Next Article in Journal
In Silico Analyses Indicate a Lower Potency for Dimerization of TLR4/MD-2 as the Reason for the Lower Pathogenicity of Omicron Compared to Wild-Type Virus and Earlier SARS-CoV-2 Variants
Previous Article in Journal
Targeting the Multiple Complex Processes of Hypoxia-Ischemia to Achieve Neuroprotection
Previous Article in Special Issue
Apoptosis Induction in HepG2 and HCT116 Cells by a Novel Quercetin-Zinc (II) Complex: Enhanced Absorption of Quercetin and Zinc (II)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Roles of Phytochemicals in Cancer Prevention and Therapeutics

by
Daniel Gabriel Pons
1,2
1
Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain
2
Grupo Multidisciplinar de Oncología Traslacional, Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain
Int. J. Mol. Sci. 2024, 25(10), 5450; https://doi.org/10.3390/ijms25105450
Submission received: 13 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024
(This article belongs to the Special Issue Roles of Phytochemicals in Cancer Prevention and Therapeutics)
This Special Issue focused on the importance of phytochemicals for their use in the prevention and treatment of cancer. The impact of publications about the effects of phytochemicals on mammal cells has grown in the last two decades due to their antioxidant, anti-inflammatory, and antimicrobial properties at low or moderate concentrations. However, there is some controversy about the effects of phytochemicals on cancer cells at these concentration levels. In fact, the mentioned properties could help cancer cells survive, conferring resistance to chemotherapeutic agents and, consequently, cell death. On the other hand, high concentrations of these compounds can trigger drastic changes in cancer cells’ physiology, directly affecting their cell viability. Considering all these facts, the study of the roles of phytochemicals in cancer prevention and therapeutics is of great interest because of the dual effects they may have on both normal and cancer cells, depending, as well, on their concentration. In this sense, the first published article of this Special Issue was a study of the effects of high concentrations of genistein, an isoflavone mainly found in soybeans [1], on the viability of colon cancer cells depending on the modulation of oxidative stress and inflammation [2]. Previous studies in breast cancer have demonstrated that genistein, at physiological concentrations, could regulate the estrogenic response by acting as a phytoestrogen, affecting mitochondrial functionality and, therefore, inflammation, oxidative stress, and cell proliferation in breast cancer cell lines with different amounts of the estrogen receptors α and β [3,4,5]. Moreover, the phytoestrogen genistein is able to modulate the effects of chemotherapeutic agents on breast cancer cells, modulating mitochondrial functionality and depending on the estrogen receptor ratio [6]. Other phytochemicals have shown different effects on mitochondrial-related parameters, indicating the importance of this organelle in the hallmarks of cancer [7,8,9,10]. There are studies indicating that the accumulation of high concentrations of genistein in certain areas of the colon mucosa could be related to its effects on colon cells [11]. Alorda-Clara et al. have demonstrated a relationship between high concentrations of genistein treatment and a decrease in cell viability through modulation of mitochondrial biogenesis, oxidative stress, and the inflammatory status of colon cancer cells [2].
Interestingly, the other five original articles published in this Special Issue have focused on different phytochemicals and their effects on different cancer types. Augustynowicz et al. studied the anticancer potential effects of rare Potentilla species extracts containing phenolic, tannin, and flavonoid compounds on colon cells [12]. Some of the extracts showed anticancer properties, damaging colon cancer cell membranes, but did not reveal any cytotoxic effect against colon epithelial cells. The same authors have reported similar results, demonstrating that all Potentilla species may be useful sources for anticancer agents against colon tumors [13].
More concretely, Ma et al. have studied the effects of mulberry Diels-Alder-type adducts (MDAAs), and specifically the Kuwanon M (KWM) from the root bark, on apoptosis and paraptosis of lung cancer cells, associated with endoplasmic reticulum stress [14]. KWM reduced cell proliferation and migration and, at the same time, increased apoptosis through the mitochondrial pathway and paraptosis through an increment in cytoplasmic vacuolation and ER stress in A549 and NCI-H292 lung cancer cells [14]. In another study, MDAAs showed that their anticancer effects increase cell apoptosis [15], but remarkably, Ma et al. determined that KWM could affect mitochondria directly, corroborating the importance of this organelle in the response to phytochemical treatment in cancer cells [14].
In addition, the other three original research papers published in this Special Issue have studied different classical phytochemicals such as caffeine, butein, and a complex quercetin-zinc(II). Eguchi et al. analyzed the increment of anticancer drug toxicity by caffeine in a spheroid model of human lung adenocarcinoma through the reduction of the protein expression of Claudin-2 and Nrf2, affecting mitochondrial respiration and ROS production [16]. Previous studies have demonstrated the relationship between oxidative stress and Nrf2 signaling, which is linked to increased chemoresistance [17]. The results confirmed the exaggeration of doxorubicin and cisplatin toxicity mediated by caffeine treatment in these spheroids [16]. On the other hand, Park et al. revealed that butein, a flavonoid identified from Butea monosperma, inhibited cell growth by blocking IL-6/IL-6Rα interaction and by regulating the IL-6/STAT3/FoxO3a pathway in human ovarian cancer cells through the higher binding affinity of butien to IL-6 [18]. The results showed a decrease in cell proliferation, migration, and invasion, as well as an increase in cell cycle arrest and apoptosis [18]. Moreover, butein caused a reduction in the tumor growth of ovarian cancer cells in mouse xenografts [18]. Many drugs were found to inhibit IL-6 signaling, but none of them had promising outcomes against ovarian cancer [19]. Park et al. have found an alternative treatment for ovarian cancer through this IL-6 pathway-inhibiting mechanism [18]. Finally, Nakamura et al. studied the apoptosis induction in hepatocellular and colorectal adenocarcinoma cell lines mediated by a novel quercetin-zinc(II) complex [20]. The main results they obtained were enhanced absorption of the complex (improved bioavailability and intracellular uptake) and an increase in the anticancer efficacy with an increment of the apoptosis levels comparing the complex with the separate compounds [20]. These results agree with others demonstrating that flavonoid metal complexes penetrate lipid bilayers through hydrophobic protein pores, increasing intracellular uptake of these complexes [21].
In this Special Issue, there were published two interesting reviews about the effects of phytochemicals in cancer prevention and treatment. One of them, carried out by Na et al., shows the importance of isothiocyanates, phytochemicals present in cruciferous vegetables, in cancer prevention and therapy. Thus, the authors split the mechanisms of isothiocyanates in cancer prevention and therapy into the following four main parts: 1. regulation of microbial homeostasis in the intestinal mucosa; 2. rearrangement of energy metabolism phenotype, with a special importance of mitochondria; 3. reconstruction of tumor microenvironment, with emphasis on inflammation status; and 4. inhibition of cancer stem cells [22]. This exciting review relates the main studies of the effects of isothiocyanates in breast, liver, gastric, bladder, prostate, lung, pancreatic, glioblastoma, endometrial, and colon cancer [23,24,25,26,27,28,29,30,31,32]. On the other hand, the other review published by Golonko et al. reveals the promising synergistic effect of different types of flavonoids in combination with anthracyclines, such as doxorubicin, daunorubicin, epirubicin, or idarubicin [33]. Anthracyclines are used in many types of cancer, including breast, lymphoma, and sarcoma [34]. The mechanism of action of anthracyclines is multifactorial, i.e., disruption of DNA integrity, binding to the cell membrane, and increasing oxidative stress by an increment in free radical production [35]. The authors of this review highlight the importance of the crosstalk between flavonoids and the molecular activity of anthracyclines, with special emphasis on the following three areas of action: 1. disruption of DNA integrity [36,37,38,39,40]; 2. modulation of antioxidant response pathways [41,42,43,44,45,46,47]; and 3. inhibition of the activity of membrane proteins responsible for the active transport of drugs and xenobiotics [48,49,50,51].
All the publications in this Special Issue highlight the importance of phytochemicals in cancer prevention and therapy. Thanks to the scientific knowledge published and reviewed in this Special Issue, we are now closer to understanding the mechanisms by which various phytochemicals can directly affect the prevention and treatment of cancer. All of this from a perspective closely related to energy metabolism and mitochondria, highlighting the role of this organelle in the response of cancer cells to anticancer treatments in combination with phytochemicals.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Ganai, A.A.; Farooqi, H. Bioactivity of Genistein: A Review of in Vitro and in Vivo Studies. Biomed. Pharmacother. 2015, 76, 30–38. [Google Scholar] [CrossRef] [PubMed]
  2. Alorda-Clara, M.; Torrens-Mas, M.; Morla-Barcelo, P.M.; Roca, P.; Sastre-Serra, J.; Pons, D.G.; Oliver, J. High Concentrations of Genistein Decrease Cell Viability Depending on Oxidative Stress and Inflammation in Colon Cancer Cell Lines. Int. J. Mol. Sci. 2022, 23, 7526. [Google Scholar] [CrossRef] [PubMed]
  3. Pons, D.G.; Nadal-Serrano, M.; Blanquer-Rossello, M.M.; Sastre-Serra, J.; Oliver, J.; Roca, P. Genistein Modulates Proliferation and Mitochondrial Functionality in Breast Cancer Cells Depending on ERalpha/ERbeta Ratio. J. Cell. Biochem. 2014, 115, 949–958. [Google Scholar] [CrossRef] [PubMed]
  4. Nadal-Serrano, M.; Pons, D.G.; Sastre-Serra, J.; Blanquer-Rossello Mdel, M.; Roca, P.; Oliver, J. Genistein Modulates Oxidative Stress in Breast Cancer Cell Lines According to ERalpha/ERbeta Ratio: Effects on Mitochondrial Functionality, Sirtuins, Uncoupling Protein 2 and Antioxidant Enzymes. Int. J. Biochem. Cell Biol. 2013, 45, 2045–2051. [Google Scholar] [CrossRef] [PubMed]
  5. Pons, D.G.; Vilanova-Llompart, J.; Gaya-Bover, A.; Alorda-Clara, M.; Oliver, J.; Roca, P.; Sastre-Serra, J. The Phytoestrogen Genistein Affects Inflammatory-Related Genes Expression Depending on the ERα/ERβ Ratio in Breast Cancer Cells. Int. J. Food Sci. Nutr. 2019, 70, 941–949. [Google Scholar] [CrossRef] [PubMed]
  6. Pons, D.G.; Nadal-Serrano, M.; Torrens-Mas, M.; Oliver, J.; Roca, P. The Phytoestrogen Genistein Affects Breast Cancer Cells Treatment Depending on the ERα/ERβ Ratio. J. Cell. Biochem. 2016, 117, 218–229. [Google Scholar] [CrossRef] [PubMed]
  7. Blanquer-Rossellõ, M.M.; Oliver, J.; Valle, A.; Roca, P. Effect of Xanthohumol and 8-Prenylnaringenin on MCF-7 Breast Cancer Cells Oxidative Stress and Mitochondrial Complexes Expression. J. Cell. Biochem. 2013, 114, 2785–2794. [Google Scholar] [CrossRef] [PubMed]
  8. Blanquer-Rosselló, M.d.M.; Hernández-López, R.; Roca, P.; Oliver, J.; Valle, A. Resveratrol Induces Mitochondrial Respiration and Apoptosis in SW620 Colon Cancer Cells. Biochim. Biophys Acta Gen. Subj. 2017, 1861, 431–440. [Google Scholar] [CrossRef] [PubMed]
  9. Sastre-Serra, J.; Ahmiane, Y.; Roca, P.; Oliver, J.; Pons, D.G. Xanthohumol, a Hop-Derived Prenylflavonoid Present in Beer, Impairs Mitochondrial Functionality of SW620 Colon Cancer Cells. Int. J. Food Sci. Nutr. 2019, 70, 396–404. [Google Scholar] [CrossRef]
  10. Torrens-Mas, M.; Alorda-Clara, M.; Martínez-Vigara, M.; Roca, P.; Sastre-Serra, J.; Oliver, J.; Pons, D.G. Xanthohumol Reduces Inflammation and Cell Metabolism in HT29 Primary Colon Cancer Cells. Int. J. Food Sci. Nutr. 2022, 73, 471–479. [Google Scholar] [CrossRef]
  11. Ahmad, A.; Hayat, I.; Arif, S.; Masud, T.; Khalid, N.; Ahmed, A. Mechanisms Involved in the Therapeutic Effects of Soybean (Glycine Max). Int. J. Food Prop. 2014, 17, 1332–1354. [Google Scholar] [CrossRef]
  12. Augustynowicz, D.; Lemieszek, M.K.; Strawa, J.W.; Wiater, A.; Tomczyk, M. Phytochemical Profiling of Extracts from Rare Potentilla Species and Evaluation of Their Anticancer Potential. Int. J. Mol. Sci. 2023, 24, 4836. [Google Scholar] [CrossRef]
  13. Augustynowicz, D.; Lemieszek, M.K.; Strawa, J.W.; Wiater, A.; Tomczyk, M. Anticancer Potential of Acetone Extracts from Selected Potentilla Species against Human Colorectal Cancer Cells. Front. Pharmacol. 2022, 13, 1027315. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, M.; Luan, X.; Zheng, H.; Wang, X.; Wang, S.; Shen, T.; Ren, D. A Mulberry Diels-Alder-Type Adduct, Kuwanon M, Triggers Apoptosis and Paraptosis of Lung Cancer Cells through Inducing Endoplasmic Reticulum Stress. Int. J. Mol. Sci. 2023, 24, 1015. [Google Scholar] [CrossRef]
  15. Shu, Y.-H.; Yuan, H.-H.; Xu, M.-T.; Hong, Y.-T.; Gao, C.-C.; Wu, Z.-P.; Han, H.-T.; Sun, X.; Gao, R.-L.; Yang, S.-F.; et al. A Novel Diels-Alder Adduct of Mulberry Leaves Exerts Anticancer Effect through Autophagy-Mediated Cell Death. Acta Pharmacol. Sin. 2021, 42, 780–790. [Google Scholar] [CrossRef]
  16. Eguchi, H.; Kimura, R.; Onuma, S.; Ito, A.; Yu, Y.; Yoshino, Y.; Matsunaga, T.; Endo, S.; Ikari, A. Elevation of Anticancer Drug Toxicity by Caffeine in Spheroid Model of Human Lung Adenocarcinoma A549 Cells Mediated by Reduction in Claudin-2 and Nrf2 Expression. Int. J. Mol. Sci. 2022, 23, 15447. [Google Scholar] [CrossRef] [PubMed]
  17. Zimta, A.A.; Cenariu, D.; Irimie, A.; Magdo, L.; Nabavi, S.M.; Atanasov, A.G.; Berindan-Neagoe, I. The Role of Nrf2 Activity in Cancer Development and Progression. Cancers 2019, 11, 1755. [Google Scholar] [CrossRef]
  18. Park, S.A.; Seo, Y.J.; Kim, L.K.; Kim, H.J.; Yoon, K.D.; Heo, T.H. Butein Inhibits Cell Growth by Blocking the IL-6/IL-6Rα Interaction in Human Ovarian Cancer and by Regulation of the IL-6/STAT3/FoxO3a Pathway. Int. J. Mol. Sci. 2023, 24, 6038. [Google Scholar] [CrossRef]
  19. Heo, T.H.; Wahler, J.; Suh, N. Potential Therapeutic Implications of IL-6/IL-6R/Gp130-Targeting Agents in Breast Cancer. Oncotarget 2016, 7, 15460–15473. [Google Scholar] [CrossRef]
  20. Nakamura, M.; Urakawa, D.; He, Z.; Akagi, I.; Hou, D.X.; Sakao, K. Apoptosis Induction in HepG2 and HCT116 Cells by a Novel Quercetin-Zinc (II) Complex: Enhanced Absorption of Quercetin and Zinc (II). Int. J. Mol. Sci. 2023, 24, 17457. [Google Scholar] [CrossRef]
  21. Tarahovsky, Y.S.; Kim, Y.A.; Yagolnik, E.A.; Muzafarov, E.N. Flavonoid–Membrane Interactions: Involvement of Flavonoid–Metal Complexes in Raft Signaling. Biochim. Et Biophys. Acta (BBA)-Biomembr. 2014, 1838, 1235–1246. [Google Scholar] [CrossRef] [PubMed]
  22. Na, G.; He, C.; Zhang, S.; Tian, S.; Bao, Y.; Shan, Y. Dietary Isothiocyanates: Novel Insights into the Potential for Cancer Prevention and Therapy. Int. J. Mol. Sci. 2023, 24, 1962. [Google Scholar] [CrossRef] [PubMed]
  23. Li, Y.; Zhang, T.; Korkaya, H.; Liu, S.; Lee, H.F.; Newman, B.; Yu, Y.; Clouthier, S.G.; Schwartz, S.J.; Wicha, M.S.; et al. Sulforaphane, a Dietary Component of Broccoli/Broccoli Sprouts, Inhibits Breast Cancer Stem Cells. Clin. Cancer Res. 2010, 16, 2580–2590. [Google Scholar] [CrossRef] [PubMed]
  24. Jeon, Y.K.; Yoo, D.R.; Jang, Y.H.; Jang, S.Y.; Nam, M.J. Sulforaphane Induces Apoptosis in Human Hepatic Cancer Cells through Inhibition of 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase4, Mediated by Hypoxia Inducible Factor-1-Dependent Pathway. Biochim. Et Biophys. Acta (BBA)-Proteins Proteom. 2011, 1814, 1340–1348. [Google Scholar] [CrossRef] [PubMed]
  25. Keenan, J.I.; Salm, N.; Wallace, A.J.; Hampton, M.B. Using Food to Reduce H. Pylori-Associated Inflammation. Phytother. Res. 2012, 26, 1620–1625. [Google Scholar] [CrossRef] [PubMed]
  26. He, C.; Huang, L.; Lei, P.; Liu, X.; Li, B.; Shan, Y. Sulforaphane Normalizes Intestinal Flora and Enhances Gut Barrier in Mice with BBN-Induced Bladder Cancer. Mol. Nutr. Food Res. 2018, 62, 1800427. [Google Scholar] [CrossRef] [PubMed]
  27. Singh, K.B.; Hahm, E.R.; Alumkal, J.J.; Foley, L.M.; Hitchens, T.K.; Shiva, S.S.; Parikh, R.A.; Jacobs, B.L.; Singh, S.V. Reversal of the Warburg Phenomenon in Chemoprevention of Prostate Cancer by Sulforaphane. Carcinogenesis 2019, 40, 1545–1556. [Google Scholar] [CrossRef] [PubMed]
  28. Yan, Y.; Zhou, Y.; Li, J.; Zheng, Z.; Hu, Y.; Li, L.; Wu, W. Sulforaphane Downregulated Fatty Acid Synthase and Inhibited Microtubule-Mediated Mitophagy Leading to Apoptosis. Cell Death Dis. 2021, 12, 10. [Google Scholar] [CrossRef] [PubMed]
  29. Li, S.H.; Fu, J.; Watkins, D.N.; Srivastava, R.K.; Shankar, S. Sulforaphane Regulates Self-Renewal of Pancreatic Cancer Stem Cells through the Modulation of Sonic Hedgehog-GLI Pathway. Mol. Cell. Biochem. 2013, 373, 217–227. [Google Scholar] [CrossRef]
  30. Kumar, R.; De Mooij, T.; Peterson, T.E.; Kaptzan, T.; Johnson, A.J.; Daniels, D.J.; Parney, I.F. Modulating Glioma-Mediated Myeloid-Derived Suppressor Cell Development with Sulforaphane. PLoS ONE 2017, 12, e0179012. [Google Scholar] [CrossRef]
  31. Rai, R.; Essel, K.G.; Benbrook, D.M.; Garland, J.; Zhao, Y.D.; Chandra, V. Preclinical Efficacy and Involvement of AKT, MTOR, and ERK Kinases in the Mechanism of Sulforaphane against Endometrial Cancer. Cancers 2020, 12, 1273. [Google Scholar] [CrossRef] [PubMed]
  32. Bao, Y.; Wang, W.; Zhou, Z.; Sun, C. Benefits and Risks of the Hormetic Effects of Dietary Isothiocyanates on Cancer Prevention. PLoS ONE 2014, 9, e114764. [Google Scholar] [CrossRef] [PubMed]
  33. Golonko, A.; Olichwier, A.J.; Swislocka, R.; Szczerbinski, L.; Lewandowski, W. Why Do Dietary Flavonoids Have a Promising Effect as Enhancers of Anthracyclines? Hydroxyl Substituents, Bioavailability and Biological Activity. Int. J. Mol. Sci. 2023, 24, 391. [Google Scholar] [CrossRef]
  34. Sobczuk, P.; Czerwińska, M.; Kleibert, M.; Cudnoch-Jędrzejewska, A. Anthracycline-Induced Cardiotoxicity and Renin-Angiotensin-Aldosterone System-from Molecular Mechanisms to Therapeutic Applications. Heart. Fail. Rev. 2022, 27, 295–319. [Google Scholar] [CrossRef] [PubMed]
  35. Dhingra, R.; Margulets, V.; Kirshenbaum, L.A. Molecular Mechanisms Underlying Anthracycline Cardiotoxicity: Challenges in Cardio-Oncology. In Cardio-Oncology; Academic Press: Cambridge, MA, USA, 2017; pp. 25–34. [Google Scholar] [CrossRef]
  36. Das, A.; Majumder, D.; Saha, C. Correlation of Binding Efficacies of DNA to Flavonoids and Their Induced Cellular Damage. J. Photochem. Photobiol. B 2017, 170, 256–262. [Google Scholar] [CrossRef] [PubMed]
  37. Nafisi, S.; Hashemi, M.; Rajabi, M.; Tajmir-Riahi, H.A. DNA Adducts with Antioxidant Flavonoids: Morin, Apigenin, and Naringin. DNA Cell Biol. 2008, 27, 433–442. [Google Scholar] [CrossRef] [PubMed]
  38. Waihenya, S.; Şenel, P.; Osonga, F.J.; Erdoǧan, T.; Altay, F.; Gölcü, A.; Sadik, O.A. Mechanism of Interactions of DsDNA Binding with Apigenin and Its Sulfamate Derivatives Using Multispectroscopic, Voltammetric, and Molecular Docking Studies. ACS Omega 2021, 6, 5124–5137. [Google Scholar] [CrossRef] [PubMed]
  39. Kumar, S.; Nair, M.S. Deciphering the Interaction of Flavones with Calf Thymus DNA and Octamer DNA Sequence (CCAATTGG)2. RSC Adv. 2021, 11, 29354–29371. [Google Scholar] [CrossRef]
  40. Sha, Y.; Chen, X.; Niu, B.; Chen, Q. The Interaction Mode of Groove Binding Between Quercetin and Calf Thymus DNA Based on Spectrometry and Simulation. Chem. Biodivers. 2017, 14, e1700133. [Google Scholar] [CrossRef]
  41. Gao, A.M.; Ke, Z.P.; Shi, F.; Sun, G.C.; Chen, H. Chrysin Enhances Sensitivity of BEL-7402/ADM Cells to Doxorubicin by Suppressing PI3K/Akt/Nrf2 and ERK/Nrf2 Pathway. Chem. Biol. Interact. 2013, 206, 100–108. [Google Scholar] [CrossRef]
  42. Sabzichi, M.; Mohammadian, J.; Bazzaz, R.; Pirouzpanah, M.B.; Shaaker, M.; Hamishehkar, H.; Chavoshi, H.; Salehi, R.; Samadi, N. Chrysin Loaded Nanostructured Lipid Carriers (NLCs) Triggers Apoptosis in MCF-7 Cancer Cells by Inhibiting the Nrf2 Pathway. Process Biochem. 2017, 60, 84–91. [Google Scholar] [CrossRef]
  43. Wang, J.; Wang, H.; Sun, K.; Wang, X.; Pan, H.; Zhu, J.; Ji, X.; Li, X. Chrysin Suppresses Proliferation, Migration, and Invasion in Glioblastoma Cell Lines via Mediating the ERK/Nrf2 Signaling Pathway. Drug Des. Devel. Ther. 2018, 12, 721–733. [Google Scholar] [CrossRef] [PubMed]
  44. Gao, A.-M.; Zhang, X.-Y.; Ke, Z.-P.; Gao, A.-M.; Zhang, X.-Y.; Ke, Z.-P. Apigenin Sensitizes BEL-7402/ADM Cells to Doxorubicin through Inhibiting MiR-101/Nrf2 Pathway. Oncotarget 2017, 8, 82085–82091. [Google Scholar] [CrossRef] [PubMed]
  45. Fouzder, C.; Mukhuty, A.; Kundu, R. Kaempferol Inhibits Nrf2 Signalling Pathway via Downregulation of Nrf2 MRNA and Induces Apoptosis in NSCLC Cells. Arch Biochem. Biophys 2021, 697, 108700. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, F.; Wang, L.; Qu, C.; Chen, L.; Geng, Y.; Cheng, C.; Yu, S.; Wang, D.; Yang, L.; Meng, Z.; et al. Kaempferol Induces ROS-Dependent Apoptosis in Pancreatic Cancer Cells via TGM2-Mediated Akt/MTOR Signaling. BMC Cancer 2021, 21, 396. [Google Scholar] [CrossRef]
  47. De Prax, M.C.A.; Ferro, K.P.V.; Santos, I.; Torello, C.O.; Salazar-Terreros, M.; Olalla Saad, S.T. NRF2 Is Targeted By the Polyphenol Quercetin and Induces Apoptosis, in Part, through up Regulation of Pro Apoptotic Mirs. Blood 2019, 134, 2529. [Google Scholar] [CrossRef]
  48. Choi, S.J.; Shin, S.C.; Choi, J.S. Effects of Myricetin on the Bioavailability of Doxorubicin for Oral Drug Delivery in Rats: Possible Role of CYP3A4 and P-Glycoprotein Inhibition by Myricetin. Arch. Pharm. Res. 2011, 34, 309–315. [Google Scholar] [CrossRef]
  49. Choi, J.S.; Piao, Y.J.; Kang, K.W. Effects of Quercetin on the Bioavailability of Doxorubicin in Rats: Role of CYP3A4 and P-Gp Inhibition by Quercetin. Arch. Pharm. Res. 2011, 34, 607–613. [Google Scholar] [CrossRef]
  50. Kimura, Y.; Ito, H.; Ohnishi, R.; Hatano, T. Inhibitory Effects of Polyphenols on Human Cytochrome P450 3A4 and 2C9 Activity. Food Chem. Toxicol. 2010, 48, 429–435. [Google Scholar] [CrossRef]
  51. Mustapíc, D.Š.; Debeljak, Ž.; Maleš, Ž.; Bojíc, M. The Inhibitory Effect of Flavonoid Aglycones on the Metabolic Activity of CYP3A4 Enzyme. Molecules 2018, 23, 2553. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pons, D.G. Roles of Phytochemicals in Cancer Prevention and Therapeutics. Int. J. Mol. Sci. 2024, 25, 5450. https://doi.org/10.3390/ijms25105450

AMA Style

Pons DG. Roles of Phytochemicals in Cancer Prevention and Therapeutics. International Journal of Molecular Sciences. 2024; 25(10):5450. https://doi.org/10.3390/ijms25105450

Chicago/Turabian Style

Pons, Daniel Gabriel. 2024. "Roles of Phytochemicals in Cancer Prevention and Therapeutics" International Journal of Molecular Sciences 25, no. 10: 5450. https://doi.org/10.3390/ijms25105450

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop