Google Scholar
[29] Evans, E.Structure and deformation properties of red blood cells - Concepts and quantitative methods,” Methods In Enzymology, vol. 173, pp. 335, 1989.Google Scholar
[30] Mow, V. C. Guilak, F. Tran-Son-Tay, R., and Hochmuth, R. in Cell Mechanics and Cellular Engineering. New York: Springer-Verlag, 1994.Google Scholar
[31] Evans, E. and Skalak, R. Mechanics and thermodynamics of biomembranes. Boca Raton Florida: CRC, 1980.Google Scholar
[32] Evans, E. “Micromethods for measurement of deformability and adhesivity properties of blood cells and synthetic membrane vesicles,” in Physical Basis of Cell-Cell Adhesion, Bongrand, P. Ed. Boca Raton, Fla.: CRC Press, Inc., 1988.Google Scholar
[33] Evans, E. and Needham, D.Physical properties of surfactant bilayer membranes: thermal transitions, elasticity, rigidity, cohesion, and colloidal interactions,” J. Phys. Chem., vol. 91, pp. 42194228, 1987.Google Scholar
[34] Evans, E. and Needham, D.Giant vesicle bilayers composed of mixtures of lipids, cholesterol and polypeptides,” Faraday Discuss. Chem. Soc., vol. 81, pp. 267280, 1986.Google Scholar
[35] Evans, E. and Hochmuth, R. M. “Mechanochemical properties of membranes,” in Current Topics in Membranes and Transport, vol. 10, Bonner, F. and Kleinzeller, A. Eds. New York: Academic Press, 1978, pp. 1.Google Scholar
[36] Discher, D. E. Boal, D. H. and Boey, S. K.Simulations of the erythrocyte cytoskeleton at large deformation. II. Micropipette aspiration,” Biophys J, vol. 75, pp. 1584–97, 1998.Google Scholar
[37] Hochmuth, R. M. Ting-Beall, H. P., Beaty, B. B. Needham, D. and Tran-Son-Tay, R., “Viscosity of passive human neutrophils undergoing small deformations,” Biophys J, vol. 64, pp. 1596–601, 1993.Google Scholar
[38] Needham, D. and Hochmuth, R. M.Rapid flow of passive neutrophils into a 4 microns pipet and measurement of cytoplasmic viscosity,” J Biomech Eng, vol. 112, pp. 269–76, 1990.Google Scholar
[39] Hochmuth, R. M. and Needham, D.The viscosity of neutrophils and their transit times through small pores,” Biorheology, vol. 27, pp. 817–28, 1990.Google Scholar
[40] Chodniewicz, D. and Zhelev, D. V. “Novel pathways of F-actin polymerization in the human neutrophil,” Blood, 2003.Google Scholar
[41] Needham, D. and Nunn, R. S.Elastic deformation and failure of lipid bilayer membranes containing cholesterol,” Biophys. J., vol. 58, pp. 9971009, 1990.Google Scholar
[42] Needham, D. and Zhelev, D. V. “Mechanochemistry of lipid vesicles examined by micropipet manipulation,” in Vesicles, Rosoff, M. Ed. New York and Basel: Marcel Dekker, 1996.Google Scholar
[43] Evans, E. and Rawicz, W.Entropy-driven tension and bending elasticity in condensedfluid membranes,” Phys. Rev. Letts., vol. 64, pp. 20942097, 1990.Google Scholar
[44] Olbrich, K. Rawicz, W. Needham, D. and Evans, E.Water permeability and mechanical strength of polyunsaturated lipid bilayers,” Biophys J, vol. 79, pp. 321–7, 2000.Google Scholar
[45] Evans, E. and Ludwig, F.Dynamic strengths of molecular anchoring and material cohesion in fluid biomembranes,” Journal of Physics-Condensed Matter, vol. 12, pp. A315–A320, 2000.Google Scholar
[46] Evans, E.Probing the relation between force - Lifetime - and chemistry in single molecular bonds,” Annual Review of Biophysics and Biomolecular Structure, vol. 30, pp. 105128, 2001.Google Scholar
[47] Luzzatti, V. in Biological Membranes, Chapman, D. Ed. New York: Academic Press, 1968, pp. 71.Google Scholar
[48] Luzzatti, V. and Tardieu, A. Annu. Rev. Phys. Chem., vol. 25, pp. 79, 1974.Google Scholar
[49] Rand, R. P. Chapman, D. and Larsson, K.Tilted hydrocarbon chains of dipalmitoyl lecithin become perpendicular to the bilayer before melting,” Biophys J, vol. 15, pp. 1117–24, 1975.Google Scholar
[50] Janiak, M. J. Small, D. M. and Shipley, G. G.Nature of the thermal pretransition of synthetic phospholipids: dimyristolyl- and dipalmitoyllecithin,” Biochemistry, vol. 15, pp. 4575–80, 1976.Google Scholar
[51] Janiak, M. J. Small, D. M. and Shipley, G. G.Temperature and compositional dependence of the structure of hydrated dimyristoyl lecithin,” J Biol Chem, vol. 254, pp. 6068–78, 1979.Google Scholar
[52] Needham, D. McIntosh, T. J. and Evans, E.Thermomechanical and transition properties of dimyristoylphosphatidylcholine/cholesterol bilayers,” Biochemistry, vol. 27, pp. 46684673, 1988.Google Scholar
[53] Sarpal, R. and Needham, D.Binding of paclitaxel to lipid interfaces: Correlations with interface compliance,” J. Liposome Research, vol. 8, pp. 147163, 1998.Google Scholar
[54] Needham, D. Stoicheva, N. and Zhelev, D. V.Exchange of monooleoylphosphatidycholine as monomer and micelle with membranes containing grafted polyethyleneglycol,” Biophys. J., vol. 73, pp. 26152629, 1997.Google Scholar
[55] Gregoriadis, G.Engineering liposomes for drug delivery: progress and problems,” Trends Biotechnol, vol. 13, pp. 527–37, 1995.Google Scholar
[56] Gregoriadis, G. and Ryman, B. E.Fate of protein-containing liposomes injected into rats. An approach to the treatment of storage diseases,” Eur J Biochem, vol. 24, pp. 485–91, 1972.Google Scholar
[57] Bradley, A. J. and Devine, D. V.The complement system in liposome clearance: Can complement deposition be inhibited?,” Adv Drug Deliv Rev, vol. 32, pp. 1929, 1998.Google Scholar
[58] Chonn, A. Semple, S. C. and Cullis, P. R.Association of blood proteins with large unilamellar liposomes in vivo. Relation to circulation lifetimes,” J Biol Chem, vol. 267, pp. 18759–65, 1992.Google Scholar
[59] Harasym, T. O. Cullis, P. R. and Bally, M. B.Intratumor distribution of doxorubicin following i.v. administration of drug encapsulated in egg phosphatidylcholine/cholesterol liposomes,” Cancer Chemother Pharmacol, vol. 40, pp. 309–17, 1997.Google Scholar
[60] Semple, S. C. Chonn, A. and Cullis, P. R.Influence of cholesterol on the association of plasma proteins with liposomes,” Biochemistry, vol. 35, pp. 2521–5, 1996.Google Scholar
[61] Lasic, D. D. and Needham, D.The Stealth Liposome: A Prototypical Biomaterial,” Chemical Reviews, vol. 95, pp. 26012628, 1995.Google Scholar
[62] Yuan, F. Leunig, M. Huang, S. K. Berk, D. A. Papahadjopoulos, D. and Jain, R. K.Microvascular permeability and interstitial penetration of sterically stabilized (stealth) liposomes in a human tumor xenograft,” Cancer Res, vol. 54, pp. 3352–6, 1994.Google Scholar
[63] Gabizon, A. and Martin, F.Polyethylene glycol-coated (pegylated) liposomal doxorubicin. Rationale for use in solid tumours,” Drugs, vol. 54 Suppl 4, pp. 1521, 1997.Google Scholar
[64] Gabizon, A. A.Pegylated liposomal doxorubicin: metamorphosis of an old drug into a new form of chemotherapy,” Cancer Invest, vol. 19, pp. 424–36, 2001.Google Scholar
[65] Needham, D. McIntosh, T. J. and Zhelev, D. V. “Surface Chemistry of the Sterically Stabilized PEG-Liposome: General Principles,” in Liposomes: Rational Design, Janoff, A. Ed. New York: Marcel Dekker, Inc., 1998, pp. 1362.Google Scholar
[66] Needham, D. and Dewhirst, M. W.The development and testing of a new temperaturesensitive drug delivery system for the treatment of solid tumors,” Adv Drug Deliv Rev, vol. 53, pp. 285305, 2001.Google Scholar
[67] Mouritsen, O. G. and Zuckermann, M. J.Model of interfacial melting,” Physical Review Letters, vol. 58, pp. 389392, 1987.Google Scholar
[68] Mouritsen, O. G.Theoretical models of phospholipid phase transitions,” Chem Phys Lipids, vol. 57, pp. 179–94, 1991.Google Scholar
[69] Nielsen, M. Miao, L. Ipsen, J. H. Jorgensen, K. Zuckermann, M. J. and Mouritsen, O. G.Model of a sub-main transition in phospholipid bilayers,” Biochim Biophys Acta, vol. 1283, pp. 170–6, 1996.Google Scholar
[70] Papahadjopoulos, D. Jacobson, K. Nir, S. and Isac, T.Phase transitions in phospholipid vesicles. Fluorescence polarization and permeability measurements concerning the effect of temperature and cholesterol,” Biochim Biophys Acta, vol. 311, pp. 330–48, 1973.Google Scholar
[71] Anyarambhatla, G. R. and Needham, D.Enhancement of the phase transition permeability of DPPC liposomes by incorporation of MPPC: A new temperaturesensitive liposome for use with mild hyperthermia,” Journal of Liposome Research, vol. 9, pp. 491506, 1999.Google Scholar
[72] Mills, J. K. and Needham, D. “The materials engineering of temperature sensitive liposomes,” Methods Enzymol. in press, 2003.Google Scholar
[73] Mills, J. K. and Needham, D.Targeted drug delivery,” Expert Opinion on Therapeutic Patents, vol. 9, pp. 14991513, 1999.Google Scholar
[74] Kong, G. Anyarambhatla, G. Petros, W. P. Braun, R. D. Colvin, O. M. Needham, D. and Dewhirst, M. W.Efficacy of liposomes and hyperthermia in a human tumor xenograft model: importance of triggered drug release,” Cancer Res, vol. 60, pp. 6950–7, 2000.Google Scholar
[75] Kong, G. and Dewhirst, M. W.Hyperthermia and liposomes,” Int J Hyperthermia, vol. 15, pp. 345–70, 1999.Google Scholar
[76] Tozer, G. M. Prise, V. E. Wilson, J. Cemazar, M. Shan, S. Dewhirst, M. W. Barber, P. R. Vojnovic, B. and Chaplin, D. J.Mechanisms associated with tumor vascular shut-down induced by combretastatin A-4 phosphate: intravital microscopy and measurement of vascular permeability,” Cancer Res, vol. 61, pp. 6413–22, 2001.Google Scholar
[77] Wilson, D. F. Evans, S. M. Jenkins, W. T. Vinogradov, S. A. Ong, E. and Dewhirst, M. W.Oxygen distributions within R3230Ac tumors growing in dorsal flap window chambers in rats,” Adv Exp Med Biol, vol. 454, pp. 603–9, 1998.Google Scholar
[78] Dewhirst, M. W. Ong, E. T. Braun, R. D. Smith, B. Klitzman, B. Evans, S. M. and Wilson, D.Quantification of longitudinal tissue pO2 gradients in window chamber tumours: impact on tumour hypoxia,” Br J Cancer, vol. 79, pp. 1717–22, 1999.Google Scholar
[79] Kong, G. Braun, R. D. and Dewhirst, M. W.Characterization of the effect of hyperthermia on nanoparticle extravasation from tumor vasculature,” Cancer Res, vol. 61, pp. 3027–32, 2001.Google Scholar
[80] Needham, D. Anyarambhatla, G. Kong, G. and Dewhirst, M. W.A new temperaturesensitive liposome for use with mild hyperthermia: characterization and testing in a human tumor xenograft model,” Cancer Res, vol. 60, pp. 1197–201, 2000.Google Scholar
[81] Matteucci, M. L. Anyarambhatla, G. Rosner, G. Azuma, C. Fisher, P. E. Dewhirst, M. W. Needham, D. and Thrall, D. E.Hyperthermia increases accumulation of technetium-99m-labeled liposomes in feline sarcomas,” Clin Cancer Res, vol. 6, pp. 3748–55, 2000.Google Scholar
[82] Hobbs, S. K. Monsky, W. L. Yuan, F. Roberts, W. G. Griffith, L. Torchilin, V. P. and Jain, R. K.Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment,” Proc Natl Acad Sci U S A, vol. 95, pp. 4607–12, 1998.Google Scholar
[83] Monsky, W. L. Fukumura, D. Gohongi, T. Ancukiewcz, M. Weich, H. A. Torchilin, V. P. Yuan, F. and Jain, R. K.Augmentation of transvascular transport of macromolecules and nanoparticles in tumors using vascular endothelial growth factor,” Cancer Res, vol. 59, pp. 4129–35, 1999.Google Scholar
[84] Wu, N. Z. Klitzman, B. Rosner, G. Needham, D. and Dewhirst, M. W.Measurement of material extravasation in microvascular networks using fluorescence video-microscopy,” Microvasc Res, vol. 46, pp. 231–53, 1993.Google Scholar
[85] Wu, N. Z. Braun, R. D. Gaber, M. H. Lin, G. M. Ong, E. T. Shan, S. Papahadjopoulos, D. and Dewhirst, M. W.Simultaneous measurement of liposome extravasation and content release in tumors,” Microcirculation, vol. 4, pp. 83101, 1997.Google Scholar
[86] Gaber, M. H. Wu, N. Z. Hong, K. Huang, S. K. Dewhirst, M. W. and Papahadjopoulos, D.Thermosensitive liposomes: extravasation and release of contents in tumor microvascular networks,” Int J Radiat Oncol Biol Phys, vol. 36, pp. 1177–87, 1996.Google Scholar
[87] Kong, G. Braun, R. D. and Dewhirst, M. W.Hyperthermia enables tumor-specific nanoparticle delivery: Effect of particle size,” Cancer Res, vol. 60, pp. 4440–5, 2000.Google Scholar