There is a significant concentration of bioactive substances in the cashew plant, which is known scientifically as Anacardium occidentale L. These compounds, which are primarily found in the spongy shells of cashew nuts, include phenolic lipids. The liquid that is taken from the shell of the cashew nut contains a lot of cardanol, which is a chemical that has many different applications, the most important of which is that it is a versatile building block that can be used in a variety of different sectors. In addition to their application in industry, cashews and the bioactive components that they contain are well-known for their important biological impacts. In this work, the anticancer potential of cardanol, which is a primary component of cashew nut shell liquid, is evaluated through the use of in silico analysis. This analysis is essential for determining the principal gene targets and cellular signaling pathways that are affected by cardanol in the setting of oral cancer. During the research, 23 genes were discovered to be involved in a wide range of biological processes. These processes include cell migration, proliferation, apoptosis regulation, PI3K-AKT pathway regulation, and cell cycle control. The genes PIK3CA, CCND1, MMP1, and MTOR were the ones that were implicated in the top 10 pathways the most frequently. These genes are all closely related to the PI3K signaling pathway, which suggests that this system may be essential for Cardanol’s potential therapeutic effects in treating oral cancer. Cardanol has been shown to have a substantial anti-proliferative effect on oral cancer, which indicates that it has the potential to be used as a therapeutic agent, according to the findings of the in-silico assessment. These findings may contribute to developing oral cancer treatments that are more effective and suited to the patient’s specific needs, ultimately resulting in improved treatment outcomes.
Jadhav V, Maini A, Ladke VS. Investigating the anti-cancer activity of cardanol a component of cashew nut shell liquid in oral cancer using in silico analysis. J Appl Pharm Sci. 2025. Online First. http://doi.org/10.7324/JAPS.2025.215258
1. Bray F, Laversanne M, Weiderpass E, Soerjomataram I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer. 2021 Aug;127(16):3029-30. https://doi.org/10.1002/cncr.33587 | |
2. Chen Z, Zhang P, Xu Y, Yan J, Liu Z, Lau WB, et al. Surgical stress and cancer progression: the twisted tango. Mol Cancer. 2019 Sep;118(1):132. https://doi.org/10.1186/s12943-019-1058-3 | |
3. Büchner FL, Bueno-de-Mesquita HB, Linseisen J, Boshuizen HC, Kiemeney LALM, Ros MM, et al. Fruits and vegetables consumption and the risk of histological subtypes of lung cancer in the European prospective investigation into cancer and nutrition (EPIC). Cancer Causes Control. 2010;21(3):357-71. https://doi.org/10.1007/s10552-009-9468-y | |
4. Soerjomataram I, Oomen D, Lemmens V, Oenema A, Benetou V, Trichopoulou A, et al. Increased consumption of fruit and vegetables and future cancer incidence in selected European countries. Eur J Cancer. 2010;46(14):2563-80. https://doi.org/10.1016/j.ejca.2010.07.026 | |
5. Ogunsina BS, Bamgboye AI. Pre-shelling parameters and conditions that influence the whole kernel out-turn of steam-boiled cashew nuts. J Saudi Soc Agric Sci. 2014 Jan;13(1):29-34. https://doi.org/10.1016/j.jssas.2012.12.005 | |
6. Kubo I, Masuoka N, Ha TJ, Tsujimoto K. Antioxidant activity of anacardic acids. Food Chem. 2006;99(3):555-62. https://doi.org/10.1016/j.foodchem.2005.08.023 | |
7. Balachandran VS, Jadhav SR, Vemula PK, John G. Recent advances in cardanol chemistry in a nutshell: from a nut to nanomaterials. Chem Soc Rev. 2013;42(2):427-38. https://doi.org/10.1039/C2CS35344J | |
8. Bastos FA, Tubino M. The use of the liquid from cashew nut shells as an antioxidant in biodiesel. J Braz Chem Soc. 2017;28(5):747-55. https://doi.org/10.21577/0103-5053.20160223 | |
9. Voirin C, Caillol S, Sadavarte NV, Tawade BV, Boutevin B, Wadgaonkar PP. Functionalization of cardanol: towards biobased polymers and additives. Polym Chem. 2014 Apr;5(9):3142-62. https://doi.org/10.1039/C3PY01194A | |
10. Itokawa H, Totsuka N, Nakahara K, Takeya K, Lepoittevin JP, Asakawa Y. Antitumor principles from Ginkgo biloba L. Chem Pharm Bull (Tokyo). 1987;35(7):3016-20. https://doi.org/10.1248/cpb.35.3016 | |
11. Zou Y, Zhao Q, Hu H, Hu L, Yu S, Xu M, et al. Synthesis and in vitro antitumor activities of xanthone derivatives containing 1,4-disubstituted-1,2,3-triazole moiety. Arch Pharm Res. 2012 Dec;35(12):2093-104. https://doi.org/10.1007/s12272-012-1206-4 | |
12. Narsimha S, Satheesh Kumar N, Kumara Swamy B, Vasudeva Reddy N, Althaf Hussain SK, Srinivasa Rao M. Indole-2-carboxylic acid derived mono and bis 1,4-disubstituted 1,2,3-triazoles: synthesis, characterization and evaluation of anticancer, antibacterial, and DNA-cleavage activities. Bioorg Med Chem Lett. 2016;26(6):1639-44. https://doi.org/10.1016/j.bmcl.2016.01.055 | |
13. Chen Y, Lopez-Sanchez M, Savoy DN, Billadeau DD, Dow GS, Kozikowski AP. A series of potent and selective, triazolylphenyl-based histone deacetylases inhibitors with activity against pancreatic cancer cells and Plasmodium falciparum. J Med Chem. 2008 Jun;51(12):3437-48. https://doi.org/10.1021/jm701606b | |
14. Bratsos I, Urankar D, Zangrando E, Genova-Kalou P, Košmrlj J, Alessio E, et al. 1-(2-Picolyl)-substituted 1,2,3-triazole as novel chelating ligand for the preparation of ruthenium complexes with potential anticancer activity. Dalton Trans. 2011 May;40(19):5188-99. https://doi.org/10.1039/c0dt01807d | |
15. Teerasripreecha D, Phuwapraisirisan P, Puthong S, Kimura K, Okuyama M, Mori H, et al. In vitro antiproliferative/cytotoxic activity on cancer cell lines of a cardanol and a cardol enriched from Thai Apis mellifera propolis. BMC Complement Altern Med. 2012 Mar;12:27. https://doi.org/10.1186/1472-6882-12-27 | |
16. Trevisan MTS, Pfundstein B, Haubner R, Würtele G, Spiegelhalder B, Bartsch H, et al. Characterization of alkyl phenols in cashew (Anacardium occidentale) products and assay of their antioxidant capacity. Food Chem Toxicol. 2006 Feb;44(2):188-97. https://doi.org/10.1016/j.fct.2005.06.012 | |
17. Azmi AS. Adopting network pharmacology for cancer drug discovery. Curr Drug Discov Technol. 2013;10(2):95-105. https://doi.org/10.2174/1570163811310020002 | |
18. Tang J, Aittokallio T. Network pharmacology strategies toward multi-target anticancer therapies: from computational models to experimental design principles. Curr Pharm Des. 2014;20(1):23-36. https://doi.org/10.2174/13816128113199990470 | |
19. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017 Mar;7:42717. https://doi.org/10.1038/srep42717 | |
20. Daina A, Michielin O, Zoete V. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019 Jul;47(W1):W357-3664. https://doi.org/10.1093/nar/gkz382 | |
21. Buva K, Kumbhar GM, Deshmukh A, Ladke VS. The assessment of the mechanism of action of lauric acid in the context of oral cancer through integrative approach combining network pharmacology and molecular docking technology. J Complement Integr Med. 2024 Mar;21(1):101-12. https://doi.org/10.1515/jcim-2023-0262 | |
22. Kumbhar GM, Jadhav AD, Kheur S, Ladke VS. Andrographolide demonstrates anti-proliferative activity in oral cancer by promoting apoptosis, the programmed cell death process. Iran J Basic Med Sci. 2024;27(10):1300-8. | |
23. Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021 Jan;49(D1):D545-51. https://doi.org/10.1093/nar/gkaa970 | |
24. Ladke VS, Kumbhar GM, Joshi K, Kheur S. Systemic explanation of Glycyrrhiza glabra's analyzed compounds and anti-cancer mechanism based on network pharmacology in oral cancer. J Oral Biosci. 2022;64(4):452-60. https://doi.org/10.1016/j.job.2022.09.002 | |
25. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000 May;25(1):25-9. https://doi.org/10.1038/75556 | |
26. Tang D, Chen M, Huang X, Zhang G, Zeng L, Zhang G, et al. SRplot: a free online platform for data visualization and graphing. PLoS One. 2023 Nov;18(11):e0294236. https://doi.org/10.1371/journal.pone.0294236 | |
27. Lin JS, Lai EM. Protein-protein interactions: co-immunoprecipitation. Methods Mol Biol. 2017;1615:211-9. https://doi.org/10.1007/978-1-4939-7033-9_17 | |
28. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003 Nov;13(11):2498-504. https://doi.org/10.1101/gr.1239303 | |
29. Liu Y, Grimm M, Dai W, Hou MC, Xiao ZX, Cao Y. CB-Dock: a web server for cavity detection-guided protein-ligand blind docking. Acta Pharmacol Sin. 2020 Jan;41(1):138-44. https://doi.org/10.1038/s41401-019-0228-6 | |
30. Chandrashekar DS, Karthikeyan SK, Korla PK, Patel H, Shovon AR, Athar M, et al. UALCAN: an update to the integrated cancer data analysis platform. Neoplasia. 2022 Mar;25:18-27. https://doi.org/10.1016/j.neo.2022.01.001 | |
31. Ghafouri-Fard S, Noie Alamdari A, Noee Alamdari Y, Abak A, Hussen BM, Taheri M, et al. Role of PI3K/AKT pathway in squamous cell carcinoma with an especial focus on head and neck cancers. Cancer Cell Int. 2022 Aug;22(1):254. https://doi.org/10.1186/s12935-022-02676-x | |
32. Argiris A, Brockstein BE, Haraf DJ, Stenson KM, Mittal BB, Kies MS, et al. Competing causes of death and second primary tumors in patients with locoregionally advanced head and neck cancer treated with chemoradiotherapy. Clin Cancer Res. 2004 Mar;10(6):1956-62. https://doi.org/10.1158/1078-0432.CCR-03-1077 | |
33. Vokes EE, Weichselbaum RR, Lippman SM, Hong WK. Head and neck cancer. N Engl J Med. 1993;328(3):184-94. https://doi.org/10.1056/NEJM199301213280306 | |
34. Lemes LFN, De Andrade Ramos G, De Oliveira AS, Da Silva FMR, De Castro Couto G, Da Silva Boni M, et al. Cardanol-derived AChE inhibitors: towards the development of dual binding derivatives for Alzheimer's disease. Eur J Med Chem. 2016 Jan;108:687-700. https://doi.org/10.1016/j.ejmech.2015.12.024 | |
35. Okoye FAN, Amaefule CC. Evaluation of the cytotoxicity and genotoxicity of aqueous leaf extracts of Azadirachta indica A. Juss using the Allium test. J Med Plants Res. 2012 Jun;6(22):3898-907. https://doi.org/10.5897/JMPR12.427 | |
36. de Sousa Leite A, Vieira Gomes DC, da Silva Oliveira GL, Correia Jardim Paz MF, Melo de Carvalho R, Oliveira Ferreira da Mata AM, et al. Net effects of cashew nuts in Saccharomyces cerevisiae front to damage induced by hydrogen peroxide. Int Arch Med. 2016;9(134):1-13. https://doi.org/10.3823/2005 | |
37. Edziri H, Mastouri M, Aouni M, Anthonissen R, Verschaeve L. Investigation on the genotoxicity of extracts from Cleome amblyocarpa Barr. and Murb, an important Tunisian medicinal plant. South Afr J Bot. 2013;84:102-3. https://doi.org/10.1016/j.sajb.2012.10.005 | |
38. Park S, Ohta T, Kumazawa S, Jun M, Ahn MR. Korean propolis suppresses angiogenesis through inhibition of tube formation and endothelial cell proliferation. Nat Prod Commun. 2014;9(4):555-60. https://doi.org/10.1177/1934578X1400900434 | |
39. Melo Cavalcante AA, Rubensam G, Picada JN, Gomes da Silva EG, Fonseca Moreira JC, Henriques JAP. Mutagenicity, antioxidant potential, and antimutagenic activity against hydrogen peroxide of cashew (Anacardium occidentale) apple juice and cajuina. Environ Mol Mutagen. 2003;41(5):360-9. https://doi.org/10.1002/em.10158 | |
40. Rodrigues FHA, Feitosa JPA, Ricardo NMPS, De França FCF, Carioca JOB. Antioxidant activity of cashew nut shell liquid (CNSL) derivatives on the thermal oxidation of synthetic cis-1,4-polyisoprene. J Braz Chem Soc. 2006;17(2):265-71. https://doi.org/10.1590/S0103-50532006000200008 | |
41. De Lima SG, Feitosa CM, Citó AM, Moita Neto JM, Lopes JA, Leite AS, et al. Effects of immature cashew nut-shell liquid (Anacardium occidentale) against oxidative damage in Saccharomyces cerevisiae and inhibition of acetylcholinesterase activity. Genet Mol Res. 2008;7(3):806-18. https://doi.org/10.4238/vol7-3gmr473 | |
42. Neto L, Matos N, Gonzaga W, Romeiro L, Santos M, Santos D, et al. Characterization of cytotoxic activity of compounds derived from anacardic acid, cardanol and cardol in oral squamous cell carcinoma. BMC Proc. 2014 Oct;8(Suppl 4):P30. https://doi.org/10.1186/1753-6561-8-S4-P30 |
Year
Month