Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

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

General Research Article

Novel Polymethoxylated Chalcones as Potential Compounds Against KRAS-Mutant Colorectal Cancers

Author(s): Alaa Mahmoud, Dana Elkhalifa*, Feras Alali, Ala-Eddin Al Moustafa and Ashraf Khalil*

Volume 26, Issue 14, 2020

Page: [1622 - 1633] Pages: 12

DOI: 10.2174/1381612826666200206095400

Price: $65

Abstract

Background/Objective: KRAS-mutant colorectal cancers (CRC) are tumors that are associated with poor prognosis. However, no effective treatments are available to target them. Therefore, we designed and synthesized novel chalcone analogs, small organic molecules, to investigate their effects on KRAS-mutant CRC cells.

Methods: Fourteen new chalcone analogs were synthesized, optimized, characterized, and tested against two KRAS-mutant CRC cell lines (HCT-116 and LoVo), one p-53 and BRAF mutant CRC cell line (HT-29) and one normal immortalized colon cells (NCE-1 E6/E7). Effects on cell viability, apoptosis, cell cycle, migration, colony formation, EMT, and angiogenesis were investigated.

Results: Compounds 3 and 14 were the most effective. Compound 3 showed potent activity against HCT-116 and LoVo cell lines (GI50 of 6.10 μM and 7.00 μM, respectively). While compound 14 showed GI50 of 8.60 μM and 8.80 μM on HCT-116 and LoVo cell lines, respectively. Both compounds were approximately 2-3 times more selective toward cancer cells rather than normal colon cells. Compound 3 was effective in inducing apoptosis in HCT-116 cells via Bax upregulation and Bcl-2 downregulation. Invasion and metastasis of KRAS-mutant cells were modulated by compounds 3 and 14 through significant inhibition of cell migration and the prevention of colony formation. In addition, they reversed EMT by downregulation of EMT markers (vimentin, fascin, and β- catenin) and upregulation of cell-cell adhesion marker, E-cadherin. Furthermore, compounds 3 and 14 had significantly inhibited angiogenesis in ovo.

Conclusion: Compounds 3 and 14 represent potent and selective leads for KRAS-mutant CRC cells, thus, further in vitro and in vivo studies are necessary to confirm their effect on KRAS-mutant CRCs.

Keywords: Chalcone, analogs, epithelial-mesenchymal transition (EMT), metastasis, colorectal cancer, KRAS mutation.

« Previous
[1]
Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019; 144(8): 1941-53.
[http://dx.doi.org/10.1002/ijc.31937] [PMID: 30350310]
[2]
Keum N, Giovannucci E. Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol 2019; 16(12): 713-32.
[http://dx.doi.org/10.1038/s41575-019-0189-8] [PMID: 31455888]
[3]
Yang J, Yu J. The association of diet, gut microbiota and colorectal cancer: what we eat may imply what we get. Protein Cell 2018; 9(5): 474-87.
[http://dx.doi.org/10.1007/s13238-018-0543-6] [PMID: 29713943]
[4]
Vamanu E. Complementary functional strategy for modulation of human gut microbiota. Curr Pharm Des 2018; 24(35): 4144-9.
[http://dx.doi.org/10.2174/1381612824666181001154242] [PMID: 30277147]
[5]
Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology 2010; 138(6): 2044-58.
[http://dx.doi.org/10.1053/j.gastro.2010.01.054] [PMID: 20420945]
[6]
Jauhri M, Bhatnagar A, Gupta S, et al. Prevalence and coexistence of KRAS, BRAF, PIK3CA, NRAS, TP53, and APC mutations in Indian colorectal cancer patients: Next-generation sequencing-based cohort study. Tumour Biol 2017; 39(2) 1010428317692265
[http://dx.doi.org/10.1177/1010428317692265] [PMID: 28222664]
[7]
Tan C, Du X. KRAS mutation testing in metastatic colorectal cancer. World J Gastroenterol 2012; 18(37): 5171-80.
[PMID: 23066310]
[8]
Issa J-P. Colon cancer: it’s CIN or CIMP. Clin Cancer Res 2008; 14(19): 5939-40.
[http://dx.doi.org/10.1158/1078-0432.CCR-08-1596] [PMID: 18829469]
[9]
Hutchins G, Southward K, Handley K, et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol 2011; 29(10): 1261-70.
[http://dx.doi.org/10.1200/JCO.2010.30.1366] [PMID: 21383284]
[10]
Imamura Y, Morikawa T, Liao X, et al. Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res 2012; 18(17): 4753-63.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-3210] [PMID: 22753589]
[11]
Bruera G, Cannita K, Di Giacomo D, et al. Worse prognosis of KRAS c.35 G > A mutant metastatic colorectal cancer (MCRC) patients treated with intensive triplet chemotherapy plus bevacizumab (FIr-B/FOx). BMC Med 2013; 11(1): 59.
[http://dx.doi.org/10.1186/1741-7015-11-59] [PMID: 23497191]
[12]
Phipps AI, Buchanan DD, Makar KW, et al. KRAS-mutation status in relation to colorectal cancer survival: the joint impact of correlated tumour markers. Br J Cancer 2013; 108(8): 1757-64.
[http://dx.doi.org/10.1038/bjc.2013.118] [PMID: 23511557]
[13]
Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 2008; 359(17): 1757-65.
[http://dx.doi.org/10.1056/NEJMoa0804385] [PMID: 18946061]
[14]
Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 2008; 26(10): 1626-34.
[http://dx.doi.org/10.1200/JCO.2007.14.7116] [PMID: 18316791]
[15]
Van Cutsem E, Köhne C-H, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 2009; 360(14): 1408-17.
[http://dx.doi.org/10.1056/NEJMoa0805019] [PMID: 19339720]
[16]
Bokemeyer C, Bondarenko I, Makhson A, et al. Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol 2008; 10(27): 663-71.
[http://dx.doi.org/10.1200/jco.2008.26.15_suppl.4000] [PMID: 19114683]
[17]
Douillard JY, Siena S, Cassidy J, et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol 2010; 28(31): 4697-705.
[http://dx.doi.org/10.1200/JCO.2009.27.4860] [PMID: 20921465]
[18]
Peeters M, Price TJ, Cervantes A, et al. Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. J Clin Oncol 2010; 28(31): 4706-13.
[http://dx.doi.org/10.1200/JCO.2009.27.6055] [PMID: 20921462]
[19]
Rizzo S, Bronte G, Fanale D, et al. Prognostic vs predictive molecular biomarkers in colorectal cancer: is KRAS and BRAF wild type status required for anti-EGFR therapy? Cancer Treat Rev 2010; 36(Suppl. 3): S56-61.
[http://dx.doi.org/10.1016/S0305-7372(10)70021-9] [PMID: 21129611]
[20]
Vakiani E, Solit DB. KRAS and BRAF: drug targets and predictive biomarkers. J Pathol 2011; 223(2): 219-29.
[http://dx.doi.org/10.1002/path.2796] [PMID: 21125676]
[21]
Nobili S, Lippi D, Witort E, et al. Natural compounds for cancer treatment and prevention. Pharmacol Res 2009; 59(6): 365-78.
[http://dx.doi.org/10.1016/j.phrs.2009.01.017] [PMID: 19429468]
[22]
Li Y, Zhang T, Chen GY. Flavonoids and colorectal cancer prevention. Antioxidants 2018; 7(12) E187
[http://dx.doi.org/10.3390/antiox7120187] [PMID: 30544686]
[23]
Hosseini S, Chamani J, Hadipanah MR, et al. Nano-curcumin’s suppression of breast cancer cells (MCF7) through the inhibition of cyclinD1 expression. Breast Cancer (Dove Med Press) 2019; 11: 137-42.
[http://dx.doi.org/10.2147/BCTT.S195800] [PMID: 30936742]
[24]
Chen XR, Lu R, Dan HX, et al. Honokiol: a promising small molecular weight natural agent for the growth inhibition of oral squamous cell carcinoma cells. Int J Oral Sci 2011; 3(1): 34-42.
[http://dx.doi.org/10.4248/IJOS11014] [PMID: 21449214]
[25]
Theodoratou E, Kyle J, Cetnarskyj R, et al. Dietary flavonoids and the risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev 2007; 16(4): 684-93.
[http://dx.doi.org/10.1158/1055-9965.EPI-06-0785] [PMID: 17416758]
[26]
Yao LH, Jiang YM, Shi J, et al. Flavonoids in food and their health benefits. Plant Foods Hum Nutr 2004; 59(3): 113-22.
[http://dx.doi.org/10.1007/s11130-004-0049-7] [PMID: 15678717]
[27]
Mahapatra DK, Bharti SK, Asati V. Anti-cancer chalcones: Structural and molecular target perspectives. Eur J Med Chem 2015; 98: 69-114.
[http://dx.doi.org/10.1016/j.ejmech.2015.05.004] [PMID: 26005917]
[28]
Sahu NK, Balbhadra SS, Choudhary J, Kohli DV. Exploring pharmacological significance of chalcone scaffold: a review. Curr Med Chem 2012; 19(2): 209-25.
[http://dx.doi.org/10.2174/092986712803414132] [PMID: 22320299]
[29]
Karthikeyan C, Moorthy NS, Ramasamy S, et al. Advances in chalcones with anticancer activities. Recent Patents Anticancer Drug Discov 2015; 10(1): 97-115.
[http://dx.doi.org/10.2174/1574892809666140819153902] [PMID: 25138130]
[30]
Rejhová A, Opattová A, Čumová A, Slíva D, Vodička P. Natural compounds and combination therapy in colorectal cancer treatment. Eur J Med Chem 2018; 144: 582-94.
[http://dx.doi.org/10.1016/j.ejmech.2017.12.039] [PMID: 29289883]
[31]
Elkhalifa D, Alali F, Al Moustafa AE, Khalil A. Targeting triple negative breast cancer heterogeneity with chalcones: a molecular insight. J Drug Target 2019; 27(8): 830-8.
[http://dx.doi.org/10.1080/1061186X.2018.1561889] [PMID: 30582377]
[32]
Walle T. Methoxylated flavones, a superior cancer chemopreventive flavonoid subclass? Semin Cancer Biol 2007; 17(5): 354-62.
[http://dx.doi.org/10.1016/j.semcancer.2007.05.002] [PMID: 17574860]
[33]
Chahar MK, Sharma N, Dobhal MP, Joshi YC. Flavonoids: A versatile source of anticancer drugs. Pharmacogn Rev 2011; 5(9): 1-12.
[http://dx.doi.org/10.4103/0973-7847.79093] [PMID: 22096313]
[34]
Orlikova B, Menezes JC, Ji S, Kamat SP, Cavaleiro JA, Diederich M. Methylenedioxy flavonoids: assessment of cytotoxic and anti-cancer potential in human leukemia cells. Eur J Med Chem 2014; 84: 173-80.
[http://dx.doi.org/10.1016/j.ejmech.2014.07.003] [PMID: 25016375]
[35]
Keri RS, Chand K, Budagumpi S, Balappa Somappa S, Patil SA, Nagaraja BM. An overview of benzo[b]thiophene-based medicinal chemistry. Eur J Med Chem 2017; 138: 1002-33.
[http://dx.doi.org/10.1016/j.ejmech.2017.07.038] [PMID: 28759875]
[36]
Ricciardi R, Ghabreau L, Yasmeen A, Darnel AD, Akil N, Al Moustafa AE. Role of E6/E7 onco-proteins of high-risk human papillomaviruses in human colorectal carcinogenesis. Cell Cycle 2009; 8(12): 1964-5.
[http://dx.doi.org/10.4161/cc.8.12.8618] [PMID: 19411835]
[37]
Ghasemzadeh A, Jaafar HZ, Karimi E. Involvement of salicylic acid on antioxidant and anticancer properties, anthocyanin production and chalcone synthase activity in ginger (Zingiber officinale Roscoe) varieties. Int J Mol Sci 2012; 13(11): 14828-44.
[http://dx.doi.org/10.3390/ijms131114828] [PMID: 23203096]
[38]
Borowicz S, Van Scoyk M, Avasarala S, et al. The soft agar colony formation assay. J Vis Exp 2014; (92): e51998
[PMID: 25408172]
[39]
Roman D, Yasmeen A, Mireuta M, Stiharu I, Al Moustafa A-E. Significant toxic role for single-walled carbon nanotubes during normal embryogenesis. Nanomedicine (Lond) 2013; 9(7): 945-50.
[http://dx.doi.org/10.1016/j.nano.2013.03.010] [PMID: 23563045]
[40]
Soulières D, Greer W, Magliocco AM, et al. KRAS mutation testing in the treatment of metastatic colorectal cancer with anti-EGFR therapies. Current oncology (Toronto, Ont) 2010; 17 Suppl 1(Suppl 1): S31-40.
[41]
Ahmed D, Eide PW, Eilertsen IA, et al. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2013; 2 e71
[http://dx.doi.org/10.1038/oncsis.2013.35] [PMID: 24042735]
[42]
Shin SY, Ahn S, Koh D, Lim Y. p53-dependent and -independent mechanisms are involved in (E)-1-(2-hydroxyphenyl)-3-(2-methoxynaphthalen-1-yl)prop-2-en-1-one (HMP)-induced apoptosis in HCT116 colon cancer cells. Biochem Biophys Res Commun 2016; 479(4): 913-9.
[http://dx.doi.org/10.1016/j.bbrc.2016.09.067] [PMID: 27641669]
[43]
Semaan J, Pinon A, Rioux B, et al. Resistance to 3-HTMC-induced apoptosis through activation of PI3K/Akt, MEK/ERK, and p38/COX-2/PGE2 pathways in human HT-29 and HCT116 colorectal cancer cells. J Cell Biochem 2016; 117(12): 2875-85.
[http://dx.doi.org/10.1002/jcb.25600] [PMID: 27192488]
[44]
Karthikeyan C, Moorthy NSH, Ramasamy S, et al. Advances in Chalcones with Anticancer Activities 2015; 10(1): 97-115.
[45]
Takac P, Kello M, Pilatova MB, et al. New chalcone derivative exhibits antiproliferative potential by inducing G2/M cell cycle arrest, mitochondrial-mediated apoptosis and modulation of MAPK signalling pathway. Chem Biol Interact 2018; 292: 37-49.
[http://dx.doi.org/10.1016/j.cbi.2018.07.005] [PMID: 29981726]
[46]
Jin H, Seo GS, Lee SH. Isoliquiritigenin-mediated p62/SQSTM1 induction regulates apoptotic potential through attenuation of caspase-8 activation in colorectal cancer cells. Eur J Pharmacol 2018; 841: 90-7.
[http://dx.doi.org/10.1016/j.ejphar.2018.10.015] [PMID: 30339814]
[47]
Jin H, Kim HS, Seo GS, Lee SH. A new chalcone derivative, 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one), inhibits phorbol ester-induced metastatic activity of colorectal cancer cells through upregulation of heme oxygenase-1. Eur J Pharmacol 2018; 841: 1-9.
[http://dx.doi.org/10.1016/j.ejphar.2018.10.011] [PMID: 30321531]
[48]
Brandão P, Loureiro JB, Carvalho S, et al. Targeting the MDM2-p53 protein-protein interaction with prenylchalcones: Synthesis of a small library and evaluation of potential antitumor activity. Eur J Med Chem 2018; 156: 711-21.
[http://dx.doi.org/10.1016/j.ejmech.2018.07.037] [PMID: 30041135]
[49]
Wang G, Peng Z, Zhang J, Qiu J, Xie Z, Gong Z. Synthesis, biological evaluation and molecular docking studies of aminochalcone derivatives as potential anticancer agents by targeting tubulin colchicine binding site. Bioorg Chem 2018; 78: 332-40.
[http://dx.doi.org/10.1016/j.bioorg.2018.03.028] [PMID: 29627654]
[50]
Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer 2004; 4(4): 253-65.
[http://dx.doi.org/10.1038/nrc1317] [PMID: 15057285]
[51]
Boumendjel A, Boccard J, Carrupt PA, et al. Antimitotic and antiproliferative activities of chalcones: forward structure-activity relationship. J Med Chem 2008; 51(7): 2307-10.
[http://dx.doi.org/10.1021/jm0708331] [PMID: 18293907]
[52]
Ducki S, Mackenzie G, Greedy B, et al. Combretastatin-like chalcones as inhibitors of microtubule polymerisation. Part 2: Structure-based discovery of alpha-aryl chalcones. Bioorg Med Chem 2009; 17(22): 7711-22.
[http://dx.doi.org/10.1016/j.bmc.2009.09.044] [PMID: 19837594]
[53]
Ducki S, Rennison D, Woo M, et al. Combretastatin-like chalcones as inhibitors of microtubule polymerization. Part 1: synthesis and biological evaluation of antivascular activity. Bioorg Med Chem 2009; 17(22): 7698-710.
[http://dx.doi.org/10.1016/j.bmc.2009.09.039] [PMID: 19837593]
[54]
Romagnoli R, Baraldi PG, Carrion MD, et al. Design, synthesis, and biological evaluation of thiophene analogues of chalcones. Bioorg Med Chem 2008; 16(10): 5367-76.
[http://dx.doi.org/10.1016/j.bmc.2008.04.026] [PMID: 18440234]
[55]
Vitorović-Todorović MD, Erić-Nikolić A, Kolundžija B, et al. (E)-4-aryl-4-oxo-2-butenoic acid amides, chalcone-aroylacrylic acid chimeras: design, antiproliferative activity and inhibition of tubulin polymerization. Eur J Med Chem 2013; 62: 40-50.
[http://dx.doi.org/10.1016/j.ejmech.2013.01.006] [PMID: 23353745]
[56]
Hsu YL, Chia CC, Chen PJ, Huang SE, Huang SC, Kuo PL. Shallot and licorice constituent isoliquiritigenin arrests cell cycle progression and induces apoptosis through the induction of ATM/p53 and initiation of the mitochondrial system in human cervical carcinoma HeLa cells. Mol Nutr Food Res 2009; 53(7): 826-35.
[http://dx.doi.org/10.1002/mnfr.200800288] [PMID: 19536869]
[57]
Shen KH, Chang JK, Hsu YL, Kuo PL. Chalcone arrests cell cycle progression and induces apoptosis through induction of mitochondrial pathway and inhibition of nuclear factor kappa B signalling in human bladder cancer cells. Basic Clin Pharmacol Toxicol 2007; 101(4): 254-61.
[http://dx.doi.org/10.1111/j.1742-7843.2007.00120.x] [PMID: 17845507]
[58]
Kim N. Butein sensitizes human leukemia cells to apoptosis induced by tumor necrosis factor-related apoptosis inducing ligand (TRAIL). Arch Pharm Res 2008; 31(9): 1179-86.
[http://dx.doi.org/10.1007/s12272-001-1286-2] [PMID: 18806962]
[59]
Yoshida T, Horinaka M, Takara M, et al. Combination of isoliquiritigenin and tumor necrosis factor-related apoptosis-inducing ligand induces apoptosis in colon cancer HT29 cells. Environ Health Prev Med 2008; 13(5): 281-7.
[http://dx.doi.org/10.1007/s12199-008-0041-1] [PMID: 19568915]
[60]
Entschladen F, Drell TL IV, Lang K, Joseph J, Zaenker KS. Tumour-cell migration, invasion, and metastasis: navigation by neurotransmitters. Lancet Oncol 2004; 5(4): 254-8.
[http://dx.doi.org/10.1016/S1470-2045(04)01431-7] [PMID: 15050959]
[61]
Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 1990; 82(1): 4-6.
[http://dx.doi.org/10.1093/jnci/82.1.4] [PMID: 1688381]
[62]
Al-Abd AM, Alamoudi AJ, Abdel-Naim AB, Neamatallah TA, Ashour OM. Anti-angiogenic agents for the treatment of solid tumors: Potential pathways, therapy and current strategies - A review. J Adv Res 2017; 8(6): 591-605.
[http://dx.doi.org/10.1016/j.jare.2017.06.006] [PMID: 28808589]
[63]
Elkhalifa D, Siddique AB, Qusa M, et al. Design, synthesis, and validation of novel nitrogen-based chalcone analogs against triple negative breast cancer. Eur J Med Chem 2020; 187 111954
[http://dx.doi.org/10.1016/j.ejmech.2019.111954] [PMID: 31838326]
[64]
Sil Lee Y, Lim S, Hyun Shin K, Shik Kim Y, Ohuchi K, Hoon Jung S. Anti-angiogenic and anti-tumor activities of 2'-hydroxy-4'- methoxychalcone 2006; 1028-31.

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