]. J. You, P. A. Jones, T. J. Harris, and F. Mccormick, Cancer Genetics and Epigenetics: Two Sides of the Same Coin? Cancer Cell The molecular pathology of cancer, Nat. Rev. Clin, vol.22, issue.1, 2012.

D. Hanahan and R. A. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, issue.5, pp.251-265, 2010.
DOI : 10.1016/j.cell.2011.02.013

]. R. Wong, Apoptosis in cancer: from pathogenesis to treatment, Journal of Experimental & Clinical Cancer Research, vol.30, issue.1
DOI : 10.1016/j.bbrc.2007.04.134

URL : https://jeccr.biomedcentral.com/track/pdf/10.1186/1756-9966-30-87?site=jeccr.biomedcentral.com

L. Zitvogel, L. Galluzzi, and M. J. Smyth, Mechanism of Action of Conventional and Targeted Anticancer Therapies: Reinstating Immunosurveillance, Immunity, vol.39, issue.1, pp.74-88, 2013.
DOI : 10.1016/j.immuni.2013.06.014

T. Otto and P. Sicinski, Cell cycle proteins as promising targets in cancer therapy, Nature Reviews Cancer, vol.56, issue.2, 2017.
DOI : 10.1038/35092592

URL : http://europepmc.org/articles/pmc5345933?pdf=render

L. Galluzzi, O. Kepp, and M. G. Heiden, Metabolic targets for cancer therapy, Nature Reviews Drug Discovery, vol.79, issue.11, pp.829-846, 2013.
DOI : 10.1016/j.bcp.2009.12.003

G. Housman, S. Byler, and S. Heerboth, Drug resistance in cancer: an overview, Cancers (Basel), pp.1769-1792, 2014.
DOI : 10.3390/cancers6031769

URL : http://www.mdpi.com/2072-6694/6/3/1769/pdf

I. A. Cree and P. Charlton, Molecular chess? Hallmarks of anti-cancer drug resistance, BMC Cancer, vol.9, issue.1, 2017.
DOI : 10.2217/bmm.14.84

URL : https://bmccancer.biomedcentral.com/track/pdf/10.1186/s12885-016-2999-1?site=bmccancer.biomedcentral.com

C. Holohan, S. Van-schaeybroeck, and D. B. Longley, Cancer drug resistance: an evolving paradigm, Nature Reviews Cancer, vol.14, issue.10, pp.714-726, 2013.
DOI : 10.1093/neuonc/nos158

S. Shukla and A. Mehta, Anticancer potential of medicinal plants and their phytochemicals: a review, Brazilian Journal of Botany, vol.10, issue.2, pp.199-210, 2015.
DOI : 10.1046/j.1460-9568.1998.00393.x

V. Ruiz-torres, J. A. Encinar, and M. Herranz-lópez, An Updated Review on Marine Anticancer Compounds: The Use of Virtual Screening for the Discovery of Small-Molecule Cancer Drugs, Molecules, vol.15, issue.7, p.22, 2017.
DOI : 10.1039/b808331m

C. Calcabrini, E. Catanzaro, and A. Bishayee, Marine Sponge Natural Products with Anticancer Potential: An Updated Review, Marine Drugs, vol.65, issue.10, 2017.
DOI : 10.1016/j.cbpc.2008.01.005

URL : http://www.mdpi.com/1660-3397/15/10/310/pdf

R. Kotecha, A. Takami, and J. L. Espinoza, Dietary phytochemicals and cancer chemoprevention: a review of the clinical evidence, Oncotarget, vol.7, issue.32, pp.52517-52529, 2016.
DOI : 10.18632/oncotarget.9593

URL : http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=9593&path%5B%5D=30041

H. Wang, T. O. Khor, and L. Shu, Plants Against Cancer: A Review on Natural Phytochemicals in Preventing and Treating Cancers and Their Druggability, Anticancer Agents Med. Chem, pp.12-1281, 2012.
DOI : 10.2174/187152012803833026

B. S. Vinod, T. T. Maliekal, and R. J. Anto, Phytochemicals As Chemosensitizers: From Molecular Mechanism to Clinical Significance, Antioxidants & Redox Signaling, vol.18, issue.11, 2013.
DOI : 10.1089/ars.2012.4573

P. Dandawate, S. Padhye, and A. Ahmad, Novel strategies targeting cancer stem cells through phytochemicals and their analogs, Drug Delivery and Translational Research, vol.25, issue.Suppl 1, pp.165-182, 2013.
DOI : 10.1096/fj.10-167924

URL : http://europepmc.org/articles/pmc3783342?pdf=render

G. Jacquemin, S. Shirley, and O. Micheau, Combining naturally occurring polyphenols with TNF-related apoptosis-inducing ligand: a promising approach to kill resistant cancer cells?, Cellular and Molecular Life Sciences, vol.31, issue.18, pp.3115-3130, 2010.
DOI : 10.1002/jps.21880

URL : https://hal.archives-ouvertes.fr/inserm-00491262

S. Gupta, R. Kannappan, and S. Reuter, Chemosensitization of tumors by resveratrol, Annals of the New York Academy of Sciences, vol.101, issue.1, pp.1215-150, 2011.
DOI : 10.1182/blood-2002-07-2130

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1749-6632.2010.05852.x/pdf

D. Moher, A. Liberati, and J. Tetzlaff, Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement, PLoS Med, vol.6, 2009.
DOI : 10.1016/j.ijsu.2010.02.007

URL : https://doi.org/10.1016/j.ijsu.2010.02.007

C. R. Hooijmans, M. M. Rovers, and R. B. De-vries, SYRCLE???s risk of bias tool for animal studies, BMC Medical Research Methodology, vol.920, issue.3, pp.43-53, 2014.
DOI : 10.1016/0005-2760(87)90105-6

URL : https://bmcmedresmethodol.biomedcentral.com/track/pdf/10.1186/1471-2288-14-43?site=bmcmedresmethodol.biomedcentral.com

P. S. Siqueira-lima, J. C. Silva, and J. S. Quintans, Natural products assessed in animal models for orofacial pain ??? a systematic review, Revista Brasileira de Farmacognosia, vol.27, issue.1
DOI : 10.1016/j.bjp.2016.06.005

URL : https://doi.org/10.1016/j.bjp.2016.06.005

Y. Z. Liu, C. M. Yang, and J. Y. Chen, Alpha-carotene inhibits metastasis in Lewis lung carcinoma in vitro, and suppresses lung metastasis and tumor growth in combination with taxol in tumor xenografted C57BL/6 mice, The Journal of Nutritional Biochemistry, vol.26, issue.6
DOI : 10.1016/j.jnutbio.2014.12.012

Y. Zhang, X. Zhu, and T. Huang, ?-Carotene synergistically enhances the antitumor effect of 5-fluorouracil on esophageal squamous cell carcinoma in vivo and in vitro, Toxicol. Lett, vol.261, 2016.

S. M. Kim, J. H. Lee, and G. Sethi, Bergamottin, a natural furanocoumarin obtained from grapefruit juice induces chemosensitization and apoptosis through the inhibition of STAT3 signaling pathway in tumor cells, Cancer Letters, vol.354, issue.1, 2014.
DOI : 10.1016/j.canlet.2014.08.002

G. Stammler and M. Volm, Green tea catechins (EGCG and EGC) have modulating effects on the activity of doxorubicin in drug-resistant cell lines, Anti-Cancer Drugs, vol.8, issue.3, pp.265-268, 1997.
DOI : 10.1097/00001813-199703000-00007

S. Dhanalakshmi, P. Agarwal, and L. M. Glode, Silibinin sensitizes human prostate carcinoma DU145 cells to cisplatin- and carboplatin-induced growth inhibition and apoptotic death, International Journal of Cancer, vol.11, issue.5, pp.699-705, 2003.
DOI : 10.1093/ndt/11.1.55

URL : http://onlinelibrary.wiley.com/doi/10.1002/ijc.11299/pdf

K. Chisholm, B. J. Bray, and R. J. Rosengren, Tamoxifen and epigallocatechin gallate are synergistically cytotoxic to MDA-MB-231 human breast cancer cells, Anti-Cancer Drugs, vol.15, issue.9, pp.889-897, 2004.
DOI : 10.1097/00001813-200410000-00010

X. H. Peng, P. Karna, and R. M. O-'regan, Down-Regulation of Inhibitor of Apoptosis Proteins by Deguelin Selectively Induces Apoptosis in Breast Cancer Cells, Molecular Pharmacology, vol.71, issue.1, pp.71-101, 2007.
DOI : 10.1124/mol.106.027367

C. Y. Jin, C. Park, and H. J. Hwang, Naringenin up-regulates the expression of death receptor 5 and enhances TRAIL-induced apoptosis in human lung cancer A549 cells, Molecular Nutrition & Food Research, vol.94, issue.2, pp.300-309, 2011.
DOI : 10.1016/S0092-8674(00)81590-1

M. E. Stearns and M. Wang, Synergistic Effects of the Green Tea Extract Epigallocatechin-3-gallate and Taxane in Eradication of Malignant Human Prostate Tumors, Translational Oncology, vol.4, issue.3, pp.147-156, 2011.
DOI : 10.1593/tlo.10286

A. S. Hönicke, S. A. Ender, and J. Radons, Combined administration of EGCG and IL-1 receptor antagonist efficiently downregulates IL-1-induced tumorigenic factors in U-2 OS human osteosarcoma cells, International Journal of Oncology, vol.41, issue.2, pp.753-758, 2012.
DOI : 10.3892/ijo.2012.1498

H. Wu, Y. Xin, and C. Xu, Capecitabine combined with (-)-epigallocatechin-3-gallate inhibits angiogenesis and tumor growth in nude mice with gastric cancer xenografts, Experimental and Therapeutic Medicine, vol.3, issue.4, pp.650-654, 2012.
DOI : 10.3892/etm.2012.448

T. W. Kwak, D. H. Kim, and C. W. Chung, Synergistic Anticancer Effects of Vorinostat and Epigallocatechin-3-Gallate against HuCC-T1 Human Cholangiocarcinoma Cells, Evid. Based Complementary Altern, 2013.
DOI : 10.1155/2013/185158

URL : http://doi.org/10.1155/2013/185158

R. Suzuki, Y. Kang, and X. Li, Genistein potentiates the antitumor effect of 5- Fluorouracil by inducing apoptosis and autophagy in human pancreatic cancer cells, Anticancer Res, vol.34, pp.4685-4692, 2014.

L. Wang, J. Feng, and X. Chen, Myricetin enhance chemosensitivity of 5-fluorouracil on esophageal carcinoma in vitro and in vivo, Cancer Cell International, vol.14, issue.1, 2014.
DOI : 10.1038/sj.onc.1207113

URL : https://cancerci.biomedcentral.com/track/pdf/10.1186/s12935-014-0071-2?site=cancerci.biomedcentral.com

M. S. Abaza, K. Y. Orabi, and E. Al-quattan, Growth inhibitory and chemosensitization effects of naringenin, a natural flavanone purified from Thymus vulgaris, on human breast and colorectal cancer, Cancer Cell Int, vol.15, issue.46, 2015.
DOI : 10.1186/s12935-015-0194-0

URL : https://cancerci.biomedcentral.com/track/pdf/10.1186/s12935-015-0194-0?site=cancerci.biomedcentral.com

P. Wang, S. M. Henning, and D. Heber, Sensitization to docetaxel in prostate cancer cells by green tea and quercetin, The Journal of Nutritional Biochemistry, vol.26, issue.4, pp.408-415, 2015.
DOI : 10.1016/j.jnutbio.2014.11.017

URL : http://europepmc.org/articles/pmc4375039?pdf=render

J. A. García-vilas, A. R. Quesada, and M. A. Medina, Screening of synergistic interactions of epigallocatechin-3-gallate with antiangiogenic and antitumor compounds, Synergy, vol.3, issue.2
DOI : 10.1016/j.synres.2016.05.001

T. Krajnovi?, G. N. Kalu?erovi?, and L. A. Wessjohann, Versatile antitumor potential of isoxanthohumol: Enhancement of paclitaxel activity in vivo, Pharmacological Research, vol.105, 2016.
DOI : 10.1016/j.phrs.2016.01.011

S. Lin, K. Lei, and W. Du, Enhancement of oxaliplatin sensitivity in human colorectal cancer by hypericin mediated photodynamic therapy via ROS-related mechanism, The International Journal of Biochemistry & Cell Biology, vol.71, pp.24-34, 2016.
DOI : 10.1016/j.biocel.2015.12.003

S. Lin, L. Yang, and H. Shi, Endoplasmic reticulum-targeting photosensitizer Hypericin confers chemo-sensitization towards oxaliplatin through inducing prodeath autophagy, Int. J. Biochem. Cell Biol, vol.87, 2017.
DOI : 10.1016/j.biocel.2017.04.001

S. Anuchapreeda, P. Leechanachai, and M. M. Smith, Modulation of P-glycoprotein expression and function by curcumin in multidrug-resistant human KB cells, Biochemical Pharmacology, vol.64, issue.4, pp.573-582, 2002.
DOI : 10.1016/S0006-2952(02)01224-8

T. C. Hour, J. Chen, and C. Y. Huang, Curcumin enhances cytotoxicity of chemotherapeutic agents in prostate cancer cells by inducing p21(WAF1/CIP1) and C/EBPbeta expressions and suppressing NF-kappaB activation, Prostate, pp.51-211, 2002.
DOI : 10.1002/pros.10089

S. Fulda and K. M. Debatin, Sensitization for anticancer drug-induced apoptosis by the chemopreventive agent resveratrol, Oncogene, vol.23, issue.40, pp.6702-6711, 2004.
DOI : 10.1007/s000180050270

URL : http://www.nature.com/onc/journal/v23/n40/pdf/1207630a.pdf

S. L. Wu, Z. J. Sun, and L. Yu, Effect of resveratrol and in combination with 5-FU on murine liver cancer, World Journal of Gastroenterology, vol.10, issue.20, pp.3048-3052, 2004.
DOI : 10.1006/bbrc.2000.3378

URL : https://doi.org/10.3748/wjg.v10.i20.3048

B. B. Aggarwal, S. Shishodia, and Y. Takada, Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB pathway in breast cancer cells and inhibits lung metastasis of human breast cancer in nude mice, Clin. Cancer Res

K. S. Chen, Y. C. Hsiao, and D. Y. Kuo, Tannic acid-induced apoptosis and -enhanced sensitivity to arsenic trioxide in human leukemia HL-60 cells, Leukemia Research, vol.33, issue.2
DOI : 10.1016/j.leukres.2008.08.006

K. B. Harikumar, A. B. Kunnumakkara, and G. Sethi, Resveratrol, a multitargeted agent, can enhance antitumor activity of gemcitabine in vitro and in orthotopic mouse model of human pancreatic cancer, Int. J. Cancer, vol.127, pp.257-268, 2010.

A. B. Kunnumakkara, P. Diagaradjane, and P. Anand, Curcumin sensitizes human colorectal cancer to capecitabine by modulation of cyclin D1, COX-2, MMP-9, VEGF and CXCR4 expression in an orthotopic mouse model, International Journal of Cancer, vol.67, issue.9
DOI : 10.1016/j.ijrobp.2006.08.010

Y. Yu, S. S. Kanwar, and B. B. Patel, Elimination of Colon Cancer Stem-Like Cells by the Combination of Curcumin and FOLFOX, Translational Oncology, vol.2, issue.4, pp.321-328, 2009.
DOI : 10.1593/tlo.09193

W. Hartojo, A. L. Silvers, and D. G. Thomas, Curcumin Promotes Apoptosis, Increases Chemosensitivity, and Inhibits Nuclear Factor ??B in Esophageal Adenocarcinoma, Translational Oncology, vol.3, issue.2, pp.99-108, 2010.
DOI : 10.1593/tlo.09235

URL : https://doi.org/10.1593/tlo.09235

S. V. Bava, C. N. Sreekanth, and A. K. Thulasidasan, Akt is upstream and MAPKs are downstream of NF-??B in paclitaxel-induced survival signaling events, which are down-regulated by curcumin contributing to their synergism, The International Journal of Biochemistry & Cell Biology, vol.43, issue.3, pp.331-341, 2011.
DOI : 10.1016/j.biocel.2010.09.011

C. N. Sreekanth, S. Bava, and E. Sreekumar, Molecular evidences for the chemosensitizing efficacy of liposomal curcumin in paclitaxel chemotherapy in mouse models of cervical cancer, Oncogene, vol.272, issue.28, pp.30-3139, 2011.
DOI : 10.1074/jbc.272.24.15174

A. M. Osman, H. M. Bayoumi, and S. E. Harthi, Modulation of doxorubicin cytotoxicity by resveratrol in a human breast cancer cell line, Cancer Cell International, vol.12, issue.1, pp.47-57, 2012.
DOI : 10.1093/jnci/85.8.632

URL : https://cancerci.biomedcentral.com/track/pdf/10.1186/1475-2867-12-47?site=cancerci.biomedcentral.com

E. M. Saleh, R. A. El-awady, and N. A. Eissa, Antagonism between curcumin and the topoisomerase II inhibitor etoposide, Cancer Biology & Therapy, vol.26, issue.11, pp.13-1058, 2012.
DOI : 10.1016/j.bbrc.2008.01.030

URL : http://www.tandfonline.com/doi/pdf/10.4161/cbt.21078?needAccess=true

H. Wang, Q. R. Geng, and L. Wang, -asparaginase via inhibition of the AKT signaling pathway in acute lymphoblastic leukemia, Leukemia & Lymphoma, vol.22, issue.7, pp.53-1376, 2012.
DOI : 10.1158/0008-5472.CAN-06-4257

F. Amiri, A. H. Zarnani, and H. Zand, Synergistic anti-proliferative effect of resveratrol and etoposide on human hepatocellular and colon cancer cell lines, European Journal of Pharmacology, vol.718, issue.1-3, 2013.
DOI : 10.1016/j.ejphar.2013.09.020

J. Díaz-chávez, M. A. Fonseca-sánchez, and E. Arechaga-ocampo, Proteomic Profiling Reveals That Resveratrol Inhibits HSP27 Expression and Sensitizes Breast Cancer Cells to Doxorubicin Therapy, PLoS ONE, vol.14, issue.3, pp.1-11, 2013.
DOI : 10.1371/journal.pone.0064378.t002

M. Shakibaei, A. Mobasheri, and C. Lueders, Curcumin Enhances the Effect of Chemotherapy against Colorectal Cancer Cells by Inhibition of NF-??B and Src Protein Kinase Signaling Pathways, PLoS ONE, vol.279, issue.2, pp.1-13, 2013.
DOI : 10.1371/journal.pone.0057218.g010

L. J. Carlson, B. Cote, and A. W. Alani, Polymeric Micellar Co-delivery of Resveratrol and Curcumin to Mitigate In Vitro Doxorubicin-Induced Cardiotoxicity, Journal of Pharmaceutical Sciences, vol.103, issue.8, pp.2315-2322, 2014.
DOI : 10.1002/jps.24042

H. Qian, Y. Yang, and X. Wang, Curcumin enhanced adriamycin-induced human liver-derived Hepatoma G2 cell death through activation of mitochondriamediated apoptosis and autophagy, Eur, J. Pharm. Sci, vol.43, pp.125-131, 2011.
DOI : 10.1016/j.ejps.2011.04.002

C. Buhrmann, P. Shayan, and P. Kraehe, Resveratrol induces chemosensitization to 5-fluorouracil through up-regulation of intercellular junctions, Epithelial-to-mesenchymal transition and apoptosis in colorectal cancer, Biochemical Pharmacology, vol.98, issue.1, pp.51-58, 2015.
DOI : 10.1016/j.bcp.2015.08.105

B. Cote, L. J. Carlson, and D. A. Rao, Combinatorial resveratrol and quercetin [97 Enhancement of cisplatin-induced colon cancer cells apoptosis by shikonin, a natural inducer of ROS in vitro and in vivo, Biochem. Biophys. Res. Commun, pp.469-1075, 2016.

J. Song, Z. Zhao, and X. Fan, Shikonin potentiates the effect of arsenic trioxide against human hepatocellular carcinoma <i>in vitro</i> and <i>in vivo</i>, Oncotarget, vol.7, issue.43, 2016.
DOI : 10.18632/oncotarget.12041

Y. Wang, H. L. Lu, and Y. D. Liu, Cryptotanshinone sensitizes antitumor effect of paclitaxel on tongue squamous cell carcinoma growth by inhibiting the JAK/STAT3 signaling pathway, Biomedicine & Pharmacotherapy, vol.95, pp.1388-1396, 2017.
DOI : 10.1016/j.biopha.2017.09.062

C. H. Choi, G. Kang, and Y. D. Min, Reversal of P-Glycoprotein-Mediated Multidrug Resistance by Protopanaxatriol Ginsenosides from Korean Red Ginseng, Planta Medica, vol.69, issue.3, pp.235-240, 2003.
DOI : 10.1055/s-2003-38483

S. M. Kim, S. Y. Lee, and J. S. Cho, Hong, Combination of ginsenoside Rg3 with docetaxel enhances the susceptibility of prostate cancer cells via inhibition of NF-?B, Eur. J. Pharmacol, vol.631, 2010.

Z. J. Ming, Y. Hu, and Y. H. Qiu, Synergistic effects of ??-aescin and 5-fluorouracil in human hepatocellular carcinoma SMMC-7721 cells, Phytomedicine, vol.17, issue.8-9, 2010.
DOI : 10.1016/j.phymed.2009.12.009

Y. W. Wang, S. J. Wang, and Y. N. Zhou, Escin augments the efficacy of gemcitabine through down-regulation of nuclear factor-?B and nuclear factor?B-regulated gene products in pancreatic cancer both in vitro and in vivo, J

H. Wang, Z. Zhai, and N. Li, Steroidal saponin of Trillium tschonoskii. Reverses multidrug resistance of hepatocellular carcinoma, Phytomedicine, vol.20, issue.11, 2013.
DOI : 10.1016/j.phymed.2013.04.014

L. Chang, B. Huo, and Y. Lv, Ginsenoside Rg3 enhances the inhibitory effects of chemotherapy on esophageal squamous cell carcinoma in mice, Molecular and Clinical Oncology, vol.2, issue.6
DOI : 10.3892/mco.2014.355

F. F. El-senduny, F. A. Badria, and A. M. El-waseef, Approach for chemosensitization of cisplatin-resistant ovarian cancer by cucurbitacin B, Tumor Biol, pp.685-698, 2016.
DOI : 10.1007/s13277-015-3773-8

X. Li, F. Zhu, and J. Jiang, Synergistic antitumor activity of withaferin A combined with oxaliplatin triggers reactive oxygen species-mediated inactivation of the PI3K/AKT pathway in human pancreatic cancer cells, Cancer Letters, vol.357, issue.1, 2015.
DOI : 10.1016/j.canlet.2014.11.026

M. Ben-eltriki, S. Deb, and H. Adomat, Calcitriol and 20( S )-protopanaxadiol synergistically inhibit growth and induce apoptosis in human prostate cancer cells, The Journal of Steroid Biochemistry and Molecular Biology, vol.158, pp.158-207, 2016.
DOI : 10.1016/j.jsbmb.2015.12.002

M. L. Holland, J. A. Panetta, and J. M. Hoskins, The effects of cannabinoids on P-glycoprotein transport and expression in multidrug resistant cells, Biochemical Pharmacology, vol.71, issue.8
DOI : 10.1016/j.bcp.2005.12.033

S. Sieber, G. Gdynia, and W. Roth, Combination treatment of malignant B cells using the anti-CD20 antibody rituximab and the anti-malarial artesunate, Int. J. Oncol, pp.35-149, 2009.

Y. He, F. Liu, and L. Zhang, Growth Inhibition and Apoptosis Induced by Lupeol, a Dietary Triterpene, in Human Hepatocellular Carcinoma Cells, Biological & Pharmaceutical Bulletin, vol.34, issue.4
DOI : 10.1248/bpb.34.517

URL : https://www.jstage.jst.go.jp/article/bpb/34/4/34_4_517/_pdf

R. Kannaiyan, H. S. Hay, and P. Rajendran, Celastrol inhibits proliferation and induces chemosensitization through down-regulation of NF-??B and STAT3 regulated gene products in multiple myeloma cells, British Journal of Pharmacology, vol.28, issue.5, pp.1506-1521, 2011.
DOI : 10.3109/07357900903095664

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1476-5381.2011.01449.x/pdf

S. Torres, M. Lorente, and F. Rodriguez-fornes, A Combined Preclinical Therapy of Cannabinoids and Temozolomide against Glioma, Molecular Cancer Therapeutics, vol.10, issue.1, pp.90-103, 2011.
DOI : 10.1158/1535-7163.MCT-10-0688

URL : http://mct.aacrjournals.org/content/molcanther/10/1/90.full.pdf

S. Prasad, V. R. Yadav, and B. Sung, Ursolic Acid Inhibits Growth and Metastasis of Human Colorectal Cancer in an Orthotopic Nude Mouse Model by Targeting Multiple Cell Signaling Pathways: Chemosensitization with Capecitabine, Clinical Cancer Research, vol.18, issue.18, pp.4942-4953, 2012.
DOI : 10.1158/1078-0432.CCR-11-2805

URL : http://clincancerres.aacrjournals.org/content/clincanres/18/18/4942.full.pdf

J. Tong, S. Yin, and Y. Dong, Pseudolaric Acid B Induces Caspase-Dependent Apoptosis and Autophagic Cell Death in Prostate Cancer Cells, Phytotherapy Research, vol.26, issue.4, pp.885-891, 2013.
DOI : 10.1016/j.tiv.2012.02.004

E. Butturini, A. C. Prati, and G. Chiavegato, Mild oxidative stress induces Sglutathionylation of STAT3 and enhances chemosensitivity of tumoural cells to chemotherapeutic drugs, Free Radic, Biol. Med, pp.65-1322, 2013.
DOI : 10.1016/j.freeradbiomed.2013.09.015

F. Liu, Y. He, and Y. Liang, PI3-kinase inhibition synergistically promoted the anti-tumor effect of lupeol in hepatocellular carcinoma, Cancer Cell International, vol.13, issue.1, pp.10-1186, 2013.
DOI : 10.1158/1078-0432.CCR-11-0916

URL : https://cancerci.biomedcentral.com/track/pdf/10.1186/1475-2867-13-108?site=cancerci.biomedcentral.com

O. A. Bamodu, W. C. Huang, and D. T. Tzeng, Ovatodiolide sensitizes aggressive breast cancer cells to doxorubicin, eliminates their cancer stem cell-like phenotype, and reduces doxorubicin-associated toxicity, Cancer Letters, vol.364, issue.2, pp.125-134, 2015.
DOI : 10.1016/j.canlet.2015.05.006

Y. Liu, T. Bi, and W. Dai, Lupeol enhances inhibitory effect of 5-fluorouracil on human gastric carcinoma cells, Naunyn-Schmiedeberg's Archives of Pharmacology, vol.17, issue.4, pp.477-484, 2016.
DOI : 10.1002/ptr.1140

M. A. Reis, O. B. Ahmed, and G. Spengler, Jatrophane diterpenes and cancer multidrug resistance ??? ABCB1 efflux modulation and selective cell death induction, Phytomedicine, vol.23, issue.9, pp.968-978, 2016.
DOI : 10.1016/j.phymed.2016.05.007

URL : http://publicatio.bibl.u-szeged.hu/10172/1/3102606.pdf

B. C. Baguley, Multiple Drug Resistance Mechanisms in Cancer, Molecular Biotechnology, vol.142, issue.9, pp.308-316, 2010.
DOI : 10.4161/cc.7.19.6780

Q. Wu, Z. Yang, and Y. Nie, Multi-drug resistance in cancer chemotherapeutics: Mechanisms and lab approaches, Cancer Letters, vol.347, issue.2, 2014.
DOI : 10.1016/j.canlet.2014.03.013

W. Y. Wu, J. J. Hou, and H. L. Long, TCM-based new drug discovery and development in China, Chin, J. Nat. Med, vol.12, pp.241-250, 2014.

J. Liu, S. Wang, and Y. Zhang, Traditional Chinese medicine and cancer: History, present situation, and development, Thoracic Cancer, vol.24, issue.6216 Suppl., pp.561-569, 2015.
DOI : 10.1007/s11916-012-0275-x

URL : http://onlinelibrary.wiley.com/doi/10.1111/1759-7714.12270/pdf

A. Richmond and Y. Su, Mouse xenograft models vs GEM models for human cancer therapeutics, Disease Models and Mechanisms, vol.1, issue.2-3, 2008.
DOI : 10.1242/dmm.000976

URL : http://dmm.biologists.org/content/dmm/1/2-3/78.full.pdf

J. Jung, Human Tumor Xenograft Models for Preclinical Assessment of Anticancer Drug Development, Toxicological Research, vol.58, issue.1, 2014.
DOI : 10.1158/1078-0432.CCR-08-0138

URL : http://www.ndsl.kr/soc_img/society/ksot/DSHHBQ/2014/v30n1/DSHHBQ_2014_v30n1_1.pdf

M. F. Festing and D. G. Altman, Guidelines for the Design and Statistical Analysis of Experiments Using Laboratory Animals, ILAR Journal, vol.78, issue.2, pp.244-258, 2002.
DOI : 10.1093/ilar.41.2.59

J. A. Hirst, J. Howick, and J. K. Aronson, The Need for Randomization in Animal Trials: An Overview of Systematic Reviews, PLoS ONE, vol.35, issue.6
DOI : 10.1371/journal.pone.0098856.s002

P. J. Karanicolas, F. Farrokhyar, and M. Bhandari, Blinding: Who, what, when, why, how? Can, J. Surg, vol.53, pp.345-348, 2010.

J. E. Aguilar-nascimento, Fundamental steps in experimental design for animal studies, Acta Cirurgica Brasileira, vol.335, issue.1, pp.2-8, 2005.
DOI : 10.1016/0140-6736(90)90014-V

URL : http://www.scielo.br/pdf/acb/v20n1/23280.pdf

V. S. Govindarajan, Turmeric ??? chemistry, technology, and quality, C R C Critical Reviews in Food Science and Nutrition, vol.7, issue.2, pp.199-301, 1980.
DOI : 10.1111/j.1365-2621.1942.tb17237.x

N. Chainani-wu, ), The Journal of Alternative and Complementary Medicine, vol.9, issue.1, pp.161-168, 2003.
DOI : 10.1089/107555303321223035

B. Chandran and A. Goel, A Randomized, Pilot Study to Assess the Efficacy and Safety of Curcumin in Patients with Active Rheumatoid Arthritis, Phytotherapy Research, vol.30, issue.Suppl. 1, pp.1719-1725, 2012.
DOI : 10.1002/ptr.4639

S. C. Gupta, S. Patchva, and B. B. Aggarwal, Therapeutic Roles of Curcumin: Lessons Learned from Clinical Trials, The AAPS Journal, vol.15, issue.1, pp.195-218, 2013.
DOI : 10.1208/s12248-012-9432-8

URL : http://europepmc.org/articles/pmc3535097?pdf=render

M. Karin, Nuclear factor-??B in cancer development and progression, Nature, vol.12, issue.suppl., pp.431-436, 2006.
DOI : 10.1016/S1097-2765(03)00358-7

I. Vivanco and C. L. Sawyers, The phosphatidylinositol 3-Kinase???AKT pathway in human cancer, Nature Reviews Cancer, vol.287, issue.7, pp.489-501, 2002.
DOI : 10.1126/science.287.5455.1040

N. C. Whitlock and S. J. Baek, The Anticancer Effects of Resveratrol: Modulation of Transcription Factors, Nutrition and Cancer, vol.5, issue.4, 2012.
DOI : 10.1158/0008-5472.CAN-07-5953

J. K. Aluyen, Q. N. Ton, and T. Tran, Resveratrol: Potential as Anticancer Agent, Journal of Dietary Supplements, vol.390, issue.5, pp.45-56, 2011.
DOI : 10.1016/j.ijpharm.2009.10.011

J. Acunzo, C. Andrieu, and V. Baylot, Hsp27 as a Therapeutic Target in Cancers, Current Drug Targets, vol.15, issue.4, pp.423-431, 2014.
DOI : 10.2174/13894501113146660230

A. Satelli and S. Li, Vimentin in cancer and its potential as a molecular target for cancer therapy, Cellular and Molecular Life Sciences, vol.50, issue.6, pp.3033-3046, 2011.
DOI : 10.1146/annurev.pharmtox.010909.105547

URL : http://europepmc.org/articles/pmc3162105?pdf=render

Y. Sakamoto, N. Terashita, and T. Muraguchi, Effects of Epigallocatechin-3-gallate (EGCG) on A549 Lung Cancer Tumor Growth and Angiogenesis, Bioscience, Biotechnology, and Biochemistry, vol.77, issue.9, pp.77-1799, 2013.
DOI : 10.1038/nrclinonc.2012.120

H. Fujiki, S. Yoshizawa, and T. Horiuchi, Anticarcinogenic effects of (???)-epigallocatechin gallate, Preventive Medicine, vol.21, issue.4, pp.503-509, 1992.
DOI : 10.1016/0091-7435(92)90057-O

C. Braicu, C. D. Gherman, and A. Irimie, Epigallocatechin-3-Gallate (EGCG) Inhibits Cell Proliferation and Migratory Behaviour of Triple Negative Breast Cancer Cells, Journal of Nanoscience and Nanotechnology, vol.13, issue.1, pp.13-632, 2013.
DOI : 10.1166/jnn.2013.6882

S. Shankar, S. Ganapathy, and S. R. Hingorani, EGCG inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer, Frontiers in Bioscience, vol.13, issue.13, pp.440-452, 2008.
DOI : 10.2741/2691

J. D. Lambert and C. S. Yang, Mechanisms of Cancer Prevention by Tea Constituents, The Journal of Nutrition, vol.8, issue.10, pp.3262-3267, 2013.
DOI : 10.1016/S0378-4347(01)00285-7

URL : https://academic.oup.com/jn/article-pdf/133/10/3262S/24019745/4w10030s3262.pdf