Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Targeting Cancer using Curcumin Encapsulated Vesicular Drug Delivery Systems

Author(s): Joel Hardwick, Jack Taylor, Meenu Mehta, Saurabh Satija, Keshav R. Paudel, Philip M. Hansbro, Dinesh K. Chellappan, Mary Bebawy* and Kamal Dua*

Volume 27, Issue 1, 2021

Published on: 28 July, 2020

Page: [2 - 14] Pages: 13

DOI: 10.2174/1381612826666200728151610

Price: $65

Abstract

Curcumin is a major curcuminoid present in turmeric. The compound is attributed to various therapeutic properties, which include anti-oxidant, anti-inflammatory, anti-bacterial, anti-malarial, and neuroprotection. Due to its therapeutic potential, curcumin has been employed for centuries in treating different ailments. Curcumin has been investigated lately as a novel therapeutic agent in the treatment of cancer. However, the mechanisms by which curcumin exerts its cytotoxic effects on malignant cells are still not fully understood. One of the main limiting factors in the clinical use of curcumin is its poor bioavailability and rapid elimination. Advancements in drug delivery systems such as nanoparticle-based vesicular drug delivery platforms have improved several parameters, namely, drug bioavailability, solubility, stability, and controlled release properties. The use of curcumin-encapsulated niosomes to improve the physical and pharmacokinetic properties of curcumin is one such approach. This review provides an up-to-date summary of nanoparticle-based vesicular drug carriers and their therapeutic applications. Specifically, we focus on niosomes as novel drug delivery formulations and their potential in improving the delivery of challenging small molecules, including curcumin. Overall, the applications of such carriers will provide a new direction for novel pharmaceutical drug delivery, as well as for biotechnology, nutraceutical, and functional food industries.

Keywords: Cancer, curcumin, exosomes, liposomes, nanoformulations, niosomes, pro-niosomes, vesicular.

[1]
Prasher P, Sharma M, Mehta M, et al. Plants derived therapeutic strategies targeting chronic respiratory diseases: Chemical and immunological perspective. Chem Biol Interact 2020; 325109125
[http://dx.doi.org/10.1016/j.cbi.2020.109125] [PMID: 32376238]
[2]
Chin LH, Hon CM, Chellappan DK, et al. Molecular mechanisms of action of naringenin in chronic airway diseases. Eur J Pharmacol 2020; 879173139
[http://dx.doi.org/10.1016/j.ejphar.2020.173139] [PMID: 32343971]
[3]
Panth N, Manandhar B, Paudel KR. Anticancer activity of Punica granatum (Pomegranate): A Review. Phytother Res 2017; 31(4): 568-78.
[http://dx.doi.org/10.1002/ptr.5784] [PMID: 28185340]
[4]
Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the Indian solid gold The molecular targets and therapeutic uses of curcumin in health and disease Springer 2007; 1-515.
[http://dx.doi.org/10.1007/978-0-387-46401-5_1]
[5]
Kaur R, Satija S, Kalsi V, Mehta M, Gupta P. Comparative study of analgesic and antipyretic activity of Curcuma caesia and Curcuma amada roxb. Rhizomes. Ethnopharmacology 2013; 2: 441.
[6]
Ng ZY, Wong JY, Panneerselvam J, et al. Assessing the potential of liposomes loaded with curcumin as a therapeutic intervention in asthma. Colloids Surf B Biointerfaces 2018; 172: 51-9.
[http://dx.doi.org/10.1016/j.colsurfb.2018.08.027] [PMID: 30134219]
[7]
Martí Coma-Cros E, Biosca A, Lantero E, et al. Antimalarial activity of orally administered curcumin incorporated in eudragit®-containing liposomes. Int J Mol Sci 2018; 19(5): 19.
[http://dx.doi.org/10.3390/ijms19051361] [PMID: 29734652]
[8]
Bannuru RR, Osani MC, Al-Eid F, Wang C. Efficacy of curcumin and Boswellia for knee osteoarthritis: Systematic review and meta-analysis. Semin Arthritis Rheum 2018; 48(3): 416-29.
[http://dx.doi.org/10.1016/j.semarthrit.2018.03.001] [PMID: 29622343]
[9]
Peddada KV, Brown A, Verma V, Nebbioso M. Therapeutic potential of curcumin in major retinal pathologies. Int Ophthalmol 2018; 39(3): 725-34.
[PMID: 29404861]
[10]
Heger M, van Golen RF, Broekgaarden M, Michel MC. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol Rev 2013; 66(1): 222-307.
[http://dx.doi.org/10.1124/pr.110.004044] [PMID: 24368738]
[11]
Jiang K, Shen M, Xu W. Arginine, glycine, aspartic acid peptide-modified paclitaxel and curcumin co-loaded liposome for the treatment of lung cancer: in vitro/vivo evaluation. Int J Nanomedicine 2018; 13: 2561-9.
[http://dx.doi.org/10.2147/IJN.S157746] [PMID: 29731631]
[12]
Zhao M, Zhao M, Fu C, Yu Y, Fu A. Targeted therapy of intracranial glioma model mice with curcumin nanoliposomes. Int J Nanomedicine 2018; 13: 1601-10.
[http://dx.doi.org/10.2147/IJN.S157019] [PMID: 29588587]
[13]
Sesarman A, Tefas L, Sylvester B, et al. Anti-angiogenic and anti-inflammatory effects of long-circulating liposomes co-encapsulating curcumin and doxorubicin on C26 murine colon cancer cells. Pharmacol Rep 2017; 70(2): 331-9.
[PMID: 29477042]
[14]
Garg M, Lata K, Satija S. Cytotoxic potential of few Indian fruit peels through 3-(4,5-dimethylthiazol-yl)-2,5-diphenyltetrazolium bromide assay on HepG2 cells. Indian J Pharmacol 2016; 48(1): 64-8.
[http://dx.doi.org/10.4103/0253-7613.174552] [PMID: 26997725]
[15]
Shehzad A, Lee J, Lee YS. Curcumin in various cancers. Biofactors 2013; 39(1): 56-68.
[http://dx.doi.org/10.1002/biof.1068] [PMID: 23303705]
[16]
Shehzad A, Ha T, Subhan F, Lee YS. New mechanisms and the anti-inflammatory role of curcumin in obesity and obesity-related metabolic diseases. Eur J Nutr 2011; 50(3): 151-61.
[http://dx.doi.org/10.1007/s00394-011-0188-1] [PMID: 21442412]
[17]
Duan Y, Wang J, Yang X, Du H, Xi Y, Zhai G. Curcumin-loaded mixed micelles: preparation, optimization, physicochemical properties and cytotoxicity in vitro. Drug Deliv 2015; 22(1): 50-7.
[http://dx.doi.org/10.3109/10717544.2013.873501] [PMID: 24417664]
[18]
Yen F-L, Wu T-H, Tzeng C-W, Lin L-T, Lin C-C. Curcumin nanoparticles improve the physicochemical properties of curcumin and effectively enhance its antioxidant and antihepatoma activities. J Agric Food Chem 2010; 58(12): 7376-82.
[http://dx.doi.org/10.1021/jf100135h] [PMID: 20486686]
[19]
Chellappan DK, Ng ZY, Wong JY, et al. Immunological axis of curcumin-loaded vesicular drug delivery systems. Future Med Chem 2018; 10(8): 839-44.
[http://dx.doi.org/10.4155/fmc-2017-0245] [PMID: 29620416]
[20]
Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery systems. Pharmacol Rep 2012; 64(5): 1020-37.
[http://dx.doi.org/10.1016/S1734-1140(12)70901-5] [PMID: 23238461]
[21]
Dua K, Rapalli VK, Shukla SD, et al. Multi-drug resistant Mycobacterium tuberculosis & oxidative stress complexity: Emerging need for novel drug delivery approaches. Biomed Pharmacother 2018; 107: 1218-29.
[http://dx.doi.org/10.1016/j.biopha.2018.08.101] [PMID: 30257336]
[22]
Xu Y-Q, Chen W-R, Tsosie JK, et al. Niosome Encapsulation of Curcumin. J Nanomater 2016; 2016: 15.
[http://dx.doi.org/10.1155/2016/6365295]
[23]
Khalil NM, do Nascimento TCF, Casa DM, et al. Pharmacokinetics of curcumin-loaded PLGA and PLGA-PEG blend nanoparticles after oral administration in rats. Colloids Surf B Biointerfaces 2013; 101: 353-60.
[http://dx.doi.org/10.1016/j.colsurfb.2012.06.024] [PMID: 23010041]
[24]
Mulik RS, Mönkkönen J, Juvonen RO, Mahadik KR, Paradkar AR. Transferrin mediated solid lipid nanoparticles containing curcumin: enhanced in vitro anticancer activity by induction of apoptosis. Int J Pharm 2010; 398(1-2): 190-203.
[http://dx.doi.org/10.1016/j.ijpharm.2010.07.021] [PMID: 20655375]
[25]
Mehta M, Satija S, Nanda A, Garg M. Nanotechnologies for boswellic acids. Am J Drug Discov Dev 2014; 4: 1-11.
[http://dx.doi.org/10.3923/ajdd.2014.1.11]
[26]
Mehta M, Deeksha Sharma N, et al. Interactions with the macrophages: An emerging targeted approach using novel drug delivery systems in respiratory diseases. Chem Biol Interact 2019; 304: 10-9.
[http://dx.doi.org/10.1016/j.cbi.2019.02.021] [PMID: 30849336]
[27]
Chellappan DK, Yee LW, Xuan KY, et al. Targeting neutrophils using novel drug delivery systems in chronic respiratory diseases. Drug Dev Res 2020; 81(4): 419-36.
[http://dx.doi.org/10.1002/ddr.21648] [PMID: 32048757]
[28]
Yang H. Targeted nanosystems: Advances in targeted dendrimers for cancer therapy. Nanomedicine (Lond) 2016; 12(2): 309-16.
[http://dx.doi.org/10.1016/j.nano.2015.11.012] [PMID: 26706410]
[29]
Xia T, He Q, Shi K, et al. Losartan loaded liposomes improve the antitumor efficacy of liposomal paclitaxel modified with pH sensitive peptides by inhibition of collagen in breast cancer. Pharm Dev Technol 2018; 23(1): 13-21.
[http://dx.doi.org/10.1080/10837450.2016.1265553] [PMID: 27884084]
[30]
Fouda NH, Abdelrehim RT, Hegazy DA, Habib BA. Sustained ocular delivery of Dorzolamide-HCl via proniosomal gel formulation: in-vitro characterization, statistical optimization, and in-vivo pharmacodynamic evaluation in rabbits. Drug Deliv 2018; 25(1): 1340-9.
[http://dx.doi.org/10.1080/10717544.2018.1477861] [PMID: 29869516]
[31]
Mehta M, Garg M. Proniosomal gel: A promising drug carrier for boswellic acids. J Med Sci 2015; 15: 130-4.
[http://dx.doi.org/10.3923/jms.2015.130.134]
[32]
Mehta M, Dureja H, Garg M. Development and optimization of boswellic acid-loaded proniosomal gel. Drug Deliv 2016; 23(8): 3072-81.
[http://dx.doi.org/10.3109/10717544.2016.1149744] [PMID: 26953869]
[33]
Mathure D, Madan JR, Gujar KN, Tupsamundre A, Ranpise HA, Dua K. Formulation and evaluation of niosomal in situ nasal gel of a serotonin receptor agonist, buspirone hydrochloride for the brain delivery via intranasal route. Pharm Nanotechnol 2018; 6(1): 69-78.
[http://dx.doi.org/10.2174/2211738506666180130105919] [PMID: 29380709]
[34]
Baskaran R, Madheswaran T, Sundaramoorthy P, Kim HM, Yoo BK. Entrapment of curcumin into monoolein-based liquid crystalline nanoparticle dispersion for enhancement of stability and anticancer activity. Int J Nanomedicine 2014; 9: 3119-30.
[PMID: 25061290]
[35]
Chan Y, Ng SW, Chellappan DK, et al. Celastrol-loaded liquid crystalline nanoparticles as an anti-inflammatory intervention for the treatment of asthma International J Polymeric Materi Polymeric Biomater 2020; 1-10.
[http://dx.doi.org/10.1080/00914037.2020.1765350]
[36]
Cho E, Nam G-H, Hong Y, et al. Comparison of exosomes and ferritin protein nanocages for the delivery of membrane protein therapeutics. J Control Release 2018; 279: 326-35.
[http://dx.doi.org/10.1016/j.jconrel.2018.04.037] [PMID: 29679665]
[37]
Kamerkar S, LeBleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017; 546(7659): 498-503.
[http://dx.doi.org/10.1038/nature22341] [PMID: 28607485]
[38]
Rajeshkumar S, Menon S, Venkat Kumar S, et al. Antibacterial and antioxidant potential of biosynthesized copper nanoparticles mediated through Cissus arnotiana plant extract. J Photochem Photobiol B 2019; 197111531
[http://dx.doi.org/10.1016/j.jphotobiol.2019.111531] [PMID: 31212244]
[39]
Nadzir MM. FEN T, Mohamed AR, Hisham SF. Size and stability of curcumin niosomes from combinations of tween 80 and span 80. Sains Malays 2017; 46: 2455-60.
[http://dx.doi.org/10.17576/jsm-2017-4612-22]
[40]
Amiri B, Ahmadvand H, Farhadi A, Najmafshar A, Chiani M, Norouzian D. Delivery of vinblastine-containing niosomes results in potent in vitro/in vivo cytotoxicity on tumor cells. Drug Dev Ind Pharm 2018; 44(8): 1371-6.
[http://dx.doi.org/10.1080/03639045.2018.1451880] [PMID: 29532687]
[41]
Mehta M, Deeksha Tewari D, et al. Oligonucleotide therapy: An emerging focus area for drug delivery in chronic inflammatory respiratory diseases. Chem Biol Interact 2019; 308: 206-15.
[http://dx.doi.org/10.1016/j.cbi.2019.05.028] [PMID: 31136735]
[42]
Jyoti K, Pandey RS, Madan J, Jain UK. Inhalable cationic niosomes of curcumin enhanced drug delivery and apoptosis in lung cancer cells. Indian J Pharma Education Res 2016; 50: S21-31.
[43]
Salem HF, Kharshoum RM, Abo El-Ela FI. F AG, Abdellatif KRA. Evaluation and optimization of pH-responsive niosomes as a carrier for efficient treatment of breast cancer. Drug Deliv Transl Res 2018; 8(3): 633-44.
[http://dx.doi.org/10.1007/s13346-018-0499-3] [PMID: 29488171]
[44]
Sharma V, Anandhakumar S, Sasidharan M. Self-degrading niosomes for encapsulation of hydrophilic and hydrophobic drugs: An efficient carrier for cancer multi-drug delivery. Mater Sci Eng C 2015; 56: 393-400.
[http://dx.doi.org/10.1016/j.msec.2015.06.049] [PMID: 26249606]
[45]
Das MK, Kumar R. Development of Curcumin nanoniosomes for skin cancer chemoprevention. Int J Chemtech Res 2015; 7: 747-54.
[46]
Bisht S, Feldmann G, Soni S, et al. Polymeric nanoparticle-encapsulated curcumin (“nanocurcumin”): a novel strategy for human cancer therapy. J Nanobiotechnology 2007; 5: 3.
[http://dx.doi.org/10.1186/1477-3155-5-3] [PMID: 17439648]
[47]
Chellappan DK, Hansbro PM, Dua K, et al. Vesicular Systems containing Curcumin and their applications in respiratory disorders-A Mini Review. Pharm Nanotechnol 2017; 5(4): 250-4.
[PMID: 28786351]
[48]
Chellappan DK, Ng ZY, Wong J-Y, et al. Immunological axis of curcumin-loaded vesicular drug delivery systems. Future Science 2018; 10(8)
[http://dx.doi.org/10.4155/fmc-2017-0245]
[49]
Mandal S, Banerjee C, Ghosh S, Kuchlyan J, Sarkar N. Modulation of the photophysical properties of curcumin in nonionic surfactant (Tween-20) forming micelles and niosomes: a comparative study of different microenvironments. J Phys Chem B 2013; 117(23): 6957-68.
[http://dx.doi.org/10.1021/jp403724g] [PMID: 23682632]
[50]
Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. The essential medicinal chemistry of curcumin: miniperspective. J Med Chem 2017; 60(5): 1620-37.
[http://dx.doi.org/10.1021/acs.jmedchem.6b00975] [PMID: 28074653]
[51]
Priyadarsini KI. Chemical and structural features influencing the biological activity of curcumin. Curr Pharm Des 2013; 19(11): 2093-100.
[PMID: 23116315]
[52]
Kou J, Xin TY, McCarron P, et al. Going beyond antibiotics: Natural plant extracts as an emergent strategy to combat biofilm-associated infections. J Environ Pathol Toxicol Oncol 2020; 39(2): 125-36.
[http://dx.doi.org/10.1615/JEnvironPatholToxicolOncol.2020032665]
[53]
Ghalandarlaki N, Alizadeh AM, Ashkani-Esfahani S. Nanotechnology-applied curcumin for different diseases therapy. BioMed research international 2014; 2014.
[http://dx.doi.org/10.1155/2014/394264]
[54]
Kumar P, Mehta M, Satija S, Garg M. Enzymatic in vitro anti-diabetic activity of few traditional Indian medicinal plants. J Biol Sci 2013; 13: 540-4.
[http://dx.doi.org/10.3923/jbs.2013.540.544]
[55]
Lee W-H, Loo C-Y, Young PM, Traini D, Mason RS, Rohanizadeh R. Recent advances in curcumin nanoformulation for cancer therapy. Expert Opin Drug Deliv 2014; 11(8): 1183-201.
[http://dx.doi.org/10.1517/17425247.2014.916686] [PMID: 24857605]
[56]
Hinge N, Pandey MM, Singhvi G, et al. Nanomedicine advances in cancer therapy Advanced 3D-printed systems and nanosystems for drug delivery and tissue engineering. Elsevier 2020.
[http://dx.doi.org/10.1016/B978-0-12-818471-4.00008-X]
[57]
Samuel M. Targeting the seven cancer hallmarks by modulation of oxidative stress-induced inflammation and immune activation: a radical therapeutic approach. J Lipid Res 2012; 53(9): 1767-82.
[58]
Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009; 30(7): 1073-81.
[http://dx.doi.org/10.1093/carcin/bgp127] [PMID: 19468060]
[59]
da Costa PL, Sirois P, Tannock IF, Chammas R. The role of kinin receptors in cancer and therapeutic opportunities. Cancer Lett 2014; 345(1): 27-38.
[http://dx.doi.org/10.1016/j.canlet.2013.12.009] [PMID: 24333733]
[60]
Mehta M, Dhanjal DS, Paudel KR, et al. Cellular signalling pathways mediating the pathogenesis of chronic inflammatory respiratory diseases: an update. Inflammopharmacology 2020; 1-23.
[http://dx.doi.org/10.1007/s10787-020-00698-3] [PMID: 32189104]
[61]
Housman G, Byler S, Heerboth S, et al. Drug resistance in cancer: an overview. Cancers (Basel) 2014; 6(3): 1769-92.
[http://dx.doi.org/10.3390/cancers6031769] [PMID: 25198391]
[62]
Aljabali AAA, Bakshi HA, Hakkim FL, et al. Albumin nano-encapsulation of piceatannol enhances its anticancer potential in colon cancer via downregulation of nuclear p65 and HIF-1α. Cancers (Basel) 2020; 12(1): 12.
[http://dx.doi.org/10.3390/cancers12010113] [PMID: 31906321]
[63]
Malyla V, Paudel KR, Shukla SD, et al. Recent advances in experimental animal models of lung cancer. Future Med Chem 2020; 12(7)
[http://dx.doi.org/10.4155/fmc-2019-0338]
[64]
Doello K, Ortiz R, Alvarez PJ, Melguizo C, Cabeza L, Prados J. Latest in vitro and in vivo assay, clinical trials and patents in cancer treatment using curcumin: a literature review. Nutr Cancer 2018; 70(4): 569-78.
[http://dx.doi.org/10.1080/01635581.2018.1464347] [PMID: 29708445]
[65]
Haroyan A, Mukuchyan V, Mkrtchyan N, et al. Efficacy and safety of curcumin and its combination with boswellic acid in osteoarthritis: a comparative, randomized, double-blind, placebo-controlled study. BMC Complement Altern Med 2018; 18(1): 7.
[http://dx.doi.org/10.1186/s12906-017-2062-z] [PMID: 29316908]
[66]
Hariri M, Haghighatdoost F. Effect of curcumin on anthropometric measures: a systematic review on randomized clinical trials. J Am Coll Nutr 2018; 37(3): 215-22.
[http://dx.doi.org/10.1080/07315724.2017.1392263] [PMID: 29313748]
[67]
Barber-Chamoux N, Milenkovic D, Verny MA, et al. Substantial variability across individuals in the vascular and nutrigenomic response to an acute intake of curcumin: a randomized controlled trial. Mol Nutr Food Res 2018; 62(5)1700418
[http://dx.doi.org/10.1002/mnfr.201700418] [PMID: 29034576]
[68]
Oliveira AS, Sousa E, Vasconcelos MH, Pinto M. Curcumin: a natural lead for potential new drug candidates. Curr Med Chem 2015; 22(36): 4196-232.
[http://dx.doi.org/10.2174/0929867322666151029104611] [PMID: 26511469]
[69]
Gdowski A, Johnson K, Shah S, Gryczynski I, Vishwanatha J, Ranjan A. Optimization and scale up of microfluidic nanolipomer production method for preclinical and potential clinical trials. J Nanobiotechnology 2018; 16(1): 12.
[http://dx.doi.org/10.1186/s12951-018-0339-0] [PMID: 29433518]
[70]
Alizadeh F, Javadi M, Karami AA, Gholaminejad F, Kavianpour M, Haghighian HK. Curcumin nanomicelle improves semen parameters, oxidative stress, inflammatory biomarkers, and reproductive hormones in infertile men: A randomized clinical trial. Phytother Res 2018; 32(3): 514-21.
[http://dx.doi.org/10.1002/ptr.5998] [PMID: 29193350]
[71]
Storka A, Vcelar B, Klickovic U, et al. Safety, tolerability and pharmacokinetics of liposomal curcumin in healthy humans. Int J Clin Pharmacol Ther 2015; 53(1): 54-65.
[http://dx.doi.org/10.5414/CP202076] [PMID: 25500488]
[72]
Naksuriya O, Okonogi S, Schiffelers RM, Hennink WE. Curcumin nanoformulations: a review of pharmaceutical properties and preclinical studies and clinical data related to cancer treatment. Biomaterials 2014; 35(10): 3365-83.
[http://dx.doi.org/10.1016/j.biomaterials.2013.12.090] [PMID: 24439402]
[73]
Martin TA, Ye L, Sanders AJ, Lane J, Jiang WG. Cancer invasion and metastasis: molecular and cellular perspective. Madame Curie Bioscience Database 2013.
[74]
Deng YI, Verron E, Rohanizadeh R. Molecular mechanisms of anti-metastatic activity of curcumin. Anticancer Res 2016; 36(11): 5639-47.
[http://dx.doi.org/10.21873/anticanres.11147] [PMID: 27793885]
[75]
Anand P, Sundaram C, Jhurani S, Kunnumakkara AB, Aggarwal BB. Curcumin and cancer: an “old-age” disease with an “age-old” solution. Cancer Lett 2008; 267(1): 133-64.
[http://dx.doi.org/10.1016/j.canlet.2008.03.025] [PMID: 18462866]
[76]
Chen H-W, Lee J-Y, Huang J-Y, et al. Curcumin inhibits lung cancer cell invasion and metastasis through the tumor suppressor HLJ1. Cancer Res 2008; 68(18): 7428-38.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-6734] [PMID: 18794131]
[77]
Sun M, Estrov Z, Ji Y, Coombes KR, Harris DH, Kurzrock R. Curcumin (diferuloylmethane) alters the expression profiles of microRNAs in human pancreatic cancer cells. Mol Cancer Ther 2008; 7(3): 464-73.
[http://dx.doi.org/10.1158/1535-7163.MCT-07-2272] [PMID: 18347134]
[78]
Zong H, Wang F, Fan QX, Wang LX. Curcumin inhibits metastatic progression of breast cancer cell through suppression of urokinase-type plasminogen activator by NF-kappa B signaling pathways. Mol Biol Rep 2012; 39(4): 4803-8.
[http://dx.doi.org/10.1007/s11033-011-1273-5] [PMID: 21947854]
[79]
Lin S-S, Lai K-C, Hsu S-C, et al. Curcumin inhibits the migration and invasion of human A549 lung cancer cells through the inhibition of matrix metalloproteinase-2 and -9 and Vascular Endothelial Growth Factor (VEGF). Cancer Lett 2009; 285(2): 127-33.
[http://dx.doi.org/10.1016/j.canlet.2009.04.037] [PMID: 19477063]
[80]
Kim J-M, Noh E-M, Kwon K-B, et al. Curcumin suppresses the TPA-induced invasion through inhibition of PKCα-dependent MMP-expression in MCF-7 human breast cancer cells. Phytomedicine 2012; 19(12): 1085-92.
[http://dx.doi.org/10.1016/j.phymed.2012.07.002] [PMID: 22921746]
[81]
Wang S, Yu S, Shi W, et al. Curcumin inhibits the migration and invasion of mouse hepatoma Hca-F cells through down-regulating caveolin-1 expression and epidermal growth factor receptor signaling. IUBMB Life 2011; 63(9): 775-82.
[http://dx.doi.org/10.1002/iub.507] [PMID: 22362715]
[82]
Yang C-L, Liu Y-Y, Ma Y-G, et al. Curcumin blocks small cell lung cancer cells migration, invasion, angiogenesis, cell cycle and neoplasia through Janus kinase-STAT3 signalling pathway. PLoS One 2012; 7(5)e37960
[http://dx.doi.org/10.1371/journal.pone.0037960] [PMID: 22662257]
[83]
Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies. Anticancer Res 2003; 23(1A): 363-98.
[PMID: 12680238]
[84]
Mehta M, Satija S, Paudel KR, et al. Incipient need of targeting airway remodeling using advanced drug delivery in chronic respiratory diseases. Future Science 2020; 12(10)
[http://dx.doi.org/10.4155/fmc-2020-0091]
[85]
Mehta M, Chellappan DK, Wich PR, Hansbro NG, Hansbro PM, Dua K. miRNA nanotherapeutics: potential and challenges in respiratory disorders. In: Future Science. 2020; 12.(11)
[86]
Singh H, Satija S, Kaur H, et al. novel drug delivery approaches for guggul. Plant Arch 2019; 19: 983-93.
[87]
Dua K, Wadhwa R, Singhvi G, et al. The potential of siRNA based drug delivery in respiratory disorders: Recent advances and progress. Drug Dev Res 2019; 80(6): 714-30.
[http://dx.doi.org/10.1002/ddr.21571] [PMID: 31691339]
[88]
Kumar GP, Rajeshwarrao P. Nonionic surfactant vesicular systems for effective drug delivery-an overview. Acta Pharm Sin B 2011; 1: 208-19.
[http://dx.doi.org/10.1016/j.apsb.2011.09.002]
[89]
Wadhwa R, Pandey P, Gupta G, et al. Emerging complexity and the need for advanced drug delivery in targeting candida species. Curr Top Med Chem 2019; 19(28): 2593-609.
[http://dx.doi.org/10.2174/1568026619666191026105308] [PMID: 31746290]
[90]
Bulbake U, Doppalapudi S, Kommineni N, Khan W. Liposomal formulations in clinical use: an updated review. Pharmaceutics 2017; 9(2): 12.
[http://dx.doi.org/10.3390/pharmaceutics9020012] [PMID: 28346375]
[91]
Ong HX, Benaouda F, Traini D, et al. In vitro and ex vivo methods predict the enhanced lung residence time of liposomal ciprofloxacin formulations for nebulisation. Eur J Pharm Biopharm 2014; 86(1): 83-9.
[http://dx.doi.org/10.1016/j.ejpb.2013.06.024] [PMID: 23851077]
[92]
Ong HX, Traini D, Cipolla D, et al. Liposomal nanoparticles control the uptake of ciprofloxacin across respiratory epithelia. Pharm Res 2012; 29(12): 3335-46.
[http://dx.doi.org/10.1007/s11095-012-0827-0] [PMID: 22833052]
[93]
Hasan M, Elkhoury K, Kahn CJF, Arab-Tehrany E, Linder M. Preparation, characterization, and release kinetics of chitosan-coated nanoliposomes encapsulating curcumin in simulated environments. Molecules 2019; 24(10): 24.
[http://dx.doi.org/10.3390/molecules24102023] [PMID: 31137865]
[94]
Karimi M, Gheybi F, Zamani P, et al. Preparation and characterization of stable nanoliposomal formulations of curcumin with high loading efficacy: In vitro and in vivo anti-tumor study. Int J Pharm 2020; 580119211
[http://dx.doi.org/10.1016/j.ijpharm.2020.119211] [PMID: 32156530]
[95]
Li Z, Xiong X, Peng S, Chen X, Liu W, Liu C. Novel folated pluronic F127 modified liposomes for delivery of curcumin: preparation, release, and cytotoxicity. J Microencapsul 2020; 37(3): 220-9.
[http://dx.doi.org/10.1080/02652048.2020.1720030] [PMID: 32039640]
[96]
Jadia R, Kydd J, Piel B, Rai P. Liposomes aid curcumin’s combat with cancer in a breast tumor model. Oncomedicine 2018; 3: 94-109.
[http://dx.doi.org/10.7150/oncm.27938]
[97]
Wang WY, Cao YX, Zhou X, Wei B. Delivery of folic acid-modified liposomal curcumin for targeted cervical carcinoma therapy. Drug Des Devel Ther 2019; 13: 2205-13.
[http://dx.doi.org/10.2147/DDDT.S205787] [PMID: 31308632]
[98]
Cheng Y, Zhao P, Wu S, et al. Cisplatin and curcumin co-loaded nano-liposomes for the treatment of hepatocellular carcinoma. Int J Pharm 2018; 545(1-2): 261-73.
[http://dx.doi.org/10.1016/j.ijpharm.2018.05.007] [PMID: 29730175]
[99]
De Leo V, Milano F, Mancini E, et al. Encapsulation of curcumin-loaded liposomes for colonic drug delivery in a ph-responsive polymer cluster using a ph-driven and organic solvent-free process. Molecules 2018; 23(4): 739.
[http://dx.doi.org/10.3390/molecules23040739] [PMID: 29570636]
[100]
Sesarman A, Muntean D, Abrudan B, et al. Improved pharmacokinetics and reduced side effects of doxorubicin therapy by liposomal co-encapsulation with curcumin. J Liposome Res 2019; 1-10.
[http://dx.doi.org/10.1080/08982104.2019.1682604] [PMID: 31631726]
[101]
Jose A, Labala S, Venuganti VV. Co-delivery of curcumin and STAT3 siRNA using deformable cationic liposomes to treat skin cancer. J Drug Target 2017; 25(4): 330-41.
[http://dx.doi.org/10.1080/1061186X.2016.1258567] [PMID: 27819148]
[102]
Alshamsan A, Hamdy S, Samuel J, El-Kadi AO, Lavasanifar A, Uludağ H. The induction of tumor apoptosis in B16 melanoma following STAT3 siRNA delivery with a lipid-substituted polyethylenimine. Biomaterials 2010; 31(6): 1420-8.
[http://dx.doi.org/10.1016/j.biomaterials.2009.11.003] [PMID: 19913908]
[103]
Yeh C-C, Su Y-H, Lin Y-J, et al. Evaluation of the protective effects of curcuminoid (curcumin and bisdemethoxycurcumin)-loaded liposomes against bone turnover in a cell-based model of osteoarthritis. Drug Des Devel Ther 2015; 9: 2285-300.
[PMID: 25945040]
[104]
Ibrahim S, Tagami T, Kishi T, Ozeki T. Curcumin marinosomes as promising nano-drug delivery system for lung cancer. Int J Pharm 2018; 540(1-2): 40-9.
[http://dx.doi.org/10.1016/j.ijpharm.2018.01.051] [PMID: 29408473]
[105]
Zhang T, Chen Y, Ge Y, Hu Y, Li M, Jin Y. Inhalation treatment of primary lung cancer using liposomal curcumin dry powder inhalers. Acta Pharm Sin B 2018; 8(3): 440-8.
[http://dx.doi.org/10.1016/j.apsb.2018.03.004] [PMID: 29881683]
[106]
Abdel-Hafez SM, Hathout RM, Sammour OA. Curcumin-loaded ultradeformable nanovesicles as a potential delivery system for breast cancer therapy. Colloids Surf B Biointerfaces 2018; 167: 63-72.
[http://dx.doi.org/10.1016/j.colsurfb.2018.03.051] [PMID: 29626721]
[107]
Anirudhan TS, Nair AS, Bino SJ. Nanoparticle assisted solvent selective transdermal combination therapy of curcumin and 5-flurouracil for efficient cancer treatment. Carbohydr Polym 2017; 173: 131-42.
[http://dx.doi.org/10.1016/j.carbpol.2017.05.045] [PMID: 28732851]
[108]
Ma Q, Qian W, Tao W, Zhou Y, Xue B. Delivery of curcumin nanoliposomes using surface modified with CD133 aptamers for prostate cancer. Drug Des Devel Ther 2019; 13: 4021-33.
[http://dx.doi.org/10.2147/DDDT.S210949] [PMID: 31819373]
[109]
Uchegbu IF, Vyas SP. Non-ionic surfactant based vesicles (niosomes) in drug delivery. Int J Pharm 1998; 172: 33-70.
[http://dx.doi.org/10.1016/S0378-5173(98)00169-0]
[110]
Yeo PL, Lim CL, Chye SM, Ling APK, Koh RY. Niosomes: a review of their structure, properties, methods of preparation, and medical applications. Asian Biomed 2017; 11: 301-14.
[http://dx.doi.org/10.1515/abm-2018-0002]
[111]
Behroozeh A, Mazloumi Tabrizi M, Kazemi SM, et al. Evaluation the anti-cancer effect of pegylated nano-niosomal gingerol, on breast cancer cell lines (T47D), in-vitro. Asian Pac J Cancer Prev 2018; 19(3): 645-8.
[PMID: 29580033]
[112]
Sasani E, Shahi Malmir H, Daneshmand F, Majdizadeh M, Haghiralsadat BF. A new study on synthesize and optimization of PEGylated LipoNiosomal nanocarriers containing curcumin for use in cancer chemotherapy. SSU J 2018; 26: 528-41.
[113]
Rungphanichkul N, Nimmannit U, Muangsiri W, Rojsitthisak P. Preparation of curcuminoid niosomes for enhancement of skin permeation. Pharmazie 2011; 66(8): 570-5.
[PMID: 21901978]
[114]
Tavano L, Muzzalupo R, Picci N, de Cindio B. Co-encapsulation of antioxidants into niosomal carriers: gastrointestinal release studies for nutraceutical applications. Colloids Surf B Biointerfaces 2014; 114: 82-8.
[http://dx.doi.org/10.1016/j.colsurfb.2013.09.058] [PMID: 24176886]
[115]
Danafar H, Sharafi A, Kheiri S, Kheiri Manjili H. Co-delivery of sulforaphane and curcumin with PEGylated iron oxide-gold core shell nanoparticles for delivery to breast cancer cell line. Iran J Pharm Res 2018; 17(2): 480-94.
[PMID: 29881406]
[116]
Bhatt H, Rompicharla SVK, Komanduri N, et al. Development of curcumin-loaded solid lipid nanoparticles utilizing glyceryl monostearate as single lipid using QbD approach: Characterization and Evaluation of anticancer activity against human breast cancer cell line. Curr Drug Deliv 2018; 15(9): 1271-83.
[http://dx.doi.org/10.2174/1567201815666180503120113] [PMID: 29732970]
[117]
Alemi A, Zavar Reza J, Haghiralsadat F, et al. Paclitaxel and curcumin coadministration in novel cationic PEGylated niosomal formulations exhibit enhanced synergistic antitumor efficacy. J Nanobiotechnology 2018; 16(1): 28.
[http://dx.doi.org/10.1186/s12951-018-0351-4] [PMID: 29571289]
[118]
Huang F, Gao Y, Zhang Y, et al. Silver-decorated polymeric micelles combined with curcumin for enhanced antibacterial activity. ACS Appl Mater Interfaces 2017; 9(20): 16880-9.
[http://dx.doi.org/10.1021/acsami.7b03347] [PMID: 28481077]
[119]
Yan J-K, Qiu W-Y, Wang Y-Y, Wu J-Y. Biocompatible polyelectrolyte complex nanoparticles from lactoferrin and pectin as potential vehicles for antioxidative curcumin. J Agric Food Chem 2017; 65(28): 5720-30.
[http://dx.doi.org/10.1021/acs.jafc.7b01848] [PMID: 28657749]
[120]
Bagheri R, Sanaat Z, Zarghami N. Synergistic effect of free and nano-encapsulated chrysin-curcumin on inhibition of htert gene expression in SW480 colorectal cancer cell line. Drug Res 2018.
[http://dx.doi.org/10.1055/s-0043-121338]
[121]
Mao K-L, Fan Z-L, Yuan J-D, et al. Skin-penetrating polymeric nanoparticles incorporated in silk fibroin hydrogel for topical delivery of curcumin to improve its therapeutic effect on psoriasis mouse model. Colloids Surf B Biointerfaces 2017; 160: 704-14.
[http://dx.doi.org/10.1016/j.colsurfb.2017.10.029] [PMID: 29035818]
[122]
Huang N, Lu S, Liu X-G, Zhu J, Wang Y-J, Liu R-T. PLGA nanoparticles modified with a BBB-penetrating peptide co-delivering Aβ generation inhibitor and curcumin attenuate memory deficits and neuropathology in Alzheimer’s disease mice. Oncotarget 2017; 8(46): 81001-13.
[http://dx.doi.org/10.18632/oncotarget.20944] [PMID: 29113362]
[123]
Khatoon M, Shah KU, Din FU, et al. Proniosomes derived niosomes: recent advancements in drug delivery and targeting. Drug Deliv 2017; 24(sup1): 56-69.
[http://dx.doi.org/10.1080/10717544.2017.1384520] [PMID: 29130758]
[124]
Abdelbary GA, Amin MM, Zakaria MY. Ocular ketoconazole-loaded proniosomal gels: formulation, ex vivo corneal permeation and in vivo studies. Drug Deliv 2017; 24(1): 309-19.
[http://dx.doi.org/10.1080/10717544.2016.1247928] [PMID: 28165809]
[125]
Khalil RM, Abd-Elbary G, Basha M, Awad GE, Elhashemy HA. Proniosomes as a carrier for ocular drug delivery. World Academy of Science, Engineering and Technology Int J Pharmacol Pharm Sci 2017; 4
[126]
Madni A, Rahim MA, Mahmood MA, et al. Enhancement of dissolution and skin permeability of pentazocine by proniosomes and niosomal gel. AAPS PharmSciTech 2018; 19(4): 1544-53.
[http://dx.doi.org/10.1208/s12249-018-0967-6] [PMID: 29470828]
[127]
Liu H, Tu L, Zhou Y, et al. Improved bioavailability and antitumor effect of docetaxel by TPGS modified proniosomes: in Vitro and in Vivo evaluations. Sci Rep 2017; 7: 43372.
[http://dx.doi.org/10.1038/srep43372] [PMID: 28266539]
[128]
Kulhari H, Pooja D, Shrivastava S, et al. Trastuzumab-grafted PAMAM dendrimers for the selective delivery of anticancer drugs to HER2-positive breast cancer. Sci Rep 2016; 6: 23179.
[http://dx.doi.org/10.1038/srep23179] [PMID: 27052896]
[129]
Shi W, Dolai S, Rizk S, et al. Synthesis of monofunctional curcumin derivatives, clicked curcumin dimer, and a PAMAM dendrimer curcumin conjugate for therapeutic applications. Org Lett 2007; 9(26): 5461-4.
[http://dx.doi.org/10.1021/ol702370m] [PMID: 18020348]
[130]
Mollazade M, Nejati-Koshki K, Akbarzadeh A, et al. PAMAM dendrimers augment inhibitory effects of curcumin on cancer cell proliferation: possible inhibition of telomerase. Asian Pac J Cancer Prev 2013; 14(11): 6925-8.
[http://dx.doi.org/10.7314/APJCP.2013.14.11.6925] [PMID: 24377627]
[131]
Sharma AK, Gupta L, Sahu H, et al. Chitosan engineered PAMAM dendrimers as nanoconstructs for the enhanced anti-cancer potential and improved in vivo brain pharmacokinetics of temozolomide. Pharm Res 2018; 35(1): 9.
[http://dx.doi.org/10.1007/s11095-017-2324-y] [PMID: 29294212]
[132]
Song Z, Zhu W, Song J, et al. Linear-dendrimer type methoxy-poly (ethylene glycol)-b-poly (ε-caprolactone) copolymer micelles for the delivery of curcumin. Drug Deliv 2015; 22(1): 58-68.
[http://dx.doi.org/10.3109/10717544.2014.901436] [PMID: 24725028]
[133]
Wang L, Xu X, Zhang Y, et al. Encapsulation of curcumin within poly(amidoamine) dendrimers for delivery to cancer cells. J Mater Sci Mater Med 2013; 24(9): 2137-44.
[http://dx.doi.org/10.1007/s10856-013-4969-3] [PMID: 23779153]
[134]
Masunaga S-i. Tumor microenvironment and hyperthermia. 151-69. 2016
[http://dx.doi.org/10.1007/978-981-10-0719-4_14]
[135]
Montazerabadi A, Beik J, Irajirad R, et al. Folate-modified and curcumin-loaded dendritic magnetite nanocarriers for the targeted thermo-chemotherapy of cancer cells. Artif Cells Nanomed Biotechnol 2019; 47(1): 330-40.
[http://dx.doi.org/10.1080/21691401.2018.1557670] [PMID: 30688084]
[136]
Suo A, Qian J, Xu M, Xu W, Zhang Y, Yao Y. Folate-decorated PEGylated triblock copolymer as a pH/reduction dual-responsive nanovehicle for targeted intracellular co-delivery of doxorubicin and Bcl-2 siRNA. Mater Sci Eng C 2017; 76: 659-72.
[http://dx.doi.org/10.1016/j.msec.2017.03.124] [PMID: 28482576]
[137]
Ghaffari M, Dehghan G, Baradaran B, et al. Co-delivery of curcumin and Bcl-2 siRNA by PAMAM dendrimers for enhancement of the therapeutic efficacy in HeLa cancer cells. Colloids Surf B Biointerfaces 2020; 188110762
[http://dx.doi.org/10.1016/j.colsurfb.2019.110762] [PMID: 31911391]
[138]
Ghalandarlaki N, Alizadeh AM, Ashkani-Esfahani S. Nanotechnology-applied curcumin for different diseases therapy. BioMed Res Int 2014; 2014394264
[http://dx.doi.org/10.1155/2014/394264] [PMID: 24995293]
[139]
Sharma P, Mehta M, Dhanjal DS, et al. Emerging trends in the novel drug delivery approaches for the treatment of lung cancer. Chem Biol Interact 2019; 309108720
[http://dx.doi.org/10.1016/j.cbi.2019.06.033] [PMID: 31226287]
[140]
De Silva L, Goh B-H, Lee L-H, Chuah L-H. Curcumin-loaded nanoparticles and their potential as anticancer agents in breast cancernatural bio-active compounds. Springer 2019.
[http://dx.doi.org/10.1007/978-981-13-7205-6_7]
[141]
Mehanny M, Hathout RM, Geneidi AS, Mansour S. Exploring the use of nanocarrier systems to deliver the magical molecule; Curcumin and its derivatives. J Control Release 2016; 225: 1-30.
[http://dx.doi.org/10.1016/j.jconrel.2016.01.018] [PMID: 26778694]
[142]
Shang L, Nienhaus K, Nienhaus GU. Engineered nanoparticles interacting with cells: size matters. J Nanobiotechnology 2014; 12: 5.
[http://dx.doi.org/10.1186/1477-3155-12-5] [PMID: 24491160]
[143]
Jourghanian P, Ghaffari S, Ardjmand M, Haghighat S, Mohammadnejad M. Sustained release curcumin loaded solid lipid nanoparticles. Adv Pharm Bull 2016; 6(1): 17-21.
[http://dx.doi.org/10.15171/apb.2016.04] [PMID: 27123413]
[144]
Yallapu MM, Khan S, Maher DM, et al. Anti-cancer activity of curcumin loaded nanoparticles in prostate cancer. Biomaterials 2014; 35(30): 8635-48.
[http://dx.doi.org/10.1016/j.biomaterials.2014.06.040] [PMID: 25028336]
[145]
Rejinold NS, Muthunarayanan M, Divyarani VV, et al. Curcumin-loaded biocompatible thermoresponsive polymeric nanoparticles for cancer drug delivery. J Colloid Interface Sci 2011; 360(1): 39-51.
[http://dx.doi.org/10.1016/j.jcis.2011.04.006] [PMID: 21549390]
[146]
Gera M, Sharma N, Ghosh M, et al. Nanoformulations of curcumin: an emerging paradigm for improved remedial application. Oncotarget 2017; 8(39): 66680-98.
[http://dx.doi.org/10.18632/oncotarget.19164] [PMID: 29029547]
[147]
Yallapu MM, Ebeling MC, Khan S, et al. Novel curcumin-loaded magnetic nanoparticles for pancreatic cancer treatment. Mol Cancer Ther 2013; 12(8): 1471-80.
[http://dx.doi.org/10.1158/1535-7163.MCT-12-1227] [PMID: 23704793]
[148]
Li L, Xiang D, Shigdar S, et al. Epithelial cell adhesion molecule aptamer functionalized PLGA-lecithin-curcumin-PEG nanoparticles for targeted drug delivery to human colorectal adenocarcinoma cells. Int J Nanomedicine 2014; 9: 1083-96.
[PMID: 24591829]
[149]
Wickens JM, Alsaab HO, Kesharwani P, et al. Recent advances in hyaluronic acid-decorated nanocarriers for targeted cancer therapy. Drug Discov Today 2017; 22(4): 665-80.
[http://dx.doi.org/10.1016/j.drudis.2016.12.009] [PMID: 28017836]
[150]
Nam NH, Doan DH, Nhung HTM, et al. Folate attached, curcumin loaded Fe3O4 nanoparticles: A novel multifunctional drug delivery system for cancer treatment. Mater Chem Phys 2016; 172: 98-104.
[http://dx.doi.org/10.1016/j.matchemphys.2015.12.065]
[151]
Metwally AA, El-Ahmady SH, Hathout RM. Selecting optimum protein nano-carriers for natural polyphenols using chemoinformatics tools. Phytomedicine 2016; 23(14): 1764-70.
[http://dx.doi.org/10.1016/j.phymed.2016.10.020] [PMID: 27912878]
[152]
Thulasidasan AKT, Retnakumari AP, Shankar M, et al. Folic acid conjugation improves the bioavailability and chemosensitizing efficacy of curcumin-encapsulated PLGA-PEG nanoparticles towards paclitaxel chemotherapy. Oncotarget 2017; 8(64): 107374-89.
[http://dx.doi.org/10.18632/oncotarget.22376] [PMID: 29296172]
[153]
Abdel-Hafez SM, Hathout RM, Sammour OA. Tracking the transdermal penetration pathways of optimized curcumin-loaded chitosan nanoparticles via confocal laser scanning microscopy. Int J Biol Macromol 2018; 108: 753-64.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.10.170] [PMID: 29104049]
[154]
He X, Li Q, Liu X, Wu G, Zhai G. Curcumin-loaded lipid cubic liquid crystalline nanoparticles: preparation, optimization, physicochemical properties and oral absorption. J Nanosci Nanotechnol 2015; 15(8): 5559-65.
[http://dx.doi.org/10.1166/jnn.2015.10311] [PMID: 26369117]
[155]
de Souza JF, da Silva Pontes K, Alves TFR, et al. Structural comparison, physicochemical properties, and in vitro release profile of curcumin-loaded lyotropic liquid crystalline nanoparticle: Influence of hydrotrope as interface stabilizers. J Mol Liq 2020.112861
[http://dx.doi.org/10.1016/j.molliq.2020.112861]
[156]
Song Z, Lu Y, Zhang X, Wang H, Han J, Dong C. Novel curcumin-loaded human serum albumin nanoparticles surface functionalized with folate: characterization and in vitro/vivo evaluation. Drug Des Devel Ther 2016; 10: 2643-9.
[http://dx.doi.org/10.2147/DDDT.S112039] [PMID: 27574403]
[157]
Saleh T, Soudi T, Shojaosadati SA. Aptamer functionalized curcumin-loaded human serum albumin (HSA) nanoparticles for targeted delivery to HER-2 positive breast cancer cells. Int J Biol Macromol 2019; 130: 109-16.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.02.129] [PMID: 30802519]
[158]
Song W, Su X, Gregory DA, Li W, Cai Z, Zhao X. Magnetic alginate/chitosan nanoparticles for targeted delivery of curcumin into human breast cancer cells. Nanomaterials (Basel) 2018; 8(11): 8.
[http://dx.doi.org/10.3390/nano8110907] [PMID: 30400634]
[159]
Chen S, Han Y, Huang J, et al. Fabrication and characterization of layer-by-layer composite nanoparticles based on zein and hyaluronic acid for codelivery of curcumin and quercetagetin. ACS Appl Mater Interfaces 2019; 11(18): 16922-33.
[http://dx.doi.org/10.1021/acsami.9b02529] [PMID: 30985111]
[160]
Song W, Muthana M, Mukherjee J, Falconer RJ, Biggs CA, Zhao X. Magnetic-silk core-Shell nanoparticles as potential carriers for targeted delivery of curcumin into human breast cancer cells. ACS Biomater Sci Eng 2017; 3: 1027-38.
[http://dx.doi.org/10.1021/acsbiomaterials.7b00153]
[161]
Nosrati H, Charmi J, Salehiabar M, Abhari F, Danafar H. Tumor targeted albumin coated bismuth sulfide nanoparticles (Bi2S3) as radiosensitizers and carriers of curcumin for enhanced chemoradiation therapy. ACS Biomater Sci Eng 2019; 5: 4416-24.
[http://dx.doi.org/10.1021/acsbiomaterials.9b00489]
[162]
Huang P, Zeng B, Mai Z, et al. Novel drug delivery nanosystems based on out-inside bifunctionalized mesoporous silica yolk-shell magnetic nanostars used as nanocarriers for curcumin. J Mater Chem B Mater Biol Med 2016; 4(1): 46-56.
[http://dx.doi.org/10.1039/C5TB02184G] [PMID: 32262808]
[163]
Ma’mani L, Nikzad S, Kheiri-Manjili H, et al. Curcumin-loaded guanidine functionalized PEGylated I3ad mesoporous silica nanoparticles KIT-6: practical strategy for the breast cancer therapy. Eur J Med Chem 2014; 83: 646-54.
[http://dx.doi.org/10.1016/j.ejmech.2014.06.069] [PMID: 25014638]
[164]
Farooqi AA, Desai NN, Qureshi MZ, et al. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol Adv 2017; 36(1): 328-64.
[PMID: 29248680]
[165]
Aqil F, Munagala R, Jeyabalan J, Agrawal AK, Gupta R. Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J 2017; 19(6): 1691-702.
[http://dx.doi.org/10.1208/s12248-017-0154-9] [PMID: 29047044]
[166]
Choi ES, Kang YY, Mok H. Evaluation of the enhanced antioxidant activity of curcumin within exosomes by fluorescence monitoring. Biotechnol Bioprocess Eng; BBE 2018; 23: 150-7.
[http://dx.doi.org/10.1007/s12257-018-0058-2]
[167]
Soung YH, Ford S, Zhang V, Chung J. Exosomes in cancer diagnostics. Cancers (Basel) 2017; 9(1): 8.
[http://dx.doi.org/10.3390/cancers9010008] [PMID: 28085080]
[168]
Rupp A-K, Rupp C, Keller S, et al. Loss of EpCAM expression in breast cancer derived serum exosomes: role of proteolytic cleavage. Gynecol Oncol 2011; 122(2): 437-46.
[http://dx.doi.org/10.1016/j.ygyno.2011.04.035] [PMID: 21601258]
[169]
Moon P-G, Lee J-E, Cho Y-E, et al. Fibronectin on circulating extracellular vesicles as a liquid biopsy to detect breast cancer. Oncotarget 2016; 7(26): 40189-99.
[http://dx.doi.org/10.18632/oncotarget.9561] [PMID: 27250024]
[170]
Moon P-G, Lee J-E, Cho Y-E, et al. Identification of developmental endothelial locus-1 on circulating extracellular vesicles as a novel biomarker for early breast cancer detection. Clin Cancer Res 2016; 22(7): 1757-66.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-0654] [PMID: 26603257]
[171]
Vashisht M, Rani P, Onteru SK, Singh D. Curcumin encapsulated in milk exosomes resists human digestion and possesses enhanced intestinal permeability in vitro. Appl Biochem Biotechnol 2017; 183(3): 993-1007.
[http://dx.doi.org/10.1007/s12010-017-2478-4] [PMID: 28466459]
[172]
Lao CD, Ruffin MT IV, Normolle D, et al. Dose escalation of a curcuminoid formulation. BMC Complement Altern Med 2006; 6: 10.
[http://dx.doi.org/10.1186/1472-6882-6-10] [PMID: 16545122]
[173]
Vareed SK, Kakarala M, Ruffin MT, et al. Pharmacokinetics of curcumin conjugate metabolites in healthy human subjects. Cancer Epidemiol Biomarkers Prev 2008; 17(6): 1411-7.
[http://dx.doi.org/10.1158/1055-9965.EPI-07-2693] [PMID: 18559556]
[174]
Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm 2007; 4(6): 807-18.
[http://dx.doi.org/10.1021/mp700113r] [PMID: 17999464]
[175]
Kanai M, Otsuka Y, Otsuka K, et al. A phase I study investigating the safety and pharmacokinetics of highly bioavailable curcumin (Theracurmin) in cancer patients. Cancer Chemother Pharmacol 2013; 71(6): 1521-30.
[http://dx.doi.org/10.1007/s00280-013-2151-8] [PMID: 23543271]
[176]
Kanai M, Imaizumi A, Otsuka Y, et al. Dose-escalation and pharmacokinetic study of nanoparticle curcumin, a potential anticancer agent with improved bioavailability, in healthy human volunteers. Cancer Chemother Pharmacol 2012; 69(1): 65-70.
[http://dx.doi.org/10.1007/s00280-011-1673-1] [PMID: 21603867]
[177]
Patra JK, Das G, Fraceto LF, et al. Sharma SJJon. Nano based drug delivery systems: recent developments and future prospects 2018; 61: 71.
[178]
Pang Y, Mai Z, Wang B, et al. Artesunate-modified nano-graphene oxide for chemo-photothermal cancer therapy. Oncotarget 2017; 8(55): 93800-12.
[http://dx.doi.org/10.18632/oncotarget.21191] [PMID: 29212190]
[179]
Chen H, Zhen Z, Todd T, Chu PK, Xie JJMS, Reports ER. Nanoparticles for improving cancer diagnosis. Mater Sci Eng Rep 2013; 74: 35-69.
[http://dx.doi.org/10.1016/j.mser.2013.03.001]
[180]
Bahadur D, Giri J. Biomaterials and magnetism. Sadhana 2003; 28: 639-56.
[http://dx.doi.org/10.1007/BF02706451]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy