Please wait a minute...
Frontiers of Chemical Science and Engineering

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

邮发代号 80-969

2019 Impact Factor: 3.552

Frontiers of Chemical Science and Engineering  2016, Vol. 10 Issue (3): 383-388   https://doi.org/10.1007/s11705-016-1574-2
  本期目录
Curcumin based combination therapy for anti-breast cancer: from in vitro drug screening to in vivo efficacy evaluation
Sunhui Chen1,Qiuling Liang1,Shuping Xie1,Ergang Liu2,Zhili Yu1,Lu Sun1,Meong Cheol Shin3,Seung Jin Lee4,Huining He1,*(),Victor C. Yang1,5,6,*()
1. Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
2. Collaborative Innovation Center of Chemical Science and Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
3. College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinjudaero, Jinju, Gyeongnam 660-751, Republic of Korea
4. Department of Pharmacy, Ewha Womans University, 11-1 Daehyun-dong, Seodaemun-gu, Seoul 120-750, Republic of Korea
5. Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor 48109-1065, MI, USA
6. Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
 全文: PDF(223 KB)   HTML
文章导读  
Abstract

While drug resistance appears to be an inevitable problem of an increasing number of anticancer drugs in monotherapy, combination drug therapy has become a prosperous method to reduce the administered total drug dosages as well as overcome the drug resistance of carcinoma cells. Curcumin, considered to possess multi-faceted roles in cancer treatment according to its multiple anti-neoplastic mechanisms as a depressor of chemo-resistance, can significantly facilitate its anti-cancer functions and improve therapeutic effects via combination usage with a variety of other drugs with different reaction mechanisms. To explore this possibility, four anti-cancer chemotherapeutic agents that all possess a certain degree of drug resistance problems, including three tyrosine kinase inhibitors (erlotinib, sunitinib and sorafenib) that are acting on different cell pathways and a typical anticancer drug doxorubicin, were combined with curcumin individually to examine the synergistic anti-tumor effect both in vitro and in vivo. Results revealed that sunitinib combined with curcumin at the molar ratio of 0.46 yielded the most potent synergistic effect in vitro, and was therefore chosen for further animal evaluation. To further enhance the anti-cancer effect, bovine serum albumin (BSA) nanoparticles were utilized as a carrier to deliver the selected drug combination in situ. Preliminary in vivo findings confirmed our hypothesis of being able to maintain a similar injected drug ratio for prolonged time periods in tested animals by our approach, thereby maximizing the therapeutic potency yet minimizing the toxicity of these drugs. This work could open up a new avenue on combination drug therapy and realization the clinical utility of such drugs.

Key wordscurcumin    sunitinib    synergistic effect    bovine serum albumin (BSA)    nano-carriers
收稿日期: 2016-04-08      出版日期: 2016-08-23
Corresponding Author(s): Huining He,Victor C. Yang   
 引用本文:   
. [J]. Frontiers of Chemical Science and Engineering, 2016, 10(3): 383-388.
Sunhui Chen,Qiuling Liang,Shuping Xie,Ergang Liu,Zhili Yu,Lu Sun,Meong Cheol Shin,Seung Jin Lee,Huining He,Victor C. Yang. Curcumin based combination therapy for anti-breast cancer: from in vitro drug screening to in vivo efficacy evaluation. Front. Chem. Sci. Eng., 2016, 10(3): 383-388.
 链接本文:  
https://academic.hep.com.cn/fcse/CN/10.1007/s11705-016-1574-2
https://academic.hep.com.cn/fcse/CN/Y2016/V10/I3/383
Fig.1  
Fig.2  
Fig.3  
Fig.4  
1 Basnet P, Skalko-Basnet N. Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules (Basel, Switzerland), 2011, 16(12): 4567–4598
https://doi.org/10.3390/molecules16064567
2 Boyanapalli S S, Kong A N. Curcumin, the king of spices: Epigenetic regulatory mechanisms in the prevention of cancer, neurological and inflammatory diseases. Current Pharmacology Reports, 2015, 1(2): 129–139
https://doi.org/10.1007/s40495-015-0018-x
3 Teiten M H, Dicato M, Diederich M. Curcumin as a regulator of epigenetic events. Molecular Nutrition & Food Research, 2013, 57(9): 1619–1629
https://doi.org/10.1002/mnfr.201300201
4 Shehzad A, Lee Y S. Molecular mechanisms of curcumin action: Signal transduction. BioFactors (Oxford, England), 2013, 39(1): 27–36
https://doi.org/10.1002/biof.1065
5 Bose S, Panda A K, Mukherjee S, Sa G. Curcumin and tumor immune-editing: Resurrecting the immune system. Cell Division, 2015, 10(1): 6
https://doi.org/10.1186/s13008-015-0012-z
6 Li Y, Zhang T. Targeting cancer stem cells by curcumin and clinical applications. Cancer Letters, 2014, 346(2): 197–205
https://doi.org/10.1016/j.canlet.2014.01.012
7 Bordoloi D, Roy N K, Monisha J, Padmavathi G, Kunnumakkara A B. Multi-targeted agents in cancer cell chemosensitization: What we learnt from curcumin thus far. Recent Patents on Anti-cancer Drug Discovery, 2016, 11(1): 67–97
https://doi.org/10.2174/1574892810666151020101706
8 Saha S, Adhikary A, Bhattacharyya P, Das T, Sa G. Death by design: Where curcumin sensitizes drug-resistant tumors. Anticancer Research, 2012, 32(7): 2567–2584
9 Chang R, Sun L, Webster T J. Selective inhibition of MG-63 osteosarcoma cell proliferation induced by curcumin-loaded self-assembled arginine-rich-RGD nanospheres. International Journal of Nanomedicine, 2015, 10: 3351–3365
10 Collins H M, Abdelghany M K, Messmer M, Yue B, Deeves S E, Kindle K B, Mantelingu K, Aslam A, Winkler G S, Kundu T K, Heery D M. Differential effects of garcinol and curcumin on histone and p53 modifications in tumor cells. BMC Cancer, 2013, 13(1): 37
https://doi.org/10.1186/1471-2407-13-37
11 Bozic I, Reiter J G, Allen B, Antal T, Chatterjee K, Shah P, Moon Y S, Yaqubie A, Kelly N, Le D T, Lipson E J, Chapman P B, Diaz L A Jr, Vogelstein B, Nowak M A. Evolutionary dynamics of cancer in response to targeted combination therapy. eLife, 2013, 2: e00747
https://doi.org/10.7554/eLife.00747
12 Brahmbhatt M, Gundala S R, Asif G, Shamsi S A, Aneja R. Ginger phytochemicals exhibit synergy to inhibit prostate cancer cell proliferation. Nutrition and Cancer, 2013, 65(2): 263–272
https://doi.org/10.1080/01635581.2013.749925
13 Gotink K J, Verheul H M. Anti-angiogenic tyrosine kinase inhibitors: What is their mechanism of action? Angiogenesis, 2010, 13(1): 1–14
https://doi.org/10.1007/s10456-009-9160-6
14 Shen G, Liu T, Wang Q, Jiang M, Shi J. Spectroscopic and molecular docking studies of binding interaction of gefitinib, lapatinib and sunitinib with bovine serum albumin (BSA). Journal of Photochemistry and Photobiology B: Biology, 2015, 153: 380–390
https://doi.org/10.1016/j.jphotobiol.2015.10.023
15 Cao H, Wang Y, He X, Zhang Z, Yin Q, Chen Y, Yu H, Huang Y, Chen L, Xu M, Gu W, Li Y. Codelivery of sorafenib and curcumin by directed self-assembled nanoparticles enhances therapeutic effect on hepatocellular carcinoma. Molecular Pharmaceutics, 2015, 12(3): 922–931
https://doi.org/10.1021/mp500755j
16 Llovet J M, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J F, de Oliveira A C, Santoro A, Raoul J L, Forner A, Schwartz M, Porta C, Zeuzem S, Bolondi L, Greten T F, Galle P R, Seitz J F, Borbath I, Häussinger D, Giannaris T, Shan M, Moscovici M, Voliotis D, Bruix J. Sorafenib in advanced hepatocellular carcinoma. New England Journal of Medicine, 2008, 359(4): 378–390
https://doi.org/10.1056/NEJMoa0708857
17 Desandes E. Survival from adolescent cancer. Cancer Treatment Reviews, 2007, 33(7): 609–615
https://doi.org/10.1016/j.ctrv.2006.12.007
18 Li S, Sun W, Wang H, Zuo D, Hua Y, Cai Z. Research progress on the multidrug resistance mechanism of osteosarcoma chemotherapy and reversal. Tumour Biology, 2015, 36(3): 1329–1338
https://doi.org/10.1007/s13277-015-3181-0
19 Nedelcu T, Kubista B, Koller A, Sulzbacher I, Mosberger I, Arrich F, Trieb K, Kotz R, Toma C D. Livin and Bcl-2 expression in high-grade osteosarcoma. Journal of Cancer Research and Clinical Oncology, 2007, 134(2): 237–244
https://doi.org/10.1007/s00432-007-0276-z
20 Rajkumar T, Yamuna M. Multiple pathways are involved in drug resistance to doxorubicin in an osteosarcoma cell line. Anti-Cancer Drugs, 2008, 19(3): 257–265
https://doi.org/10.1097/CAD.0b013e3282f435b6
21 Guo L, Shen Y, Zhao X, Guo L, Yu Z, Wang D, Liu L, Liu J. Curcumin combined with oxaliplatin effectively suppress colorectal carcinoma in vivo through inducing apoptosis. Phytotherapy Research, 2015, 29(3): 357–365
https://doi.org/10.1002/ptr.5257
22 Jonker M J, Svendsen C, Bedaux J J, Bongers M, Kammenga J E. Significance testing of synergistic/antagonistic, dose level-dependent, or dose ratio-dependent effects in mixture dose-response analysis. Environmental Toxicology and Chemistry, 2005, 24(10): 2701–2713
https://doi.org/10.1897/04-431R.1
23 Hu C, Aryal S, Zhang L. Nanoparticle-assisted combination therapies for effective cancer treatment. Therapeutic Delivery, 2010, 1(2): 323–334
https://doi.org/10.4155/tde.10.13
24 Ashton J C. Drug combination studies and their synergy quantification using the Chou-Talalay Method-Letter. Cancer Research, 2015, 75(11): 2400
https://doi.org/10.1158/0008-5472.CAN-14-3763
25 Tardi P, Johnstone S, Harasym N, Xie S, Harasym T, Zisman N, Harvie P, Bermudes D, Mayer L. In vivo maintenance of synergistic cytarabine: Daunorubicin ratios greatly enhances therapeutic efficacy. Leukemia Research, 2009, 33(1): 129–139
https://doi.org/10.1016/j.leukres.2008.06.028
26 Mayer L D, Harasym T O, Tardi P G, Harasym N L, Shew C R, Johnstone S A, Ramsay E C, Bally M B, Janoff A S. Ratiometric dosing of anticancer drug combinations: Controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice. Molecular Cancer Therapeutics, 2006, 5(7): 1854–1863
https://doi.org/10.1158/1535-7163.MCT-06-0118
27 Farokhzad O C, Langer R. Impact of nanotechnology on drug delivery. ACS Nano, 2009, 3(1): 16–20
https://doi.org/10.1021/nn900002m
28 He H, David A, Chertok B, Cole A, Lee K, Zhang J, Wang J, HuangY, Yang V C. Magnetic nanoparticles for tumor imaging and therapy: A so-called theranostic system. Pharmaceutical Research, 2013, 30(10): 2445–2458
https://doi.org/10.1007/s11095-013-0982-y
29 He H, Ye J, Sheng J, Wang J, Huang Y, Chen G, Wang J, Yang V C. Overcoming oral insulin delivery barriers: Application of cell penetrating peptide and silica-based nanoporous composites. Frontiers of Chemical Science and Engineering, 2013, 7(1): 9–19
https://doi.org/10.1007/s11705-013-1306-9
30 He H, Liang Q, Shin M C, Lee K, Gong J, Ye J, Liu Q, Wang J, Yang V C. Significance and strategies in developing delivery systems for bio-macromolecular drugs. Frontiers of Chemical Science and Engineering, 2013, 7(4): 496–507
https://doi.org/10.1007/s11705-013-1362-1
31 Parhi P, Mohanty C, Sahoo S K. Nanotechnology-based combinational drug delivery: An emerging approach for cancer therapy. Drug Discovery Today, 2012, 17(17-18): 1044–1052
https://doi.org/10.1016/j.drudis.2012.05.010
32 Yamasaki K, Chuang V T, Maruyama T, Otagiri M. Albumin-drug interaction and its clinical implication. Biochimica et Biophysica Acta, 2013, 1830(12): 5435–5443
https://doi.org/10.1016/j.bbagen.2013.05.005
33 Lee H J, Bergen P J, Bulitta J B, Tsuji B, Forrest A, Nation R L, Li J. Synergistic activity of colistin and rifampin combination against multidrug-resistant Acinetobacter baumannii in an in vitro pharmacokinetic/pharmacodynamic model. Antimicrobial Agents and Chemotherapy, 2013, 57(8): 3738–3745
https://doi.org/10.1128/AAC.00703-13
34 Allen T M, Cullis P R. Drug delivery systems: Entering the mainstream. Science, 2004, 303(5665): 1818–1822
https://doi.org/10.1126/science.1095833
35 Torchilin V P. Targeted pharmaceutical nanocarriers for cancer therapy and imaging. AAPS Journal, 2007, 9(2): E128–E147
https://doi.org/10.1208/aapsj0902015
36 Kratz F. Albumin as a drug carrier: Design of prodrugs, drug conjugates and nanoparticles. Journal of Controlled Release, 2008, 132(3): 171–183
https://doi.org/10.1016/j.jconrel.2008.05.010
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed