Generic placeholder image

Current Nanomaterials

Editor-in-Chief

ISSN (Print): 2405-4615
ISSN (Online): 2405-4623

Review Article

Lipid Based Nanoparticles: Current Strategies for Brain Tumor Targeting

Author(s): Bibhash C. Mohanta, Narahari N. Palei*, Vijayaraj Surendran, Subas C. Dinda, Jayaraman Rajangam, Jyotirmoy Deb and Biswa M. Sahoo

Volume 4, Issue 2, 2019

Page: [84 - 100] Pages: 17

DOI: 10.2174/2405461504666190510121911

Abstract

Brain tumors arise from an uncontrolled proliferation of neural tissue cells or supportive glial tissue cells within the brain. The diagnosis and therapy of brain tumor is an extremely challenging task. Moreover, absence of early stage symptoms and consequently delays in diagnosis and therapy worsen its severity. Though in the present days, chemotherapeutic approach is the most common therapeutic approach; still it is linked with several precincts. The blood-brain barrier (BBB) is the main hurdle in delivering most of the chemotherapeutic agents as well as imaging agent that leads to insufficient accumulation of therapeutic / imaging agents at tumor site, and prevents adequate destruction of malignant cells. Recently, lipid based nanoparticles are gaining much more interest and are preferred over polymeric nanoparticles owing to their biodegradability, non-toxicity, excellent tumortargeting ability and ease of surface modification. Certain receptors are over expressed in brain tumor cells which confer an opportunity to the researchers for delivering the chemotherapeutic as well as imaging agent particularly to the tumor cells through the surface modification approach of nanoparticles. Ligands like proteins/peptides, carbohydrates, aptamers, antibodies, and antibody fragments are generally conjugated to the surface of the nanoparticles that bind specifically to an over expressed target on the brain tumor cell surface. In the present review, we discuss the diagnostic and therapeutic application of various types of lipid based nanoparticles such as liposomes, niosomes, solid lipid nanoparticles, nanostructured lipid carrier, lipid nanocapsule, and lipid polymer hybrid nanocarriers along with their various surface modified forms for targeting brain tumor.

Keywords: BBB, brain tumor, lipid nanoparticles, chemotherapy, diagnosis, hybrid nanocarriers.

Graphical Abstract
[1]
Kelava I, Lancaster MA. Dishing out mini-brains: Current progress and future prospects in brain organoid research. Dev Biol 2016; 420(2): 199-209.
[2]
Mutha PK, Haaland KY, Sainburg RL. The effects of brain lateralization on motor control and adaptation. J Mot Behav 2012; 44(6): 455-69.
[3]
Guertin PA. Central pattern generator for locomotion: anatomical, physiological, and pathophysiological considerations. Front Neurol 2013; 3: 183.
[4]
Stockley C, Oxlade C, Wertheim J. The Usborne illustrated dictionary of science.In: Usborn London. 1999; pp. 302-9.
[5]
Martin E. A dictionary of science. 6th ed. In: Oxford University Press. 2010; pp. 109-10.
[6]
Velasco-Aguirre C, Morales F, Gallardo-Toledo E, et al. Peptides and proteins used to enhance gold nanoparticle delivery to the brain: preclinical approaches. Int J Nanomedicine 2015; 10: 4919-36.
[7]
Dinda SC, Pattnaik G. Nanobiotechnology-based drug delivery in brain targeting. Curr Pharm Biotechnol 2013; 14(15): 1264-74.
[8]
Butt AM, Jones HC, Abbott NJ. Electrical resistance across the blood-brain barrier in anaesthetized rats: a developmental study. J Physiol 1990; 429: 47-62.
[9]
de Vries HE, Kuiper J, de Boer AG, Van Berkel TJ, Breimer DD. The blood-brain barrier in neuroinflammatory diseases. Pharmacol Rev 1997; 49(2): 143-55.
[10]
Ballabh P, Braun A, Nedergaard M. The blood-brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis 2004; 16(1): 1-13.
[11]
Stamatovic SM, Keep RF, Andjelkovic AV. Brain endothelial cell-cell junctions: how to “open” the blood brain barrier. Curr Neuropharmacol 2008; 6(3): 179-92.
[12]
Ronaldson PT, Davis TP. Blood-brain barrier integrity and glial support: mechanisms that can be targeted for novel therapeutic approaches in stroke. Curr Pharm Des 2012; 18(25): 3624-44.
[13]
Nuriya M, Shinotsuka T, Yasui M. Diffusion properties of molecules at the blood-brain interface: potential contributions of astrocyte endfeet to diffusion barrier functions. Cereb Cortex 2013; 23(9): 2118-26.
[14]
Armulik A, Genové G, Mäe M, et al. Pericytes regulate the blood-brain barrier. Nature 2010; 468(7323): 557-61.
[15]
Kesari S. Understanding glioblastoma tumor biology: the potential to improve current diagnosis and treatments. Semin Oncol 2011; 38(4)(Suppl. 4): S2-S10.
[16]
Stummer W, Reulen HJ, Meinel T, et al. Extent of resection and survival in glioblastoma multiforme: identification of and adjustment for bias. Neurosurgery 2008; 62(3): 564-76.
[17]
Portnow J, Badie B, Chen M, Liu A, Blanchard S, Synold TW. The neuropharmacokinetics of temozolomide in patients with resectable brain tumors: potential implications for the current approach to chemoradiation. Clin Cancer Res 2009; 15(22): 7092-8.
[18]
Pardridge WM. Alzheimer’s disease drug development and the problem of the blood-brain barrier. Alzheimers Dement 2009; 5(5): 427-32.
[19]
Neuwelt EA, Bauer B, Fahlke C, et al. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 2011; 12(3): 169-82.
[20]
Kuhn JG. Influence of anticonvulsants on the metabolism and elimination of irinotecan. a north american brain tumor consortium preliminary report. Oncology 2002; 16(8): 33-40.
[21]
Haar CP, Hebbar P, Wallace GC IV, et al. Drug resistance in glioblastoma: a mini review. Neurochem Res 2012; 37(6): 1192-200.
[22]
Martins SM, Sarmento B, Nunes C, Lúcio M, Reis S, Ferreira DC. Brain targeting effect of camptothecin-loaded solid lipid nanoparticles in rat after intravenous administration. Eur J Pharm Biopharm 2013; 85(3 Pt A): 488-502.
[23]
Saraiva C, Praça C, Ferreira R, Santos T, Ferreira L, Bernardino L. Nanoparticle-mediated brain drug delivery: overcoming blood-brain barrier to treat neurodegenerative diseases. J Control Release 2016; 235: 34-47.
[24]
Biddlestone-Thorpe L, Marchi N, Guo K, et al. Nanomaterial-mediated CNS delivery of diagnostic and therapeutic agents. Adv Drug Deliv Rev 2012; 64(7): 605-13.
[25]
Grover A, Hirani A, Sutariya VK. Nanoparticle-based brain targeted delivery systems. J Biomol Res Ther 2013; 2(2): 1-3.
[26]
Demeule M, Régina A, Jodoin J, et al. Drug transport to the brain: key roles for the efflux pump P-glycoprotein in the blood-brain barrier. Vascul Pharmacol 2002; 38(6): 339-48.
[27]
Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRx 2005; 2(1): 1-12.
[28]
Mousa SA, Bharali DJ. Nanotechnology-based detection and targeted therapy in cancer: nano-bio paradigms and applications. Cancers 2011; 3(3): 2888-903.
[29]
Antunes AM, Alencar MS, da Silva CH, Nunes J, Mendes FM. Trends in nanotechnology patents applied to the health sector. Recent Pat Nanotechnol 2012; 6(1): 29-43.
[30]
Lueshen E, Venugopal I, Soni T, Alaraj A, Linninger A. Implant-assisted intrathecal magnetic drug targeting to aid in therapeutic nanoparticle localization for potential treatment of central nervous system disorders. J Biomed Nanotechnol 2015; 11(2): 253-61.
[31]
Pardeshi CV, Belgamwar VS. Direct nose to brain drug delivery via integrated nerve pathways bypassing the blood-brain barrier: an excellent platform for brain targeting. Expert Opin Drug Deliv 2013; 10(7): 957-72.
[32]
Kozlovskaya L, Abou-Kaoud M, Stepensky D. Quantitative analysis of drug delivery to the brain via nasal route. J Control Release 2014; 189: 133-40.
[33]
Kozler P, Pokorny J. Effect of methylprednisolone on the axonal impairment accompanying cellular brain oedema induced by water intoxication in rats. Neuroendocrinol Lett 2012; 33(8): 782-6.
[34]
Foley CP, Nishimura N, Neeves KB, Schaffer CB, Olbricht WL. Real-time imaging of perivascular transport of nanoparticles during convection-enhanced delivery in the rat cortex. Ann Biomed Eng 2012; 40(2): 292-303.
[35]
Alkins RD, Brodersen PM, Sodhi RN, Hynynen K. Enhancing drug delivery for boron neutron capture therapy of brain tumors with focused ultrasound. Neuro-oncol 2013; 15(9): 1225-35.
[36]
Aryal M, Vykhodtseva N, Zhang YZ, McDannold N. Multiple sessions of liposomal doxorubicin delivery via focused ultrasound mediated blood-brain barrier disruption: a safety study. J Control Release 2015; 204: 60-9.
[37]
Shinkai M, Yanase M, Suzuki M, et al. Intracellular hyperthermia for cancer using magnetite cationic liposomes. J Magn Magn Mater 1999; 194(1-3): 176-84.
[38]
Diaz RJ, McVeigh PZ, O’Reilly MA, et al. Focused ultrasound delivery of Raman nanoparticles across the blood-brain barrier: potential for targeting experimental brain tumors. Nanomedicine 2014; 10(5): 1075-87.
[39]
Eugenin EA, Clements JE, Zink MC, Berman JW. Human immunodeficiency virus infection of human astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. J Neurosci 2011; 31(26): 9456-65.
[40]
Balducci A, Wen Y, Zhang Y, et al. A novel probe for the non-invasive detection of tumor-associated inflammation. OncoImmunology 2013; 2(2)e23034
[41]
Wen Y, Meng WS. Recent In vivo evidences of particle-based delivery of small-interfering rna (sirna) into solid tumors. J Pharm Innov 2014; 9(2): 158-73.
[42]
Kuo YC, Ko HF. Targeting delivery of saquinavir to the brain using 83-14 monoclonal antibody-grafted solid lipid nanoparticles. Biomaterials 2013; 34(20): 4818-30.
[43]
Muller RH, Keck CM. Challenges and solutions for the delivery of biotech drugs--a review of drug nanocrystal technology and lipid nanoparticles. J Biotechnol 2004; 113(1-3): 151-70.
[44]
Narvekar M, Xue HY, Eoh JY, Wong HL. Nanocarrier for poorly water-soluble anticancer drugs barriers of translation and solutions. AAPS PharmSciTech 2014; 15(4): 822-33.
[45]
Liu D, Liu C, Zou W. Enhanced gastrointestinal absorption of N3-O-toluyl-fluorouracil by cationic solid lipid nanoparticles. J Nano Res 2009; 1: 1-10.
[46]
Estella-Hermoso de Mendoza A, Campanero MA, et al. Complete inhibition of extranodal dissemination of lymphoma by edelfosine-loaded lipid nanoparticles. Nanomedicine 2012; 7(5): 679-90.
[47]
Lasa-Saracibar B, Estella-Hermoso de Mendoza A, Guada M, Dios-Vieitez C, Blanco-Prieto MJ. Lipid nanoparticles for cancer therapy: state of the art and future prospects. Expert Opin Drug Deliv 2012; 9(10): 1245-61.
[48]
Pardridge WM. Blood-brain barrier drug targeting: the future of brain drug development. Mol Interv 2003; 3(2): 90-105. : 51
[49]
Chopineau J, Robert S, Fénart L, et al. Monoacylation of ribonuclease A enables its transport across an in vitro model of the blood-brain barrier. J Control Release 1998; 56(1-3): 231-7.
[50]
Prokai-Tatrai K, Prokai L. Prodrugs of thyrotropin-releasing hormone and related peptides as central nervous system agents. Molecules 2009; 14(2): 633-54.
[51]
Fenart L, Casanova A, Dehouck B, et al. Evaluation of effect of charge and lipid coating on ability of 60-nm nanoparticles to cross an in vitro model of the blood-brain barrier. J Pharmacol Exp Ther 1999; 291(3): 1017-22.
[52]
Samad A, Sultana Y, Aqil M. Liposomal drug delivery systems: an update review. Curr Drug Deliv 2007; 4(4): 297-305.
[53]
Peira E, Marzola P, Podio V, Aime S, Sbarbati A, Gasco MR. In vitro and in vivo study of solid lipid nanoparticles loaded with superparamagnetic iron oxide. J Drug Target 2003; 11(1): 19-24.
[54]
Banks WA. Characteristics of compounds that cross the blood-brain barrier. BMC Neurol 2009; 9(1): S3.
[55]
Vlieghe P, Khrestchatisky M. Medicinal chemistry based approaches and nanotechnology-based systems to improve CNS drug targeting and delivery. Med Res Rev 2013; 33(3): 457-516.
[56]
Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett 2013; 8(1): 102.
[57]
Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine 2015; 10: 975-99.
[58]
Ordikhani F, Erdem Arslan M, Marcelo R, et al. Drug delivery approaches for the treatment of cervical cancer. Pharmaceutics 2016; 8(3): 1-15.
[59]
Noble GT, Stefanick JF, Ashley JD, Kiziltepe T, Bilgicer B. Ligand-targeted liposome design: challenges and fundamental considerations. Trends Biotechnol 2014; 32(1): 32-45.
[60]
Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov 2005; 4(2): 145-60.
[61]
Robinson RF, Nahata MC. A comparative review of conventional and lipid formulations of amphotericin B. J Clin Pharm Ther 1999; 24(4): 249-57.
[62]
Sawant RR, Torchilin VP. Challenges in development of targeted liposomal therapeutics. AAPS J 2012; 14(2): 303-15.
[63]
Lee N, Yoo D, Ling D, Cho MH, Hyeon T, Cheon J. Iron oxide based nanoparticles for multimodal imaging and magnetoresponsive therapy. Chem Rev 2015; 115(19): 10637-89.
[64]
Martina MS, Fortin JP, Ménager C, et al. Generation of superparamagnetic liposomes revealed as highly efficient MRI contrast agents for in vivo imaging. J Am Chem Soc 2005; 127(30): 10676-85.
[65]
Yang J, Lee T, Lee J, et al. Synthesis of ultrasensitive magnetic resonance contrast agents for cancer imaging using PEG-fatty acid. Chem Mater 2007; 19: 3870-6.
[66]
Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res Int 2014; 2014869269
[67]
Sonali, Singh RP, Singh N. Transferrin liposomes of docetaxel for brain-targeted cancer. Drug Deliv 2016; 23(4): 1261-71.
[68]
Zhou Z, Lu ZR. Gadolinium-based contrast agents for magnetic resonance cancer imaging. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2013; 5(1): 1-18.
[69]
Saito R, Bringas JR, McKnight TR, et al. Distribution of liposomes into brain and rat brain tumor models by convection-enhanced delivery monitored with magnetic resonance imaging. Cancer Res 2004; 64(7): 2572-9.
[70]
Xie F, Yao N, Qin Y, et al. Investigation of glucose-modified liposomes using polyethylene glycols with different chain lengths as the linkers for brain targeting. Int J Nanomedicine 2012; 7: 163-75.
[71]
Qin Y, Fan W, Chen H, et al. In vitro and in vivo investigation of glucose-mediated brain-targeting liposomes. J Drug Target 2010; 18(7): 536-49.
[72]
Li X, Qu B, Jin X, Hai L, Wu Y. Design, synthesis and biological evaluation for docetaxel-loaded brain targeting liposome with “lock-in” function. J Drug Target 2014; 22(3): 251-61.
[73]
Gao H, Xiong Y, Zhang S, Yang Z, Cao S, Jiang X. RGD and interleukin-13 peptide functionalized nanoparticles for enhanced glioblastoma cells and neovasculature dual targeting delivery and elevated tumor penetration. Mol Pharm 2014; 11(3): 1042-52.
[74]
Miura Y, Takenaka T, Toh K, et al. Cyclic RGD-linked polymeric micelles for targeted delivery of platinum anticancer drugs to glioblastoma through the blood-brain tumor barrier. ACS Nano 2013; 7(10): 8583-92.
[75]
Oehler C, von Bueren AO, Furmanova P, et al. The microtubule stabilizer patupilone (epothilone B) is a potent radiosensitizer in medulloblastoma cells. Neuro-oncol 2011; 13(9): 1000-10.
[76]
Broggini-Tenzer A, Sharma A, Nytko KJ, et al. Combined treatment strategies for microtubule stabilizing agent-resistant tumors. J Natl Cancer Inst 2015; 107(4): 1-10.
[77]
Scherzinger-Laude K, Schönherr C, Lewrick F, Süss R, Francese G, Rössler J. Treatment of neuroblastoma and rhabdomyosarcoma using RGD-modified liposomal formulations of patupilone (EPO906). Int J Nanomedicine 2013; 8: 2197-211.
[78]
Song S, Mao G, Du J, Zhu X. Novel RGD containing, temozolomide-loading nanostructured lipid carriers for glioblastoma multiforme chemotherapy. Drug Deliv 2016; 23(4): 1404-8.
[79]
Hsiao YH, Kuo SJ, Tsai HD, Chou MC, Yeh GP. Clinical application of high-intensity focused ultrasound in cancer therapy. J Cancer 2016; 7(3): 225-31.
[80]
Zhou YF. High intensity focused ultrasound in clinical tumor ablation. World J Clin Oncol 2011; 2(1): 8-27.
[81]
Dogra VS, Zhang M, Bhatt S. High-intensity focused ultrasound (hifu) therapy applications. Ultrasound Clin 2009; 4(3): 307-21.
[82]
Yang FY, Wang HE, Lin GL, et al. Micro-SPECT/CT-based pharmacokinetic analysis of 99mTc-diethylenetriaminepentaacetic acid in rats with blood-brain barrier disruption induced by focused ultrasound. J Nucl Med 2011; 52(3): 478-84.
[83]
Khaibullina A, Jang BS, Sun H, et al. Pulsed high-intensity focused ultrasound enhances uptake of radiolabeled monoclonal antibody to human epidermoid tumor in nude mice. J Nucl Med 2008; 49(2): 295-302.
[84]
Yang FY, Teng MC, Lu M, et al. Treating glioblastoma multiforme with selective high-dose liposomal doxorubicin chemotherapy induced by repeated focused ultrasound. Int J Nanomedicine 2012; 7: 965-74.
[85]
Vanpouille-Box C, Lacoeuille F, Belloche C, et al. Tumor eradication in rat glioma and bypass of immunosuppressive barriers using internal radiation with (188)Re-lipid nanocapsules. Biomaterials 2011; 32(28): 6781-90.
[86]
Phillips WT, Goins B, Bao A, et al. Rhenium-186 liposomes as convection-enhanced nanoparticle brachytherapy for treatment of glioblastoma. Neuro-oncol 2012; 14(4): 416-25.
[87]
Casacó A, López G, García I, et al. Phase I single-dose study of intracavitary-administered Nimotuzumab labeled with 188 Re in adult recurrent high-grade glioma. Cancer Biol Ther 2008; 7(3): 333-9.
[88]
Huang FYJ, Lee TW, Chang CH, et al. Evaluation of 188Re-labeled PEGylated nanoliposome as a radionuclide therapeutic agent in an orthotopic glioma-bearing rat model. Int J Nanomedicine 2015; 10: 463-73.
[89]
Moghassemi S, Hadjizadeh A. Nano-niosomes as nanoscale drug delivery systems: an illustrated review. J Control Release 2014; 185: 22-36.
[90]
Gharbavi M, Amani J, Kheiri-Manjili H, Danafar H, Sharafi A. Niosome: a promising nanocarrier for natural drug delivery through blood-brain barrier. Adv Pharmacol Sci 2018; 2018684797
[91]
Das MK, Palei NN. Sorbitan ester niosomes for topical delivery of rofecoxib. Indian J Exp Biol 2011; 49(6): 438-45.
[92]
Azmin MN, Florence AT, Handjani-Vila RM, Stuart JF, Vanlerberghe G, Whittaker JS. The effect of non-ionic surfactant vesicle (niosome) entrapment on the absorption and distribution of methotrexate in mice. J Pharm Pharmacol 1985; 37(4): 237-42.
[93]
El Maghraby GM, Williams AC. Vesicular systems for delivering conventional small organic molecules and larger macromolecules to and through human skin. Expert Opin Drug Deliv 2009; 6(2): 149-63.
[94]
Khan R, Irchhaiya R. Niosomes: a potential tool for novel drug delivery. J Pharm Investig 2016; 46(3): 195-204.
[95]
Kong M, Park H, Feng C, Hou L, Cheng X, Chen X. Construction of hyaluronic acid noisome as functional transdermal nanocarrier for tumor therapy. Carbohydr Polym 2013; 94(1): 634-41.
[96]
Ag D, Bongartz R, Dogan LE, et al. Biofunctional quantum dots as fluorescence probe for cell-specific targeting. Colloids Surf B Biointerfaces 2014; 114: 96-103.
[97]
Bragagni M, Mennini N, Ghelardini C, Mura P. Development and characterization of niosomal formulations of doxorubicin aimed at brain targeting. J Pharm Pharm Sci 2012; 15(1): 184-96.
[98]
Tavano L, Mauro L, Naimo GD, et al. Further evolution of multifunctional niosomes based on pluronic surfactant: dual active targeting and drug combination properties. Langmuir 2016; 32(35): 8926-33.
[99]
Bibhas CM, Gitanjali M, Subas CD, Narahari NP. Exploring the use of lipid based nano-formulations for the management of tuberculosis. J Nanosci Curr Res 2017; 2(3): 1-15.
[100]
Palei NP, Mohanta BC, Sabapathi ML, Das MK. Lipid-based nanoparticles for cancer diagnosis and therapy Organic Materials as Smart Nanocarriers for Drug Delivery. 1st ed. William Andrew Applied Science Publisher, Elsevier Inc. 2018; pp. 415-70.
[101]
Mohanta BC, Dinda SC, Mishra G, Palei NP, Dusthackeer VNA. Formulation, characterization, in vitro anti-tubercular activity and cytotoxicity study of solid lipid nanoparticles of isoniazid. Nano Biomed Eng 2018; 10(4): 379-91.
[102]
Das S, Chaudhury A. Recent advances in lipid nanoparticle formulations with solid matrix for oral drug delivery. AAPS PharmSciTech 2011; 12(1): 62-76.
[103]
Kuo YC, Liang CT. Inhibition of human brain malignant glioblastoma cells using carmustine-loaded catanionic solid lipid nanoparticles with surface anti-epithelial growth factor receptor. Biomaterials 2011; 32(12): 3340-50.
[104]
Bondì ML, Craparo EF, Giammona G, Drago F. Brain-targeted solid lipid nanoparticles containing riluzole: preparation, characterization and biodistribution. Nanomedicine 2010; 5(1): 25-32.
[105]
Sun C, Ding Y, Zhou L, et al. Noninvasive nanoparticle strategies for brain tumor targeting. Nanomedicine 2017; 13(8): 2605-21.
[106]
Kuo YC, Cheng SJ. Brain targeted delivery of carmustine using solid lipid nanoparticles modified with tamoxifen and lactoferrin for antitumor proliferation. Int J Pharm 2016; 499(1-2): 10-9.
[107]
Singh I, Swami R, Pooja D, Jeengar MK, Khan W, Sistla R. Lactoferrin bioconjugated solid lipid nanoparticles: a new drug delivery system for potential brain targeting. J Drug Target 2016; 24(3): 212-23.
[108]
Gomes MJ, Martins S, Sarmento B. siRNA as a tool to improve the treatment of brain diseases: mechanism, targets and delivery. Ageing Res Rev 2015; 21: 43-54.
[109]
Wei L, Guo XY, Yang T, Yu MZ, Chen DW, Wang JC. Brain tumor-targeted therapy by systemic delivery of siRNA with Transferrin receptor-mediated core-shell nanoparticles. Int J Pharm 2016; 510(1): 394-405.
[110]
Bruun J, Larsen TB, Jølck RI, et al. Investigation of enzyme-sensitive lipid nanoparticles for delivery of siRNA to blood-brain barrier and glioma cells. Int J Nanomedicine 2015; 10: 5995-6008.
[111]
Jaiswal P, Gidwani B, Vyas A. Nanostructured lipid carriers and their current application in targeted drug delivery. Artif Cells Nanomed Biotechnol 2016; 44(1): 27-40.
[112]
Frias I, Neves AR, Pinheiro M, Reis S. Design, development, and characterization of lipid nanocarriers-based epigallocatechin gallate delivery system for preventive and therapeutic supplementation. Drug Des Devel Ther 2016; 10: 3519-28.
[113]
Palei NP, Mohanta BC, Sabapathi ML, Das MK. Lornoxicam loaded nanostructured lipid carriers for topical delivery: Optimization, skin uptake and in vivostudies. J Drug Deliv Sci Technol 2017; 39: 490-500.
[114]
Shah NV, Seth AK, Balaraman R, Aundhia CJ, Maheshwari RA, Parmar GR. Nanostructured lipid carriers for oral bioavailability enhancement of raloxifene: Design and in vivo study. J Adv Res 2016; 7(3): 423-34.
[115]
Naseri N, Valizadeh H, Zakeri-Milani P. Solid lipid nanoparticles and nanostructured lipid carriers: structure, preparation and application. Adv Pharm Bull 2015; 5(3): 305-13.
[116]
Gaba B, Fazil M, Ali A, Baboota S, Sahni JK, Ali J. Nanostructured lipid (NLCs) carriers as a bioavailability enhancement tool for oral administration. Drug Deliv 2015; 22(6): 691-700.
[117]
Yallapu MM, Jaggi M, Chauhan SC. Curcumin nanomedicine: a road to cancer therapeutics. Curr Pharm Des 2013; 19(11): 1994-2010.
[118]
Wang P, Zhang L, Peng H, Li Y, Xiong J, Xu Z. The formulation and delivery of curcumin with solid lipid nanoparticles for the treatment of on non-small cell lung cancer both in vitro and in vivo. Mater Sci Eng C 2013; 33(8): 4802-8.
[119]
Chen Y, Pan L, Jiang M, Li D, Jin L. Nanostructured lipid carriers enhance the bioavailability and brain cancer inhibitory efficacy of curcumin both in vitro and in vivo. Drug Deliv 2016; 23(4): 1383-92.
[120]
Zhang C, Peng F, Liu W, et al. Nanostructured lipid carriers as a novel oral delivery system for triptolide: induced changes in pharmacokinetics profile associated with reduced toxicity in male rats. Int J Nanomedicine 2014; 9: 1049-63.
[121]
Tosi G, Musumeci T, Ruozi B, et al. The “fate” of polymeric and lipid nanoparticles for brain delivery and targeting: strategies and mechanism of blood–brain barrier crossing and trafficking into the central nervous system. J Drug Deliv Sci Technol 2016; 32: 66-76.
[122]
Qu J, Zhang L, Chen Z, et al. Nanostructured lipid carriers, solid lipid nanoparticles, and polymeric nanoparticles: which kind of drug delivery system is better for glioblastoma chemotherapy? Drug Deliv 2016; 23(9): 3408-16.
[123]
Wu M, Fan Y, Lv S, Xiao B, Ye M, Zhu X. Vincristine and temozolomide combined chemotherapy for the treatment of glioma: a comparison of solid lipid nanoparticles and nanostructured lipid carriers for dual drugs delivery. Drug Deliv 2016; 23(8): 2720-5.
[124]
Sharma G, Lakkadwala S, Modgil A, Singh J. The role of cell-penetrating peptide and transferrin on enhanced delivery of drug to brain. Int J Mol Sci 2016; 17(6): 2-18.
[125]
Zheng C, Ma C, Bai E, Yang K, Xu R. Transferrin and cell-penetrating peptide dual-functioned liposome for targeted drug delivery to glioma. Int J Clin Exp Med 2015; 8(2): 1658-68.
[126]
Xu S, Olenyuk BZ, Okamoto CT, Hamm-Alvarez SF. Targeting receptor-mediated endocytotic pathways with nanoparticles: rationale and advances. Adv Drug Deliv Rev 2013; 65(1): 121-38.
[127]
Emami J, Rezazadeh M, Sadeghi H, Khadivar K. Development and optimization of transferrin-conjugated nanostructured lipid carriers for brain delivery of paclitaxel using Box-Behnken design. Pharm Dev Technol 2017; 22(3): 370-82.
[128]
Huynh NT, Morille M, Bejaud J, et al. Treatment of 9L gliosarcoma in rats by ferrociphenol-loaded lipid nanocapsules based on a passive targeting strategy via the EPR effect. Pharm Res 2011; 28(12): 3189-98.
[129]
Roger M, Clavreul A, Venier-Julienne MC, et al. Mesenchymal stem cells as cellular vehicles for delivery of nanoparticles to brain tumors. Biomaterials 2010; 31(32): 8393-401.
[130]
Roger M, Clavreul A, Huynh NT, et al. Ferrociphenol lipid nanocapsule delivery by mesenchymal stromal cells in brain tumor therapy. Int J Pharm 2012; 423(1): 63-8.
[131]
Huynh NT, Passirani C, Allard-Vannier E, et al. Administration-dependent efficacy of ferrociphenol lipid nanocapsules for the treatment of intracranial 9L rat gliosarcoma. Int J Pharm 2012; 423(1): 55-62.
[132]
Wakaskar RR. General overview of lipid-polymer hybrid nanoparticles, dendrimers, micelles, liposomes, spongosomes and cubosomes. J Drug Target 2018; 26(4): 311-8.
[133]
Li J, Wang Y, Zhu Y, Oupický D. Recent advances in delivery of drug-nucleic acid combinations for cancer treatment. J Control Release 2013; 172(2): 589-600.
[134]
Küçüktürkmen B, Devrim B, Saka OM, Yilmaz Ş, Arsoy T, Bozkir A. Co-delivery of pemetrexed and miR-21 antisense oligonucleotide by lipid-polymer hybrid nanoparticles and effects on glioblastoma cells. Drug Dev Ind Pharm 2017; 43(1): 12-21.

© 2024 Bentham Science Publishers | Privacy Policy