Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica

ilustraciones, fotografías, graficas

Autores:
Velandia Paris, María Angélica
Tipo de recurso:
Fecha de publicación:
2021
Institución:
Universidad Nacional de Colombia
Repositorio:
Universidad Nacional de Colombia
Idioma:
spa
OAI Identifier:
oai:repositorio.unal.edu.co:unal/80864
Acceso en línea:
https://repositorio.unal.edu.co/handle/unal/80864
https://repositorio.unal.edu.co/
Palabra clave:
610 - Medicina y salud::615 - Farmacología y terapéutica
Sistemas particulados
Polímero
Lípido
Péptido sintético
Doble emulsión
Evaporación del solvente
Híbridos
Lipopoliméricos
Particulate systems
Polymer
Lipid
Synthetic peptide
Double emulsion
Solvent evaporation
Hybrids
Lipopolymerics
Polímero
Polymers
Compuesto orgánico
Rights
openAccess
License
Atribución-NoComercial-CompartirIgual 4.0 Internacional
id UNACIONAL2_f16b9baca967827cda365e4539726f12
oai_identifier_str oai:repositorio.unal.edu.co:unal/80864
network_acronym_str UNACIONAL2
network_name_str Universidad Nacional de Colombia
repository_id_str
dc.title.spa.fl_str_mv Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
dc.title.translated.eng.fl_str_mv Design and development of a lipopolymeric microparticular system for the administration od a synthetic peptide of hydrophilic nature
title Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
spellingShingle Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
610 - Medicina y salud::615 - Farmacología y terapéutica
Sistemas particulados
Polímero
Lípido
Péptido sintético
Doble emulsión
Evaporación del solvente
Híbridos
Lipopoliméricos
Particulate systems
Polymer
Lipid
Synthetic peptide
Double emulsion
Solvent evaporation
Hybrids
Lipopolymerics
Polímero
Polymers
Compuesto orgánico
title_short Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
title_full Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
title_fullStr Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
title_full_unstemmed Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
title_sort Diseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílica
dc.creator.fl_str_mv Velandia Paris, María Angélica
dc.contributor.advisor.none.fl_str_mv Rosas Pérez, Jaiver Eduardo
dc.contributor.author.none.fl_str_mv Velandia Paris, María Angélica
dc.contributor.researchgroup.spa.fl_str_mv Sistemas para liberación controlada de moléculas biológicamente activas (SILICOMOBA)
dc.subject.ddc.spa.fl_str_mv 610 - Medicina y salud::615 - Farmacología y terapéutica
topic 610 - Medicina y salud::615 - Farmacología y terapéutica
Sistemas particulados
Polímero
Lípido
Péptido sintético
Doble emulsión
Evaporación del solvente
Híbridos
Lipopoliméricos
Particulate systems
Polymer
Lipid
Synthetic peptide
Double emulsion
Solvent evaporation
Hybrids
Lipopolymerics
Polímero
Polymers
Compuesto orgánico
dc.subject.proposal.spa.fl_str_mv Sistemas particulados
Polímero
Lípido
Péptido sintético
Doble emulsión
Evaporación del solvente
Híbridos
Lipopoliméricos
dc.subject.proposal.eng.fl_str_mv Particulate systems
Polymer
Lipid
Synthetic peptide
Double emulsion
Solvent evaporation
Hybrids
Lipopolymerics
dc.subject.unesco.none.fl_str_mv Polímero
Polymers
Compuesto orgánico
description ilustraciones, fotografías, graficas
publishDate 2021
dc.date.issued.none.fl_str_mv 2021
dc.date.accessioned.none.fl_str_mv 2022-02-03T15:42:48Z
dc.date.available.none.fl_str_mv 2022-02-03T15:42:48Z
dc.type.spa.fl_str_mv Trabajo de grado - Maestría
dc.type.driver.spa.fl_str_mv info:eu-repo/semantics/masterThesis
dc.type.version.spa.fl_str_mv info:eu-repo/semantics/acceptedVersion
dc.type.content.spa.fl_str_mv Text
dc.type.redcol.spa.fl_str_mv http://purl.org/redcol/resource_type/TM
status_str acceptedVersion
dc.identifier.uri.none.fl_str_mv https://repositorio.unal.edu.co/handle/unal/80864
dc.identifier.instname.spa.fl_str_mv Universidad Nacional de Colombia
dc.identifier.reponame.spa.fl_str_mv Repositorio Institucional Universidad Nacional de Colombia
dc.identifier.repourl.spa.fl_str_mv https://repositorio.unal.edu.co/
url https://repositorio.unal.edu.co/handle/unal/80864
https://repositorio.unal.edu.co/
identifier_str_mv Universidad Nacional de Colombia
Repositorio Institucional Universidad Nacional de Colombia
dc.language.iso.spa.fl_str_mv spa
language spa
dc.relation.indexed.spa.fl_str_mv LaReferencia
dc.relation.references.spa.fl_str_mv 1. Lau JL, Dunn MK. Therapeutic peptides: Historical perspectives, current development trends, and future directions. Bioorganic Med Chem [Internet]. 2018;26(10):2700–7. Available from: https://doi.org/10.1016/j.bmc.2017.06.052
2. Sachdeva S. Peptides as ‘ Drugs ’: The Journey so Far. Int J Pept Res Ther. 2016;(May).
3. Peptides C. Derek N. Woolfson 1,2 1 2. 2009;94(1):118–27.
4. Grant GA. Synthetic peptides : a user’s guide. 2nd ed. Grant GA, editor. New York: Oxford Univesity Press; 2002.
5. Vlieghe P, Lisowski V, Martinez J, Khrestchatisky M. Synthetic therapeutic peptides : science and market. 2010;15(January).
6. Zhang J, Desale SS, Bronich TK. Polymer-based vehicles for therapeutic peptide delivery. Ther Deliv. 2015;6(11):1279–96.
7. Couvreur P, Puisieux F. Nano- and microparticles for the delivery of polypeptides and proteins. Adv Drug Deliv Rev. 1993;10(2–3):141–62.
8. Bruschi M. Modification of drug release. Strateg to Modify Drug Release from Pharm Syst. 2015;15–28.
9. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in Medicine : Therapeutic Applications and Developments. 2008;83(5):761–9.
10. Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics : an emerging treatment modality for cancer. 2008;7(SEPTEMbER):771–82
11. Devrim B, Kara A, Vural İ, Bozkır A. Lysozyme-loaded lipid-polymer hybrid nanoparticles: preparation, characterization and colloidal stability evaluation. Drug Dev Ind Pharm. 2016;42(11):1865–76.
12. Zhang L, Granick S. How to Stabilize Phospholipid Liposomes ( Using Nanoparticles ). 2006;0–4.
13. Zhang L, Zhang LF. Lipid–Polymer Hybrid Nanoparticles: Synthesis,Characterization and Applications. Nano Life. 2010;01(01n02):163–73.
14. Garud A, Singh D, Garud N. Solid Lipid Nanoparticles (SLN): Method, Characterization and Applications. Int Curr Pharm J. 2012;1(11):384–93.
15. Ansari MJ, Anwer MK, Jamil S, Al-Shdefat R, Ali BE, Ahmad MM, et al. Enhanced oral bioavailability of insulin-loaded solid lipid nanoparticles: pharmacokinetic bioavailability of insulin-loaded solid lipid nanoparticles in diabetic rats. Drug Deliv [Internet]. 2016;23(6):1972–9. Available from: http://dx.doi.org/10.3109/10717544.2015.1039666
16. Chan JM, Zhang L, Yuet KP, Liao G, Rhee J, Langer R, et al. Biomaterials PLGA – lecithin – PEG core – shell nanoparticles for controlled drug delivery. Biomaterials [Internet]. 2009;30(8):1627–34. Available from: http://dx.doi.org/10.1016/j.biomaterials.2008.12.013
17. Thevenot J, Troutier A, David L, Delair T, Pascal B. Steric Stabilization of Lipid / Polymer Particle Assemblies by Poly ( ethylene glycol ) -Lipids. 2007;3651–60.
18. Grigoras AG. Polymer-lipid hybrid systems used as carriers for insulin delivery. Nanomedicine Nanotechnology, Biol Med [Internet]. 2017;13(8):2425–37. Available from: https://doi.org/10.1016/j.nano.2017.08.005
19. Walton HF. General considerations. Ligand Exch Chromatogr. 2018;7–30.
20. Ito Y. Drug delivery systems. Photochemistry for Biomedical Applications: From Device Fabrication to Diagnosis and Therapy. 2018. 231–275 p
21. Nastruzzi C. Lipospheres in drug targets and delivery. 2005.
22. Manjunath K, Reddy JS, Venkateswarlu V. Solid lipid nanoparticles as drug delivery systems. Methods Find Exp Clin Pharmacol [Internet]. 2005;27(2):127. Available from: http://journals.prous.com/journals/servlet/xmlxsl/pk_journals.xml_summary_pr?p_J ournalId=6&p_RefId=876286&p_IsPs=N
23. Matougui N, Boge L, Groo AC, Umerska A, Ringstad L, Bysell H, et al. Lipid-based nanoformulations for peptide delivery. Int J Pharm [Internet]. 2016;502(1–2):80–97. Available from: http://dx.doi.org/10.1016/j.ijpharm.2016.02.019
24. Shukla T, Upmanyu N, Pandey SP, Gosh D. Chapter 1. Lipid nanocarriers [Internet]. Lipid Nanocarriers for Drug Targeting. Elsevier Inc.; 2018. 1–48 p. Available from: http://dx.doi.org/10.1016/B978-0-12-813687-4.00001-3
25. Rawal SU, Patel MM. Chapter 2. Lipid nanoparticulate systems: Modern versatile drug carriers [Internet]. Lipid Nanocarriers for Drug Targeting. Elsevier Inc.; 2018. 49–138 p. Available from: http://dx.doi.org/10.1016/B978-0-12-813687-4.00002-5
26. Abbasi E, Aval SF, Akbarzadeh A, Milani M, Nasrabadi HT, Joo SW, et al. Dendrimers: Synthesis, applications, and properties. Vol. 9, Nanoscale Research Letters. Springer New York LLC; 2014. p. 1–10.
27. Jyothi NVN, Prasanna PM, Sakarkar SN, Prabha KS, Ramaiah PS, Srawan GY. Microencapsulation techniques, factors influencing encapsulation efficiency. J Microencapsul. 2010;27(3):187–97
28. Freiberg S, Zhu XX. Polymer microspheres for controlled drug release. Int J Pharm. 2004;282(1–2):1–18.
29. Zhang F, Fan J bing, Wang S. Interfacial Polymerization: From Chemistry to Functional Materials. Angew Chemie - Int Ed. 2020;59(49):21840–56.
30. Kwak H-S. Nano- and Microencapsulation. 2014.
31. JONG; H. G. BDHRK. Coacervation (Partial miscibility in colloid systems). (Preliminary Communication). 1929; Available from: https://www.dwc.knaw.nl/DL/publications/PU00015781.pdf
32. Ghosh SK. Functional Coatings and Microencapsulation: A General Perspective. Funct Coatings By Polym Microencapsul. 2006;1–28
33. Zuidam NJ, Nedović VA. Encapsulation technologies for active food ingredients and food processing. Encapsulation Technol Act Food Ingredients Food Process. 2010;1–400.
34. Freitas S, Merkle HP, Gander B. Microencapsulation by solvent extraction/evaporation: reviewing the state of the art of microsphere preparation process technology. J Control Release [Internet]. 2005 Feb;102(2):313–32. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0168365904004845
35. Hallan SS, Kaur P, Kaur V, Mishra N, Vaidya B. Lipid polymer hybrid as emerging tool in nanocarriers for oral drug delivery. Artif Cells, Nanomedicine Biotechnol. 2016;44(1):334–49.
36. Hadinoto K, Sundaresan A, Cheow WS. Lipid-polymer hybrid nanoparticles as a new generation therapeutic delivery platform: A review. Eur J Pharm Biopharm [Internet]. 2013;85(3 PART A):427–43. Available from: http://dx.doi.org/10.1016/j.ejpb.2013.07.002
37. Dong W, Wang X, Liu C, Zhang X, Zhang X, Chen X, et al. Chitosan based polymerlipid hybrid nanoparticles for oral delivery of enoxaparin. Int J Pharm [Internet]. 2018;547(1–2):499–505. Available from: https://doi.org/10.1016/j.ijpharm.2018.05.076
38. Sengel-Turk CT, Gumustas M, Uslu B, Ozkan SA. A novel approach for drug targeting: Core-shell type lipid-polymer hybrid nanocarriers. Core-shell type lipidpolymer hybrid nanocarriers. [Internet]. Design of Nanostructures for Theranostics Applications. Elsevier Inc.; 2018. 69–107 p. Available from: http://dx.doi.org/10.1016/B978-0-12-813669-0.00003-8
39. Jain A, Jain A, Gulbake A, Shilpi S, Hurkat P, Jain SK. Peptide and Protein Delivery Using New Drug Delivery Systems. Crit Rev Ther Drug Carrier Syst. 2013;30(4):293–329.
40. McClements DJ. Encapsulation, protection, and delivery of bioactive proteins and peptides using nanoparticle and microparticle systems: A review. Adv Colloid Interface Sci [Internet]. 2018;253(2017):1–22. Available from: https://doi.org/10.1016/j.cis.2018.02.002
41. Pinto Reis C, Neufeld RJ, Ribeiro AJ, Veiga F. Nanoencapsulation II. Biomedical applications and current status of peptide and protein nanoparticulate delivery systems. Nanomedicine Nanotechnology, Biol Med. 2006;2(2):53–65.
42. Kovalainen M, Mönkäre J, Riikonen J, Pesonen U, Vlasova M, Salonen J. Novel Delivery Systems for Improving the Clinical Use of Peptides. 2015;(July):541–61.
43. Patarroyo ME, Romero P, Torres ML, Clavijo P, Moreno A, Martínez A, et al. Induction of protective immunity against experimental infection with malaria using synthetic peptides. Nature. 1988;328(6131):629–32.
44. Lozano JM, Patarroyo ME. A rational strategy for a malarial vaccine development. Microbes Infect. 2007;9(6):751–60.
45. McClements DJ. Encapsulation, protection, and delivery of bioactive proteins and peptides using nanoparticle and microparticle systems: A review. Adv Colloid Interface Sci [Internet]. 2018 Mar 1 [cited 2020 Mar 20];253:1–22. Available from: https://www.sciencedirect.com/science/article/pii/S0001868617305171
46. Bolhassani A. Improvements in chemical carriers of proteins and peptides. 2019;43:437–52
47. Gasco AR. Biodegradable PLGA microspheres as a delivery system for malaria synthetic peptide SPf66. 2001;19:4445–51.
48. Dave V, Tak K, Sohgaura A, Gupta A, Sadhu V. Lipid-polymer hybrid nanoparticles : Synthesis strategies and biomedical applications. J Microbiol Methods [Internet]. 2019;160(March):130–42. Available from: https://doi.org/10.1016/j.mimet.2019.03.017
49. Samad A, Tariq M, Alam MI, Akhter MS. Microsphere: A novel drug delivery system. Colloids Drug Deliv. 2016;11(4):455–78
50. Acar H, Ting JM, Srivastava S, LaBelle JL, Tirrell M V. Molecular engineering solutions for therapeutic peptide delivery. Chem Soc Rev [Internet]. 2017;46(21):6553–69. Available from: http://dx.doi.org/10.1039/C7CS00536A
51. Blasi P. Poly(lactic acid)/poly(lactic-co-glycolic acid)-based microparticles: an overview. J Pharm Investig [Internet]. 2019;49(4):337–46. Available from: https://doi.org/10.1007/s40005-019-00453-z
52. Hajavi J, Ebrahimian M, Sankian M, Khakzad MR, Hashemi M. Optimization of PLGA formulation containing protein or peptide-based antigen: Recent advances. J Biomed Mater Res - Part A. 2018;106(9):2540–51.
53. Qi F, Wu J, Li H, Ma G. Recent research and development of PLGA/PLA microspheres/nanoparticles: A review in scientific and industrial aspects. Front Chem Sci Eng. 2019;13(1):14–27.
54. Schoubben A, Ricci M, Giovagnoli S. Meeting the unmet: from traditional to cuttingedge techniques for poly lactide and poly lactide-co-glycolide microparticle manufacturing. J Pharm Investig [Internet]. 2019;49(4):381–404. Available from: https://doi.org/10.1007/s40005-019-00446-y
55. Zhong H, Chan G, Hu Y, Hu H, Ouyang D. A comprehensive map of FDA-approved pharmaceutical products. Pharmaceutics. 2018;10(4):1–19.
56. Jain A, Kunduru KR, Basu A, Mizrahi B, Domb AJ, Khan W. Injectable formulations of poly(lactic acid) and its copolymers in clinical use. Adv Drug Deliv Rev [Internet]. 2016;107:213–27. Available from: http://dx.doi.org/10.1016/j.addr.2016.07.002
57. Igartua M, Hernández RM, Rosas JE, Patarroyo ME, Pedraz JL. γ-Irradiation effects on biopharmaceutical properties of PLGA microspheres loaded with SPf66 synthetic vaccine. Eur J Pharm Biopharm [Internet]. 2008 Jun 1 [cited 2020 Jan 23];69(2):519– 26. Available from: https://www.sciencedirect.com/science/article/pii/S0939641107004201?via%3Dihu b
58. Carcaboso A., Hernández R., Igartua M, Rosas J., Patarroyo M., Pedraz J. Potent, long lasting systemic antibody levels and mixed Th1/Th2 immune response after nasal immunization with malaria antigen loaded PLGA microparticles. Vaccine [Internet]. 2004 Mar 29 [cited 2020 Jan 23];22(11–12):1423–32. Available from: https://www.sciencedirect.com/science/article/pii/S0264410X03007679?via%3Dihu b
59. Yang YY, Chung TS, Ping Ng N. Morphology, drug distribution, and in vitro release profiles of biodegradable polymeric microspheres containing protein fabricated by double-emulsion solvent extraction/evaporation method. Biomaterials. 2001;22(3):231–41.
60. D’Souza SS, Faraj JA, DeLuca PP. A Model-dependent Approach to Correlate Accelerated With Real-Time Release From Biodegradable Microspheres. AAPS PharmSciTech. 2005;6(4):553–64.
61. Martinez NY, Andrade PF, Durán N, Cavalitto S. Development of double emulsion nanoparticles for the encapsulation of bovine serum albumin. Colloids Surfaces B Biointerfaces [Internet]. 2017;158:190–6. Available from: http://dx.doi.org/10.1016/j.colsurfb.2017.06.033
62. Agrawal A, Rellegadla S, Jain S. Biomedical applications of PLGA particles [Internet]. Materials for Biomedical Engineering: Nanomaterials-based Drug Delivery. Elsevier Inc.; 2019. 87–129 p. Available from: http://dx.doi.org/10.1016/B978-0-12-816913-1.00004-0
63. Note T. Measuring Zeta Potential – Laser Doppler Electrophoresis. Malvern Guid. 2015;1–2.
64. Bhattacharjee S. DLS and zeta potential – What they are and what they are not? J Control Release [Internet]. 2016 Aug 10 [cited 2020 Apr 7];235:337–51. Available from: https://www.sciencedirect.com/science/article/abs/pii/S0168365916303832?via%3 Dihub
65. Agrawal A, Rellegadla S, Jain S. Biomedical applications of PLGA particles. Mater Biomed Eng [Internet]. 2019 Jan 1 [cited 2020 Mar 20];87–129. Available from: https://www.sciencedirect.com/science/article/pii/B9780128169131000040
66. Mundargi RC, Babu VR, Rangaswamy V, Patel P, Aminabhavi TM. Nano/micro technologies for delivering macromolecular therapeutics using poly(d,l-lactide-coglycolide) and its derivatives. J Control Release. 2008;125(3):193–209.
67. Saez, Vivian; Hernández, José R; Peniche C. Microspheres as delivery systems for the controlled release of peptides and proteins. Biotecnol Apl. 2007;24(2):108–16.
68. Busatto C, Pesoa J, Helbling I, Luna J, Estenoz D. Effect of particle size, polydispersity and polymer degradation on progesterone release from PLGA microparticles: Experimental and mathematical modeling. Int J Pharm [Internet]. 2018;536(1):360–9. Available from: http://dx.doi.org/10.1016/j.ijpharm.2017.12.006
69. Giteau A, Venier-julienne MC, Benoit JP. How to achieve sustained and complete protein release from PLGA-based microparticles ? 2008;350:14–26.
70. Fredenberg S, Wahlgren M, Reslow M, Axelsson A. The mechanisms of drug release in poly ( lactic-co-glycolic acid ) -based drug delivery systems — A review. Int J Pharm [Internet]. 2011;415(1–2):34–52. Available from: http://dx.doi.org/10.1016/j.ijpharm.2011.05.049
71. Gohla S, Ma K, Mu RH. Solid lipid nanoparticles ( SLN ) for controlled drug delivery ± a review of the state of the art. 2000;50.
72. Gallarate M, Battaglia L, Peira E, Trotta M. Peptide-loaded solid lipid nanoparticles prepared through coacervation technique. Int J Chem Eng. 2011;2011.
73. Battaglia L, Trotta M, Gallarate M, Carlotti ME, Zara GP, Bargoni A. Solid lipid nanoparticles formed by solvent-in-water emulsion-diffusion technique: Development and influence on insulin stability. J Microencapsul. 2007;24(7):672–84.
74. Bakliwal A, Bedse S, Talele S, Chaudhari G. LIPOSPHERES: A NOVEL APPROACH FOR DRUG DELIVERY. Int J Res Pharm Nano Sci. 2015;4(5):298– 306.
75. Cortesi R, Esposito E, Luca G, Nastruzzi C. Production of lipospheres as carriers for bioactive compounds. Biomaterials. 2002;23(11):2283–94.
76. Patil P, Harak K, Saudagar R. Solid Lipid Nanoparticles : A Review. 2019;9(3):525– 30.
77. Mishra H, Mishra D, Mishra PK, Nahar M, Dubey V, Jain NK. Evaluation of solid lipid nanoparticles as carriers for delivery of hepatitis b surface antigen for vaccination using subcutaneous route. J Pharm Pharm Sci. 2010;13(4):495–509.
78. Reithmeier H, Herrmann J, Göpferich A. Development and characterization of lipid microparticles as a drug carrier for somatostatin. Int J Pharm. 2001;218(1–2):133– 43.
79. KH R, SA S, SN D. Solid lipid nanoparticles- A review. Int J Appl Pharm. 2013;5(2):8– 18.
80. Patel MR, San Martin-Gonzalez MF. Characterization of ergocalciferol loaded solid lipid nanoparticles. J Food Sci. 2012;77(1):8–13.
81. Prombutara P, Kulwatthanasal Y, Supaka N, Sramala I, Chareonpornwattana S. Production of nisin-loaded solid lipid nanoparticles for sustained antimicrobial activity. Food Control [Internet]. 2012;24(1–2):184–90. Available from: http://dx.doi.org/10.1016/j.foodcont.2011.09.025
82. Pinho S. Viability of the microencapsulation of a casein hydrolysate in lipid microparticles of cupuacu butter and stearic acid. Int J Food Stud. 2013;2(1):48–59.
83. Health N and. Novata® BD PH: Product data sheet [Internet]. [cited 2020 Apr 16]. Available from: https://e-applications.basf-ag.de/data/basf-pcan/pds2/pds2- web.nsf/CA7971BA2D4E244AC12573B100597C04/$File/NOVATA_r__BD_PH_E. pdf
84. Reithmeier H, Herrmann J, Gopferich A. Lipid microparticles as a parenteral controlled release device for peptides. 2001;73:339–50.
85. Qi C, Chen Y, Huang J, Jin Q. Preparation and characterization of catalase-loaded solid lipid nanoparticles based on soybean phosphatidylcholine. 2012;(February 2011):787–93.
86. Almeida AJ, Souto E. Solid lipid nanoparticles as a drug delivery system for peptides and proteins. Adv Drug Deliv Rev. 2007;59(6):478–90.
87. Mehnert W, Mäder K. Solid lipid nanoparticles: Production, characterization and applications. Adv Drug Deliv Rev [Internet]. 2012;64(SUPPL.):83–101. Available from: http://dx.doi.org/10.1016/j.addr.2012.09.021
88. Gordillo-Galeano A, Ponce A, Mora-Huertas CE. Surface structural characteristics of some colloidal lipid systems used in pharmaceutics. J Drug Deliv Sci Technol [Internet]. 2021;62(January):102345. Available from: https://doi.org/10.1016/j.jddst.2021.102345
89. Alexandridis P, Holzwarth JF, Hatton TA. Micellization of Poly(ethylene oxide)- Poly(propylene oxide)-Poly(ethylene oxide) Triblock Copolymers in Aqueous Solutions: Thermodynamics of Copolymer Association. Macromolecules. 1994;27(9):2414–25.
90. Mandal B, Bhattacharjee H, Mittal N, Sah H, Balabathula P, Thoma LA, et al. Core – shell-type lipid – polymer hybrid nanoparticles as a drug delivery platform. Nanomedicine Nanotechnology, Biol Med [Internet]. 2013;9(4):474–91. Available from: http://dx.doi.org/10.1016/j.nano.2012.11.010
91. Garg NK, Singh B, Kushwah V, Tyagi RK, Sharma R, Jain S, et al. The ligand (s) anchored lipobrid nanoconstruct mediated delivery of methotrexate: An effective approach in breast cancer therapeutics. Nanomedicine Nanotechnology, Biol Med [Internet]. 2016;12(7):2043–60. Available from: http://dx.doi.org/10.1016/j.nano.2016.05.008
92. Mukherjee A, Waters AK, Kalyan P, Achrol AS, Kesari S, Yenugonda VM. Lipidpolymer hybrid nanoparticles as a nextgeneration drug delivery platform: State of the art, emerging technologies, and perspectives. Int J Nanomedicine. 2019;14:1937– 52.
93. Chaudhary Z, Ahmed N, .ur.Rehman A, Khan GM. Lipid polymer hybrid carrier systems for cancer targeting: A review. Int J Polym Mater Polym Biomater [Internet]. 2018;67(2):86–100. Available from: https://doi.org/10.1080/00914037.2017.1300900
94. Garg NK, Tandel N, Jadon RS, Tyagi RK, Katare OP. Lipid–polymer hybrid nanocarrier-mediated cancer therapeutics: current status and future directions. Drug Discov Today [Internet]. 2018;23(9):1610–21. Available from: https://doi.org/10.1016/j.drudis.2018.05.033
95. Beija M, Salvayre R, Viguerie NL, Marty J. Colloidal systems for drug delivery : from design to therapy. Trends Biotechnol [Internet]. 2012;30(9):485–96. Available from: http://dx.doi.org/10.1016/j.tibtech.2012.04.008
96. Wong HL, Rauth AM, Bendayan R, Manias JL, Ramaswamy M, Liu Z, et al. Research Paper A New Polymer Y Lipid Hybrid Nanoparticle System Increases Cytotoxicity of Doxorubicin Against Multidrug-Resistant Human Breast Cancer Cells. 2006;23(7):1574–85.
97. Wong HL, Bendayan R, Mike A, Yu X. Simultaneous delivery of doxorubicin and GG918 ( Elacridar ) by new Polymer-Lipid Hybrid Nanoparticles ( PLN ) for enhanced treatment of multidrug-resistant breast cancer. 2006;116:275–84.
98. Li Y, Lun H, Shuhendler AJ, Rauth AM, Yu X. Molecular interactions , internal structure and drug release kinetics of rationally developed polymer – lipid hybrid nanoparticles. 2008;128:60–70.
99. Mandal B, Mittal NK, Balabathula P, Thoma LA, Wood GC. Development and in vitro evaluation of core-shell type lipid-polymer hybrid nanoparticles for the delivery of erlotinib in non-small cell lung cancer. Eur J Pharm Sci [Internet]. 2016;81:162–71. Available from: http://dx.doi.org/10.1016/j.ejps.2015.10.021
100. Devrim B, Kara A, Vural İ, Bozkır A. Lysozyme-loaded lipid-polymer hybrid nanoparticles: preparation, characterization and colloidal stability evaluation. Drug Dev Ind Pharm. 2016;42(11):1865–76.
101. Wu C, Baldursdottir S, Yang M, Mu H. Lipid and PLGA hybrid microparticles as carriers for protein delivery. J Drug Deliv Sci Technol [Internet]. 2018;43:65–72. Available from: https://doi.org/10.1016/j.jddst.2017.09.006
102. Bose RJC, Lee SH, Park H. Lipid polymer hybrid nanospheres encapsulating antiproliferative agents for stent applications. J Ind Eng Chem [Internet]. 2016;36:284–92. Available from: http://dx.doi.org/10.1016/j.jiec.2016.02.015
103. Li H, Teng Y, Sun J, Liu J. Inhibition of hemangioma growth using polymer–lipid hybrid nanoparticles for delivery of rapamycin. Biomed Pharmacother [Internet]. 2017;95:875–84. Available from: https://doi.org/10.1016/j.biopha.2017.08.035
104. K.S. J, Snigdha SS, Kalarikkal N, Pothen LA, Thomas S. Gelatin modified lipid nanoparticles for anti- viral drug delivery. Chem Phys Lipids [Internet]. 2017;207:24– 37. Available from: http://dx.doi.org/10.1016/j.chemphyslip.2017.07.002
105. Rose F, Wern JE, Gavins F, Andersen P, Follmann F, Foged C. A strong adjuvant based on glycol-chitosan-coated lipid-polymer hybrid nanoparticles potentiates mucosal immune responses against the recombinant Chlamydia trachomatis fusion antigen CTH522. J Control Release [Internet]. 2018;271(December 2017):88–97. Available from: https://doi.org/10.1016/j.jconrel.2017.12.003
106. Li J, Ma YJ, Wang Y, Chen BZ, Guo XD, Zhang CY. Dual redox/pH-responsive hybrid polymer-lipid composites: Synthesis, preparation, characterization and application in drug delivery with enhanced therapeutic efficacy. Chem Eng J [Internet]. 2018;341(November 2017):450–61. Available from: https://doi.org/10.1016/j.cej.2018.02.055
107. Toragall V, Jayapala N, Vallikannan B. Chitosan-oleic acid-sodium alginate a hybrid nanocarrier as an efficient delivery system for enhancement of lutein stability and bioavailability. Int J Biol Macromol [Internet]. 2020;150:578–94. Available from: https://doi.org/10.1016/j.ijbiomac.2020.02.104
108. Patel G, Thakur NS, Kushwah V, Patil MD, Nile SH, Jain S, et al. Mycophenolate coadministration with quercetin via lipid-polymer hybrid nanoparticles for enhanced breast cancer management. Nanomedicine Nanotechnology, Biol Med [Internet]. 2020;24:102147. Available from: https://doi.org/10.1016/j.nano.2019.102147
109. Thanki K, van Eetvelde D, Geyer A, Fraire J, Hendrix R, Van Eygen H, et al. Mechanistic profiling of the release kinetics of siRNA from lipidoid-polymer hybrid nanoparticles in vitro and in vivo after pulmonary administration. J Control Release [Internet]. 2019;310(August):82–93. Available from: https://doi.org/10.1016/j.jconrel.2019.08.004
110. Leng D, Thanki K, Fattal E, Foged C, Yang M. Engineering of budesonide-loaded lipid-polymer hybrid nanoparticles using a quality-by-design approach. Int J Pharm [Internet]. 2018;548(2):740–6. Available from: https://doi.org/10.1016/j.ijpharm.2017.08.094
111. Bachhav SS, Dighe VD, Kotak D, Devarajan P V. Rifampicin Lipid-Polymer hybrid nanoparticles (LIPOMER) for enhanced Peyer’s patch uptake. Int J Pharm [Internet]. 2017;532(1):612–22. Available from: http://dx.doi.org/10.1016/j.ijpharm.2017.09.040
112. Tahir N, Madni A, Balasubramanian V, Rehman M, Correia A, Kashif PM, et al. Development and optimization of methotrexate-loaded lipid-polymer hybrid nanoparticles for controlled drug delivery applications. Int J Pharm [Internet]. 2017;533(1):156–68. Available from: http://dx.doi.org/10.1016/j.ijpharm.2017.09.061
113. Belletti D, Grabrucker AM, Pederzoli F, Menerath I, Vandelli MA, Tosi G, et al. Hybrid nanoparticles as a new technological approach to enhance the delivery of cholesterol into the brain. Int J Pharm. 2018;543(1–2):300–10.
114. Yalcin TE, Ilbasmis-Tamer S, Takka S. Development and characterization of gemcitabine hydrochloride loaded lipid polymer hybrid nanoparticles (LPHNs) using central composite design. Int J Pharm [Internet]. 2018;548(1):255–62. Available from: https://doi.org/10.1016/j.ijpharm.2018.06.063
115. Li C, Ge X, Wang L. Construction and comparison of different nanocarriers for codelivery of cisplatin and curcumin: A synergistic combination nanotherapy for cervical cancer. Biomed Pharmacother [Internet]. 2017;86(27):628–36. Available from: http://dx.doi.org/10.1016/j.biopha.2016.12.042
116. Zhang Y, Zhang P, Zhu T. Ovarian carcinoma biological nanotherapy: Comparison of the advantages and drawbacks of lipid, polymeric, and hybrid nanoparticles for cisplatin delivery. Biomed Pharmacother [Internet]. 2019;109(1):475–83. Available from: https://doi.org/10.1016/j.biopha.2018.10.158
117. Ma P, Li T, Xing H, Wang S, Sun Y, Sheng X, et al. Local anesthetic effects of bupivacaine loaded lipid-polymer hybrid nanoparticles: In vitro and in vivo evaluation. Biomed Pharmacother [Internet]. 2017;89(October 2011):689–95. Available from: http://dx.doi.org/10.1016/j.biopha.2017.01.175
18. Qiu J, Cai G, Liu X, Ma D. αvβ3 integrin receptor specific peptide modified, salvianolic acid B and panax notoginsenoside loaded nanomedicine for the combination therapy of acute myocardial ischemia. Biomed Pharmacother [Internet]. 2017;96(89):1418–26. Available from: https://doi.org/10.1016/j.biopha.2017.10.086
119. Song Z, Shi Y, Han Q, Dai G. Endothelial growth factor receptor-targeted and reactive oxygen species-responsive lung cancer therapy by docetaxel and resveratrol encapsulated lipid-polymer hybrid nanoparticles. Biomed Pharmacother [Internet]. 2018;105(28):18–26. Available from: https://doi.org/10.1016/j.biopha.2018.05.095
120. Yugui F, Wang H, Sun D, Zhang X. Nasopharyngeal cancer combination chemoradiation therapy based on folic acid modified, gefitinib and yttrium 90 coloaded, core-shell structured lipid-polymer hybrid nanoparticles. Biomed Pharmacother. 2019;114(27):0–6.
121. Wang J, Su G, Yin X, Luo J, Gu R, Wang S, et al. Non-small cell lung cancer-targeted, redox-sensitive lipid-polymer hybrid nanoparticles for the delivery of a second-generation irreversible epidermal growth factor inhibitor—Afatinib: In vitro and in vivo evaluation. Biomed Pharmacother [Internet]. 2019;120(September):109493. Available from: https://doi.org/10.1016/j.biopha.2019.109493
122. Pang J, Xing H, Sun Y, Feng S, Wang S. Non-small cell lung cancer combination therapy: Hyaluronic acid modified, epidermal growth factor receptor targeted, pH sensitive lipid-polymer hybrid nanoparticles for the delivery of erlotinib plus bevacizumab. Biomed Pharmacother [Internet]. 2020;125(December 2019):109861. Available from: https://doi.org/10.1016/j.biopha.2020.109861
123. Seedat N, Kalhapure RS, Mocktar C, Vepuri S, Jadhav M, Soliman M, et al. Coencapsulation of multi-lipids and polymers enhances the performance of vancomycin in lipid-polymer hybrid nanoparticles: In vitro and in silico studies. Mater Sci Eng C [Internet]. 2016;61:616–30. Available from: http://dx.doi.org/10.1016/j.msec.2015.12.053
124. Xue HY, Tran N, Wong HL. A biodistribution study of solid lipid-polyethyleneimine hybrid nanocarrier for cancer RNAi therapy. Eur J Pharm Biopharm [Internet]. 2016;108:68–75. Available from: http://dx.doi.org/10.1016/j.ejpb.2016.08.014
125. Zhao J, Zhang X, Sun X, Zhao M, Yu C, Lee RJ, et al. Dual-functional lipid polymeric hybrid pH-responsive nanoparticles decorated with cell penetrating peptide and folate for therapy against rheumatoid arthritis. Eur J Pharm Biopharm [Internet]. 2018;130(June):39–47. Available from: https://doi.org/10.1016/j.ejpb.2018.06.020
126. Jansen MAA, Klausen LH, Thanki K, Lyngsø J, Skov Pedersen J, Franzyk H, et al. Lipidoid-polymer hybrid nanoparticles loaded with TNF siRNA suppress inflammation after intra-articular administration in a murine experimental arthritis model. Eur J Pharm Biopharm [Internet]. 2019;142(November 2018):38–48. Available from: https://doi.org/10.1016/j.ejpb.2019.06.009
127. Wu C, Luo X, Baldursdottir SG, Yang M, Sun X, Mu H. In vivo evaluation of solid lipid microparticles and hybrid polymer-lipid microparticles for sustained delivery of leuprolide. Eur J Pharm Biopharm [Internet]. 2019;142(July):315–21. Available from: https://doi.org/10.1016/j.ejpb.2019.07.010
128. Zhang J, Hu J, Chan HF, Skibba M, Liang G, Chen M. iRGD decorated lipid-polymer hybrid nanoparticles for targeted co-delivery of doxorubicin and sorafenib to enhance anti-hepatocellular carcinoma efficacy. Nanomedicine Nanotechnology, Biol Med [Internet]. 2016;12(5):1303–11. Available from: http://dx.doi.org/10.1016/j.nano.2016.01.017
129. Liu Y, Jiang Z, Hou X, Xie X, Shi J, Shen J, et al. Functional lipid polymeric nanoparticles for oral drug delivery: Rapid mucus penetration and improved cell entry and cellular transport. Nanomedicine Nanotechnology, Biol Med [Internet]. 2019;21:102075. Available from: https://doi.org/10.1016/j.nano.2019.102075
130. Boushra M, Tous S, Fetih G, Xue HY, Wong HL. Development of bi-polymer lipid hybrid nanocarrier (BLN) to improve the entrapment and stability of insulin for efficient oral delivery. J Drug Deliv Sci Technol [Internet]. 2019;49(July 2018):632– 41. Available from: https://doi.org/10.1016/j.jddst.2019.01.007
131. Meraj Anjum M, Kanoujia J, Parashar P, Arya M, K. Yadav A, A. Saraf S. Evaluation of a Polymer-Lipid-Polymer System Utilising Hybrid Nanoparticles of Dapsone as a Novel Antiacne Agent. Curr Drug ther. 2016;11(2):86–100
132. Arya RKK, Juyal V. Polymer -Lipid Hybrid Nanoparticles for Brain Targeting Through Intranasal Delivery. J Drug Deliv Ther. 2017;7(4):129–36.
133. Tahir N, Madni A, Kashif PM, Rehman M, Raza A, Khan MI, et al. Formulation and compatibility assessment of PLGA/lecithin based lipid-polymer hybrid nanoparticles containing doxorubicin. Acta Pol Pharm - Drug Res. 2017;74(5):1563–72.
134. Tahir N, Madni A, Correia A, Rehman M, Balasubramanian V, Khan MM, et al. Lipidpolymer hybrid nanoparticles for controlled delivery of hydrophilic and lipophilic doxorubicin for breast cancer therapy. Int J Nanomedicine. 2019;14:4961–74.
135. Zhao W, Zhang C, Li B, Zhang X, Luo X, Zeng C, et al. Lipid Polymer Hybrid Nanomaterials for mRNA Delivery. Cell Mol Bioeng. 2018;11(5):397–406.
136. Gu J, Chen Y, Tong L, Wang X, Yu D, Wu H. Astaxanthin-loaded polymer-lipid hybrid nanoparticles (ATX-LPN): Assessment of potential otoprotective effects. J Nanobiotechnology [Internet]. 2020;18(1):1–17. Available from: https://doi.org/10.1186/s12951-020-00600-x
137. Wang Q, Alshaker H, Böhler T, Srivats S, Chao Y, Cooper C, et al. Core shell lipidpolymer hybrid nanoparticles with combined docetaxel and molecular targeted therapy for the treatment of metastatic prostate cancer. Sci Rep. 2017;7(1):1–8.
138. Abdou EM, Fayed MAA, Helal D, Ahmed KA. Assessment of the hepatoprotective effect of developed lipid-polymer hybrid nanoparticles (LPHNPs) encapsulating naturally extracted β-Sitosterol against CCl4 induced hepatotoxicity in rats. Sci Rep [Internet]. 2019;9(1):1–14. Available from: http://dx.doi.org/10.1038/s41598-019- 56320-2
139. Cao S, Slack SD, Levy CN, Hughes SM, Jiang Y, Yogodzinski C, et al. Hybrid nanocarriers incorporating mechanistically distinct drugs for lymphatic CD4 + T cell activation and HIV-1 latency reversal. Sci Adv. 2019;5(3):1–13.
140. Garg NK, Tyagi RK, Sharma G, Jain A, Singh B, Jain S, et al. Functionalized LipidPolymer Hybrid Nanoparticles Mediated Codelivery of Methotrexate and Aceclofenac: A Synergistic Effect in Breast Cancer with Improved Pharmacokinetics Attributes. Mol Pharm. 2017;14(6):1883–97.
141. Wang J, Zhang L, Chi H, Wang S. An alternative choice of lidocaine-loaded liposomes: lidocaine-loaded lipid–polymer hybrid nanoparticles for local anesthetic therapy. Drug Deliv. 2016;23(4):1254–60.
142. Chen W, Guo M, Wang S. Anti prostate cancer using PEGylated bombesin containing, cabazitaxel loading nano-sized drug delivery system. Drug Dev Ind Pharm. 2016;42(12):1968–76.
143. 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
144. Shao Y, Luo W, Guo Q, Li X, Zhang Q, Li J. In vitro and in vivo effect of hyaluronic acid modified, doxorubicin and gallic acid co-delivered lipid-polymeric hybrid nanosystem for leukemia therapy. Drug Des Devel Ther. 2019;13:2043–55.
145. Wang J. Combination treatment of cervical cancer using folate-decorated, phsensitive, carboplatin and paclitaxel co-loaded lipid-polymer hybrid nanoparticles. Drug Des Devel Ther. 2020;14:823–32.
146. Yu Z, Chen F, Qi X, Dong Y, Zhang Y, Ge Z, et al. Epidermal growth factor receptor aptamer-conjugated polymer-lipid hybrid nanoparticles enhance salinomycin delivery to osteosarcoma and cancer stem cells. Exp Ther Med. 2018;15(2):1247– 56.
147. Gao F, Zhang J, Fu C, Xie X, Peng F, You J, et al. iRGD-modified lipid–polymer hybrid nanoparticles loaded with isoliquiritigenin to enhance anti-breast cancer effect and tumor-targeting ability. Int J Nanomedicine. 2017;12:4147–62.
148. Khan MM, Madni A, Torchilin V, Filipczak N, Pan J, Tahir N, et al. Lipid-chitosan hybrid nanoparticles for controlled delivery of cisplatin. Drug Deliv [Internet]. 2019;26(1):765–72. Available from: https://doi.org/10.1080/10717544.2019.1642420
149. Sun S, Liang N, Yamamoto H, Kawashima Y, Cui F, Yan P. pH-sensitive poly(lactideco-glycolide) nanoparticle Composite Microcapsules for Oral Delivery of Insulin. Int J Nanomedicine. 2015;10:3489–98.
150. Tng DJH, Song P, Lin G, Soehartono AM, Yang G, Yang C, et al. Synthesis and characterization of multifunctional hybrid-polymeric nanoparticles for drug delivery and multimodal imaging of cancer. Int J Nanomedicine. 2015;10:5771–86.
151. Wu B, Lu ST, Deng K, Yu H, Cui C, Zhang Y, et al. MRI-guided targeting delivery of doxorubicin with reduction-responsive lipid-polymer hybrid nanoparticles. Int J Nanomedicine. 2017;12:6871–82.
152. Li J, Xu W, Yuan X, Chen H, Song H, Wang B, et al. Polymer-lipid hybrid anti-HER 2 nanoparticles for targeted salinomycin delivery to HER 2-positive breast cancer stem cells and cancer cells. Int J Nanomedicine. 2017;12:6909–21.
153. Wu B, Lu ST, Zhang LJ, Zhuo RX, Xu HB, Huang SW. Codelivery of doxorubicin and triptolide with reduction-sensitive lipid–polymer hybrid nanoparticles for in vitro and in vivo synergistic cancer treatment. Int J Nanomedicine. 2017;12:1853–62.
154. Men W, Zhu P, Dong S, Liu W, Zhou K, Bai Y, et al. Fabrication of dual pH/redoxresponsive lipid-polymer hybrid nanoparticles for anticancer drug delivery and controlled release. Int J Nanomedicine. 2019;14:8001–11.
155. Hu Y, Hoerle R, Ehrich M, Zhang C. Engineering the lipid layer of lipid–PLGA hybrid nanoparticles for enhanced in vitro cellular uptake and improved stability. Acta Biomater [Internet]. 2015 Dec;28(1):149–59. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1742706115301252
156. Hu Y, Zhao Z, Ehrich M, Fuhrman K, Zhang C. In vitro controlled release of antigen in dendritic cells using pH-sensitive liposome-polymeric hybrid nanoparticles. Polymer (Guildf). 2015;80:171–9.
157. Dehaini D, Fang RH, Luk BT, Pang Z, Hu CMJ, Kroll A V., et al. Ultra-small lipidpolymer hybrid nanoparticles for tumor-penetrating drug delivery. Nanoscale. 2016;8(30):14411–9.
158. Hu Y, Smith D, Frazier E, Hoerle R, Ehrich M, Zhang C. The next-generation nicotine vaccine: a novel and potent hybrid nanoparticle-based nicotine vaccine. Biomaterials [Internet]. 2016 Nov;106(12):228–39. Available from: https://linkinghub.elsevier.com/retrieve/pii/S014296121630415X
159. Golan‐Paz S, Frizzell H, Woodrow KA. Cross‐Platform Comparison of Therapeutic Delivery from Multilamellar Lipid‐Coated Polymer Nanoparticles. Macromol Biosci [Internet]. 2019 Apr 27;19(4):1800362. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1002/mabi.201800362
160. Wei J, Sun J, Liu Y. Enhanced targeting of prostate cancer-initiating cells by salinomycin-encapsulated lipid-PLGA nanoparticles linked with CD44 antibodies. Oncol Lett. 2019;17(4):4024–33.
161. Bhardwaj A, Mehta S, Yadav S, Singh SK, Grobler A, Goyal AK, et al. Pulmonary delivery of antitubercular drugs using spray-dried lipid–polymer hybrid nanoparticles. Artif Cells, Nanomedicine Biotechnol. 2016;44(6):1544–55.
162. Liang J, Liu Y, Liu J, Li Z, Fan Q, Jiang Z, et al. Chitosan-functionalized lipid-polymer hybrid nanoparticles for oral delivery of silymarin and enhanced lipid-lowering effect in NAFLD. J Nanobiotechnology [Internet]. 2018;16(1):1–12. Available from: https://doi.org/10.1186/s12951-018-0391-9
163. Yasar H, Biehl A, De Rossi C, Koch M, Murgia X, Loretz B, et al. Kinetics of mRNA delivery and protein translation in dendritic cells using lipid-coated PLGA nanoparticles. J Nanobiotechnology [Internet]. 2018;16(1):1–19. Available from: https://doi.org/10.1186/s12951-018-0401-y
164. Jin Z, Lv Y, Cao H, Yao J, Zhou J, He W, et al. Core-shell nanocarriers with high paclitaxel loading for passive and active targeting. Nat Publ Gr [Internet]. 2016;(February):1–10. Available from: http://dx.doi.org/10.1038/srep27559
165. Ishak RAH, Mostafa NM, Kamel AO. Stealth lipid polymer hybrid nanoparticles loaded with rutin for effective brain delivery – comparative study with the gold standard (Tween 80): Optimization, characterization and biodistribution. Drug Deliv [Internet]. 2017;24(1):1874–90. Available from: https://doi.org/10.1080/10717544.2017.1410263
166. Yan J, Wang Y, Zhang X, Liu S, Tian C, Wang H. Targeted nanomedicine for prostate cancer therapy: docetaxel and curcumin co-encapsulated lipid–polymer hybrid nanoparticles for the enhanced anti-tumor activity in vitro and in vivo. Drug Deliv. 2016;23(5):1757–62.
167. Tawfik MA, Tadros MI, Mohamed MI. Lipomers (Lipid-polymer Hybrid Particles) of Vardenafil Hydrochloride: a Promising Dual Platform for Modifying the Drug Release Rate and Enhancing Its Oral Bioavailability. AAPS PharmSciTech. 2018;19(8):3650– 60.
168. Thakur K, Sharma G, Singh B, Chhibber S, Katare OP. Nano-engineered lipidpolymer hybrid nanoparticles of fusidic acid: an investigative study on dermatokinetics profile and MRSA-infected burn wound model. Drug Deliv Transl Res. 2019;9(4):748–63.
dc.rights.coar.fl_str_mv http://purl.org/coar/access_right/c_abf2
dc.rights.license.spa.fl_str_mv Atribución-NoComercial-CompartirIgual 4.0 Internacional
dc.rights.uri.spa.fl_str_mv http://creativecommons.org/licenses/by-nc-sa/4.0/
dc.rights.accessrights.spa.fl_str_mv info:eu-repo/semantics/openAccess
rights_invalid_str_mv Atribución-NoComercial-CompartirIgual 4.0 Internacional
http://creativecommons.org/licenses/by-nc-sa/4.0/
http://purl.org/coar/access_right/c_abf2
eu_rights_str_mv openAccess
dc.format.extent.spa.fl_str_mv xiii, 146 paginas
dc.format.mimetype.spa.fl_str_mv application/pdf
dc.publisher.spa.fl_str_mv Universidad Nacional de Colombia
dc.publisher.program.spa.fl_str_mv Bogotá - Ciencias - Maestría en Ciencias Farmacéuticas
dc.publisher.department.spa.fl_str_mv Departamento de Farmacia
dc.publisher.faculty.spa.fl_str_mv Facultad de Administración
dc.publisher.place.spa.fl_str_mv Bogotá, Colombia
dc.publisher.branch.spa.fl_str_mv Universidad Nacional de Colombia - Sede Bogotá
institution Universidad Nacional de Colombia
bitstream.url.fl_str_mv https://repositorio.unal.edu.co/bitstream/unal/80864/1/1032480023.2021%20-%20Maria%20Angelica%20Velandia%20Paris.pdf
https://repositorio.unal.edu.co/bitstream/unal/80864/2/license.txt
https://repositorio.unal.edu.co/bitstream/unal/80864/3/1032480023.2021%20-%20Maria%20Angelica%20Velandia%20Paris.pdf.jpg
bitstream.checksum.fl_str_mv 65c8f18d62e1834b2a974e48e6dbc681
8153f7789df02f0a4c9e079953658ab2
71f12ff5f9dea73f691536f0672b1a83
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
repository.name.fl_str_mv Repositorio Institucional Universidad Nacional de Colombia
repository.mail.fl_str_mv repositorio_nal@unal.edu.co
_version_ 1806885925540593664
spelling Atribución-NoComercial-CompartirIgual 4.0 Internacionalhttp://creativecommons.org/licenses/by-nc-sa/4.0/info:eu-repo/semantics/openAccesshttp://purl.org/coar/access_right/c_abf2Rosas Pérez, Jaiver Eduardod998f772bc3d8f602a106bc9e589eac8Velandia Paris, María Angélica7853ac119a9bf4c236bb7540b4e65917Sistemas para liberación controlada de moléculas biológicamente activas (SILICOMOBA)2022-02-03T15:42:48Z2022-02-03T15:42:48Z2021https://repositorio.unal.edu.co/handle/unal/80864Universidad Nacional de ColombiaRepositorio Institucional Universidad Nacional de Colombiahttps://repositorio.unal.edu.co/ilustraciones, fotografías, graficasEn los últimos años se ha despertado un gran interés por los sistemas para la entrega de fármacos (SEF) mediante liberación controlada, debido a que permiten superar ciertas limitaciones de algunos ingredientes farmacéuticos activos (IFAs). Entre estos IFAs, se destacan los péptidos que, a pesar de ser degradados rápidamente, son moléculas esenciales en la vida debido a la posibilidad que tienen de cumplir múltiples funciones en el organismo, lo que ha permitido que sean considerados como IFA de elección para tratar numerosas enfermedades. Se ha reportado que los SEF particulados son capaces de proteger a moléculas lábiles como los péptidos de agentes externos como las enzimas, haciendo que se prolongue su tiempo de vida media y por tanto mejorando su actividad. Teniendo en cuenta esto se han diseñado diferentes SEF, entre ellos los liposomas, las partículas poliméricas y las partículas sólidas lipídicas, siendo estos sistemas los que han mostrado de manera general los mejores resultados como transportadores de IFAs. Producto de estas investigaciones, se ha mostrado un gran interés en desarrollar un SEF que permita reunir las ventajas de las partículas poliméricas y las partículas sólidas lipídicas, planteándose de esta forma el diseño y desarrollo de los sistemas híbridos lipopoliméricos. Con el propósito de contribuir en el campo de la investigación de SEF, en el marco de este trabajo se planteó el desarrollo una metodología para la obtención de tres sistemas particulados: micropartículas poliméricas, partículas sólidas lipídicas y micropartículas lipopoliméricas, empleando como material polimérico PLGA 50:50 (Viscosidad inherente: 0,8dL/g) y como material lipídico una mezcla de mono, di y triglicéridos C12-C18. Todos los sistemas se obtuvieron vacíos y cargados con un péptido sintético modelo de naturaleza hidrofílica, mediante la metodología de doble emulsión – evaporación del solvente Todos los sistemas fueron caracterizados en cuanto a morfología, tamaño, potencial Z, eficiencia de encapsulación y perfil de liberación del péptido. Como resultado, se logró obtener partículas en su mayoría esféricas de tamaño micrométrico para los sistemas poliméricos (3,08-5,60 µm) y lipopoliméricos (3,22-3,93 µm), de tamaño nanométrico para el sistema sólido lipídico (135,8-162,9 nm) y con un potencial Z entre -18,5mV y -24,5mV para las micropartículas poliméricas, -15,5mV y -29,9mV para las partículas sólidas lipídicas, -20,0mV y -26,1mV para las micropartículas lipopoliméricas. En cuanto a la eficiencia de encapsulación se determinó que el mejor sistema fue el de las micropartículas poliméricas con una E.E. promedio del 57,42% seguido de las micropartículas lipopoliméricas cuya E.E. promedio fue de 43,15% y las partículas sólidas lipídicas con una E.E. promedio del 40,40%. En referencia al perfil de liberación, se observó que las micropartículas lipopoliméricas tienden a mostrar un comportamiento intermedio entre los observados en las micropartículas poliméricas y en las partículas lipídicas. En conclusión, en este trabajo se desarrolló una metodología para la obtención y caracterización de un sistema micropartícular lipopolimérico que permite la encapsulación de un péptido sintético modelo de naturaleza hidrofílica con características tanto de micropartículas poliméricas (Tamaño y morfología) como de partículas sólidas lipídicas (E.E. y perfil de liberación). (texto tomado de la fuente)In recent years, controlled release drug delivery systems (DDS) have had great interest due they make possible to overcome certain limitations of some active pharmaceutical ingredients (APIs), between these APIs stand out peptides that despite of fast degradation, are essential molecules in life due to the possibility to accomplish multiple functions in the body, this has allowed to be considerate as choice API to treat numerous diseases. Particulate DDS have been shown to be able to protect labile molecules such as peptides from external agents such as enzymes, prolonging their half-life and therefore improving their activity. Bearing this in mind, different DDS have been designed, including liposomes, polymeric particles, and solid lipid particles, these being the ones that in general have shown better results as APIs carriers. As product of these research a great interest has been shown in developing a DDS that allows to have advantages of polymeric particles and solid lipid particles, proposing in this way the design and development of hybrid lipopolymeric systems. With the purpose of contribute to research field of DDS, within the framework of this work, the development of a methodology for the obtention of three particulate systems: Polymeric microparticles, solid lipid particles and lipopolymeric particles was proposed, using as polymeric material PLGA 50:50 (Inherent Viscosity 0,8 dL/g) and as lipidic material a mixture of mono, di and triglycerides C12-C18. All systems were obtained empty and loaded with a model synthetic peptide of hydrophilic nature using a methodology of double emulsion – solvent evaporation. All systems obtained were characterized in terms of morphology, size, Z potential, encapsulation efficiency and release profile. As result, was achieved particles mostly spherical, of micrometrical size for polymeric systems (3,08-5,60 µm) and lipopolymeric (3,22-3,93 µm), nanometrical size for the solid lipid system (135,8-162,9 nm), with a Z potential between -18,5mV -and -24,5mV for the polymeric particles, -15,5mV and -29,9mV for the solid lipid particles and -20,0mV and -26,1mV for the lipopolymeric particles. Regarding encapsulation efficiency, it was observed that lipopolymeric particles tend to show an intermediate behavior between the behavior observed in polymeric particles and in lipid particles. In conclusion, in this work a method was developed to obtain and characterize a lipopolymeric microparticle system that allows the encapsulation of a model synthetic peptide of a hydrophilic nature with characteristics of both polymeric particles (size and morphology) and solid lipid particles (E.E. and release profile).MaestríaMagíster en Ciencias FarmacéuticasTecnología Farmacéutica: Sistemas micro y nano particularesxiii, 146 paginasapplication/pdfspaUniversidad Nacional de ColombiaBogotá - Ciencias - Maestría en Ciencias FarmacéuticasDepartamento de FarmaciaFacultad de AdministraciónBogotá, ColombiaUniversidad Nacional de Colombia - Sede Bogotá610 - Medicina y salud::615 - Farmacología y terapéuticaSistemas particuladosPolímeroLípidoPéptido sintéticoDoble emulsiónEvaporación del solventeHíbridosLipopoliméricosParticulate systemsPolymerLipidSynthetic peptideDouble emulsionSolvent evaporationHybridsLipopolymericsPolímeroPolymersCompuesto orgánicoDiseño y desarrollo de un sistema microparticular lipopolimérico para la administración de un péptido sintético modelo de naturaleza hidrofílicaDesign and development of a lipopolymeric microparticular system for the administration od a synthetic peptide of hydrophilic natureTrabajo de grado - Maestríainfo:eu-repo/semantics/masterThesisinfo:eu-repo/semantics/acceptedVersionTexthttp://purl.org/redcol/resource_type/TMLaReferencia1. Lau JL, Dunn MK. Therapeutic peptides: Historical perspectives, current development trends, and future directions. Bioorganic Med Chem [Internet]. 2018;26(10):2700–7. Available from: https://doi.org/10.1016/j.bmc.2017.06.0522. Sachdeva S. Peptides as ‘ Drugs ’: The Journey so Far. Int J Pept Res Ther. 2016;(May).3. Peptides C. Derek N. Woolfson 1,2 1 2. 2009;94(1):118–27.4. Grant GA. Synthetic peptides : a user’s guide. 2nd ed. Grant GA, editor. New York: Oxford Univesity Press; 2002.5. Vlieghe P, Lisowski V, Martinez J, Khrestchatisky M. Synthetic therapeutic peptides : science and market. 2010;15(January).6. Zhang J, Desale SS, Bronich TK. Polymer-based vehicles for therapeutic peptide delivery. Ther Deliv. 2015;6(11):1279–96.7. Couvreur P, Puisieux F. Nano- and microparticles for the delivery of polypeptides and proteins. Adv Drug Deliv Rev. 1993;10(2–3):141–62.8. Bruschi M. Modification of drug release. Strateg to Modify Drug Release from Pharm Syst. 2015;15–28.9. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in Medicine : Therapeutic Applications and Developments. 2008;83(5):761–9.10. Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics : an emerging treatment modality for cancer. 2008;7(SEPTEMbER):771–8211. Devrim B, Kara A, Vural İ, Bozkır A. Lysozyme-loaded lipid-polymer hybrid nanoparticles: preparation, characterization and colloidal stability evaluation. Drug Dev Ind Pharm. 2016;42(11):1865–76.12. Zhang L, Granick S. How to Stabilize Phospholipid Liposomes ( Using Nanoparticles ). 2006;0–4.13. Zhang L, Zhang LF. Lipid–Polymer Hybrid Nanoparticles: Synthesis,Characterization and Applications. Nano Life. 2010;01(01n02):163–73.14. Garud A, Singh D, Garud N. Solid Lipid Nanoparticles (SLN): Method, Characterization and Applications. Int Curr Pharm J. 2012;1(11):384–93.15. Ansari MJ, Anwer MK, Jamil S, Al-Shdefat R, Ali BE, Ahmad MM, et al. Enhanced oral bioavailability of insulin-loaded solid lipid nanoparticles: pharmacokinetic bioavailability of insulin-loaded solid lipid nanoparticles in diabetic rats. Drug Deliv [Internet]. 2016;23(6):1972–9. Available from: http://dx.doi.org/10.3109/10717544.2015.103966616. Chan JM, Zhang L, Yuet KP, Liao G, Rhee J, Langer R, et al. Biomaterials PLGA – lecithin – PEG core – shell nanoparticles for controlled drug delivery. Biomaterials [Internet]. 2009;30(8):1627–34. Available from: http://dx.doi.org/10.1016/j.biomaterials.2008.12.01317. Thevenot J, Troutier A, David L, Delair T, Pascal B. Steric Stabilization of Lipid / Polymer Particle Assemblies by Poly ( ethylene glycol ) -Lipids. 2007;3651–60.18. Grigoras AG. Polymer-lipid hybrid systems used as carriers for insulin delivery. Nanomedicine Nanotechnology, Biol Med [Internet]. 2017;13(8):2425–37. Available from: https://doi.org/10.1016/j.nano.2017.08.00519. Walton HF. General considerations. Ligand Exch Chromatogr. 2018;7–30.20. Ito Y. Drug delivery systems. Photochemistry for Biomedical Applications: From Device Fabrication to Diagnosis and Therapy. 2018. 231–275 p21. Nastruzzi C. Lipospheres in drug targets and delivery. 2005.22. Manjunath K, Reddy JS, Venkateswarlu V. Solid lipid nanoparticles as drug delivery systems. Methods Find Exp Clin Pharmacol [Internet]. 2005;27(2):127. Available from: http://journals.prous.com/journals/servlet/xmlxsl/pk_journals.xml_summary_pr?p_J ournalId=6&p_RefId=876286&p_IsPs=N23. Matougui N, Boge L, Groo AC, Umerska A, Ringstad L, Bysell H, et al. Lipid-based nanoformulations for peptide delivery. Int J Pharm [Internet]. 2016;502(1–2):80–97. Available from: http://dx.doi.org/10.1016/j.ijpharm.2016.02.01924. Shukla T, Upmanyu N, Pandey SP, Gosh D. Chapter 1. Lipid nanocarriers [Internet]. Lipid Nanocarriers for Drug Targeting. Elsevier Inc.; 2018. 1–48 p. Available from: http://dx.doi.org/10.1016/B978-0-12-813687-4.00001-325. Rawal SU, Patel MM. Chapter 2. Lipid nanoparticulate systems: Modern versatile drug carriers [Internet]. Lipid Nanocarriers for Drug Targeting. Elsevier Inc.; 2018. 49–138 p. Available from: http://dx.doi.org/10.1016/B978-0-12-813687-4.00002-526. Abbasi E, Aval SF, Akbarzadeh A, Milani M, Nasrabadi HT, Joo SW, et al. Dendrimers: Synthesis, applications, and properties. Vol. 9, Nanoscale Research Letters. Springer New York LLC; 2014. p. 1–10.27. Jyothi NVN, Prasanna PM, Sakarkar SN, Prabha KS, Ramaiah PS, Srawan GY. Microencapsulation techniques, factors influencing encapsulation efficiency. J Microencapsul. 2010;27(3):187–9728. Freiberg S, Zhu XX. Polymer microspheres for controlled drug release. Int J Pharm. 2004;282(1–2):1–18.29. Zhang F, Fan J bing, Wang S. Interfacial Polymerization: From Chemistry to Functional Materials. Angew Chemie - Int Ed. 2020;59(49):21840–56.30. Kwak H-S. Nano- and Microencapsulation. 2014.31. JONG; H. G. BDHRK. Coacervation (Partial miscibility in colloid systems). (Preliminary Communication). 1929; Available from: https://www.dwc.knaw.nl/DL/publications/PU00015781.pdf32. Ghosh SK. Functional Coatings and Microencapsulation: A General Perspective. Funct Coatings By Polym Microencapsul. 2006;1–2833. Zuidam NJ, Nedović VA. Encapsulation technologies for active food ingredients and food processing. Encapsulation Technol Act Food Ingredients Food Process. 2010;1–400.34. Freitas S, Merkle HP, Gander B. Microencapsulation by solvent extraction/evaporation: reviewing the state of the art of microsphere preparation process technology. J Control Release [Internet]. 2005 Feb;102(2):313–32. Available from: https://linkinghub.elsevier.com/retrieve/pii/S016836590400484535. Hallan SS, Kaur P, Kaur V, Mishra N, Vaidya B. Lipid polymer hybrid as emerging tool in nanocarriers for oral drug delivery. Artif Cells, Nanomedicine Biotechnol. 2016;44(1):334–49.36. Hadinoto K, Sundaresan A, Cheow WS. Lipid-polymer hybrid nanoparticles as a new generation therapeutic delivery platform: A review. Eur J Pharm Biopharm [Internet]. 2013;85(3 PART A):427–43. Available from: http://dx.doi.org/10.1016/j.ejpb.2013.07.00237. Dong W, Wang X, Liu C, Zhang X, Zhang X, Chen X, et al. Chitosan based polymerlipid hybrid nanoparticles for oral delivery of enoxaparin. Int J Pharm [Internet]. 2018;547(1–2):499–505. Available from: https://doi.org/10.1016/j.ijpharm.2018.05.07638. Sengel-Turk CT, Gumustas M, Uslu B, Ozkan SA. A novel approach for drug targeting: Core-shell type lipid-polymer hybrid nanocarriers. Core-shell type lipidpolymer hybrid nanocarriers. [Internet]. Design of Nanostructures for Theranostics Applications. Elsevier Inc.; 2018. 69–107 p. Available from: http://dx.doi.org/10.1016/B978-0-12-813669-0.00003-839. Jain A, Jain A, Gulbake A, Shilpi S, Hurkat P, Jain SK. Peptide and Protein Delivery Using New Drug Delivery Systems. Crit Rev Ther Drug Carrier Syst. 2013;30(4):293–329.40. McClements DJ. Encapsulation, protection, and delivery of bioactive proteins and peptides using nanoparticle and microparticle systems: A review. Adv Colloid Interface Sci [Internet]. 2018;253(2017):1–22. Available from: https://doi.org/10.1016/j.cis.2018.02.00241. Pinto Reis C, Neufeld RJ, Ribeiro AJ, Veiga F. Nanoencapsulation II. Biomedical applications and current status of peptide and protein nanoparticulate delivery systems. Nanomedicine Nanotechnology, Biol Med. 2006;2(2):53–65.42. Kovalainen M, Mönkäre J, Riikonen J, Pesonen U, Vlasova M, Salonen J. Novel Delivery Systems for Improving the Clinical Use of Peptides. 2015;(July):541–61.43. Patarroyo ME, Romero P, Torres ML, Clavijo P, Moreno A, Martínez A, et al. Induction of protective immunity against experimental infection with malaria using synthetic peptides. Nature. 1988;328(6131):629–32.44. Lozano JM, Patarroyo ME. A rational strategy for a malarial vaccine development. Microbes Infect. 2007;9(6):751–60.45. McClements DJ. Encapsulation, protection, and delivery of bioactive proteins and peptides using nanoparticle and microparticle systems: A review. Adv Colloid Interface Sci [Internet]. 2018 Mar 1 [cited 2020 Mar 20];253:1–22. Available from: https://www.sciencedirect.com/science/article/pii/S000186861730517146. Bolhassani A. Improvements in chemical carriers of proteins and peptides. 2019;43:437–5247. Gasco AR. Biodegradable PLGA microspheres as a delivery system for malaria synthetic peptide SPf66. 2001;19:4445–51.48. Dave V, Tak K, Sohgaura A, Gupta A, Sadhu V. Lipid-polymer hybrid nanoparticles : Synthesis strategies and biomedical applications. J Microbiol Methods [Internet]. 2019;160(March):130–42. Available from: https://doi.org/10.1016/j.mimet.2019.03.01749. Samad A, Tariq M, Alam MI, Akhter MS. Microsphere: A novel drug delivery system. Colloids Drug Deliv. 2016;11(4):455–7850. Acar H, Ting JM, Srivastava S, LaBelle JL, Tirrell M V. Molecular engineering solutions for therapeutic peptide delivery. Chem Soc Rev [Internet]. 2017;46(21):6553–69. Available from: http://dx.doi.org/10.1039/C7CS00536A51. Blasi P. Poly(lactic acid)/poly(lactic-co-glycolic acid)-based microparticles: an overview. J Pharm Investig [Internet]. 2019;49(4):337–46. Available from: https://doi.org/10.1007/s40005-019-00453-z52. Hajavi J, Ebrahimian M, Sankian M, Khakzad MR, Hashemi M. Optimization of PLGA formulation containing protein or peptide-based antigen: Recent advances. J Biomed Mater Res - Part A. 2018;106(9):2540–51.53. Qi F, Wu J, Li H, Ma G. Recent research and development of PLGA/PLA microspheres/nanoparticles: A review in scientific and industrial aspects. Front Chem Sci Eng. 2019;13(1):14–27.54. Schoubben A, Ricci M, Giovagnoli S. Meeting the unmet: from traditional to cuttingedge techniques for poly lactide and poly lactide-co-glycolide microparticle manufacturing. J Pharm Investig [Internet]. 2019;49(4):381–404. Available from: https://doi.org/10.1007/s40005-019-00446-y55. Zhong H, Chan G, Hu Y, Hu H, Ouyang D. A comprehensive map of FDA-approved pharmaceutical products. Pharmaceutics. 2018;10(4):1–19.56. Jain A, Kunduru KR, Basu A, Mizrahi B, Domb AJ, Khan W. Injectable formulations of poly(lactic acid) and its copolymers in clinical use. Adv Drug Deliv Rev [Internet]. 2016;107:213–27. Available from: http://dx.doi.org/10.1016/j.addr.2016.07.00257. Igartua M, Hernández RM, Rosas JE, Patarroyo ME, Pedraz JL. γ-Irradiation effects on biopharmaceutical properties of PLGA microspheres loaded with SPf66 synthetic vaccine. Eur J Pharm Biopharm [Internet]. 2008 Jun 1 [cited 2020 Jan 23];69(2):519– 26. Available from: https://www.sciencedirect.com/science/article/pii/S0939641107004201?via%3Dihu b58. Carcaboso A., Hernández R., Igartua M, Rosas J., Patarroyo M., Pedraz J. Potent, long lasting systemic antibody levels and mixed Th1/Th2 immune response after nasal immunization with malaria antigen loaded PLGA microparticles. Vaccine [Internet]. 2004 Mar 29 [cited 2020 Jan 23];22(11–12):1423–32. Available from: https://www.sciencedirect.com/science/article/pii/S0264410X03007679?via%3Dihu b59. Yang YY, Chung TS, Ping Ng N. Morphology, drug distribution, and in vitro release profiles of biodegradable polymeric microspheres containing protein fabricated by double-emulsion solvent extraction/evaporation method. Biomaterials. 2001;22(3):231–41.60. D’Souza SS, Faraj JA, DeLuca PP. A Model-dependent Approach to Correlate Accelerated With Real-Time Release From Biodegradable Microspheres. AAPS PharmSciTech. 2005;6(4):553–64.61. Martinez NY, Andrade PF, Durán N, Cavalitto S. Development of double emulsion nanoparticles for the encapsulation of bovine serum albumin. Colloids Surfaces B Biointerfaces [Internet]. 2017;158:190–6. Available from: http://dx.doi.org/10.1016/j.colsurfb.2017.06.03362. Agrawal A, Rellegadla S, Jain S. Biomedical applications of PLGA particles [Internet]. Materials for Biomedical Engineering: Nanomaterials-based Drug Delivery. Elsevier Inc.; 2019. 87–129 p. Available from: http://dx.doi.org/10.1016/B978-0-12-816913-1.00004-063. Note T. Measuring Zeta Potential – Laser Doppler Electrophoresis. Malvern Guid. 2015;1–2.64. Bhattacharjee S. DLS and zeta potential – What they are and what they are not? J Control Release [Internet]. 2016 Aug 10 [cited 2020 Apr 7];235:337–51. Available from: https://www.sciencedirect.com/science/article/abs/pii/S0168365916303832?via%3 Dihub65. Agrawal A, Rellegadla S, Jain S. Biomedical applications of PLGA particles. Mater Biomed Eng [Internet]. 2019 Jan 1 [cited 2020 Mar 20];87–129. Available from: https://www.sciencedirect.com/science/article/pii/B978012816913100004066. Mundargi RC, Babu VR, Rangaswamy V, Patel P, Aminabhavi TM. Nano/micro technologies for delivering macromolecular therapeutics using poly(d,l-lactide-coglycolide) and its derivatives. J Control Release. 2008;125(3):193–209.67. Saez, Vivian; Hernández, José R; Peniche C. Microspheres as delivery systems for the controlled release of peptides and proteins. Biotecnol Apl. 2007;24(2):108–16.68. Busatto C, Pesoa J, Helbling I, Luna J, Estenoz D. Effect of particle size, polydispersity and polymer degradation on progesterone release from PLGA microparticles: Experimental and mathematical modeling. Int J Pharm [Internet]. 2018;536(1):360–9. Available from: http://dx.doi.org/10.1016/j.ijpharm.2017.12.00669. Giteau A, Venier-julienne MC, Benoit JP. How to achieve sustained and complete protein release from PLGA-based microparticles ? 2008;350:14–26.70. Fredenberg S, Wahlgren M, Reslow M, Axelsson A. The mechanisms of drug release in poly ( lactic-co-glycolic acid ) -based drug delivery systems — A review. Int J Pharm [Internet]. 2011;415(1–2):34–52. Available from: http://dx.doi.org/10.1016/j.ijpharm.2011.05.04971. Gohla S, Ma K, Mu RH. Solid lipid nanoparticles ( SLN ) for controlled drug delivery ± a review of the state of the art. 2000;50.72. Gallarate M, Battaglia L, Peira E, Trotta M. Peptide-loaded solid lipid nanoparticles prepared through coacervation technique. Int J Chem Eng. 2011;2011.73. Battaglia L, Trotta M, Gallarate M, Carlotti ME, Zara GP, Bargoni A. Solid lipid nanoparticles formed by solvent-in-water emulsion-diffusion technique: Development and influence on insulin stability. J Microencapsul. 2007;24(7):672–84.74. Bakliwal A, Bedse S, Talele S, Chaudhari G. LIPOSPHERES: A NOVEL APPROACH FOR DRUG DELIVERY. Int J Res Pharm Nano Sci. 2015;4(5):298– 306.75. Cortesi R, Esposito E, Luca G, Nastruzzi C. Production of lipospheres as carriers for bioactive compounds. Biomaterials. 2002;23(11):2283–94.76. Patil P, Harak K, Saudagar R. Solid Lipid Nanoparticles : A Review. 2019;9(3):525– 30.77. Mishra H, Mishra D, Mishra PK, Nahar M, Dubey V, Jain NK. Evaluation of solid lipid nanoparticles as carriers for delivery of hepatitis b surface antigen for vaccination using subcutaneous route. J Pharm Pharm Sci. 2010;13(4):495–509.78. Reithmeier H, Herrmann J, Göpferich A. Development and characterization of lipid microparticles as a drug carrier for somatostatin. Int J Pharm. 2001;218(1–2):133– 43.79. KH R, SA S, SN D. Solid lipid nanoparticles- A review. Int J Appl Pharm. 2013;5(2):8– 18.80. Patel MR, San Martin-Gonzalez MF. Characterization of ergocalciferol loaded solid lipid nanoparticles. J Food Sci. 2012;77(1):8–13.81. Prombutara P, Kulwatthanasal Y, Supaka N, Sramala I, Chareonpornwattana S. Production of nisin-loaded solid lipid nanoparticles for sustained antimicrobial activity. Food Control [Internet]. 2012;24(1–2):184–90. Available from: http://dx.doi.org/10.1016/j.foodcont.2011.09.02582. Pinho S. Viability of the microencapsulation of a casein hydrolysate in lipid microparticles of cupuacu butter and stearic acid. Int J Food Stud. 2013;2(1):48–59.83. Health N and. Novata® BD PH: Product data sheet [Internet]. [cited 2020 Apr 16]. Available from: https://e-applications.basf-ag.de/data/basf-pcan/pds2/pds2- web.nsf/CA7971BA2D4E244AC12573B100597C04/$File/NOVATA_r__BD_PH_E. pdf84. Reithmeier H, Herrmann J, Gopferich A. Lipid microparticles as a parenteral controlled release device for peptides. 2001;73:339–50.85. Qi C, Chen Y, Huang J, Jin Q. Preparation and characterization of catalase-loaded solid lipid nanoparticles based on soybean phosphatidylcholine. 2012;(February 2011):787–93.86. Almeida AJ, Souto E. Solid lipid nanoparticles as a drug delivery system for peptides and proteins. Adv Drug Deliv Rev. 2007;59(6):478–90.87. Mehnert W, Mäder K. Solid lipid nanoparticles: Production, characterization and applications. Adv Drug Deliv Rev [Internet]. 2012;64(SUPPL.):83–101. Available from: http://dx.doi.org/10.1016/j.addr.2012.09.02188. Gordillo-Galeano A, Ponce A, Mora-Huertas CE. Surface structural characteristics of some colloidal lipid systems used in pharmaceutics. J Drug Deliv Sci Technol [Internet]. 2021;62(January):102345. Available from: https://doi.org/10.1016/j.jddst.2021.10234589. Alexandridis P, Holzwarth JF, Hatton TA. Micellization of Poly(ethylene oxide)- Poly(propylene oxide)-Poly(ethylene oxide) Triblock Copolymers in Aqueous Solutions: Thermodynamics of Copolymer Association. Macromolecules. 1994;27(9):2414–25.90. Mandal B, Bhattacharjee H, Mittal N, Sah H, Balabathula P, Thoma LA, et al. Core – shell-type lipid – polymer hybrid nanoparticles as a drug delivery platform. Nanomedicine Nanotechnology, Biol Med [Internet]. 2013;9(4):474–91. Available from: http://dx.doi.org/10.1016/j.nano.2012.11.01091. Garg NK, Singh B, Kushwah V, Tyagi RK, Sharma R, Jain S, et al. The ligand (s) anchored lipobrid nanoconstruct mediated delivery of methotrexate: An effective approach in breast cancer therapeutics. Nanomedicine Nanotechnology, Biol Med [Internet]. 2016;12(7):2043–60. Available from: http://dx.doi.org/10.1016/j.nano.2016.05.00892. Mukherjee A, Waters AK, Kalyan P, Achrol AS, Kesari S, Yenugonda VM. Lipidpolymer hybrid nanoparticles as a nextgeneration drug delivery platform: State of the art, emerging technologies, and perspectives. Int J Nanomedicine. 2019;14:1937– 52.93. Chaudhary Z, Ahmed N, .ur.Rehman A, Khan GM. Lipid polymer hybrid carrier systems for cancer targeting: A review. Int J Polym Mater Polym Biomater [Internet]. 2018;67(2):86–100. Available from: https://doi.org/10.1080/00914037.2017.130090094. Garg NK, Tandel N, Jadon RS, Tyagi RK, Katare OP. Lipid–polymer hybrid nanocarrier-mediated cancer therapeutics: current status and future directions. Drug Discov Today [Internet]. 2018;23(9):1610–21. Available from: https://doi.org/10.1016/j.drudis.2018.05.03395. Beija M, Salvayre R, Viguerie NL, Marty J. Colloidal systems for drug delivery : from design to therapy. Trends Biotechnol [Internet]. 2012;30(9):485–96. Available from: http://dx.doi.org/10.1016/j.tibtech.2012.04.00896. Wong HL, Rauth AM, Bendayan R, Manias JL, Ramaswamy M, Liu Z, et al. Research Paper A New Polymer Y Lipid Hybrid Nanoparticle System Increases Cytotoxicity of Doxorubicin Against Multidrug-Resistant Human Breast Cancer Cells. 2006;23(7):1574–85.97. Wong HL, Bendayan R, Mike A, Yu X. Simultaneous delivery of doxorubicin and GG918 ( Elacridar ) by new Polymer-Lipid Hybrid Nanoparticles ( PLN ) for enhanced treatment of multidrug-resistant breast cancer. 2006;116:275–84.98. Li Y, Lun H, Shuhendler AJ, Rauth AM, Yu X. Molecular interactions , internal structure and drug release kinetics of rationally developed polymer – lipid hybrid nanoparticles. 2008;128:60–70.99. Mandal B, Mittal NK, Balabathula P, Thoma LA, Wood GC. Development and in vitro evaluation of core-shell type lipid-polymer hybrid nanoparticles for the delivery of erlotinib in non-small cell lung cancer. Eur J Pharm Sci [Internet]. 2016;81:162–71. Available from: http://dx.doi.org/10.1016/j.ejps.2015.10.021100. Devrim B, Kara A, Vural İ, Bozkır A. Lysozyme-loaded lipid-polymer hybrid nanoparticles: preparation, characterization and colloidal stability evaluation. Drug Dev Ind Pharm. 2016;42(11):1865–76.101. Wu C, Baldursdottir S, Yang M, Mu H. Lipid and PLGA hybrid microparticles as carriers for protein delivery. J Drug Deliv Sci Technol [Internet]. 2018;43:65–72. Available from: https://doi.org/10.1016/j.jddst.2017.09.006102. Bose RJC, Lee SH, Park H. Lipid polymer hybrid nanospheres encapsulating antiproliferative agents for stent applications. J Ind Eng Chem [Internet]. 2016;36:284–92. Available from: http://dx.doi.org/10.1016/j.jiec.2016.02.015103. Li H, Teng Y, Sun J, Liu J. Inhibition of hemangioma growth using polymer–lipid hybrid nanoparticles for delivery of rapamycin. Biomed Pharmacother [Internet]. 2017;95:875–84. Available from: https://doi.org/10.1016/j.biopha.2017.08.035104. K.S. J, Snigdha SS, Kalarikkal N, Pothen LA, Thomas S. Gelatin modified lipid nanoparticles for anti- viral drug delivery. Chem Phys Lipids [Internet]. 2017;207:24– 37. Available from: http://dx.doi.org/10.1016/j.chemphyslip.2017.07.002105. Rose F, Wern JE, Gavins F, Andersen P, Follmann F, Foged C. A strong adjuvant based on glycol-chitosan-coated lipid-polymer hybrid nanoparticles potentiates mucosal immune responses against the recombinant Chlamydia trachomatis fusion antigen CTH522. J Control Release [Internet]. 2018;271(December 2017):88–97. Available from: https://doi.org/10.1016/j.jconrel.2017.12.003106. Li J, Ma YJ, Wang Y, Chen BZ, Guo XD, Zhang CY. Dual redox/pH-responsive hybrid polymer-lipid composites: Synthesis, preparation, characterization and application in drug delivery with enhanced therapeutic efficacy. Chem Eng J [Internet]. 2018;341(November 2017):450–61. Available from: https://doi.org/10.1016/j.cej.2018.02.055107. Toragall V, Jayapala N, Vallikannan B. Chitosan-oleic acid-sodium alginate a hybrid nanocarrier as an efficient delivery system for enhancement of lutein stability and bioavailability. Int J Biol Macromol [Internet]. 2020;150:578–94. Available from: https://doi.org/10.1016/j.ijbiomac.2020.02.104108. Patel G, Thakur NS, Kushwah V, Patil MD, Nile SH, Jain S, et al. Mycophenolate coadministration with quercetin via lipid-polymer hybrid nanoparticles for enhanced breast cancer management. Nanomedicine Nanotechnology, Biol Med [Internet]. 2020;24:102147. Available from: https://doi.org/10.1016/j.nano.2019.102147109. Thanki K, van Eetvelde D, Geyer A, Fraire J, Hendrix R, Van Eygen H, et al. Mechanistic profiling of the release kinetics of siRNA from lipidoid-polymer hybrid nanoparticles in vitro and in vivo after pulmonary administration. J Control Release [Internet]. 2019;310(August):82–93. Available from: https://doi.org/10.1016/j.jconrel.2019.08.004110. Leng D, Thanki K, Fattal E, Foged C, Yang M. Engineering of budesonide-loaded lipid-polymer hybrid nanoparticles using a quality-by-design approach. Int J Pharm [Internet]. 2018;548(2):740–6. Available from: https://doi.org/10.1016/j.ijpharm.2017.08.094111. Bachhav SS, Dighe VD, Kotak D, Devarajan P V. Rifampicin Lipid-Polymer hybrid nanoparticles (LIPOMER) for enhanced Peyer’s patch uptake. Int J Pharm [Internet]. 2017;532(1):612–22. Available from: http://dx.doi.org/10.1016/j.ijpharm.2017.09.040112. Tahir N, Madni A, Balasubramanian V, Rehman M, Correia A, Kashif PM, et al. Development and optimization of methotrexate-loaded lipid-polymer hybrid nanoparticles for controlled drug delivery applications. Int J Pharm [Internet]. 2017;533(1):156–68. Available from: http://dx.doi.org/10.1016/j.ijpharm.2017.09.061113. Belletti D, Grabrucker AM, Pederzoli F, Menerath I, Vandelli MA, Tosi G, et al. Hybrid nanoparticles as a new technological approach to enhance the delivery of cholesterol into the brain. Int J Pharm. 2018;543(1–2):300–10.114. Yalcin TE, Ilbasmis-Tamer S, Takka S. Development and characterization of gemcitabine hydrochloride loaded lipid polymer hybrid nanoparticles (LPHNs) using central composite design. Int J Pharm [Internet]. 2018;548(1):255–62. Available from: https://doi.org/10.1016/j.ijpharm.2018.06.063115. Li C, Ge X, Wang L. Construction and comparison of different nanocarriers for codelivery of cisplatin and curcumin: A synergistic combination nanotherapy for cervical cancer. Biomed Pharmacother [Internet]. 2017;86(27):628–36. Available from: http://dx.doi.org/10.1016/j.biopha.2016.12.042116. Zhang Y, Zhang P, Zhu T. Ovarian carcinoma biological nanotherapy: Comparison of the advantages and drawbacks of lipid, polymeric, and hybrid nanoparticles for cisplatin delivery. Biomed Pharmacother [Internet]. 2019;109(1):475–83. Available from: https://doi.org/10.1016/j.biopha.2018.10.158117. Ma P, Li T, Xing H, Wang S, Sun Y, Sheng X, et al. Local anesthetic effects of bupivacaine loaded lipid-polymer hybrid nanoparticles: In vitro and in vivo evaluation. Biomed Pharmacother [Internet]. 2017;89(October 2011):689–95. Available from: http://dx.doi.org/10.1016/j.biopha.2017.01.17518. Qiu J, Cai G, Liu X, Ma D. αvβ3 integrin receptor specific peptide modified, salvianolic acid B and panax notoginsenoside loaded nanomedicine for the combination therapy of acute myocardial ischemia. Biomed Pharmacother [Internet]. 2017;96(89):1418–26. Available from: https://doi.org/10.1016/j.biopha.2017.10.086119. Song Z, Shi Y, Han Q, Dai G. Endothelial growth factor receptor-targeted and reactive oxygen species-responsive lung cancer therapy by docetaxel and resveratrol encapsulated lipid-polymer hybrid nanoparticles. Biomed Pharmacother [Internet]. 2018;105(28):18–26. Available from: https://doi.org/10.1016/j.biopha.2018.05.095120. Yugui F, Wang H, Sun D, Zhang X. Nasopharyngeal cancer combination chemoradiation therapy based on folic acid modified, gefitinib and yttrium 90 coloaded, core-shell structured lipid-polymer hybrid nanoparticles. Biomed Pharmacother. 2019;114(27):0–6.121. Wang J, Su G, Yin X, Luo J, Gu R, Wang S, et al. Non-small cell lung cancer-targeted, redox-sensitive lipid-polymer hybrid nanoparticles for the delivery of a second-generation irreversible epidermal growth factor inhibitor—Afatinib: In vitro and in vivo evaluation. Biomed Pharmacother [Internet]. 2019;120(September):109493. Available from: https://doi.org/10.1016/j.biopha.2019.109493122. Pang J, Xing H, Sun Y, Feng S, Wang S. Non-small cell lung cancer combination therapy: Hyaluronic acid modified, epidermal growth factor receptor targeted, pH sensitive lipid-polymer hybrid nanoparticles for the delivery of erlotinib plus bevacizumab. Biomed Pharmacother [Internet]. 2020;125(December 2019):109861. Available from: https://doi.org/10.1016/j.biopha.2020.109861123. Seedat N, Kalhapure RS, Mocktar C, Vepuri S, Jadhav M, Soliman M, et al. Coencapsulation of multi-lipids and polymers enhances the performance of vancomycin in lipid-polymer hybrid nanoparticles: In vitro and in silico studies. Mater Sci Eng C [Internet]. 2016;61:616–30. Available from: http://dx.doi.org/10.1016/j.msec.2015.12.053124. Xue HY, Tran N, Wong HL. A biodistribution study of solid lipid-polyethyleneimine hybrid nanocarrier for cancer RNAi therapy. Eur J Pharm Biopharm [Internet]. 2016;108:68–75. Available from: http://dx.doi.org/10.1016/j.ejpb.2016.08.014125. Zhao J, Zhang X, Sun X, Zhao M, Yu C, Lee RJ, et al. Dual-functional lipid polymeric hybrid pH-responsive nanoparticles decorated with cell penetrating peptide and folate for therapy against rheumatoid arthritis. Eur J Pharm Biopharm [Internet]. 2018;130(June):39–47. Available from: https://doi.org/10.1016/j.ejpb.2018.06.020126. Jansen MAA, Klausen LH, Thanki K, Lyngsø J, Skov Pedersen J, Franzyk H, et al. Lipidoid-polymer hybrid nanoparticles loaded with TNF siRNA suppress inflammation after intra-articular administration in a murine experimental arthritis model. Eur J Pharm Biopharm [Internet]. 2019;142(November 2018):38–48. Available from: https://doi.org/10.1016/j.ejpb.2019.06.009127. Wu C, Luo X, Baldursdottir SG, Yang M, Sun X, Mu H. In vivo evaluation of solid lipid microparticles and hybrid polymer-lipid microparticles for sustained delivery of leuprolide. Eur J Pharm Biopharm [Internet]. 2019;142(July):315–21. Available from: https://doi.org/10.1016/j.ejpb.2019.07.010128. Zhang J, Hu J, Chan HF, Skibba M, Liang G, Chen M. iRGD decorated lipid-polymer hybrid nanoparticles for targeted co-delivery of doxorubicin and sorafenib to enhance anti-hepatocellular carcinoma efficacy. Nanomedicine Nanotechnology, Biol Med [Internet]. 2016;12(5):1303–11. Available from: http://dx.doi.org/10.1016/j.nano.2016.01.017129. Liu Y, Jiang Z, Hou X, Xie X, Shi J, Shen J, et al. Functional lipid polymeric nanoparticles for oral drug delivery: Rapid mucus penetration and improved cell entry and cellular transport. Nanomedicine Nanotechnology, Biol Med [Internet]. 2019;21:102075. Available from: https://doi.org/10.1016/j.nano.2019.102075130. Boushra M, Tous S, Fetih G, Xue HY, Wong HL. Development of bi-polymer lipid hybrid nanocarrier (BLN) to improve the entrapment and stability of insulin for efficient oral delivery. J Drug Deliv Sci Technol [Internet]. 2019;49(July 2018):632– 41. Available from: https://doi.org/10.1016/j.jddst.2019.01.007131. Meraj Anjum M, Kanoujia J, Parashar P, Arya M, K. Yadav A, A. Saraf S. Evaluation of a Polymer-Lipid-Polymer System Utilising Hybrid Nanoparticles of Dapsone as a Novel Antiacne Agent. Curr Drug ther. 2016;11(2):86–100132. Arya RKK, Juyal V. Polymer -Lipid Hybrid Nanoparticles for Brain Targeting Through Intranasal Delivery. J Drug Deliv Ther. 2017;7(4):129–36.133. Tahir N, Madni A, Kashif PM, Rehman M, Raza A, Khan MI, et al. Formulation and compatibility assessment of PLGA/lecithin based lipid-polymer hybrid nanoparticles containing doxorubicin. Acta Pol Pharm - Drug Res. 2017;74(5):1563–72.134. Tahir N, Madni A, Correia A, Rehman M, Balasubramanian V, Khan MM, et al. Lipidpolymer hybrid nanoparticles for controlled delivery of hydrophilic and lipophilic doxorubicin for breast cancer therapy. Int J Nanomedicine. 2019;14:4961–74.135. Zhao W, Zhang C, Li B, Zhang X, Luo X, Zeng C, et al. Lipid Polymer Hybrid Nanomaterials for mRNA Delivery. Cell Mol Bioeng. 2018;11(5):397–406.136. Gu J, Chen Y, Tong L, Wang X, Yu D, Wu H. Astaxanthin-loaded polymer-lipid hybrid nanoparticles (ATX-LPN): Assessment of potential otoprotective effects. J Nanobiotechnology [Internet]. 2020;18(1):1–17. Available from: https://doi.org/10.1186/s12951-020-00600-x137. Wang Q, Alshaker H, Böhler T, Srivats S, Chao Y, Cooper C, et al. Core shell lipidpolymer hybrid nanoparticles with combined docetaxel and molecular targeted therapy for the treatment of metastatic prostate cancer. Sci Rep. 2017;7(1):1–8.138. Abdou EM, Fayed MAA, Helal D, Ahmed KA. Assessment of the hepatoprotective effect of developed lipid-polymer hybrid nanoparticles (LPHNPs) encapsulating naturally extracted β-Sitosterol against CCl4 induced hepatotoxicity in rats. Sci Rep [Internet]. 2019;9(1):1–14. Available from: http://dx.doi.org/10.1038/s41598-019- 56320-2139. Cao S, Slack SD, Levy CN, Hughes SM, Jiang Y, Yogodzinski C, et al. Hybrid nanocarriers incorporating mechanistically distinct drugs for lymphatic CD4 + T cell activation and HIV-1 latency reversal. Sci Adv. 2019;5(3):1–13.140. Garg NK, Tyagi RK, Sharma G, Jain A, Singh B, Jain S, et al. Functionalized LipidPolymer Hybrid Nanoparticles Mediated Codelivery of Methotrexate and Aceclofenac: A Synergistic Effect in Breast Cancer with Improved Pharmacokinetics Attributes. Mol Pharm. 2017;14(6):1883–97.141. Wang J, Zhang L, Chi H, Wang S. An alternative choice of lidocaine-loaded liposomes: lidocaine-loaded lipid–polymer hybrid nanoparticles for local anesthetic therapy. Drug Deliv. 2016;23(4):1254–60.142. Chen W, Guo M, Wang S. Anti prostate cancer using PEGylated bombesin containing, cabazitaxel loading nano-sized drug delivery system. Drug Dev Ind Pharm. 2016;42(12):1968–76.143. 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):12–21144. Shao Y, Luo W, Guo Q, Li X, Zhang Q, Li J. In vitro and in vivo effect of hyaluronic acid modified, doxorubicin and gallic acid co-delivered lipid-polymeric hybrid nanosystem for leukemia therapy. Drug Des Devel Ther. 2019;13:2043–55.145. Wang J. Combination treatment of cervical cancer using folate-decorated, phsensitive, carboplatin and paclitaxel co-loaded lipid-polymer hybrid nanoparticles. Drug Des Devel Ther. 2020;14:823–32.146. Yu Z, Chen F, Qi X, Dong Y, Zhang Y, Ge Z, et al. Epidermal growth factor receptor aptamer-conjugated polymer-lipid hybrid nanoparticles enhance salinomycin delivery to osteosarcoma and cancer stem cells. Exp Ther Med. 2018;15(2):1247– 56.147. Gao F, Zhang J, Fu C, Xie X, Peng F, You J, et al. iRGD-modified lipid–polymer hybrid nanoparticles loaded with isoliquiritigenin to enhance anti-breast cancer effect and tumor-targeting ability. Int J Nanomedicine. 2017;12:4147–62.148. Khan MM, Madni A, Torchilin V, Filipczak N, Pan J, Tahir N, et al. Lipid-chitosan hybrid nanoparticles for controlled delivery of cisplatin. Drug Deliv [Internet]. 2019;26(1):765–72. Available from: https://doi.org/10.1080/10717544.2019.1642420149. Sun S, Liang N, Yamamoto H, Kawashima Y, Cui F, Yan P. pH-sensitive poly(lactideco-glycolide) nanoparticle Composite Microcapsules for Oral Delivery of Insulin. Int J Nanomedicine. 2015;10:3489–98.150. Tng DJH, Song P, Lin G, Soehartono AM, Yang G, Yang C, et al. Synthesis and characterization of multifunctional hybrid-polymeric nanoparticles for drug delivery and multimodal imaging of cancer. Int J Nanomedicine. 2015;10:5771–86.151. Wu B, Lu ST, Deng K, Yu H, Cui C, Zhang Y, et al. MRI-guided targeting delivery of doxorubicin with reduction-responsive lipid-polymer hybrid nanoparticles. Int J Nanomedicine. 2017;12:6871–82.152. Li J, Xu W, Yuan X, Chen H, Song H, Wang B, et al. Polymer-lipid hybrid anti-HER 2 nanoparticles for targeted salinomycin delivery to HER 2-positive breast cancer stem cells and cancer cells. Int J Nanomedicine. 2017;12:6909–21.153. Wu B, Lu ST, Zhang LJ, Zhuo RX, Xu HB, Huang SW. Codelivery of doxorubicin and triptolide with reduction-sensitive lipid–polymer hybrid nanoparticles for in vitro and in vivo synergistic cancer treatment. Int J Nanomedicine. 2017;12:1853–62.154. Men W, Zhu P, Dong S, Liu W, Zhou K, Bai Y, et al. Fabrication of dual pH/redoxresponsive lipid-polymer hybrid nanoparticles for anticancer drug delivery and controlled release. Int J Nanomedicine. 2019;14:8001–11.155. Hu Y, Hoerle R, Ehrich M, Zhang C. Engineering the lipid layer of lipid–PLGA hybrid nanoparticles for enhanced in vitro cellular uptake and improved stability. Acta Biomater [Internet]. 2015 Dec;28(1):149–59. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1742706115301252156. Hu Y, Zhao Z, Ehrich M, Fuhrman K, Zhang C. In vitro controlled release of antigen in dendritic cells using pH-sensitive liposome-polymeric hybrid nanoparticles. Polymer (Guildf). 2015;80:171–9.157. Dehaini D, Fang RH, Luk BT, Pang Z, Hu CMJ, Kroll A V., et al. Ultra-small lipidpolymer hybrid nanoparticles for tumor-penetrating drug delivery. Nanoscale. 2016;8(30):14411–9.158. Hu Y, Smith D, Frazier E, Hoerle R, Ehrich M, Zhang C. The next-generation nicotine vaccine: a novel and potent hybrid nanoparticle-based nicotine vaccine. Biomaterials [Internet]. 2016 Nov;106(12):228–39. Available from: https://linkinghub.elsevier.com/retrieve/pii/S014296121630415X159. Golan‐Paz S, Frizzell H, Woodrow KA. Cross‐Platform Comparison of Therapeutic Delivery from Multilamellar Lipid‐Coated Polymer Nanoparticles. Macromol Biosci [Internet]. 2019 Apr 27;19(4):1800362. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1002/mabi.201800362160. Wei J, Sun J, Liu Y. Enhanced targeting of prostate cancer-initiating cells by salinomycin-encapsulated lipid-PLGA nanoparticles linked with CD44 antibodies. Oncol Lett. 2019;17(4):4024–33.161. Bhardwaj A, Mehta S, Yadav S, Singh SK, Grobler A, Goyal AK, et al. Pulmonary delivery of antitubercular drugs using spray-dried lipid–polymer hybrid nanoparticles. Artif Cells, Nanomedicine Biotechnol. 2016;44(6):1544–55.162. Liang J, Liu Y, Liu J, Li Z, Fan Q, Jiang Z, et al. Chitosan-functionalized lipid-polymer hybrid nanoparticles for oral delivery of silymarin and enhanced lipid-lowering effect in NAFLD. J Nanobiotechnology [Internet]. 2018;16(1):1–12. Available from: https://doi.org/10.1186/s12951-018-0391-9163. Yasar H, Biehl A, De Rossi C, Koch M, Murgia X, Loretz B, et al. Kinetics of mRNA delivery and protein translation in dendritic cells using lipid-coated PLGA nanoparticles. J Nanobiotechnology [Internet]. 2018;16(1):1–19. Available from: https://doi.org/10.1186/s12951-018-0401-y164. Jin Z, Lv Y, Cao H, Yao J, Zhou J, He W, et al. Core-shell nanocarriers with high paclitaxel loading for passive and active targeting. Nat Publ Gr [Internet]. 2016;(February):1–10. Available from: http://dx.doi.org/10.1038/srep27559165. Ishak RAH, Mostafa NM, Kamel AO. Stealth lipid polymer hybrid nanoparticles loaded with rutin for effective brain delivery – comparative study with the gold standard (Tween 80): Optimization, characterization and biodistribution. Drug Deliv [Internet]. 2017;24(1):1874–90. Available from: https://doi.org/10.1080/10717544.2017.1410263166. Yan J, Wang Y, Zhang X, Liu S, Tian C, Wang H. Targeted nanomedicine for prostate cancer therapy: docetaxel and curcumin co-encapsulated lipid–polymer hybrid nanoparticles for the enhanced anti-tumor activity in vitro and in vivo. Drug Deliv. 2016;23(5):1757–62.167. Tawfik MA, Tadros MI, Mohamed MI. Lipomers (Lipid-polymer Hybrid Particles) of Vardenafil Hydrochloride: a Promising Dual Platform for Modifying the Drug Release Rate and Enhancing Its Oral Bioavailability. AAPS PharmSciTech. 2018;19(8):3650– 60.168. Thakur K, Sharma G, Singh B, Chhibber S, Katare OP. Nano-engineered lipidpolymer hybrid nanoparticles of fusidic acid: an investigative study on dermatokinetics profile and MRSA-infected burn wound model. Drug Deliv Transl Res. 2019;9(4):748–63.EstudiantesInvestigadoresPúblico generalORIGINAL1032480023.2021 - Maria Angelica Velandia Paris.pdf1032480023.2021 - Maria Angelica Velandia Paris.pdfTesis de Maestría en Ciencias Farmacéuticasapplication/pdf5216553https://repositorio.unal.edu.co/bitstream/unal/80864/1/1032480023.2021%20-%20Maria%20Angelica%20Velandia%20Paris.pdf65c8f18d62e1834b2a974e48e6dbc681MD51LICENSElicense.txtlicense.txttext/plain; charset=utf-84074https://repositorio.unal.edu.co/bitstream/unal/80864/2/license.txt8153f7789df02f0a4c9e079953658ab2MD52THUMBNAIL1032480023.2021 - Maria Angelica Velandia Paris.pdf.jpg1032480023.2021 - Maria Angelica Velandia Paris.pdf.jpgGenerated Thumbnailimage/jpeg5381https://repositorio.unal.edu.co/bitstream/unal/80864/3/1032480023.2021%20-%20Maria%20Angelica%20Velandia%20Paris.pdf.jpg71f12ff5f9dea73f691536f0672b1a83MD53unal/80864oai:repositorio.unal.edu.co:unal/808642023-07-31 23:04:37.931Repositorio Institucional Universidad Nacional de Colombiarepositorio_nal@unal.edu.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