Generic placeholder image

Current Chemical Biology

Editor-in-Chief

ISSN (Print): 2212-7968
ISSN (Online): 1872-3136

Research Article

Anticarcinogenic Effects of Capsaicin-Loaded Nanoparticles on In vitro Hepatocellular Carcinoma

Author(s): Noha M Hazem*, Wagdi F ElKashef, Ibrahim M El-Sherbiny, Ahmed A Emam, Dalia Shaalan and Mohamed Sobh

Volume 15, Issue 2, 2021

Published on: 16 November, 2020

Page: [188 - 201] Pages: 14

DOI: 10.2174/2212796814999201116211648

Price: $65

Abstract

Background: Hepatocellular Carcinoma (HCC) is the fifth most frequent cancer worldwide with a low overall survival due to high metastasis and recurrence rates. The aim of this study is to assess and compare the possible anti-neoplastic effect of capsaicin and nanoformulated capsaicin on in vitro HCC human cell line HepG2. The source of the cell line, including when and from where it was obtained. Whether the cell line has recently been authenticated and by what method. Whether the cell line has recently been tested for mycoplasma contamination.

Materials and Methods: Capsaicin-loaded Trimethyl Chitosan Nanoparticles (CL TMCS NPs) were synthesized by ionotropic gelation of cationic TMCS with capsaicin. The synthesized nanoparticles were characterized through TEM, and zeta analyzer. Human hepatocarcinoma HepG2 cell lines were cultured and treated with 50, 75 & 100 μM of Capsaicin (CAP), plain TMCS NPs and CL-NPs as well as ethanol (control) for 24h and 48h. The induced effects were investigated by flow cytometry, immunocytochemistry assay for Bcl-2, Bax, and caspase proteins and evaluating gene expression levels of Bcl-2, Bax, and MDR-1 mRNA by real-time PCR.

Results: Our results demonstrated that capsaicin- loaded NPs had the potential to significantly increase capsaicin bioactivity compared with the plain capsaicin formulation either in inducing apoptosis through altering expression of apoptotic regulators or modifying MDR-1 expression.

Conclusions: TMCs nanoparticles investigated in this study may be a good drug delivery vehicle for capsaicin. Application of capsaicin-loaded NPs in HCC management as an adjunct therapeutic approach may be a novel strategy to improve the treatment efficacy and resistance of the conventionally used chemotherapy.

Keywords: Hepatocellular carcinoma, capsaicin, trimethyl chitosan nanoparticles, anticarcinogenic, fetal bovine serum, transient receptor potential vanilloid 1.

« Previous
Graphical Abstract
[1]
Adami HO, Hunter DJ, Trichopoulos D. Textbook of cancer epidemiology New York, NY, USA: Oxford Unizversity Press. 2008.
[http://dx.doi.org/10.1093/acprof:oso/9780195311174.001.0001]
[2]
Rose PG, Piver MS, Tsukada Y, Lau T. Patterns of metastasis in uterine sarcoma. An autopsy study. Cancer 1989; 63(5): 935-8.
[http://dx.doi.org/10.1002/1097-0142(19890301)63:5<935::AID-CNCR2820630525>3.0.CO;2-9] [PMID: 2914299]
[3]
Ma C, Han M, Heinrich B, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018; 360(6391): eaan5931.
[http://dx.doi.org/10.1126/science.aan5931] [PMID: 29798856]
[4]
Xie B, Wang DH, Spechler SJ. Sorafenib for treatment of hepatocellular carcinoma: a systematic review. Dig Dis Sci 2012; 57(5): 1122-9.
[http://dx.doi.org/10.1007/s10620-012-2136-1] [PMID: 22451120]
[5]
Colombo M, Sangiovanni A. Treatment of hepatocellular carcinoma: beyond international guidelines. Liver Int 2015; 35(Suppl. 1): 129-38.
[http://dx.doi.org/10.1111/liv.12713] [PMID: 25529098]
[6]
Sora GTS, Haminiuk CWI, da Silva MV, et al. A comparative study of the capsaicinoid and phenolic contents and in vitro antioxidant activities of the peppers of the genus Capsicum: an application of chemometrics. J Food Sci Technol 2015; 52(12): 8086-94.
[http://dx.doi.org/10.1007/s13197-015-1935-8] [PMID: 26604381]
[7]
Chapa-Oliver AM, Mejía-Teniente L. Capsaicin: From Plants to a Cancer-Suppressing Agent. Molecules 2016; 21(8): 931.
[http://dx.doi.org/10.3390/molecules21080931] [PMID: 27472308]
[8]
Kang C, Wang B, Kaliannan K, et al. Gut microbiota mediates the protective effects of dietary capsaicin against chronic lowgrade inflammation and associated obesity induced by high-fat diet. MBio 2017; 8(3): e00470-17.
[http://dx.doi.org/10.1128/mBio.00470-17] [PMID: 28536285]
[9]
Zhang SS, Ni YH, Zhao CR, et al. Capsaicin enhances the antitumor activity of sorafenib in hepatocellular carcinoma cells and mouse xenograft tumors through increased ERK signaling. Acta Pharmacol Sin 2018; 39(3): 438-48.
[http://dx.doi.org/10.1038/aps.2017.156] [PMID: 29188798]
[10]
Lee JH, Kim C, Baek SH, et al. Capsazepine inhibits JAK/STAT3 signaling, tumor growth, and cell survival in prostate cancer. Oncotarget 2017; 8(11): 17700-11.
[http://dx.doi.org/10.18632/oncotarget.10775] [PMID: 27458171]
[11]
Nazıroğlu M, Çiğ B, Blum W, et al. Targeting breast cancer cells by MRS1477, a positive allosteric modulator of TRPV1 channels. PLoS One 2017; 12(6): e0179950.
[http://dx.doi.org/10.1371/journal.pone.0179950] [PMID: 28640864]
[12]
Yoshitani SI, Tanaka T, Kohno H, Takashima S. Chemoprevention of azoxymethane-induced rat colon carcinogenesis by dietary capsaicin and rotenone. Int J Oncol 2001; 19(5): 929-39.
[http://dx.doi.org/10.3892/ijo.19.5.929] [PMID: 11604990]
[13]
Kim YM, Hwang JT, Kwak DW, Lee YK, Park OJ. Involvement of AMPK signaling cascade in capsaicin-induced apoptosis of HT-29 colon cancer cells. Ann N Y Acad Sci 2007; 1095: 496-503.
[http://dx.doi.org/10.1196/annals.1397.053] [PMID: 17404062]
[14]
Lin YT, Wang HC, Hsu YC, Cho CL, Yang MY, Chien CY. Capsaicin induces autophagy and apoptosis in human nasopharyngeal carcinoma cells by downregulating the PI3K/AKT/mTOR pathway. Int J Mol Sci 2017; 18(7): E1343.
[http://dx.doi.org/10.3390/ijms18071343] [PMID: 28644386]
[15]
Wu CC, Lin JP, Yang JS, et al. Capsaicin induced cell cycle arrest and apoptosis in human esophagus epidermoid carcinoma CE 81T/VGH cells through the elevation of intracellular reactive oxygen species and Ca2+ productions and caspase-3 activation. Mutat Res 2006; 601(1-2): 71-82.
[http://dx.doi.org/10.1016/j.mrfmmm.2006.06.015] [PMID: 16942782]
[16]
Huang SP, Chen JC, Wu CC, et al. Capsaicin-induced apoptosis in human hepatoma HepG2 cells. Anticancer Res 2009; 29(1): 165-74.
[PMID: 19331147]
[17]
Chen X, Tan M, Xie Z, et al. Inhibiting ROS-STAT3-dependent autophagy enhanced capsaicin-induced apoptosis in human hepatocellular carcinoma cells. Free Radic Res 2016; 50(7): 744-55.
[http://dx.doi.org/10.3109/10715762.2016.1173689] [PMID: 27043357]
[18]
Lee JY, Lee SY, Kim GG, et al. Development of 68Ga-SCN-DOTA-Capsaicin as an imaging agent targeting apoptosis and cell cycle arrest in breast cancer. Cancer Biother Radiopharm 2017; 32(5): 169-75.
[http://dx.doi.org/10.1089/cbr.2017.2186] [PMID: 28598691]
[19]
Granato M, Gilardini Montani MS, Filardi M, Faggioni A, Cirone M. Capsaicin triggers immunogenic PEL cell death, stimulates DCs and reverts PEL-induced immune suppression. Oncotarget 2015; 6(30): 29543-54.
[http://dx.doi.org/10.18632/oncotarget.4911] [PMID: 26338963]
[20]
Weber LV, Al-Refae K, Wölk G, et al. Expression and functionality of TRPV1 in breast cancer cells. Breast Cancer (Dove Med Press) 2016; 8: 243-52.
[http://dx.doi.org/10.2147/BCTT.S121610] [PMID: 28008282]
[21]
Sánchez AM, Malagarie-Cazenave S, Olea N, Vara D, Chiloeches A, Díaz-Laviada I. Apoptosis induced by capsaicin in prostate PC-3 cells involves ceramide accumulation, neutral sphingomyelinase, and JNK activation. Apoptosis 2007; 12(11): 2013-24.
[http://dx.doi.org/10.1007/s10495-007-0119-z] [PMID: 17828457]
[22]
Li L, Wei XH, Pan YP, et al. LAPTM4B: a novel cancer-associated gene motivates multidrug resistance through efflux and activating PI3K/AKT signaling. Oncogene 2010; 29(43): 5785-95.
[http://dx.doi.org/10.1038/onc.2010.303] [PMID: 20711237]
[23]
Wang XQ, Ongkeko WM, Chen L, et al. Octamer 4 (Oct4) mediates chemotherapeutic drug resistance in liver cancer cells through a potential Oct4-AKT-ATP-binding cassette G2 pathway. Hepatology 2010; 52(2): 528-39.
[http://dx.doi.org/10.1002/hep.23692] [PMID: 20683952]
[24]
Tsang WP, Kwok TT. Riboregulator H19 induction of MDR1-associated drug resistance in human hepatocellular carcinoma cells. Oncogene 2007; 26(33): 4877-81.
[http://dx.doi.org/10.1038/sj.onc.1210266] [PMID: 17297456]
[25]
Kasting GB. Kinetics of finite dose absorption through skin 1. Vanillylnonanamide. J Pharm Sci 2001; 90(2): 202-12.
[http://dx.doi.org/10.1002/1520-6017(200102)90:2<202::AID-JPS11>3.0.CO;2-E] [PMID: 11169537]
[26]
Trubetskoy VS. Polymeric micelles as carriers of diagnostic agents. Adv Drug Deliv Rev 1999; 37(1-3): 81-8.
[http://dx.doi.org/10.1016/S0169-409X(98)00100-8] [PMID: 10837728]
[27]
Smith H, Brooks JR. Capsaicin-based therapies for pain control. Prog Drug Res 2014; 68: 129-46.
[PMID: 24941667]
[28]
Sharma SK, Vij AS, Sharma M. Mechanisms and clinical uses of capsaicin. Eur J Pharmacol 2013; 720(1-3): 55-62.
[http://dx.doi.org/10.1016/j.ejphar.2013.10.053] [PMID: 24211679]
[29]
Verheul RJ, Amidi M, van der Wal S, van Riet E, Jiskoot W, Hennink WE. Synthesis, characterization and in vitro biological properties of O-methyl free N,N,N-trimethylated chitosan. Biomaterials 2008; 29(27): 3642-9.
[http://dx.doi.org/10.1016/j.biomaterials.2008.05.026] [PMID: 18556059]
[30]
Elkholi IE, Hazem NM, ElKashef WF, et al. Evaluation of anti-cancer potential of capsaicin-loaded trimethyl chitosan-based nanoparticles in HepG2 hepatocarcinoma cells. J Nanomed Nanotechnol 2014; 5: 240.
[http://dx.doi.org/10.4172/2157-7439.1000240]
[31]
Bort A, Sánchez BG, Spínola E, Mateos-Gómez PA, Rodríguez-Henche N, Díaz-Laviada I. The red pepper’s spicy ingredient capsaicin activates AMPK in HepG2 cells through CaMKKβ. PLoS One 2019; 14(1): e0211420.
[http://dx.doi.org/10.1371/journal.pone.0211420] [PMID: 30695053]
[32]
Zhao H, Oczos J, Janowski P, et al. Rationale for the real-time and dynamic cell death assays using propidium iodide. Cytometry A 2010; 77(4): 399-405.
[http://dx.doi.org/10.1002/cyto.a.20867] [PMID: 20131407]
[33]
Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Δ Δ C(T)) Method. Methods 2001; 25(4): 402-8.
[http://dx.doi.org/10.1006/meth.2001.1262] [PMID: 11846609]
[34]
Akshatha GM, Raval SK, Arpitha GM, Raval SH, Ghodasara DJ. Immunohistochemical, histopathological study and chemoprotective effect of Solanum nigrum in N-nitrosodiethylamine-induced hepatocellular carcinoma in Wistar rats. Vet World 2018; 11(4): 402-9.
[http://dx.doi.org/10.14202/vetworld.2018.402-409]
[35]
Khan N, Adhami VM, Mukhtar H. Apoptosis by dietary agents for prevention and treatment of prostate cancer. Endocr Relat Cancer 2010; 17(1): R39-52.
[http://dx.doi.org/10.1677/ERC-09-0262] [PMID: 19926708]
[36]
Jung MY, Kang HJ, Moon A. Capsaicin-induced apoptosis in SK-Hep-1 hepatocarcinoma cells involves Bcl-2 downregulation and caspase-3 activation. Cancer Lett 2001; 165(2): 139-45.
[http://dx.doi.org/10.1016/S0304-3835(01)00426-8] [PMID: 11275362]
[37]
Kaymaz BT, Cetintaş VB, Aktan C, Kosova B. MicroRNA-520a-5p displays a therapeutic effect upon chronic myelogenous leukemia cells by targeting STAT3 and enhances the anticarcinogenic role of capsaicin. Tumour Biol 2014; 35(9): 8733-42.
[http://dx.doi.org/10.1007/s13277-014-2138-z] [PMID: 24870597]
[38]
Jin J, Lin G, Huang H, et al. Capsaicin mediates cell cycle arrest and apoptosis in human colon cancer cells via stabilizing and activating p53. Int J Biol Sci 2014; 10(3): 285-95.
[http://dx.doi.org/10.7150/ijbs.7730] [PMID: 24643130]
[39]
Moon DO, Kang CH, Kang SH, et al. Capsaicin sensitizes TRAIL-induced apoptosis through Sp1-mediated DR5 up-regulation: involvement of Ca(2+) influx. Toxicol Appl Pharmacol 2012; 259(1): 87-95.
[http://dx.doi.org/10.1016/j.taap.2011.12.010] [PMID: 22200406]
[40]
Bhutani M, Pathak AK, Nair AS, et al. Capsaicin is a novel blocker of constitutive and interleukin-6-inducible STAT3 activation. Clin Cancer Res 2007; 13(10): 3024-32.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-2575] [PMID: 17505005]
[41]
Yang J, Li TZ, Xu GH, Luo BB, Chen YX, Zhang T. Low-concentration capsaicin promotes colorectal cancer metastasis by triggering ROS production and modulating Akt/mTOR and STAT-3 pathways. Neoplasma 2013; 60(4): 364-72.
[http://dx.doi.org/10.4149/neo_2013_048] [PMID: 23581408]
[42]
Warren CFA, Wong-Brown MW, Bowden NA. BCL-2 family isoforms in apoptosis and cancer. Cell Death Dis 2019; 10(3): 177.
[http://dx.doi.org/10.1038/s41419-019-1407-6] [PMID: 30792387]
[43]
Irani K, Xia Y, Zweier JL, et al. Mitogenic signaling mediated by oxidants in Ras-transformed fibroblasts. Science 1997; 275(5306): 1649-52.
[http://dx.doi.org/10.1126/science.275.5306.1649] [PMID: 9054359]
[44]
Renschler MF. The emerging role of reactive oxygen species in cancer therapy. Eur J Cancer 2004; 40(13): 1934-40.
[http://dx.doi.org/10.1016/j.ejca.2004.02.031] [PMID: 15315800]
[45]
Ramsey MR, Sharpless NE. ROS as a tumour suppressor? Nat Cell Biol 2006; 8(11): 1213-5.
[http://dx.doi.org/10.1038/ncb1106-1213] [PMID: 17077852]
[46]
Scaffidi C, Fulda S, Srinivasan A, et al. Two CD95 (APO-1/Fas) signaling pathways. EMBO J 1998; 17(6): 1675-87.
[http://dx.doi.org/10.1093/emboj/17.6.1675] [PMID: 9501089]
[47]
Krammer PH. CD95's deadly mission in the immune system. Nature 2000; 407(6805): 789-95.
[http://dx.doi.org/10.1038/35037728] [PMID: 11048730]
[48]
Sartorius U, Schmitz I, Krammer PH. Molecular mechanisms of death-receptor-mediated apoptosis. ChemBioChem 2001; 2(1): 20-9.
[http://dx.doi.org/10.1002/1439-7633(20010105)2:1<20::AID-CBIC20>3.0.CO;2-X] [PMID: 11828422]
[49]
Kluck RM, Bossy-Wetzel E, Green DR, Newmeyer DD. The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science 1997; 275(5303): 1132-6.
[http://dx.doi.org/10.1126/science.275.5303.1132] [PMID: 9027315]
[50]
Yang J, Liu X, Bhalla K, et al. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science 1997; 275(5303): 1129-32.
[http://dx.doi.org/10.1126/science.275.5303.1129] [PMID: 9027314]
[51]
Dai N, Ye R, He Q, Guo P, Chen H, Zhang Q. Capsaicin and sorafenib combination treatment exerts synergistic anti‑hepatocellular carcinoma activity by suppressing EGFR and PI3K/Akt/mTOR signaling. Oncol Rep 2018; 40(6): 3235-48.
[PMID: 30272354]
[52]
Guan J, Chen XP, Zhu H, Luo SF, Cao B, Ding L. Involvement of extracellular signal-regulated kinase/mitogen-activated protein kinase pathway in multidrug resistance induced by HBx in hepatoma cell line. World J Gastroenterol 2004; 10(23): 3522-7.
[http://dx.doi.org/10.3748/wjg.v10.i23.3522] [PMID: 15526378]
[53]
Ng KK, Vauthey JN, Pawlik TM, et al. International Cooperative Study Group on Hepatocellular Carcinoma. Is hepatic resection for large or multinodular hepatocellular carcinoma justified? Results from a multi-institutional database. Ann Surg Oncol 2005; 12(5): 364-73.
[http://dx.doi.org/10.1245/ASO.2005.06.004] [PMID: 15915370]
[54]
Soini Y, Virkajärvi N, Raunio H, Pääkkö P. Expression of P-glycoprotein in hepatocellular carcinoma: a potential marker of prognosis. J Clin Pathol 1996; 49(6): 470-3.
[http://dx.doi.org/10.1136/jcp.49.6.470] [PMID: 8763260]
[55]
Kato A, Miyazaki M, Ambiru S, et al. Multidrug resistance gene (MDR-1) expression as a useful prognostic factor in patients with human hepatocellular carcinoma after surgical resection. J Surg Oncol 2001; 78(2): 110-5.
[http://dx.doi.org/10.1002/jso.1129] [PMID: 11579388]
[56]
Ganne-Carrié N, Trinchet JC. Systemic treatment of hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2004; 16(3): 275-81.
[http://dx.doi.org/10.1097/00042737-200403000-00005] [PMID: 15195890]
[57]
Vander Borght S, Komuta M, Libbrecht L, et al. Expression of multidrug resistance-associated protein 1 in hepatocellular carcinoma is associated with a more aggressive tumour phenotype and may reflect a progenitor cell origin. Liver Int 2008; 28(10): 1370-80.
[http://dx.doi.org/10.1111/j.1478-3231.2008.01889.x] [PMID: 19055643]
[58]
Chen CJ, Chin JE, Ueda K, et al. Internal duplication and homology with bacterial transport proteins in the mdr1 (P-glycoprotein) gene from multidrug-resistant human cells. Cell 1986; 47(3): 381-9.
[http://dx.doi.org/10.1016/0092-8674(86)90595-7] [PMID: 2876781]
[59]
Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2002; 2(1): 48-58.
[http://dx.doi.org/10.1038/nrc706] [PMID: 11902585]
[60]
Fojo T, Bates S. Strategies for reversing drug resistance. Oncogene 2003; 22(47): 7512-23.
[http://dx.doi.org/10.1038/sj.onc.1206951] [PMID: 14576855]
[61]
Gouazé V, Yu JY, Bleicher RJ, et al. Overexpression of glucosylceramide synthase and P-glycoprotein in cancer cells selected for resistance to natural product chemotherapy. Mol Cancer Ther 2004; 3(5): 633-9.
[PMID: 15141021]
[62]
Warmann S, Göhring G, Teichmann B, Geerlings H, Pietsch T, Fuchs J. P-glycoprotein modulation improves in vitro chemosensitivity in malignant pediatric liver tumors. Anticancer Res 2003; 23(6C): 4607-11.
[PMID: 14981903]
[63]
Huesker M, Folmer Y, Schneider M, Fulda C, Blum HE, Hafkemeyer P. Reversal of drug resistance of hepatocellular carcinoma cells by adenoviral delivery of anti-MDR1 ribozymes. Hepatology 2002; 36(4 Pt 1): 874-84.
[http://dx.doi.org/10.1053/jhep.2002.35619] [PMID: 12297834]
[64]
Xing F, Cheng G, Yi K, Ma L. Nanoencapsulation of capsaicin by complex coacervation of gelatin, acacia, and tannins. J Appl Polym Sci 2005; 96: 2225-9.
[http://dx.doi.org/10.1002/app.21698]
[65]
Tyagi P, Chancellor MB, Li Z, et al. Urodynamic and immunohistochemical evaluation of intravesical capsaicin delivery using thermosensitive hydrogel and liposomes. J Urol 2004; 171(1): 483-9.
[http://dx.doi.org/10.1097/01.ju.0000102360.11785.d7] [PMID: 14665960]
[66]
Tavano L, Alfano P, Muzzalupo R, de Cindio B. Niosomes vs microemulsions: new carriers for topical delivery of Capsaicin. Colloids Surf B Biointerfaces 2011; 87(2): 333-9.
[http://dx.doi.org/10.1016/j.colsurfb.2011.05.041] [PMID: 21684725]
[67]
J SJ, Jimena CF, Dalet FE, Guadalupe TJ, Antonio SM. J SJ. Scope of lipid nanoparticles in neuroscience: impact on the treatment of neurodegenerative diseases. Curr Pharm Des 2017; 23(21): 3120-33.
[http://dx.doi.org/10.2174/1381612823666170301123504] [PMID: 28260513]
[68]
Hong J, Li Y, Li Y, Xiao Y, Kuang H, Wang X. Annonaceous acetogenins nanosuspensions stabilized by PCL-PEG block polymer: significantly improved antitumor efficacy. Int J Nanomedicine 2016; 11: 3239-53.
[http://dx.doi.org/10.2147/IJN.S108143] [PMID: 27486323]
[69]
Alizadeh L, Zarebkohan A, Salehi R, Ajjoolabady A, Rahmati-Yamchi M. Chitosan-based nanotherapeutics for ovarian cancer treatment. J Drug Target 2019; 27(8): 839-52.
[http://dx.doi.org/10.1080/1061186X.2018.1564923] [PMID: 30596291]
[70]
Jia J, Zhang Y, Xin Y, Jiang C, Yan B, Zhai S. Interactions between nanoparticles and dendritic cells: From the perspective of cancer immunotherapy. Front Oncol 2018; 8: 404.
[http://dx.doi.org/10.3389/fonc.2018.00404] [PMID: 30319969]
[71]
Komenek S, Luesakul U, Ekgasit S, et al. Nanogold-gallate chitosan-targeted pulmonary delivery for treatment of lung cancer. AAPS PharmSciTech 2017; 18(4): 1104-15.
[http://dx.doi.org/10.1208/s12249-016-0644-6] [PMID: 27796908]
[72]
Siahmansouri H, Somi MH, Babaloo Z, et al. Effects of HMGA2 siRNA and doxorubicin dual delivery by chitosan nanoparticles on cytotoxicity and gene expression of HT-29 colorectal cancer cell line. J Pharm Pharmacol 2016; 68(9): 1119-30.
[http://dx.doi.org/10.1111/jphp.12593] [PMID: 27350211]
[73]
Khan MA, Zafaryab M, Mehdi SH, Ahmad I, Rizvi MMA. Characterization and anti-proliferative activity of curcumin loaded chitosan nanoparticles in cervical cancer. Int J Biol Macromol 2016; 93(Pt A): 242-53.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.08.050] [PMID: 27565296]
[74]
Anitha A, Deepa N, Chennazhi KP, Lakshmanan V-K, Jayakumar R. Combinatorial anticancer effects of curcumin and 5-fluorouracil loaded thiolated chitosan nanoparticles towards colon cancer treatment. Biochim Biophys Acta 2014; 1840(9): 2730-43. [BBA].
[http://dx.doi.org/10.1016/j.bbagen.2014.06.004] [PMID: 24946270]

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