Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

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

Review Article

Curcumin Based Drug Delivery Systems for Cancer Therapy

Author(s): Ankita Tiwari and Sanjay K. Jain*

Volume 26, Issue 42, 2020

Page: [5430 - 5440] Pages: 11

DOI: 10.2174/1381612826666200429095503

Price: $65

Abstract

Cancer accounts for the second major cause of death globally. Conventional cancer therapies lead to systemic toxicity that forbids their long term application. Besides, tumor resistance and recurrence have been observed in the majority of cases. Thus, the development of such therapy, which will pose minimum side effects, is the need of the hour. Curcumin or diferuloylmethane (CUR) is a natural polyphenol bioactive (obtained from Curcuma longa) which possesses anti-cancer and chemo-preventive activity. It acts by modulating various components of signaling cascades that are involved in cancer cell proliferation, invasion, and apoptosis process. It interacts with the adaptive and innate immune systems of our body and causes tumor regression. This may be the reason behind the attainment of in vivo anti-tumor activity at a very low concentration. Its ease of availability, safety profile, low cost, and multifaceted role in cancer prevention and treatment has made it a promising agent for chemoprevention of many cancers. Regardless of the phenomenal properties, its clinical utility is haltered due to its low aqueous solubility, poor bioavailability, rapid metabolism, and low cellular uptake. In the last few years, a variety of novel drug carriers have been fabricated to enhance the bioavailability and pharmacokinetic profile of CUR to attain better targeting of cancer. In this review, the recent developments in the arena of nanoformulations, like liposomes, polymeric NPs, solid lipid NPs (SNPs), polymeric micelles, nanoemulsions, microspheres, nanogels, etc. in anticancer therapy have been discussed along with a brief overview of the molecular targets for CUR in cancer therapy and role of CUR in cancer immunotherapy.

Keywords: Curcumin, cancer therapy, liposomes, nano-formulations, drug delivery, solubility.

[1]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69(1): 7-34.
[http://dx.doi.org/10.3322/caac.21551] [PMID: 30620402]
[2]
Tiwari A, Saraf S, Jain A, Panda PK, Verma A, Jain SK. Basics to advances in nanotherapy of colorectal cancer. Drug Deliv Transl Res 2019; 1-20.
[PMID: 31701486]
[3]
Sawyers C. Targeted cancer therapy. Nature 2004; 432(7015): 294-7.
[http://dx.doi.org/10.1038/nature03095] [PMID: 15549090]
[4]
Brown JM, Giaccia AJ. The unique physiology of solid tumors: opportunities (and problems) for cancer therapy. Cancer Res 1998; 58(7): 1408-16.
[PMID: 9537241]
[5]
Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev 2004; 56(11): 1649-59.
[http://dx.doi.org/10.1016/j.addr.2004.02.014] [PMID: 15350294]
[6]
Guillemard V, Saragovi HU. Novel approaches for targeted cancer therapy. Curr Cancer Drug Targets 2004; 4(4): 313-26.
[http://dx.doi.org/10.2174/1568009043332989] [PMID: 15180497]
[7]
Tennant DA, Durán RV, Gottlieb E. Targeting metabolic transformation for cancer therapy. Nat Rev Cancer 2010; 10(4): 267-77.
[http://dx.doi.org/10.1038/nrc2817] [PMID: 20300106]
[8]
Szakács G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006; 5(3): 219-34.
[http://dx.doi.org/10.1038/nrd1984] [PMID: 16518375]
[9]
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]
[10]
Gillet J-P, Gottesman MM. Mechanisms of multidrug resistance in cancer Multi-drug resistance in cancer. Springer 2010; pp. 47-76.
[http://dx.doi.org/10.1007/978-1-60761-416-6_4]
[11]
Ouyang L, Shi Z, Zhao S, et al. Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif 2012; 45(6): 487-98.
[http://dx.doi.org/10.1111/j.1365-2184.2012.00845.x] [PMID: 23030059]
[12]
Bianco R, Melisi D, Ciardiello F, Tortora G. Key cancer cell signal transduction pathways as therapeutic targets. Eur J Cancer 2006; 42(3): 290-4.
[http://dx.doi.org/10.1016/j.ejca.2005.07.034] [PMID: 16376541]
[13]
Ahmad MZ, Akhter S, Mohsin N, et al. Transformation of curcumin from food additive to multifunctional medicine: nanotechnology bridging the gap. Curr Drug Discov Technol 2014; 11(3): 197-213.
[http://dx.doi.org/10.2174/1570163811666140616153436] [PMID: 24934264]
[14]
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]
[15]
Mirzaei H, Masoudifar A, Sahebkar A, et al. MicroRNA: A novel target of curcumin in cancer therapy. J Cell Physiol 2018; 233(4): 3004-15.
[http://dx.doi.org/10.1002/jcp.26055] [PMID: 28617957]
[16]
Ahmad MZ, Alkahtani SA, Akhter S, et al. Progress in nanotechnology-based drug carrier in designing of curcumin nanomedicines for cancer therapy: current state-of-the-art. J Drug Target 2016; 24(4): 273-93.
[http://dx.doi.org/10.3109/1061186X.2015.1055570] [PMID: 26066739]
[17]
Tuttle S, Hertan L, Katz JS. Indian gold treating cancer in the age of nano. Cancer Biol Ther 2011; 11(5): 474-6.
[http://dx.doi.org/10.4161/cbt.11.5.14810] [PMID: 21263231]
[18]
Bisht S, Feldmann G, Soni S, et al. Polymeric nanoparticle-encapsulated curcumin (“nanocurcumin”): a novel strategy for human cancer therapy. J Nanobiotechnology 2007; 5(1): 3.
[http://dx.doi.org/10.1186/1477-3155-5-3] [PMID: 17439648]
[19]
Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol 2009; 41(1): 40-59.
[http://dx.doi.org/10.1016/j.biocel.2008.06.010] [PMID: 18662800]
[20]
Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies Anticancer Res 2003; 23(1/A): 363-98.
[21]
Basnet P, Skalko-Basnet N. Curcumin: an anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 2011; 16(6): 4567-98.
[http://dx.doi.org/10.3390/molecules16064567] [PMID: 21642934]
[22]
Wilken R, Veena MS, Wang MB, Srivatsan ES. Curcumin: A review of anti-cancer properties and therapeutic activity in head and neck squamous cell carcinoma. Mol Cancer 2011; 10(1): 12.
[http://dx.doi.org/10.1186/1476-4598-10-12] [PMID: 21299897]
[23]
Bar-Sela G, Epelbaum R, Schaffer M. Curcumin as an anti-cancer agent: review of the gap between basic and clinical applications. Curr Med Chem 2010; 17(3): 190-7.
[http://dx.doi.org/10.2174/092986710790149738] [PMID: 20214562]
[24]
Mahran RI, Hagras MM, Sun D, Brenner DE. Bringing curcumin to the clinic in cancer prevention: a review of strategies to enhance bioavailability and efficacy. AAPS J 2017; 19(1): 54-81.
[http://dx.doi.org/10.1208/s12248-016-0003-2] [PMID: 27783266]
[25]
Bansal SS, Goel M, Aqil F, Vadhanam MV, Gupta RC. Advanced drug delivery systems of curcumin for cancer chemoprevention. Cancer Prev Res (Phila) 2011; 4(8): 1158-71.
[http://dx.doi.org/10.1158/1940-6207.CAPR-10-0006] [PMID: 21546540]
[26]
Schluep T, Hwang J, Hildebrandt IJ, et al. Pharmacokinetics and tumor dynamics of the nanoparticle IT-101 from PET imaging and tumor histological measurements. Proc Natl Acad Sci USA 2009; 106(27): 11394-9.
[http://dx.doi.org/10.1073/pnas.0905487106] [PMID: 19564622]
[27]
Grabovac V, Bernkop-Schnürch A. Development and in vitro evaluation of surface modified poly(lactide-co-glycolide) nanoparticles with chitosan-4-thiobutylamidine. Drug Dev Ind Pharm 2007; 33(7): 767-74.
[http://dx.doi.org/10.1080/03639040601050163] [PMID: 17654025]
[28]
Jain A, Kumari R, Tiwari A, et al. Nanocarrier based advances in drug delivery to tumor: an overview. Curr Drug Targets 2018; 19(13): 1498-518.
[http://dx.doi.org/10.2174/1389450119666180131105822] [PMID: 29384060]
[29]
Sutton D, Nasongkla N, Blanco E, Gao J. Functionalized micellar systems for cancer targeted drug delivery. Pharm Res 2007; 24(6): 1029-46.
[http://dx.doi.org/10.1007/s11095-006-9223-y] [PMID: 17385025]
[30]
Singh R, Lillard JW Jr. Nanoparticle-based targeted drug delivery. Exp Mol Pathol 2009; 86(3): 215-23.
[http://dx.doi.org/10.1016/j.yexmp.2008.12.004] [PMID: 19186176]
[31]
Liu J, Liu J, Xu H, et al. Novel tumor-targeting, self-assembling peptide nanofiber as a carrier for effective curcumin delivery. Int J Nanomedicine 2014; 9: 197-207.
[PMID: 24399876]
[32]
Thangapazham RL, Sharma A, Maheshwari RK. Multiple molecular targets in cancer chemoprevention by curcumin. AAPS J 2006; 8(3): E443-9.
[http://dx.doi.org/10.1208/aapsj080352] [PMID: 17025261]
[33]
Willenbacher E, Khan SZ, Mujica SCA, et al. Curcumin: new insights into an ancient ingredient against cancer. Int J Mol Sci 2019; 20(8): 1808.
[http://dx.doi.org/10.3390/ijms20081808] [PMID: 31013694]
[34]
Salem M, Rohani S, Gillies ER. Curcumin, a promising anti-cancer therapeutic: a review of its chemical properties, bioactivity and approaches to cancer cell delivery. RSC Advances 2014; 4(21): 10815-29.
[http://dx.doi.org/10.1039/c3ra46396f]
[35]
Shehzad A, Lee YS. Molecular mechanisms of curcumin action: signal transduction. Biofactors 2013; 39(1): 27-36.
[http://dx.doi.org/10.1002/biof.1065] [PMID: 23303697]
[36]
Youns M, Fathy GM. Upregulation of extrinsic apoptotic pathway in curcumin-mediated antiproliferative effect on human pancreatic carcinogenesis. J Cell Biochem 2013; 114(12): 2654-65.
[http://dx.doi.org/10.1002/jcb.24612] [PMID: 23794119]
[37]
Han S-S, Keum Y-S, Seo H-J, Surh Y-J. Curcumin suppresses activation of NF-kappaB and AP-1 induced by phorbol ester in cultured human promyelocytic leukemia cells. J Biochem Mol Biol 2002; 35(3): 337-42.
[PMID: 12297018]
[38]
Balasubramanian S, Eckert RL. Curcumin suppresses AP1 transcription factor-dependent differentiation and activates apoptosis in human epidermal keratinocytes. J Biol Chem 2007; 282(9): 6707-15.
[http://dx.doi.org/10.1074/jbc.M606003200] [PMID: 17148446]
[39]
Zhou H, Beevers CS, Huang S. The targets of curcumin. Curr Drug Targets 2011; 12(3): 332-47.
[http://dx.doi.org/10.2174/138945011794815356] [PMID: 20955148]
[40]
Shehzad A, Wahid F, Lee YS. Curcumin in cancer chemoprevention: molecular targets, pharmacokinetics, bioavailability, and clinical trials. Arch Pharm (Weinheim) 2010; 343(9): 489-99.
[http://dx.doi.org/10.1002/ardp.200900319] [PMID: 20726007]
[41]
John PC, Mews M, Moore R. Cyclin/Cdk complexes: their involvement in cell cycle progression and mitotic division. Protoplasma 2001; 216(3-4): 119-42.
[http://dx.doi.org/10.1007/BF02673865] [PMID: 11732181]
[42]
Lim T-G, Lee S-Y, Huang Z, et al. Curcumin suppresses proliferation of colon cancer cells by targeting CDK2. Cancer Prev Res (Phila) 2014; 7(4): 466-74.
[http://dx.doi.org/10.1158/1940-6207.CAPR-13-0387] [PMID: 24550143]
[43]
Srivastava RK, Chen Q, Siddiqui I, Sarva K, Shankar S. Linkage of curcumin-induced cell cycle arrest and apoptosis by cyclin-dependent kinase inhibitor p21(/WAF1/CIP1). Cell Cycle 2007; 6(23): 2953-61.
[http://dx.doi.org/10.4161/cc.6.23.4951] [PMID: 18156803]
[44]
Limtrakul P, Anuchapreeda S, Lipigorngoson S, Dunn FW. Inhibition of carcinogen induced c-Ha-ras and c-fos proto-oncogenes expression by dietary curcumin. BMC Cancer 2001; 1(1): 1.
[http://dx.doi.org/10.1186/1471-2407-1-1] [PMID: 11231886]
[45]
Qiao Q, Jiang Y, Li G. Inhibition of the PI3K/AKT-NF-κB pathway with curcumin enhanced radiation-induced apoptosis in human Burkitt’s lymphoma. J Pharmacol Sci 2013; 121(4): 247-56.
[http://dx.doi.org/10.1254/jphs.12149FP] [PMID: 23603894]
[46]
Tiwari A, Saraf S, Verma A, Panda PK, Jain SK. Novel targeting approaches and signaling pathways of colorectal cancer: An insight. World J Gastroenterol 2018; 24(39): 4428-35.
[http://dx.doi.org/10.3748/wjg.v24.i39.4428] [PMID: 30357011]
[47]
Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene 2017; 36(11): 1461-73.
[http://dx.doi.org/10.1038/onc.2016.304] [PMID: 27617575]
[48]
Dou H, Shen R, Tao J, et al. Curcumin suppresses the colon cancer proliferation by inhibiting Wnt/β-catenin pathways via miR-130a. Front Pharmacol 2017; 8: 877.
[http://dx.doi.org/10.3389/fphar.2017.00877] [PMID: 29225578]
[49]
Thacker PC, Karunagaran D. Curcumin and emodin down-regulate TGF-β signaling pathway in human cervical cancer cells. PLoS One 2015; 10(3)e0120045
[http://dx.doi.org/10.1371/journal.pone.0120045] [PMID: 25786122]
[50]
Lavecchia A, Di Giovanni C, Novellino E. STAT-3 inhibitors: state of the art and new horizons for cancer treatment. Curr Med Chem 2011; 18(16): 2359-75.
[http://dx.doi.org/10.2174/092986711795843218] [PMID: 21568920]
[51]
Blasius R, Reuter S, Henry E, Dicato M, Diederich M. Curcumin regulates signal transducer and activator of transcription (STAT) expression in K562 cells. Biochem Pharmacol 2006; 72(11): 1547-54.
[http://dx.doi.org/10.1016/j.bcp.2006.07.029] [PMID: 16959222]
[52]
Chakraborty G, Jain S, Kale S, et al. Curcumin suppresses breast tumor angiogenesis by abrogating osteopontin-induced VEGF expression. Mol Med Rep 2008; 1(5): 641-6.
[http://dx.doi.org/10.3892/mmr_00000005] [PMID: 21479462]
[53]
Radhakrishnan VM, Kojs P, Young G, et al. pTyr421 cortactin is overexpressed in colon cancer and is dephosphorylated by curcumin: involvement of non-receptor type 1 protein tyrosine phosphatase (PTPN1). PLoS One 2014; 9(1)e85796
[http://dx.doi.org/10.1371/journal.pone.0085796] [PMID: 24465712]
[54]
Killian PH, Kronski E, Michalik KM, et al. Curcumin inhibits prostate cancer metastasis in vivo by targeting the inflammatory cytokines CXCL1 and -2. Carcinogenesis 2012; 33(12): 2507-19.
[http://dx.doi.org/10.1093/carcin/bgs312] [PMID: 23042094]
[55]
Park W, Amin AR, Chen ZG, Shin DM. New perspectives of curcumin in cancer prevention. Cancer Prev Res (Phila) 2013; 6(5): 387-400.
[http://dx.doi.org/10.1158/1940-6207.CAPR-12-0410] [PMID: 23466484]
[56]
Saraf S, Jain A, Tiwari A, Verma A, Panda PK, Jain SK. Advances in liposomal drug delivery to cancer: An overview. J Drug Deliv Sci Technol 2020.101549
[http://dx.doi.org/10.1016/j.jddst.2020.101549]
[57]
Yallapu MM, Jaggi M, Chauhan SC. Curcumin nanoformulations: a future nanomedicine for cancer. Drug Discov Today 2012; 17(1-2): 71-80.
[http://dx.doi.org/10.1016/j.drudis.2011.09.009] [PMID: 21959306]
[58]
Zhang X, Dai F, Chen J, et al. Antitumor effect of curcumin liposome after transcatheter arterial embolization in VX2 rabbits. Cancer Biol Ther 2019; 20(5): 642-52.
[http://dx.doi.org/10.1080/15384047.2018.1550567] [PMID: 30621501]
[59]
Sun D, Zhou J-K, Zhao L, et al. Novel curcumin liposome modified with hyaluronan targeting CD44 plays an anti-leukemic role in acute myeloid leukemia in vitro and in vivo. ACS Appl Mater Interfaces 2017; 9(20): 16857-68.
[http://dx.doi.org/10.1021/acsami.7b02863] [PMID: 28489348]
[60]
Perche F, Torchilin VP. Recent trends in multifunctional liposomal nanocarriers for enhanced tumor targeting. J Drug Delivery 2013; 2013.
[http://dx.doi.org/10.1155/2013/705265]
[61]
Sesarman A, Tefas L, Sylvester B, et al. Co-delivery of curcumin and doxorubicin in PEGylated liposomes favored the antineoplastic C26 murine colon carcinoma microenvironment. Drug Deliv Transl Res 2019; 9(1): 260-72.
[http://dx.doi.org/10.1007/s13346-018-00598-8] [PMID: 30421392]
[62]
Jiang H, Li Z-P, Tian G-X, et al. Liver-targeted liposomes for codelivery of curcumin and combretastatin A4 phosphate: preparation, characterization, and antitumor effects. Int J Nanomedicine 2019; 14: 1789-804.
[http://dx.doi.org/10.2147/IJN.S188971] [PMID: 30880980]
[63]
Lu Y, Ding N, Yang C, Huang L, Liu J, Xiang G. Preparation and in vitro evaluation of a folate-linked liposomal curcumin formulation. J Liposome Res 2012; 22(2): 110-9.
[http://dx.doi.org/10.3109/08982104.2011.627514] [PMID: 22372871]
[64]
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]
[65]
Kakkar V, Singh S, Singla D, Kaur IP. Exploring solid lipid nanoparticles to enhance the oral bioavailability of curcumin. Mol Nutr Food Res 2011; 55(3): 495-503.
[http://dx.doi.org/10.1002/mnfr.201000310] [PMID: 20938993]
[66]
Guri A, Gülseren I, Corredig M. Utilization of solid lipid nanoparticles for enhanced delivery of curcumin in cocultures of HT29-MTX and Caco-2 cells. Food Funct 2013; 4(9): 1410-9.
[http://dx.doi.org/10.1039/c3fo60180c] [PMID: 23921424]
[67]
Wang W, Chen T, Xu H, et al. Curcumin-loaded solid lipid nanoparticles enhanced anticancer efficiency in breast cancer. Molecules 2018; 23(7): 1578.
[http://dx.doi.org/10.3390/molecules23071578] [PMID: 29966245]
[68]
Saralkar P, Dash AK. Alginate nanoparticles containing curcumin and resveratrol: preparation, characterization, and in vitro evaluation against DU145 prostate cancer cell line. AAPS PharmSciTech 2017; 18(7): 2814-23.
[http://dx.doi.org/10.1208/s12249-017-0772-7] [PMID: 28397161]
[69]
Zhang J, Li J, Shi Z, et al. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater 2017; 58: 349-64.
[http://dx.doi.org/10.1016/j.actbio.2017.04.029] [PMID: 28455219]
[70]
Gianella A, Jarzyna PA, Mani V, et al. Multifunctional nanoemulsion platform for imaging guided therapy evaluated in experimental cancer. ACS Nano 2011; 5(6): 4422-33.
[http://dx.doi.org/10.1021/nn103336a] [PMID: 21557611]
[71]
Ahmed K, Li Y, McClements DJ, Xiao H. Nanoemulsion-and emulsion-based delivery systems for curcumin: Encapsulation and release properties. Food Chem 2012; 132(2): 799-807.
[http://dx.doi.org/10.1016/j.foodchem.2011.11.039] [PMID: 22868161]
[72]
Guan YB, Zhou SY, Zhang YQ, et al. Therapeutic effects of curcumin nanoemulsions on prostate cancer. J Huazhong Univ Sci Technolog Med Sci 2017; 37(3): 371-8.
[http://dx.doi.org/10.1007/s11596-017-1742-8] [PMID: 28585133]
[73]
De Matos RPA, Calmon MF, Amantino CF, et al. Effect of curcumin-nanoemulsion associated with photodynamic therapy in cervical carcinoma cell lines BioMed research international 2018 2018.
[http://dx.doi.org/10.1155/2018/4057959]
[74]
Guerrero S, Inostroza-Riquelme M, Contreras-Orellana P, et al. Curcumin-loaded nanoemulsion: a new safe and effective formulation to prevent tumor reincidence and metastasis. Nanoscale 2018; 10(47): 22612-22.
[http://dx.doi.org/10.1039/C8NR06173D] [PMID: 30484463]
[75]
Akay C, Özkan SA. Simultaneous LC determination of trimethoprim and sulphamethoxazole in pharmaceutical formulations. J Pharm Biomed Anal 2002; 30(4): 1207-13.
[http://dx.doi.org/10.1016/S0731-7085(02)00460-0] [PMID: 12408911]
[76]
Shukla M, Jaiswal S, Sharma A, et al. A combination of complexation and self-nanoemulsifying drug delivery system for enhancing oral bioavailability and anticancer efficacy of curcumin. Drug Dev Ind Pharm 2017; 43(5): 847-61.
[http://dx.doi.org/10.1080/03639045.2016.1239732] [PMID: 27648633]
[77]
Ganta S, Amiji M. Coadministration of Paclitaxel and curcumin in nanoemulsion formulations to overcome multidrug resistance in tumor cells. Mol Pharm 2009; 6(3): 928-39.
[http://dx.doi.org/10.1021/mp800240j] [PMID: 19278222]
[78]
Varde NK, Pack DW. Microspheres for controlled release drug delivery. Expert Opin Biol Ther 2004; 4(1): 35-51.
[http://dx.doi.org/10.1517/14712598.4.1.35] [PMID: 14680467]
[79]
Edlund U, Albertsson A-C. Degradable polymer microspheres for controlled drug delivery Degradable aliphatic polyesters. Springer 2002; pp. 67-112.
[http://dx.doi.org/10.1007/3-540-45734-8_3]
[80]
Deng X-Q, Zhang H-B, Wang G-F, et al. Colon-specific microspheres loaded with puerarin reduce tumorigenesis and metastasis in colitis-associated colorectal cancer. Int J Pharm 2019.570118644
[http://dx.doi.org/10.1016/j.ijpharm.2019.118644] [PMID: 31465837]
[81]
Jyoti K, Bhatia RK, Martis EAF, et al. Soluble curcumin amalgamated chitosan microspheres augmented drug delivery and cytotoxicity in colon cancer cells: In vitro and in vivo study. Colloids Surf B Biointerfaces 2016; 148: 674-83.
[http://dx.doi.org/10.1016/j.colsurfb.2016.09.044] [PMID: 27701049]
[82]
Pal K, Roy S, Parida PK, et al. Folic acid conjugated curcumin loaded biopolymeric gum acacia microsphere for triple negative breast cancer therapy in invitro and invivo model. Mater Sci Eng C 2019; 95: 204-16.
[http://dx.doi.org/10.1016/j.msec.2018.10.071] [PMID: 30573243]
[83]
Fan R, Li X, Deng J, et al. Dual drug loaded biodegradable nanofibrous microsphere for improving anti-colon cancer activity. Sci Rep 2016; 6(1): 28373.
[http://dx.doi.org/10.1038/srep28373] [PMID: 27324595]
[84]
Gong J, Chen M, Zheng Y, Wang S, Wang Y. Polymeric micelles drug delivery system in oncology. J Control Release 2012; 159(3): 312-23.
[http://dx.doi.org/10.1016/j.jconrel.2011.12.012] [PMID: 22285551]
[85]
Kwon GS, Okano T. Polymeric micelles as new drug carriers. Adv Drug Deliv Rev 1996; 21(2): 107-16.
[http://dx.doi.org/10.1016/S0169-409X(96)00401-2]
[86]
Woraphatphadung T, Sajomsang W, Rojanarata T, Ngawhirunpat T, Tonglairoum P, Opanasopit P. Development of chitosan-based pH-sensitive polymeric micelles containing curcumin for colon-targeted drug delivery. AAPS PharmSciTech 2018; 19(3): 991-1000.
[http://dx.doi.org/10.1208/s12249-017-0906-y] [PMID: 29110292]
[87]
Le TT, Kim D. Folate-PEG/Hyd-curcumin/C18-g-PSI micelles for site specific delivery of curcumin to colon cancer cells via Wnt/β-catenin signaling pathway. Mater Sci Eng C 2019; 101: 464-71.
[http://dx.doi.org/10.1016/j.msec.2019.03.100] [PMID: 31029341]
[88]
An Q, Shi C-X, Guo H, et al. Development and characterization of octreotide-modified curcumin plus docetaxel micelles for potential treatment of non-small-cell lung cancer. Pharm Dev Technol 2019; 24(9): 1164-74.
[http://dx.doi.org/10.1080/10837450.2019.1647236] [PMID: 31340709]
[89]
Tima S, Okonogi S, Ampasavate C, Berkland C, Anuchapreeda S. FLT3-specific curcumin micelles enhance activity of curcumin on FLT3-ITD overexpressing MV4-11 leukemic cells. Drug Dev Ind Pharm 2019; 45(3): 498-505.
[http://dx.doi.org/10.1080/03639045.2018.1562462] [PMID: 30572745]
[90]
Shafabakhsh R, Pourhanifeh MH, Mirzaei HR, Sahebkar A, Asemi Z, Mirzaei H. Targeting regulatory T cells by curcumin: A potential for cancer immunotherapy. Pharmacol Res 2019; 147104353
[http://dx.doi.org/10.1016/j.phrs.2019.104353] [PMID: 31306775]
[91]
Joe B, Lokesh BR. Role of capsaicin, curcumin and dietary n-3 fatty acids in lowering the generation of reactive oxygen species in rat peritoneal macrophages. Biochim Biophys Acta 1994; 1224(2): 255-63.
[http://dx.doi.org/10.1016/0167-4889(94)90198-8] [PMID: 7981240]
[92]
Kim K, Ryu K, Ko Y, Park C. Effects of nuclear factor-kappaB inhibitors and its implication on natural killer T-cell lymphoma cells. Br J Haematol 2005; 131(1): 59-66.
[http://dx.doi.org/10.1111/j.1365-2141.2005.05720.x] [PMID: 16173963]
[93]
Kim G-Y, Kim K-H, Lee S-H, et al. Curcumin inhibits immunostimulatory function of dendritic cells: MAPKs and translocation of NF-κ B as potential targets. J Immunol 2005; 174(12): 8116-24.
[http://dx.doi.org/10.4049/jimmunol.174.12.8116] [PMID: 15944320]
[94]
Han S-S, Chung S-T, Robertson DA, Ranjan D, Bondada S. Curcumin causes the growth arrest and apoptosis of B cell lymphoma by downregulation of egr-1, c-myc, bcl-XL, NF-κ B, and p53. Clin Immunol 1999; 93(2): 152-61.
[http://dx.doi.org/10.1006/clim.1999.4769] [PMID: 10527691]
[95]
Ghosh AK, Kay NE, Secreto CR, Shanafelt TD. Curcumin inhibits prosurvival pathways in chronic lymphocytic leukemia B cells and may overcome their stromal protection in combination with EGCG. Clin Cancer Res 2009; 15(4): 1250-8.
[http://dx.doi.org/10.1158/1078-0432.CCR-08-1511] [PMID: 19228728]
[96]
Ranjan D, Siquijor A, Johnston TD, Wu G, Nagabhuskahn M. The effect of curcumin on human B-cell immortalization by Epstein-Barr virus. Am Surg 1998; 64(1): 47-51.
[PMID: 9457037]
[97]
Wang Q, Redovan C, Tubbs R, et al. Selective cytokine gene expression in renal cell carcinoma tumor cells and tumor-infiltrating lymphocytes. Int J Cancer 1995; 61(6): 780-5.
[http://dx.doi.org/10.1002/ijc.2910610607] [PMID: 7790111]
[98]
Woo EY, Chu CS, Goletz TJ, et al. Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res 2001; 61(12): 4766-72.
[PMID: 11406550]
[99]
Parmiani G, Rivoltini L, Andreola G, Carrabba M. Cytokines in cancer therapy. Immunol Lett 2000; 74(1): 41-4.
[http://dx.doi.org/10.1016/S0165-2478(00)00247-9] [PMID: 10996626]
[100]
Churchill M, Chadburn A, Bilinski RT, Bertagnolli MM. Inhibition of intestinal tumors by curcumin is associated with changes in the intestinal immune cell profile. J Surg Res 2000; 89(2): 169-75.
[http://dx.doi.org/10.1006/jsre.2000.5826] [PMID: 10729246]
[101]
Bose S, Panda AK, Mukherjee S, Sa G. Curcumin and tumor immune-editing: resurrecting the immune system. Cell Div 2015; 10(1): 6.
[http://dx.doi.org/10.1186/s13008-015-0012-z] [PMID: 26464579]
[102]
Zhao GJ, Lu ZQ, Tang LM, et al. Curcumin inhibits suppressive capacity of naturally occurring CD4+CD25+ regulatory T cells in mice in vitro. Int Immunopharmacol 2012; 14(1): 99-106.
[http://dx.doi.org/10.1016/j.intimp.2012.06.016] [PMID: 22749847]
[103]
Hossain DMS, Panda AK, Manna A, et al. FoxP3 acts as a cotranscription factor with STAT3 in tumor-induced regulatory T cells. Immunity 2013; 39(6): 1057-69.
[http://dx.doi.org/10.1016/j.immuni.2013.11.005] [PMID: 24315995]
[104]
Bhattacharyya S, Md Sakib Hossain D, Mohanty S, et al. Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts. Cell Mol Immunol 2010; 7(4): 306-15.
[http://dx.doi.org/10.1038/cmi.2010.11] [PMID: 20305684]
[105]
Bhattacharyya S, Mandal D, Saha B, Sen GS, Das T, Sa G. Curcumin prevents tumor-induced T cell apoptosis through Stat-5a-mediated Bcl-2 induction. J Biol Chem 2007; 282(22): 15954-64.
[http://dx.doi.org/10.1074/jbc.M608189200] [PMID: 17392282]
[106]
Bhattacharyya S, Mandal D, Sen GS, et al. Tumor-induced oxidative stress perturbs nuclear factor-kappaB activity-augmenting tumor necrosis factor-α-mediated T-cell death: protection by curcumin. Cancer Res 2007; 67(1): 362-70.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-2583] [PMID: 17210719]
[107]
Luo F, Song X, Zhang Y, Chu Y. Low-dose curcumin leads to the inhibition of tumor growth via enhancing CTL-mediated antitumor immunity. Int Immunopharmacol 2011; 11(9): 1234-40.
[http://dx.doi.org/10.1016/j.intimp.2011.04.002] [PMID: 21497674]
[108]
Hatcher H, Planalp R, Cho J, Torti FM, Torti SV. Curcumin: from ancient medicine to current clinical trials. Cell Mol Life Sci 2008; 65(11): 1631-52.
[http://dx.doi.org/10.1007/s00018-008-7452-4] [PMID: 18324353]
[109]
Ryan JL, Heckler CE, Ling M, et al. Curcumin for radiation dermatitis: a randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer patients. Radiat Res 2013; 180(1): 34-43.
[http://dx.doi.org/10.1667/RR3255.1] [PMID: 23745991]
[110]
NCT01740323. Miller AH. Phase II study of curcumin vs placebo for chemotherapy-treated breast cancer patients undergoing radiotherapy. Available from https://clinicaltrials.gov/ct2/show/NCT01740323
[111]
NCT02556632 Prophylactic topical agents in reducing radiation-induced dermatitis in patients with non-inflammatory breast cancer Available from: https://clinicaltrials.gov/ct2/show/NCT02556632
[112]
Cruz-Correa M, Hylind LM, Marrero JH, et al. Efficacy and safety of curcumin in treatment of intestinal adenomas in patients with familial adenomatous polyposis. Gastroenterology 2018; 155(3): 668-73.
[http://dx.doi.org/10.1053/j.gastro.2018.05.031] [PMID: 29802852]
[113]
NCT00113841. Curcumin (Diferuloylmethane Derivative) with or without bioperine in patients with multiple myeloma Available from: https://clinicaltrials.gov/ct2/show/NCT00113841
[114]
NCT00365209. Phase II A trial of curcumin among patients with prevalent subclinical neoplastic lesions (Aberrant Crypt Foci). Available from: https://clinicaltrials.gov/ct2/show/NCT00365209]

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