We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Published Online:https://doi.org/10.2217/nnm.10.9

Aim: To develop a suitable formulation of curcumin-encapsulated methoxy poly(ethylene glycol) (MePEG)/poly-ε-caprolactone (PCL) diblock copolymeric micelle by varying the copolymer ratio, for achieving small sized micelles with high encapsulation of curcumin. To evaluate the micelle’s aqueous solubility and stability, efficiency of cellular uptake, cell cytotoxicity and ability to induce apoptosis on pancreatic cell lines. Method: Amphiphilic diblock copolymers (composed of MePEG and PCL) were used in various ratios for the preparation of curcumin-encapsulated micelles using a modified dialysis method. Physicochemical characterization of the formulation included size and surface charge measurement, transmission electron microscopy characterization, spectroscopic analysis, stability and in vitro release kinetics studies. The anticancer efficacy of the curcumin-encapsulated micelle formulation was compared with unmodified curcumin in terms of cellular uptake, cell cytotoxicity and apoptosis of pancreatic cell lines MIA PaCa-2 and PANC-1. Results: Physiochemical characterization of the formulations revealed that curcumin was efficiently encapsulated in all formulation of MePEG/PCL micelles; however, a 40:60 MePEG:PCL ratio micelle was chosen for experimental studies owing to its high encapsulation (∼60%) with size (∼110 nm) and negative ζ potential (∼-16 mV). Curcumin-encapsulated micelles increased the bioavailability of curcumin due to enhanced uptake (2.95 times more compared with unmodified) with comparative cytotoxic activity (by induction of apoptosis) compared with unmodified curcumin at equimolar concentrations. IC50 values for unmodified curcumin and curcumin micelles were found to be 24.75 µM and 22.8 µM for PANC-1 and 14.96 µM and 13.85 µM for MIA PaCa-2, respectively. Together the results clearly indicate the promise of a micellar system for efficient solubilization, stabilization and controlled delivery of the hydrophobic drug curcumin for cancer therapy.

Bibliography

  • Erkan M, Kleeff J, Esposito I et al.: Loss of bnip3 expression is a late event in pancreatic cancer contributing to chemoresistance and worsened prognosis. Oncogene24(27),4421–4432 (2005).
  • Fryer RA, Galustian C, Dalgelish AG: Recent advances and developments in treatment strategies against pancreatic cancer. Curr. Clin. Pharmacol.4(2),102–112 (2009).
  • Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB: Bioavailability of curcumin: problems and promises. Mol. Pharm.4(6),807–818 (2007).
  • Somasundaram S, Edmund NA, Moore DT, Small GW, Shi YY, Orlowski RZ: Dietary curcumin inhibits chemotherapy-induced apoptosis in human models of human breast cancer. Cancer Res.62(13),3868–3875 (2002).
  • Aggarwal BB, Kumar A, Bharti AC: Anticancer potential of curcumin: preclinical and clinical studies. Anticancer Res.23(1A),363–398 (2003).
  • Aggarwal BB, Sung B: Pharmacological basis for the role of curcumin in chronic diseases: an age-old spice with modern targets. Trends Pharmacol. Sci.30(2),85–94 (2009).
  • Cohen AN, Veena MS, Srivatsan ES, Wang MB: Suppression of interleukin 6 and 8 production in head and neck cancer cells with curcumin via inhibition of iκβ kinase. Arch. Otolaryngol. Head Neck Surg.135(2),190–197 (2009).
  • Farombi EO, Shrotriya S, Surh YJ: Kolaviron inhibits dimethyl nitrosamine-induced liver injury by suppressing cox-2 and inos expression via NF-κB and AP-1. Life Sci.84(5–6),149–155 (2009).
  • Owens DE 3rd, Peppas NA: Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm.307(1),93–102 (2006).
  • 10  Bisht S, Feldmann G, Soni S et al.: Polymeric nanoparticle-encapsulated curcumin (“Nanocurcumin”): a novel strategy for human cancer therapy. J. Nanobiotechnology5,3 (2007).
  • 11  Suresh D, Srinivasan K: Studies on the in vitro absorption of spice principles – curcumin, capsaicin and piperine in rat intestines. Food Chem. Toxicol.45(8),1437–1442 (2007).
  • 12  Ma Z, Haddadi A, Molavi O, Lavasanifar A, Lai R, Samuel J: Micelles of poly(ethylene oxide)-b-poly(ε-caprolactone) as vehicles for the solubilization, stabilization, and controlled delivery of curcumin. J. Biomed. Mater. Res. A86(2),300–310 (2008).
  • 13  Sahu A, Kasoju N, Bora U: Fluorescence study of the curcumin-casein micelle complexation and its application as a drug nanocarrier to cancer cells. Biomacromolecules9(10),2905–2912 (2008).
  • 14  Torchilin VP: Structure and design of polymeric surfactant-based drug delivery systems. J. Control Release73(2–3),137–172 (2001).
  • 15  Lavasanifar A, Samuel J, Kwon GS: Poly(ethylene oxide)-block-poly(l-amino acid) micelles for drug delivery. Adv. Drug Deliv. Rev.54(2),169–190 (2002).
  • 16  Creamer LK, Plowman JE, Liddell MJ, Smith MH, Hill JP: Micelle stability: κ-casein structure and function. J. Dairy Sci.81(11),3004–3012 (1998).
  • 17  Park EK, Lee SB, Lee YM: Preparation and characterization of methoxy poly(ethylene glycol)/poly(ε-caprolactone) amphiphilic block copolymeric nanospheres for tumor-specific folate-mediated targeting of anticancer drugs. Biomaterials26(9),1053–1061 (2005).
  • 18  Torchilin VP: Micellar nanocarriers: pharmaceutical perspectives. Pharm. Res.24(1),1–16 (2007).
  • 19  Berrada M, Serreqi A, Dabbarh F, Owusu A, Gupta A, Lehnert S: A novel non-toxic camptothecin formulation for cancer chemotherapy. Biomaterials26(14),2115–2120 (2005).
  • 20  Letchford K, Zastre J, Liggins R, Burt H: Synthesis and micellar characterization of short block length methoxy poly(ethylene glycol)-block-poly(caprolactone) diblock copolymers. Colloids Surf. B Biointerfaces35(2),81–91 (2004).
  • 21  Mohanty AK, Dilnawaz F, Sahoo SK: A biodegradable amphiphilic methoxy (poly ethylene glycol) and poly (ε caprolactone) copolymeric micelles loaded with etoposide as drug delivery vehicle for pancreatic cell line. Drug Delivery (2010) (In press).
  • 22  Zhou S, Deng X, Yang H: Biodegradable poly(ε-caprolactone)-poly(ethylene glycol) block copolymers: characterization and their use as drug carriers for a controlled delivery system. Biomaterials24(20),3563–3570 (2003).
  • 23  Bong PH: Spectral and photophysical behaviors of curcumin and curcuminoids. Bull. Korean Chem. Soc.21(1),81–86 (2000).
  • 24  Chignell CF, Bilski P, Reszka KJ, Motten AG, Sik RH, Dahl TA: Spectral and photochemical properties of curcumin. Photochem. Photobiol.59(3),295–302 (1994).
  • 25  Khopde SM, Priyadarsini KI, Palit DK, Mukherjee T: Effect of solvent on the excited-state photophysical properties of curcumin. Photochem. Photobiol.72(5),625–631 (2000).
  • 26  Misra R, Acharya S, Dilnawaz F, Sahoo SK: Sustained antibacterial activity of doxycycline-loaded poly(D,L-lactide-co-glycolide) and poly(ε-caprolactone) nanoparticles. Nanomedicine (Lond.)4(5),519–530 (2009).
  • 27  Acharya S, Dilnawaz F, Sahoo SK: Targeted epidermal growth factor receptor nanoparticle bioconjugates for breast cancer therapy. Biomaterials30(29),5737–5750 (2009).
  • 28  Sahoo SK, Ma W, Labhasetwar V: Efficacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer. Int. J. Cancer112(2),335–340 (2004).
  • 29  Kunwar A, Barik A, Mishra B, Rathinasamy K, Pandey R, Priyadarsini KI: Quantitative cellular uptake, localization and cytotoxicity of curcumin in normal and tumor cells. Biochim. Biophys. Acta1780(4),673–679 (2008).
  • 30  Lev-Ari S, Vexler A, Starr A et al.: Curcumin augments gemcitabine cytotoxic effect on pancreatic adenocarcinoma cell lines. Cancer Invest.25(6),411–418 (2007).
  • 31  Shin IG, Kim SY, Lee YM, Cho CS, Sung YK: Methoxy poly(ethylene glycol)/ε-caprolactone amphiphilic block copolymeric micelle containing indomethacin. I. Preparation and characterization. J. Control Release51(1),1–11 (1998).
  • 32  Shuai X, Ai H, Nasongkla N, Kim S, Gao J: Micellar carriers based on block copolymers of poly(ε-caprolactone) and poly(ethylene glycol) for doxorubicin delivery. J. Control Release98(3),415–426 (2004).
  • 33  Kim SY, Shin IG, Lee YM, Cho CS, Sung YK: Methoxy poly(ethylene glycol) and ε-caprolactone amphiphilic block copolymeric micelle containing indomethacin. II. Micelle formation and drug release behaviours. J. Control Release51(1),13–22 (1998).
  • 34  Yokoyama M, Okano T, Sakurai Y, Ekimoto H, Shibazaki C, Kataoka K: Toxicity and antitumor activity against solid tumors of micelle-forming polymeric anticancer drug and its extremely long circulation in blood. Cancer Res.51(12),3229–3236 (1991).
  • 35  Maeda H, Wu J, Sawa T, Matsumura Y, Hori K: Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J. Control Release65(1–2),271–284 (2000).
  • 36  Kim SY, Shin IG, Lee YM: Preparation and characterization of biodegradable nanospheres composed of methoxy poly(ethylene glycol) and dl-lactide block copolymer as novel drug carriers. J. Control Release56(1–3),197–208 (1998).
  • 37  Baimark Y: Surfactant-free nanospheres of methoxy poly (ethylene glycol)-b-poly (ε-caprolactone) for controlled release of ibuprofen. J. Appl. Sci.9(12),2287–2293 (2009).
  • 38  Yueying H, Yan Z, Chunhua G, Weifeng D, Meidong L: Micellar carrier based on methoxy poly(ethylene glycol)-block-poly(ε-caprolactone) block copolymers bearing ketone groups on the polyester block for doxorubicin delivery. J. Mater. Sci. Mater. Med.21(2),567–574 (2010).
  • 39  Adhikary R, Mukherjee P, Kee TW, Petrich JW: Excited-state intramolecular hydrogen atom transfer and solvation dynamics of the medicinal pigment curcumin. J. Phys. Chem. B113(15),5255–5261 (2009).
  • 40  Leung MH, Kee TW: Effective stabilization of curcumin by association to plasma proteins: human serum albumin and fibrinogen. Langmuir25(10),5773–5777 (2009).
  • 41  Wang YJ, Pan MH, Cheng AL et al.: Stability of curcumin in buffer solutions and characterization of its degradation products. J. Pharm. Biomed. Anal.15(12),1867–1876 (1997).
  • 42  Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MN: Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur. J. Pharm. Sci.37(3–4),223–230 (2009).
  • 43  Lev-Ari S, Strier L, Kazanov D et al.: Curcumin synergistically potentiates the growth-inhibitory and pro-apoptotic effects of celecoxib in osteoarthritis synovial adherent cells. Rheumatology (Oxford)45(2),171–177 (2006).
  • 44  Lev-Ari S, Strier L, Kazanov D et al.: Celecoxib and curcumin synergistically inhibit the growth of colorectal cancer cells. Clin. Cancer Res.11(18),6738–6744 (2005).
  • 45  Wang YZ, Fang XL, Li YJ, Zhang ZW, Han LM, Sha XY: preparation, characterization of paclitaxel-loaded pluronic p105 polymeric micelles and in vitro reversal of multidrug resistant tumor. Yao Xue Xue Bao43(6),640–646 (2008).
  • 46  Ganta S, Amiji M: Coadministration of paclitaxel and curcumin in nanoemulsion formulations to overcome multidrug resistance in tumor cells. Mol. Pharm.6(3),928–939 (2009).
  • 47  Jiang H, Deng CS, Zhang M, Xia J: Curcumin-attenuated trinitrobenzene sulphonic acid induces chronic colitis by inhibiting expression of cyclooxygenase-2. World J. Gastroenterol.12(24),3848–3853 (2006).
  • 48  Momand J, Zambetti GP, Olson DC, George D, Levine AJ: The MDM-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell69(7),1237–1245 (1992).
  • 49  Rayburn ER, Zhang R: Antisense, RNAi, and gene silencing strategies for therapy: mission possible or impossible? Drug Discov. Today13(11–12),513–521 (2008).
  • 50  Wang H, Yu D, Agrawal S, Zhang R: Experimental therapy of human prostate cancer by inhibiting MDM2 expression with novel mixed-backbone antisense oligonucleotides: in vitro and in vivo activities and mechanisms. Prostate54(3),194–205 (2003).
  • 51  Zhang Z, Zhang R: p53-independent activities of MDM2 and their relevance to cancer therapy. Curr. Cancer Drug Targets5(1),9–20 (2005).
  • 52  Wang W, Rayburn ER, Zhao Y, Wang H, Zhang R: Novel ginsenosides 25-OH-PPD and 25-OCH3-PPD as experimental therapy for pancreatic cancer: anticancer activity and mechanisms of action. Cancer Lett.278(2),241–248 (2009).
  • 53  Chen HW, Huang HC: Effect of curcumin on cell cycle progression and apoptosis in vascular smooth muscle cells. Br. J. Pharmacol.124(6),1029–1040 (1998).
  • 54  Hermeking H, Eick D: Mediation of c-myc-induced apoptosis by p53. Science265(5181),2091–2093 (1994).
  • 55  Sakamuro D, Eviner V, Elliott KJ, Showe L, White E, Prendergast GC: C-Myc induces apoptosis in epithelial cells by both p53-dependent and p53-independent mechanisms. Oncogene11(11),2411–2418 (1995).