Generic placeholder image

Pharmaceutical Nanotechnology

Editor-in-Chief

ISSN (Print): 2211-7385
ISSN (Online): 2211-7393

Research Article

Scalable Manufacturing Processes for Solid Lipid Nanoparticles

Author(s): Giulia Anderluzzi, Gustavo Lou, Yang Su and Yvonne Perrie*

Volume 7, Issue 6, 2019

Page: [444 - 459] Pages: 16

DOI: 10.2174/2211738507666190925112942

Abstract

Background: Solid lipid nanoparticles offer a range of advantages as delivery systems but they are limited by effective manufacturing processes.

Objective: In this study, we outline a high-throughput and scalable manufacturing process for solid lipid nanoparticles.

Methods: The solid lipid nanoparticles were formulated from a combination of tristearin and 1,2-Distearoyl-phosphatidylethanolamine-methyl-polyethyleneglycol conjugate-2000 and manufactured using the M-110P Microfluidizer processor (Microfluidics Inc, Westwood, Massachusetts, US).

Results: The manufacturing process was optimized in terms of the number of process cycles (1 to 5) and operating pressure (20,000 to 30,000 psi). The solid lipid nanoparticles were purified using tangential flow filtration and they were characterized in terms of their size, PDI, Z-potential and protein loading. At-line particle size monitoring was also incorporated within the process. Our results demonstrate that solid lipid nanoparticles can be effectively manufactured using this process at pressures of 20,000 psi with as little as 2 process passes, with purification and removal of non-entrapped protein achieved after 12 diafiltration cycles. Furthermore, the size could be effectively monitored at-line to allow rapid process control monitoring and product validation.

Conclusion: Using this method, protein-loaded solid lipid nanoparticles containing a low (1%) and high (16%) Pegylation were manufactured, purified and monitored for particle size using an at-line system demonstrating a scalable process for the manufacture of these nanoparticles.

Keywords: High-throughput manufacturing, microfluidizer processor, protein delivery, solid lipid nanoparticles, microfluidics, tangential flow filtration.

Graphical Abstract
[1]
Zur-Mühlen A. Solid lipid nanoparticles (SLN) for controlled drug delivery - drug release and release mechanism. Eur J Pharm Biopharm 1998; 422: 149-55.
[2]
Müller RH, Mehnert W, Lucks JS, et al. Solid lipid nanoparticles (SLN)-an alternative colloidal carrier system for controlled drug delivery. Eur J Pharm Biopharm 1995; 41: 62-9.
[3]
Müller RH, Radtke M, Wissing SA. Nanostructured lipid matrices for improved microencapsulation of drugs. Int J Pharm 2002; 242(1-2): 121-8.
[http://dx.doi.org/10.1016/S0378-5173(02)00180-1] [PMID: 12176234]
[4]
Siekmann B, Westesen K. Sub-micron sized parenteral carrier systems based on solid lipid. Pharm Pharmacol Lett 1992; 1: 123-6.
[5]
Müller RH, Mäder K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery - a review of the state of the art. Eur J Pharm Biopharm 2000; 50(1,3): 161-77.
[6]
Linstrom PJ, Mallard WG. NIST Chemistry WebBook. National Institute of Standards and Technology: Washington 1997.
[7]
Abou-Saleh RH, Swain M, Evans SD, Thomson NH. Poly(ethylene glycol) lipid-shelled microbubbles: abundance, stability, and mechanical properties. Langmuir 2014; 30(19): 5557-63.
[http://dx.doi.org/10.1021/la404804u] [PMID: 24758714]
[8]
Kaur R, Bramwell VW, Kirby DJ, Perrie Y. Manipulation of the surface pegylation in combination with reduced vesicle size of cationic liposomal adjuvants modifies their clearance kinetics from the injection site, and the rate and type of T cell response. J Control Release 2012; 164(3): 331-7.
[http://dx.doi.org/10.1016/j.jconrel.2012.07.012] [PMID: 22800572]
[9]
Gref R, Lück M, Quellec P, et al. ‘Stealth’ corona-core nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption. Colloids Surf B Biointerfaces 2000; 18(3-4): 301-13.
[http://dx.doi.org/10.1016/S0927-7765(99)00156-3] [PMID: 10915952]
[10]
Battaglia L, Gallarate M, Panciani PP, et al. Techniques for the preparation of solid lipid nano and microparticles. Nanomater Nanotechnol 2015; 2: 51-75.
[11]
Qi C, Chen Y, Jing QZ, Wang XG. Preparation and characterization of catalase-loaded solid lipid nanoparticles protecting enzyme against proteolysis. Int J Mol Sci 2011; 12(7): 4282-93.
[http://dx.doi.org/10.3390/ijms12074282] [PMID: 21845078]
[12]
Qi C, Chen Y, Huang JH, Jin QZ, Wang XG. Preparation and characterization of catalase-loaded solid lipid nanoparticles based on soybean phosphatidylcholine. J Sci Food Agric 2012; 92(4): 787-93.
[http://dx.doi.org/10.1002/jsfa.4646] [PMID: 22101976]
[13]
Becker Peres L, Becker Peres L, de Araújo PHH, Sayer C. Solid lipid nanoparticles for encapsulation of hydrophilic drugs by an organic solvent free double emulsion technique. Colloids Surf B Biointerfaces 2016; 140(140): 317-23.
[http://dx.doi.org/10.1016/j.colsurfb.2015.12.033] [PMID: 26764112]
[14]
Almeida AJ, Runge S, Müller RH. Peptide-loaded solid lipid nanoparticles (SLN): influence of production parameters. Int J Pharm 1997; 149: 255-65.
[http://dx.doi.org/10.1016/S0378-5173(97)04885-0]
[15]
Müller RH, Ravelli V, Runge S. Pharmaceutical cyclosporin formulation with improved biopharmaceutical properties, improved physical quality and greater stability, and method for producing said formulation United States patent US 6,551,619 . 1998.
[16]
Zhang Q, Yie G, Li Y, Q Yang, Nagai T. Studies on the cyclosporine a loaded stearic acid nanoparticles. Int J Pharm 2000; 200: 153-9.
[17]
Morel S, Ugazio E, Cavalli R, Gasco MR. Thymopentin in solid lipid nanoparticles. Int J Pharm 1996; 132: 259-61.
[http://dx.doi.org/10.1016/0378-5173(95)04388-8]
[18]
Ugazio E, Cavalli R, Gasco MR. Incorporation of cyclosporin A in solid lipid nanoparticles (SLN). Int J Pharm 2002; 241(2): 341-4.
[http://dx.doi.org/10.1016/S0378-5173(02)00268-5] [PMID: 12100861]
[19]
Preparation of solid lipid nanoparticles by microemulsion technique. Mao SR, Wang YZ, Ji HY. Bi DZ 2003 Aug; 38(8): 624-6.
[20]
Santos ID, Richard J, Pech B, Thies C, Benoit JP. Microencapsulation of protein particles within lipids using a novel supercritical fluid process. Int J Pharm 2002; 242: 69-78.
[http://dx.doi.org/10.1016/S0378-5173(02)00149-7]
[21]
Salmaso S, Bersani S, Elvassore N, Bertucco A, Caliceti P. Biopharmaceutical characterisation of insulin and recombinant human growth hormone loaded lipid submicron particles produced by supercritical gas micro-atomisation. Int J Pharm 2009; 379(1): 51-8.
[http://dx.doi.org/10.1016/j.ijpharm.2009.06.014] [PMID: 19545616]
[22]
Salmaso S, Elvassore N, Bertucco A, Caliceti P. Production of solid lipid submicron particles for protein delivery using a novel supercritical gas-assisted melting atomization process. J Pharm Sci 2009; 98(2): 640-50.
[http://dx.doi.org/10.1002/jps.21434] [PMID: 18484622]
[23]
Hu FQ, Hong Y, Yuan H. Preparation and characterization of solid lipid nanoparticles containing peptide. Int J Pharm 2004; 273(1-2): 29-35.
[http://dx.doi.org/10.1016/j.ijpharm.2003.12.016] [PMID: 15010127]
[24]
Zhang N, Ping Q, Huang G, Xu W, Cheng Y, Han X. Lectin-modified solid lipid nanoparticles as carriers for oral administration of insulin. Int J Pharm 2006; 327(1-2): 153-9.
[http://dx.doi.org/10.1016/j.ijpharm.2006.07.026] [PMID: 16935443]
[25]
Reithmeier H, Herrmann J, Göpferich A. Lipid microparticles as a parenteral controlled release device for peptides. J Control Release 2001; 73(2-3): 339-50.
[http://dx.doi.org/10.1016/S0168-3659(01)00354-6] [PMID: 11516510]
[26]
Mayhew E, Lazo R, Vail W, King J, Green A. Characterization of liposomes prepared using a microemulsifier. Biochim Biophys Acta 1984; 775: 169-74.
[http://dx.doi.org/10.1016/0005-2736(84)90167-6]
[27]
Washington C, Davis S. The production of parenteral feeding emulsions by microfluidizer. Int J Pharm 1988; 44: 169-76.
[http://dx.doi.org/10.1016/0378-5173(88)90113-5]
[28]
Saheki A, Seki J, Nakanishi T, Tamai I. Effect of back pressure on emulsification of lipid nanodispersions in a high-pressure homogenizer. Int J Pharm 2012; 422(1-2): 489-94.
[http://dx.doi.org/10.1016/j.ijpharm.2011.10.060] [PMID: 22108638]
[29]
Mahdi JS, He Y, Bhandari B. Nano-emulsion production by sonication and microfluidization - a comparison. Int J Food Prop 2006; 9: 475-85.
[http://dx.doi.org/10.1080/10942910600596464]
[30]
Sorgi FL, Huang L. Large scale production of DC-Chol cationic liposomes by microfluidization. Int J Pharm 1996; 144: 131-9.
[http://dx.doi.org/10.1016/S0378-5173(96)04733-3]
[31]
Bodmeier R, Huagang C. Indomethacin polymeric nanosuspensions prepared by microfujidization. J Control Release 1990; 12: 223-33.
[http://dx.doi.org/10.1016/0168-3659(90)90103-Z]
[32]
Sani SN, Das NG, Das SK. Effect of microfluidization parameters on the physical properties of PEG-PLGA nanoparticles prepared using high pressure microfluidization. J Microencapsul 2009; 26(6): 556-61.
[http://dx.doi.org/10.1080/02652040802500655] [PMID: 18946802]
[33]
Siqueira G, Bras J, Dufresne A. New process of chemical grafting of cellulose nanoparticles with a long chain isocyanate. Langmuir 2010; 26(1): 402-11.
[http://dx.doi.org/10.1021/la9028595] [PMID: 19921797]
[34]
Takahashi M, Kitamoto D, Asikin Y, Takara K, Wada K. Liposomes encapsulating Aloe vera leaf gel extract significantly enhance proliferation and collagen synthesis in human skin cell lines. J Oleo Sci 2009; 58(12): 643-50.
[http://dx.doi.org/10.5650/jos.58.643] [PMID: 19915322]
[35]
Thompson AK, Singh H. Preparation of liposomes from milk fat globule membrane phospholipids using a microfluidizer. J Dairy Sci 2006; 89(2): 410-9.
[http://dx.doi.org/10.3168/jds.S0022-0302(06)72105-1] [PMID: 16428611]
[36]
Laye C, McClements DJ, Weiss J. Formation of biopolymer-coated liposomes by electrostatic deposition of chitosan. J Food Sci 2008; 73(5): N7-N15.
[http://dx.doi.org/10.1111/j.1750-3841.2008.00747.x] [PMID: 18577008]
[37]
Gibis M, Zeeb B, Weiss J. Formation, characterization, and stability of encapsulated hibiscus extract in multilayered liposomes. Food Hydrocoll 2014; 38: 28-39.
[http://dx.doi.org/10.1016/j.foodhyd.2013.11.014]
[38]
Lajunen T, Hisazumi K, Kanazawa T, et al. Topical drug delivery to retinal pigment epithelium with microfluidizer produced small liposomes. Eur J Pharm Sci 2014; 62: 23-32.
[http://dx.doi.org/10.1016/j.ejps.2014.04.018] [PMID: 24810393]
[39]
Gavi E, Kubicki D, Padron GA, Özcan-Taskın NG. Breakup of nanoparticle clusters using microfluidizer M110-P. Chem Eng Res Des 2018; 132: 902-12.
[http://dx.doi.org/10.1016/j.cherd.2018.01.011]
[40]
Dalwadi G, Sunderland VB. Purification of PEGylated nanoparticles using tangential flow filtration (TFF). Drug Dev Ind Pharm 2007; 33: 1030-9.
[41]
Salminen H, Helgason T, Kristinsson B, Kristbergsson K, Weiss J. Tuning of shell thickness of solid lipid particles impacts the chemical stability of encapsulated x-3 fish oil. J Colloid Interface Sci 2017; 490: 207-16.
[42]
Asumadu-Mensah A, Smith KW, Ribeiro HS. Solid lipid dispersions: potential delivery system for functional ingredients in foods. J Food Sci 2013; 78(7): E1000-8.
[http://dx.doi.org/10.1111/1750-3841.12162] [PMID: 23865449]
[43]
Fox CB, Mulligan SK, Sung J, et al. Cryogenic transmission electron microscopy of recombinant tuberculosis vaccine antigen with anionic liposomes reveals formation of flattened liposomes. Int J Nanomedicine 2014; 9: 1367-77.
[http://dx.doi.org/10.2147/IJN.S56582] [PMID: 24648734]
[44]
Cui J, Li C, Guo W, et al. Direct comparison of two pegylated liposomal doxorubicin formulations: is AUC predictive for toxicity and efficacy? J Control Release 2007; 118(2): 204-15.
[http://dx.doi.org/10.1016/j.jconrel.2006.12.002] [PMID: 17239468]
[45]
Salminen H, Helgason T, Kristinsson B, Kristbergsson K, Weiss J. Tuning of shell thickness of solid lipid particles impacts the chemical stability of encapsulated ω-3 fish oil. J Colloid Interface Sci 2017; 490: 207-16.
[http://dx.doi.org/10.1016/j.jcis.2016.11.063] [PMID: 27912119]
[46]
He Z, Hu Y, Nie T, et al. Size-controlled lipid nanoparticle production using turbulent mixing to enhance oral DNA delivery. Acta Biomater 2018; 81: 195-207.
[http://dx.doi.org/10.1016/j.actbio.2018.09.047] [PMID: 30267888]
[47]
Dalwadi G, Sunderland VB. Purification of PEGylated nanoparticles using tangential flow filtration (TFF). Drug Dev Ind Pharm 2007; 33(9): 1030-9.
[http://dx.doi.org/10.1080/03639040601180143] [PMID: 17891590]
[48]
Dalwadi G, Benson HA, Chen Y. Comparison of diafiltration and tangential flow filtration for purification of nanoparticle suspensions. Pharm Res 2005; 22(12): 2152-62.
[http://dx.doi.org/10.1007/s11095-005-7781-z] [PMID: 16151669]
[49]
Dimov N, Kastner E, Hussain M, Perrie Y, Szita N. Formation and purification of tailored liposomes for drug delivery using a module-based micro continuous-flow system. Sci Rep 2017; 7(1): 12045.
[http://dx.doi.org/10.1038/s41598-017-11533-1] [PMID: 28935923]
[50]
Conrad ML, Yildirim AO, Sonar SS, et al. Comparison of adjuvant and adjuvant-free murine experimental asthma models. Clin Exp Allergy 2009; 39(8): 1246-54.
[http://dx.doi.org/10.1111/j.1365-2222.2009.03260.x] [PMID: 19438585]
[51]
Gupta A, Khajuria A, Singh J, Singh S, Suri KA, Qazi GN. Immunological adjuvant effect of Boswellia serrata (BOS 2000) on specific antibody and cellular response to ovalbumin in mice. Int Immunopharmacol 2011; 11: 968-75.
[52]
Almeida AJ, Runge S, Muller RH. Peptide-loaded solid lipid nanoparticles (SLN): influence of production parameters. Int J Pharm 1997; 149: 255-65.
[53]
Andreas K, Zehbe R, Kazubek M, et al. Biodegradable insulin-loaded PLGA microspheres fabricated by three different emulsification techniques: investigation for cartilage tissue engineering. Acta Biomater 2011; 7(4): 1485-95.
[http://dx.doi.org/10.1016/j.actbio.2010.12.014] [PMID: 21168535]
[54]
van de Weert M, Hennink WE, Jiskoot W. Protein instability in poly (lactic-co-glycolic acid) microparticles. Pharm Res 2000; 17(10): 1159-67.
[http://dx.doi.org/10.1023/A:1026498209874] [PMID: 11145219]
[55]
Hong SS, Kim SH, Lim SJ. Effects of triglycerides on the hydrophobic drug loading capacity of saturated phosphatidylcholine-based liposomes. Int J Pharm 2015; 483(1-2): 142-50.
[http://dx.doi.org/10.1016/j.ijpharm.2015.02.013] [PMID: 25667981]
[56]
Colletier JP, Chaize B, Winterhalter M, Fournier D. Protein encapsulation in liposomes: efficiency depends on interactions between protein and phospholipid bilayer. BMC Biotechnol 2002; 2: 9-20.
[http://dx.doi.org/10.1186/1472-6750-2-9] [PMID: 12003642]
[57]
Manjunath K, Reddy JS, Venkateswarlu V. Solid lipid nanoparticles as drug delivery systems. Methods Find Exp Clin Pharmacol 2005; 27(2): 127-44.
[http://dx.doi.org/10.1358/mf.2005.27.2.876286] [PMID: 15834465]
[58]
del Pozo-Rodríguez A, Solinís MA, Gascón AR, Pedraz JL. Short- and long-term stability study of lyophilized solid lipid nanoparticles for gene therapy. Eur J Pharm Biopharm 2009; 71(2): 181-9.
[http://dx.doi.org/10.1016/j.ejpb.2008.09.015] [PMID: 18940256]
[59]
Belliveau NM, Huft J, Lin PJ, et al. Microfluidic synthesis of highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA. Mol Ther Nucleic Acids 2012; 1 e37
[http://dx.doi.org/10.1038/mtna.2012.28] [PMID: 23344179]
[60]
Cullis PR, Hope MJ. Lipid nanoparticle systems for enabling gene therapies. Mol Ther 2017; 25(7): 1467-75.
[http://dx.doi.org/10.1016/j.ymthe.2017.03.013] [PMID: 28412170]
[61]
Kenny GD, Kamaly N, Kalber TL, et al. Novel multifunctional nanoparticle mediates siRNA tumour delivery, visualisation and therapeutic tumour reduction in vivo. J Control Release 2011; 149(2): 111-6.
[http://dx.doi.org/10.1016/j.jconrel.2010.09.020] [PMID: 20888381]
[62]
de Lima GF, de Souza AG, Rosa DS. Effect of adsorption of polyethylene glycol (PEG), in aqueous media, to improve cellulose nanostructures stability. J Mol Liq 2018; 268: 415-24.
[http://dx.doi.org/10.1016/j.molliq.2018.07.080]
[63]
Luangtana-Anan M, Limmatvapirat S, Nunthanid J, Chalongsuk R, Yamamoto K. Polyethylene glycol on stability of chitosan microparticulate carrier for protein. AAPS PharmSciTech 2010; 11(3): 1376-82.
[http://dx.doi.org/10.1208/s12249-010-9512-y] [PMID: 20821174]
[64]
Chen D-B, Yang T-Z, Lu W-L, Zhang Q. In vitro and in vivo study of two types of long-circulating solid lipid nanoparticles containing paclitaxel. Chem Pharm Bull (Tokyo) 2001; 49(11): 1444-7.
[http://dx.doi.org/10.1248/cpb.49.1444] [PMID: 11724235]
[65]
Venkateswarlu V, Manjunath K. Preparation, characterization and in vitro release kinetics of clozapine solid lipid nanoparticles. J Control Release 2004; 95(3): 627-38.
[http://dx.doi.org/10.1016/j.jconrel.2004.01.005] [PMID: 15023472]
[66]
Robinson JR, Gauger LJ. Formulation of controlled-release products. J Allergy Clin Immunol 1986; 78(4 Pt 2): 676-81.
[http://dx.doi.org/10.1016/0091-6749(86)90045-X] [PMID: 3771957]
[67]
Brillo J, Pommrich AI, Meyer A. Relation between self-diffusion and viscosity in dense liquids: new experimental results from electrostatic levitation. Phys Rev Lett 2011; 107(16)165902
[http://dx.doi.org/10.1103/PhysRevLett.107.165902] [PMID: 22107404]
[68]
Fick A. On liquid diffusion. J Membr Sci 1995; 100: 33-8.
[http://dx.doi.org/10.1016/0376-7388(94)00230-V]

© 2024 Bentham Science Publishers | Privacy Policy