Synthesis of three novel curcumin analog compounds and their activity tests against breast cancer based on in vitro, network pharmacology, and molecular docking assessments

Endang Astuti Jihan Alfiyah Kultsum Zarah Aulia Frika Rahmawari Kasta Gurning Sugeng Triono Winarto Haryadi Harno Dwi Pranowo   

Open Access   

Published:  Aug 06, 2025

DOI: 10.7324/JAPS.2025.231151
Abstract

Breast cancer, the most common disease suffered by women, causes high mortality compared to other cancer types. This study aims to synthesize mono-carbonyl curcumin analogs and test their cytotoxic activity against four breast cancer cell lines, i.e., MCF-7, T47D, 4T1, and HER2, along with bioinformatics analysis, molecular docking, and pharmacokinetic profile prediction evaluation. Curcumin analogs were produced through aldol condensation, which reacting 3-bromo-4-methoxybenzaldehyde with various ketones to form N-benzyl-4-piperidone (A), 4-piperidone (B), and N-methyl-4-piperidone (C). Pharmacokinetic profiles showed that compounds AC showed improved absorption, tissue distribution, retention, and toxicity profiles compared to curcumin. However, these compounds did not fully meet the Lipinski criteria, indicating limitations in oral bioavailability. Compound B exhibited the highest anticancer activity against T47D (IC50 = 19.20 μg/ml) and HER2 cells (IC50 = 30.70 μg/ml). Compound A was found to the best against 4T1 cells (IC50 = 174.05 μg/ml) while compound C gave the best activity on MCF-7 cells (IC50 = 110.91 μg/ml). All compounds showed low efficacy, with a selectivity index value of lower than 3. Bioinformatics studies of the network pharmacology approach on cancer pathways showed that mitogen-activated protein kinase 8 protein became the main target of compound A while AKT1 protein was for compounds B and C. Further improvement in their activity and selectivity through targeted delivery systems and further molecular studies are recommended to enhance their therapeutic potential.


Keyword:     ADMET bioinformatic breast cancer curcumin analog molecular docking


Citation:

Astuti E, Kultsum JA, Aulia Z, Rahmawari F, Gurning K, Triono S, Haryadi W, Pranowo HD. Synthesis of three novel curcumin analog compounds and their activity tests against breast cancer based on in vitro, network pharmacology, and molecular docking assessments. J Appl Pharm Sci. 2025. Article in Press. http://doi.org/10.7324/JAPS.2025.231151

Copyright: © The Author(s). This is an open-access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

HTML Full Text

Reference

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49. https://doi.org/10.3322/caac.21660

2. Thakur C, Qiu Y, Fu Y, Bi Z, Zhang W, Ji H, et al. Epigenetics and environment in breast cancer: new paradigms for anti-cancer therapies. Front Oncol. 2022;12:971288. https://doi.org/10.3389/fonc.2022.971288

3. Novitasari D, Jenie RI, Utomo RY, Kato JY, Meiyanto E. CCA-1.1, a novel curcumin analog, exerts cytotoxic anti-migratory activity toward TNBC and HER2-enriched breast cancer cells. Asian Pac J Cancer Prev. 2021;22:1827–36. https://doi.org/10.31557/APJCP.2021.22.6.1827

4. Burguin A, Diorio C, Durocher F. Breast cancer treatments: updates and new challenges. J Pers Med. 2021;11:808. https://doi.org/10.3390/jpm11080808

5. Stoicescu EA, Iancu RC, Cherecheanu AP, Iancu G. Ocular adverse effects of anti-cancer chemotherapy. J Med Life. 2023;16:818–21. https://doi.org/10.25122/jml-2023-0041

6. Khudhayer Oglah M, Fakri Mustafa Y. Curcumin analogs: synthesis and biological activities. Med Chem Res. 2020;29:479–86. https://doi.org/10.1007/s00044-019-02497-0

7. Wolosewicz K, Podgorska K, Rutkowska E, Lazny R. Synthesis of Dicarbonyl curcumin analogues containing the tropane scaffold. European J Org Chem. 2019;2019:4662–74. https://doi.org/10.1002/ejoc.201900416

8. Ardiansah B, Hardhani MR, Putera DDSR, Wukirsari T, Cahyana AH, Jia JW, et al. Design, synthesis, and antioxidant evaluation of monocarbonyl curcumin analogues tethered 1,2,3-triazole scaffold. Case Stud Chem Environ Eng. 2023;8:100425. https://doi.org/10.1016/j.cscee.2023.100425

9. Hani U, Jaswanth Gowda BH, Siddiqua A, Wahab S, Begum Y, Sathishbabu P, et al. Herbal approach for treatment of cancer using curcumin as an anticancer agent: a review on novel drug delivery systems. J Mol Liq. 2023;390:123037. https://doi.org/10.1016/j.molliq.2023.123037

10.Kunnumakkara AB, Bordoloi D, Padmavathi G, Monisha J, Roy NK, Prasad S, et al. Curcumin, the golden nutraceutical: multitargeting for multiple chronic diseases. Br J Pharmacol. 2017;174:1325–48. https://doi.org/10.1111/bph.13621

11. Al-Mulla A. A review: biological importance of heterocyclic compounds. Der Pharma Chemica. 2017;9:141–7.

12. Ansari A, Ali A, Asif M, Shamsuzzaman. Review: biologically active pyrazole derivatives. New J Chem. 2016;41:16–41. https://doi.org/10.1039/c6nj03181a

13. Al-Hujaily EM, Mohamed AG, Al-Sharif I, Youssef KM, Manogaran PS, Al-Otaibi B, et al. PAC, a novel curcumin analogue, has anti-breast cancer properties with higher efficiency on ER-negative cells. Breast Cancer Res Treat. 2011;128:97–107. https://doi.org/10.1007/s10549-010-1089-3

14. Adams BK, Cai J, Armstrong J, Herold M, Lu YJ, Sun A, et al. EF24, a novel synthetic curcumin analog, induces apoptosis in cancer cells via a redox-dependent mechanism. Anticancer Drugs. 2005;16:263– 75. https://doi.org/10.1097/00001813-200503000-00005

15. Yuan X, Li H, Bai H, Su Z, Xiang Q, Wang C, et al. Synthesis of novel curcumin analogues for inhibition of 11β-hydroxysteroid dehydrogenase type 1 with anti-diabetic properties. Eur J Med Chem. 2014;77:223–30. https://doi.org/10.1016/j.ejmech.2014.03.012

16. Gurning K, Suratno S, Astuti E, Haryadi W. Untargeted LC/HRMS metabolomics analysis and anticancer activity assay on MCF-7 and A549 cells from coleus amboinicus lour leaf extract. IJ Pharm Res. 2024;23:e143494. https://doi.org/10.5812/IJPR-143494

17. Islamie R, Iksen I, Buana BC, Gurning K, Syahputra HD, Winata HS. Construction of network pharmacology-based approach and potential mechanism from major components of Coriander sativum L. against COVID-19. Pharmacia. 2022;69:689–97. https://doi.org/10.3897/pharmacia.69.e84388

18. Haryadi W, Gurning K, Astuti E. Molecular target identification of two Coleus amboinicus leaf isolates toward lung cancer using a bioinformatic approach and molecular docking-based assessment. J Appl Pharm Sci. 2024;14:203–10. https://doi.org/10.7324/JAPS.2024.164753

19. Haryadi W, Gurning K, Fachiroh J, Astuti E. Potential of bioactive compounds in Coleus amboinicus, Lour., leaves against breast cancer by assessment using a network pharmacology approach and cytotoxic test. J Multidisciplinary Appl Nat Sci. 2025;5:267–87. https://doi.org/10.47352/JMANS.2774-3047.246

20. Liu J, Liu J, Tong X, Peng W, Wei S, Sun T, et al. Network pharmacology prediction and molecular docking-based strategy to discover the potential pharmacological mechanism of huai hua san against ulcerative colitis. Drug Des Devel Ther. 2021;15:3255–76. https://doi.org/10.2147/DDDT.S319786

21. Haryadi W, Pranowo HD. Molecular docking and dynamics analysis of halogenated imidazole chalcone as anticancer compounds. Pharmacia. 2023;70:323–9. https://doi.org/10.3897/PHARMACIA.70.E101989

22. Bitew M, Desalegn T, Demissie TB, Belayneh A, Endale M, Eswaramoorthy R. Pharmacokinetics and drug-likeness of antidiabetic flavonoids: Molecular docking and DFT study. PLoS One. 2021;16:e0260853. https://doi.org/10.1371/journal.pone.0260853

23. Hou T, Wang J, Zhang W, Xu X. ADME evaluation in drug discovery. 7. prediction of oral absorption by correlation and classification. J Chem Inf Model. 2007;47:e0260853. https://doi.org/10.1021/ci600343x

24. Smith DA, Beaumont K, Maurer TS, Di L. Volume of distribution in drug design. J Med Chem. 2015;58:5691–8. https://doi.org/10.1021/acs.jmedchem.5b00201

25. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013;138:103–41. https://doi.org/10.1016/j.pharmthera.2012.12.007

26. Bertholee D, Maring JG, van Kuilenburg ABP. Genotypes affecting the pharmacokinetics of anticancer drugs. Clin Pharmacokinet. 2017;56:317–37. https://doi.org/10.1007/s40262-016-0450-z

27. Maurya R, Vikal A, Patel P, Narang RK, Kurmi B Das. “Enhancing oral drug absorption: overcoming physiological and pharmaceutical barriers for improved bioavailability.” AAPS PharmSciTech. 2024;25:228. https://doi.org/10.1208/s12249-024-02940-5

28. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46:3–26. https://doi.org/10.1016/S0169-409X(96)00423-1

29. Liu Q, Liu N, van der Noord V, van der Stel W, van de Water B, Danen EHJ, et al. Differential response of luminal and basal breast cancer cells to acute and chronic hypoxia. Breast Cancer Res Treat. 2023;198:583–96. https://doi.org/10.1007/s10549-023-06863-w

30. Rabinovich I, De Noronha L, Sebastião APM, Lima RS, Urban CA, Schunemann E, et al. HER2-expressing breast tumors are associated with breast cancer stem-cell phenotype CD44 + /CD24. J Bras Patol Med Lab. 2018;54:310–8. https://doi.org/10.5935/1676-2444.20180052

31. Arroyo-Crespo JJ, Armiñán A, Charbonnier D, Deladriere C, Palomino-Schätzlein M, Lamas-Domingo R, et al. Characterization of triple-negative breast cancer preclinical models provides functional evidence of metastatic progression. Int J Cancer. 2019;145:2267–81. https://doi.org/10.1002/ijc.32270

32. Fernandes TB, Damião MFCB, Polli MC, Filho RP. Analysis of the applicability and use of Lipinski‘s rule for central nervous system drugs. Lett Drug Des Discov. 2016;13:999–1006. https://doi.org/10.2174/1570180813666160622092839

33. Ugwu DI, Conradie J. Anticancer properties of complexes derived from bidentate ligands. J Inorg Biochem. 2023;246:112268. https://doi.org/10.1016/j.jinorgbio.2023.112268

34. Romano JD, Tatonetti NP. Informatics and computational methods in natural product drug discovery: a review and perspectives. Front Genet. 2019;10:442506. https://doi.org/10.3389/FGENE.2019.00368/BIBTEX

35. Xu P, Zhang G, Hou S, Sha L gui. MAPK8 mediates resistance to temozolomide and apoptosis of glioblastoma cells through MAPK signaling pathway. Biomed Pharmacother. 2018;106:1419–27. https://doi.org/10.1016/J.BIOPHA.2018.06.084

36. Marinello PC, Panis C, Silva TNX, Binato R, Abdelhay E, Rodrigues JA, et al. Metformin prevention of doxorubicin resistance in MCF- 7 and MDA-MB-231 involves oxidative stress generation and modulation of cell adaptation genes. Sci Rep. 2019;9:1–11. https://doi.org/10.1038/s41598-019-42357-w

37. Wang R, Zhao Y. Effects of metformin on JNK signaling pathway and PD-L1 expression in triple negative breast cancer. Cancer Manag Res. 2024;16:259–68. https://doi.org/10.2147/CMAR.S454960

38. Zhang YH, Zeng T, Chen L, Huang T, Cai YD. Determining protein–protein functional associations by functional rules based on gene ontology and KEGG pathway. Biochimica et Biophysica Acta (BBA)—Proteins Proteomics. 2021;1869:140621. https://doi. org/10.1016/J.BBAPAP.2021.140621

39. Hinz N, Jücker M. Distinct functions of AKT isoforms in breast cancer: a comprehensive review. Cell Commun Signaling. 2019;17:1–29. https://doi.org/10.1186/S12964-019-0450-3

40. Shariati M, Meric-Bernstam F. Targeting AKT for cancer therapy. Expert Opin Investig Drugs. 2019;28:977–88. https://doi.org/10.108 0/13543784.2019.1676726

41. Miricescu D, Totan A, Stanescu-Spinu II, Badoiu SC, Stefani C, Greabu M. PI3K/AKT/mTOR Signaling pathway in breast cancer: from molecular landscape to clinical aspects. Int J Mol Sci. 2020;22:173. https://doi.org/10.3390/IJMS22010173

42. Martorana F, Motta G, Pavone G, Motta L, Stella S, Vitale SR, et al. AKT Inhibitors: new weapons in the fight against breast cancer? Front Pharmacol. 2021;12:662232. https://doi.org/10.3389/FPHAR.2021.662232/BIBTEX

43. Srinivasan M, Gangurde A, Chandane AY, Tagalpallewar A, Pawar A, Baheti AM. Integrating network pharmacology and in silico analysis deciphers Withaferin-A’s anti- breast cancer potential via hedgehog pathway and target network interplay. Brief Bioinform. 2024;25:bbae032. https://doi.org/10.1093/BIB/BBAE032

44. Khzem AH Al, Alturki MS, Almuzaini OK, Wali SM, Almaghrabi M, Aldawsari MF, et al. Isoetin from Isoetaceae exhibits superior pentatransferase inhibition in breast cancer: comparative computational profiling with FDA-approved Tucatinib. Pharmaceuticals. 2025;18:662. https://doi.org/10.3390/PH18050662

Article Metrics
4 Views 2 Downloads 6 Total

Year

Month

Related Search

By author names