Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer
Abstract
:1. Introduction
2. The Role of Hormones in Mammary Gland Development
3. The Estrogenic Signaling
3.1. The Genomic Pathway
3.2. The Non-Genomic Pathway
3.3. The Estrogen-Independent Pathway
4. Molecular Subtypes of Breast Cancer
5. Treatment for Luminal Tumors
6. Resistance to Hormone Therapy
7. Natural Compounds and Their Effects on Luminal Tumors
7.1. Flavonoids
7.2. Isoflavonoids
7.3. Alkaloids and Catechins
7.4. Lignans
7.5. Coumestans and Stilbenes
7.6. Other Compounds
8. The Challenges of Clinical Practice
9. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- World Health Organization. Breast Cancer. 2023. Available online: https://www.who.int/news-room/fact-sheets/detail/breast-cancer (accessed on 23 September 2023).
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Martei, Y.M.; Pace, L.E.; Brock, J.E.; Shulman, L.N. Breast Cancer in Low- and Middle-Income Countries: Why We Need Pathology Capability to Solve This Challenge. Clin. Lab. Med. 2018, 38, 161–173. [Google Scholar] [CrossRef] [PubMed]
- Ignatiadis, M.; Sotiriou, C. Luminal breast cancer: From biology to treatment. Nat. Rev. Clin. Oncol. 2013, 10, 494–506. [Google Scholar] [CrossRef] [PubMed]
- Fahad Ullah, M. Breast Cancer: Current Perspectives on the Disease Status. Adv. Exp. Med. Biol. 2019, 1152, 51–64. [Google Scholar] [CrossRef] [PubMed]
- Shin, S.A.; Moon, S.Y.; Kim, W.Y.; Paek, S.M.; Park, H.H.; Lee, C.S. Structure-Based Classification and Anti-Cancer Effects of Plant Metabolites. Int. J. Mol. Sci. 2018, 19, 2651. [Google Scholar] [CrossRef] [PubMed]
- Kapadia, P.; Newell, A.S.; Cunningham, J.; Roberts, M.R.; Hardy, J.G. Extraction of High-Value Chemicals from Plants for Technical and Medical Applications. Int. J. Mol. Sci. 2022, 23, 10334. [Google Scholar] [CrossRef] [PubMed]
- Shin, S.A.; Joo, B.J.; Lee, J.S.; Ryu, G.; Han, M.; Kim, W.Y.; Park, H.H.; Lee, J.H.; Lee, C.S. Phytochemicals as Anti-Inflammatory Agents in Animal Models of Prevalent Inflammatory Diseases. Molecules 2020, 25, 5932. [Google Scholar] [CrossRef]
- Bulotta, S.; Capriglione, F.; Celano, M.; Pecce, V.; Russo, D.; Maggisano, V. Phytochemicals in thyroid cancer: Analysis of the preclinical studies. Endocrine 2021, 73, 8–15. [Google Scholar] [CrossRef]
- Piasecka, D.; Braun, M.; Kitowska, K.; Mieczkowski, K.; Kordek, R.; Sadej, R.; Romanska, H. FGFs/FGFRs-dependent signalling in regulation of steroid hormone receptors—Implications for therapy of luminal breast cancer. J. Exp. Clin. Cancer Res. 2019, 38, 230. [Google Scholar] [CrossRef]
- Basu, P.; Maier, C. Phytoestrogens and breast cancer: In vitro anticancer activities of isoflavones, lignans, coumestans, stilbenes and their analogs and derivatives. Biomed. Pharmacother. 2018, 107, 1648–1666. [Google Scholar] [CrossRef]
- Alex, A.; Bhandary, E.; McGuire, K.P. Anatomy and Physiology of the Breast during Pregnancy and Lactation. Adv. Exp. Med. Biol. 2020, 1252, 3–7. [Google Scholar] [CrossRef]
- Jesinger, R.A. Breast anatomy for the interventionalist. Tech. Vasc. Interv. Radiol. 2014, 17, 3–9. [Google Scholar] [CrossRef]
- McGhee, D.E.; Steele, J.R. Breast Biomechanics: What Do We Really Know? Physiology 2020, 35, 144–156. [Google Scholar] [CrossRef]
- Inman, J.L.; Robertson, C.; Mott, J.D.; Bissell, M.J. Mammary gland development: Cell fate specification, stem cells and the microenvironment. Development 2015, 142, 1028–1042. [Google Scholar] [CrossRef]
- Lin, J.; Ye, S.; Ke, H.; Lin, L.; Wu, X.; Guo, M.; Jiao, B.; Chen, C.; Zhao, L. Changes in the mammary gland during aging and its links with breast diseases. Acta Biochim. Biophys. Sin. 2023, 55, 1001–1019. [Google Scholar] [CrossRef]
- Macias, H.; Hinck, L. Mammary gland development. Wiley Interdiscip. Rev. Dev. Biol. 2012, 1, 533–557. [Google Scholar] [CrossRef] [PubMed]
- Hilton, H.N.; Clarke, C.L.; Graham, J.D. Estrogen and progesterone signalling in the normal breast and its implications for cancer development. Mol. Cell. Endocrinol. 2018, 466, 2–14. [Google Scholar] [CrossRef] [PubMed]
- de Holanda, A.A.; Gonçalves, A.K.; de Medeiros, R.D.; de Oliveira, A.M.; Maranhão, T.M. Ultrasound findings of the physiological changes and most common breast diseases during pregnancy and lactation. Radiol. Bras. 2016, 49, 389–396. [Google Scholar] [CrossRef] [PubMed]
- Taraborrelli, S. Physiology, production and action of progesterone. Acta Obstet. Gynecol. Scand. 2015, 94 (Suppl. 161), 8–16. [Google Scholar] [CrossRef]
- Chen, W.; Wei, W.; Yu, L.; Ye, Z.; Huang, F.; Zhang, L.; Hu, S.; Cai, C. Mammary Development and Breast Cancer: A Notch Perspective. J. Mammary Gland Biol. Neoplasia 2021, 26, 309–320. [Google Scholar] [CrossRef] [PubMed]
- Slepicka, P.F.; Cyrill, S.L.; Dos Santos, C.O. Pregnancy and Breast Cancer: Pathways to Understand Risk and Prevention. Trends Mol. Med. 2019, 25, 866–881. [Google Scholar] [CrossRef] [PubMed]
- Gallez, A.; Dias Da Silva, I.; Wuidar, V.; Foidart, J.M.; Péqueux, C. Estetrol and Mammary Gland: Friends or Foes? J. Mammary Gland. Biol. Neoplasia 2021, 26, 297–308. [Google Scholar] [CrossRef] [PubMed]
- Tian, J.M.; Ran, B.; Zhang, C.L.; Yan, D.M.; Li, X.H. Estrogen and progesterone promote breast cancer cell proliferation by inducing cyclin G1 expression. Braz. J. Med. Biol. Res. 2018, 51, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Key, T.; Appleby, P.; Barnes, I.; Reeves, G. Endogenous sex hormones and breast cancer in postmenopausal women: Reanalysis of nine prospective studies. J. Natl. Cancer Inst. 2002, 94, 606–616. [Google Scholar] [CrossRef] [PubMed]
- Maynadier, M.; Chambon, M.; Basile, I.; Gleizes, M.; Nirde, P.; Gary-Bobo, M.; Garcia, M. Estrogens promote cell-cell adhesion of normal and malignant mammary cells through increased desmosome formation. Mol. Cell. Endocrinol. 2012, 364, 126–133. [Google Scholar] [CrossRef] [PubMed]
- Feng, Y.; Spezia, M.; Huang, S.; Yuan, C.; Zeng, Z.; Zhang, L.; Ji, X.; Liu, W.; Huang, B.; Luo, W.; et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018, 5, 77–106. [Google Scholar] [CrossRef]
- Ikeda, K.; Horie-Inoue, K.; Inoue, S. Functions of estrogen and estrogen receptor signaling on skeletal muscle. J. Steroid Biochem. Mol. Biol. 2019, 191, 105375. [Google Scholar] [CrossRef]
- Faltas, C.L.; LeBron, K.A.; Holz, M.K. Unconventional Estrogen Signaling in Health and Disease. Endocrinology 2020, 161, bqaa030. [Google Scholar] [CrossRef]
- Heldring, N.; Pike, A.; Andersson, S.; Matthews, J.; Cheng, G.; Hartman, J.; Tujague, M.; Ström, A.; Treuter, E.; Warner, M.; et al. Estrogen receptors: How do they signal and what are their targets. Physiol. Rev. 2007, 87, 905–931. [Google Scholar] [CrossRef]
- Paterni, I.; Granchi, C.; Katzenellenbogen, J.A.; Minutolo, F. Estrogen receptors alpha (ERα) and beta (ERβ): Subtype-selective ligands and clinical potential. Steroids 2014, 90, 13–29. [Google Scholar] [CrossRef]
- Fuentes, N.; Silveyra, P. Estrogen receptor signaling mechanisms. Adv. Protein Chem. Struct. Biol. 2019, 116, 135–170. [Google Scholar] [CrossRef] [PubMed]
- Vrtačnik, P.; Ostanek, B.; Mencej-Bedrač, S.; Marc, J. The many faces of estrogen signaling. Biochem. Med. 2014, 24, 329–342. [Google Scholar] [CrossRef] [PubMed]
- Miller, W.R. Aromatase inhibitors: Mechanism of action and role in the treatment of breast cancer. Semin. Oncol. 2003, 30, 3–11. [Google Scholar] [CrossRef]
- Barakat, R.; Oakley, O.; Kim, H.; Jin, J.; Ko, C.J. Extra-gonadal sites of estrogen biosynthesis and function. BMB Rep. 2016, 49, 488–496. [Google Scholar] [CrossRef] [PubMed]
- Mahboobifard, F.; Pourgholami, M.H.; Jorjani, M.; Dargahi, L.; Amiri, M.; Sadeghi, S.; Tehrani, F.R. Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed. Pharmacother. 2022, 156, 113808. [Google Scholar] [CrossRef]
- Nazari, E.; Suja, F. Effects of 17β-estradiol (E2) on aqueous organisms and its treatment problem: A review. Rev. Environ. Health 2016, 31, 465–491. [Google Scholar] [CrossRef]
- van der Ploeg, P.; van Lieshout, L.A.M.; van de Stolpe, A.; Bosch, S.L.; Lentjes-Beer, M.; Bekkers, R.L.M.; Piek, J.M.J. Functional estrogen receptor signaling pathway activity in high-grade serous ovarian carcinoma as compared to estrogen receptor protein expression by immunohistochemistry. Cell. Oncol. 2021, 44, 951–957. [Google Scholar] [CrossRef]
- Hah, N.; Kraus, W.L. Hormone-regulated transcriptomes: Lessons learned from estrogen signaling pathways in breast cancer cells. Mol. Cell. Endocrinol. 2014, 382, 652–664. [Google Scholar] [CrossRef]
- Eyster, K.M. The Estrogen Receptors: An Overview from Different Perspectives. In Estrogen Receptors: Methods and Protocols; Methods in Molecular Biology; Humana: New York, NY, USA, 2016; Volume 1366, pp. 1–10. [Google Scholar] [CrossRef]
- Krishnan, V.; Heath, H.; Bryant, H.U. Mechanism of action of estrogens and selective estrogen receptor modulators. Vitam. Horm. 2000, 60, 123–147. [Google Scholar] [CrossRef]
- Ruggiero, R.J.; Likis, F.E. Estrogen: Physiology, pharmacology, and formulations for replacement therapy. J. Midwifery Women’s Health 2002, 47, 130–138. [Google Scholar] [CrossRef]
- Wilkenfeld, S.R.; Lin, C.; Frigo, D.E. Communication between genomic and non-genomic signaling events coordinate steroid hormone actions. Steroids 2018, 133, 2–7. [Google Scholar] [CrossRef] [PubMed]
- Driscoll, M.D.; Sathya, G.; Muyan, M.; Klinge, C.M.; Hilf, R.; Bambara, R.A. Sequence requirements for estrogen receptor binding to estrogen response elements. J. Biol. Chem. 1998, 273, 29321–29330. [Google Scholar] [CrossRef] [PubMed]
- Glück, S. Consequences of the Convergence of Multiple Alternate Pathways on the Estrogen Receptor in the Treatment of Metastatic Breast Cancer. Clin. Breast Cancer 2017, 17, 79–90. [Google Scholar] [CrossRef]
- Klinge, C.M. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res. 2001, 29, 2905–2919. [Google Scholar] [CrossRef] [PubMed]
- Le Dily, F.; Beato, M. Signaling by Steroid Hormones in the 3D Nuclear Space. Int. J. Mol. Sci. 2018, 19, 306. [Google Scholar] [CrossRef]
- Cheung, E.; Kraus, W.L. Genomic analyses of hormone signaling and gene regulation. Annu. Rev. Physiol. 2010, 72, 191–218. [Google Scholar] [CrossRef]
- Ruhl, D.D.; Kraus, W.L. Biochemical analyses of nuclear receptor-dependent transcription with chromatin templates. Prog. Mol. Biol. Transl. Sci. 2009, 87, 137–192. [Google Scholar] [CrossRef]
- Kulkoyluoglu-Cotul, E.; Arca, A.; Madak-Erdogan, Z. Crosstalk between Estrogen Signaling and Breast Cancer Metabolism. Trends Endocrinol. Metab. 2019, 30, 25–38. [Google Scholar] [CrossRef]
- Liang, J.; Shang, Y. Estrogen and cancer. Annu. Rev. Physiol. 2013, 75, 225–240. [Google Scholar] [CrossRef]
- Saha, T.; Makar, S.; Swetha, R.; Gutti, G.; Singh, S.K. Estrogen signaling: An emanating therapeutic target for breast cancer treatment. Eur. J. Med. Chem. 2019, 177, 116–143. [Google Scholar] [CrossRef]
- Ribeiro, J.R.; Freiman, R.N. Estrogen signaling crosstalk: Implications for endocrine resistance in ovarian cancer. J. Steroid Biochem. Mol. Biol. 2014, 143, 160–173. [Google Scholar] [CrossRef] [PubMed]
- Clusan, L.; Ferrière, F.; Flouriot, G.; Pakdel, F. A Basic Review on Estrogen Receptor Signaling Pathways in Breast Cancer. Int. J. Mol. Sci. 2023, 24, 6834. [Google Scholar] [CrossRef] [PubMed]
- Marino, M.; Acconcia, F.; Trentalance, A.; Ascenzi, P.; Bocedi, A.; Spisni, E.; Tomasi, V.; Visca, P.; Fernando, R.I.; Wimalasena, J.; et al. Biphasic estradiol-induced AKT phosphorylation is modulated by PTEN via MAP kinase in HepG2 cells. Mol. Biol. Cell 2003, 14, 2583–2591. [Google Scholar] [CrossRef] [PubMed]
- Rusidzé, M.; Adlanmérini, M.; Chantalat, E.; Raymond-Letron, I.; Cayre, S.; Arnal, J.F.; Deugnier, M.A.; Lenfant, F. Estrogen receptor-α signaling in post-natal mammary development and breast cancers. Cell. Mol. Life Sci. 2021, 78, 5681–5705. [Google Scholar] [CrossRef]
- Treeck, O.; Schüler-Toprak, S.; Ortmann, O. Estrogen Actions in Triple-Negative Breast Cancer. Cells 2020, 9, 2358. [Google Scholar] [CrossRef]
- Ho, K.J.; Liao, J.K. Non-nuclear actions of estrogen: New targets for prevention and treatment of cardiovascular disease. Mol. Interv. 2002, 2, 219–228. [Google Scholar] [CrossRef]
- Schwartz, N.; Verma, A.; Bivens, C.B.; Schwartz, Z.; Boyan, B.D. Rapid steroid hormone actions via membrane receptors. Biochim. Biophys. Acta 2016, 1863, 2289–2298. [Google Scholar] [CrossRef]
- Velarde, M.C.; Turner, R.J.; Kerber, I.J. Pleiotropic actions of estrogen: A mitochondrial matter. Physiol. Genom. 2013, 45, 106–109. [Google Scholar] [CrossRef]
- Ignar-Trowbridge, D.M.; Teng, C.T.; Ross, K.A.; Parker, M.G.; Korach, K.S.; McLachlan, J.A. Peptide growth factors elicit estrogen receptor-dependent transcriptional activation of an estrogen-responsive element. Mol. Endocrinol. 1993, 7, 992–998. [Google Scholar] [CrossRef]
- Khan, M.Z.I.; Uzair, M.; Nazli, A.; Chen, J.Z. An overview on Estrogen receptors signaling and its ligands in breast cancer. Eur. J. Med. Chem. 2022, 241, 114658. [Google Scholar] [CrossRef]
- Starek-Świechowicz, B.; Budziszewska, B.; Starek, A. Endogenous estrogens—Breast cancer and chemoprevention. Pharmacol. Rep. 2021, 73, 1497–1512. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Liu, X. The role of estrogen receptor beta in breast cancer. Biomark. Res. 2020, 8, 39. [Google Scholar] [CrossRef] [PubMed]
- Murphy, L.C.; Leygue, E. The role of estrogen receptor-β in breast cancer. Semin. Reprod. Med. 2012, 30, 5–13. [Google Scholar] [CrossRef] [PubMed]
- Helguero, L.A.; Faulds, M.H.; Gustafsson, J.A.; Haldosén, L.A. Estrogen receptors alfa (ERalpha) and beta (ERbeta) differentially regulate proliferation and apoptosis of the normal murine mammary epithelial cell line HC11. Oncogene 2005, 24, 6605–6616. [Google Scholar] [CrossRef]
- Chen, P.; Li, B.; Ou-Yang, L. Role of estrogen receptors in health and disease. Front. Endocrinol. 2022, 13, 839005. [Google Scholar] [CrossRef]
- Vivacqua, A. GPER1 and microRNA: Two Players in Breast Cancer Progression. Int. J. Mol. Sci. 2020, 22, 98. [Google Scholar] [CrossRef]
- Thomas, M.; Kelly, E.D.; Abraham, J.; Kruse, M. Invasive lobular breast cancer: A review of pathogenesis, diagnosis, management, and future directions of early stage disease. Semin. Oncol. 2019, 46, 121–132. [Google Scholar] [CrossRef]
- Eroles, P.; Bosch, A.; Pérez-Fidalgo, J.A.; Lluch, A. Molecular biology in breast cancer: Intrinsic subtypes and signaling pathways. Cancer Treat. Rev. 2012, 38, 698–707. [Google Scholar] [CrossRef]
- Perou, C.M.; Sørlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
- Tsang, J.Y.S.; Tse, G.M. Molecular Classification of Breast Cancer. Adv. Anat. Pathol. 2020, 27, 27–35. [Google Scholar] [CrossRef]
- Mueller, C.; Haymond, A.; Davis, J.B.; Williams, A.; Espina, V. Protein biomarkers for subtyping breast cancer and implications for future research. Expert Rev. Proteom. 2018, 15, 131–152. [Google Scholar] [CrossRef] [PubMed]
- Rattani, N.S.; Swift-Scanlan, T. Deconstructing breast cancer heterogeneity: Clinical implications for women with Basal-like tumors. Oncol. Nurs. Forum 2014, 41, 639–646. [Google Scholar] [CrossRef] [PubMed]
- Gao, J.J.; Swain, S.M. Luminal A Breast Cancer and Molecular Assays: A Review. Oncologist 2018, 23, 556–565. [Google Scholar] [CrossRef] [PubMed]
- Tan, S.; Fu, X.; Xu, S.; Qiu, P.; Lv, Z.; Xu, Y.; Zhang, Q. Quantification of Ki67 Change as a Valid Prognostic Indicator of Luminal B Type Breast Cancer After Neoadjuvant Therapy. Pathol. Oncol. Res. 2021, 27, 1609972. [Google Scholar] [CrossRef] [PubMed]
- Tran, B.; Bedard, P.L. Luminal-B breast cancer and novel therapeutic targets. Breast Cancer Res. 2011, 13, 221. [Google Scholar] [CrossRef]
- Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef]
- Oh, D.Y.; Bang, Y.J. HER2-targeted therapies—A role beyond breast cancer. Nat. Rev. Clin. Oncol. 2020, 17, 33–48. [Google Scholar] [CrossRef]
- Yersal, O.; Barutca, S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014, 5, 412–424. [Google Scholar] [CrossRef]
- Schlam, I.; Swain, S.M. HER2-positive breast cancer and tyrosine kinase inhibitors: The time is now. npj Breast Cancer 2021, 7, 56. [Google Scholar] [CrossRef]
- Veeraraghavan, J.; Gutierrez, C.; Sethunath, V.; Mehravaran, S.; Giuliano, M.; Shea, M.J.; Mitchell, T.; Wang, T.; Nanda, S.; Pereira, R.; et al. Neratinib plus trastuzumab is superior to pertuzumab plus trastuzumab in HER2-positive breast cancer xenograft models. npj Breast Cancer 2021, 7, 63. [Google Scholar] [CrossRef]
- Li, Z.H.; Hu, P.H.; Tu, J.H.; Yu, N.S. Luminal B breast cancer: Patterns of recurrence and clinical outcome. Oncotarget 2016, 7, 65024–65033. [Google Scholar] [CrossRef] [PubMed]
- Riggio, A.I.; Varley, K.E.; Welm, A.L. The lingering mysteries of metastatic recurrence in breast cancer. Br. J. Cancer 2021, 124, 13–26. [Google Scholar] [CrossRef] [PubMed]
- Barnard, M.E.; Boeke, C.E.; Tamimi, R.M. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim. Biophys. Acta 2015, 1856, 73–85. [Google Scholar] [CrossRef] [PubMed]
- Roulot, A.; Héquet, D.; Guinebretière, J.M.; Vincent-Salomon, A.; Lerebours, F.; Dubot, C.; Rouzier, R. Tumoral heterogeneity of breast cancer. Ann. Biol. Clin. 2016, 74, 653–660. [Google Scholar] [CrossRef] [PubMed]
- Łukasiewicz, S.; Czeczelewski, M.; Forma, A.; Baj, J.; Sitarz, R.; Stanisławek, A. Breast Cancer-Epidemiology, Risk Factors, Classification, Prognostic Markers, and Current Treatment Strategies-An Updated Review. Cancers 2021, 13, 4287. [Google Scholar] [CrossRef]
- Creighton, C.J.; Kent Osborne, C.; van de Vijver, M.J.; Foekens, J.A.; Klijn, J.G.; Horlings, H.M.; Nuyten, D.; Wang, Y.; Zhang, Y.; Chamness, G.C.; et al. Molecular profiles of progesterone receptor loss in human breast tumors. Breast Cancer Res. Treat. 2009, 114, 287–299. [Google Scholar] [CrossRef]
- Li, Z.; Wei, H.; Li, S.; Wu, P.; Mao, X. The Role of Progesterone Receptors in Breast Cancer. Drug Des. Dev. Ther. 2022, 16, 305–314. [Google Scholar] [CrossRef]
- Arciero, C.A.; Guo, Y.; Jiang, R.; Behera, M.; O’Regan, R.; Peng, L.; Li, X. ER+/HER2+ Breast Cancer Has Different Metastatic Patterns and Better Survival than ER−/HER2+ Breast Cancer. Clin. Breast Cancer 2019, 19, 236–245. [Google Scholar] [CrossRef]
- Takada, M.; Toi, M. Neoadjuvant treatment for HER2-positive breast cancer. Chin. Clin. Oncol. 2020, 9, 32. [Google Scholar] [CrossRef]
- Ferraro, E.; Singh, J.; Patil, S.; Razavi, P.; Modi, S.; Chandarlapaty, S.; Barrio, A.V.; Malani, R.; Mellinghoff, I.K.; Boire, A.; et al. Incidence of brain metastases in patients with early HER2-positive breast cancer receiving neoadjuvant chemotherapy with trastuzumab and pertuzumab. npj Breast Cancer 2022, 8, 37. [Google Scholar] [CrossRef]
- Lyu, X.; Luo, B. Prognostic factors and survival prediction in HER2-positive breast cancer with bone metastases: A retrospective cohort study. Cancer Med. 2021, 10, 8114–8126. [Google Scholar] [CrossRef] [PubMed]
- Garrido-Castro, A.C.; Lin, N.U.; Polyak, K. Insights into Molecular Classifications of Triple-Negative Breast Cancer: Improving Patient Selection for Treatment. Cancer Discov. 2019, 9, 176–198. [Google Scholar] [CrossRef] [PubMed]
- Siddharth, S.; Sharma, D. Racial Disparity and Triple-Negative Breast Cancer in African-American Women: A Multifaceted Affair between Obesity, Biology, and Socioeconomic Determinants. Cancers 2018, 10, 514. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.Y.; Jiang, Z.; Ben-David, Y.; Woodgett, J.R.; Zacksenhaus, E. Molecular stratification within triple-negative breast cancer subtypes. Sci. Rep. 2019, 9, 19107. [Google Scholar] [CrossRef] [PubMed]
- Orrantia-Borunda, E.; Anchondo-Nuñez, P.; Acuña-Aguilar, L.E.; Gómez-Valles, F.O.; Ramírez-Valdespino, C.A. Subtypes of Breast Cancer. In Breast Cancer; Mayrovitz, H.N., Ed.; Exon Publications: Brisbane, Australia, 2022. [Google Scholar]
- Maeda, T.; Nakanishi, Y.; Hirotani, Y.; Fuchinoue, F.; Enomoto, K.; Sakurai, K.; Amano, S.; Nemoto, N. Immunohistochemical co-expression status of cytokeratin 5/6, androgen receptor, and p53 as prognostic factors of adjuvant chemotherapy for triple negative breast cancer. Med. Mol. Morphol. 2016, 49, 11–21. [Google Scholar] [CrossRef]
- Dietze, E.C.; Sistrunk, C.; Miranda-Carboni, G.; O’Regan, R.; Seewaldt, V.L. Triple-negative breast cancer in African-American women: Disparities versus biology. Nat. Rev. Cancer 2015, 15, 248–254. [Google Scholar] [CrossRef]
- Dalal, H.; Dahlgren, M.; Gladchuk, S.; Brueffer, C.; Gruvberger-Saal, S.K.; Saal, L.H. Clinical associations of ESR2 (estrogen receptor beta) expression across thousands of primary breast tumors. Sci. Rep. 2022, 12, 4696. [Google Scholar] [CrossRef]
- Drăgănescu, M.; Carmocan, C. Hormone Therapy in Breast Cancer. Chirurgia 2017, 112, 413–417. [Google Scholar] [CrossRef]
- Rossi, L.; Mazzara, C.; Pagani, O. Diagnosis and Treatment of Breast Cancer in Young Women. Curr. Treat. Options Oncol. 2019, 20, 86. [Google Scholar] [CrossRef]
- Sada, A.; Day, C.N.; Hoskin, T.L.; Degnim, A.C.; Habermann, E.B.; Hieken, T.J. Mastectomy and immediate breast reconstruction in the elderly: Trends and outcomes. Surgery 2019, 166, 709–714. [Google Scholar] [CrossRef]
- Paolucci, T.; Bernetti, A.; Bai, A.V.; Segatori, L.; Monti, M.; Maggi, G.; Ippolitoni, G.; Tinelli, L.; Santilli, V.; Paoloni, M.; et al. The sequelae of mastectomy and quadrantectomy with respect to the reaching movement in breast cancer survivors: Evidence for an integrated rehabilitation protocol during oncological care. Support. Care Cancer 2021, 29, 899–908. [Google Scholar] [CrossRef] [PubMed]
- Pellegrino, B.; Hlavata, Z.; Migali, C.; De Silva, P.; Aiello, M.; Willard-Gallo, K.; Musolino, A.; Solinas, C. Luminal Breast Cancer: Risk of Recurrence and Tumor-Associated Immune Suppression. Mol. Diagn. Ther. 2021, 25, 409–424. [Google Scholar] [CrossRef] [PubMed]
- Akram, M.; Iqbal, M.; Daniyal, M.; Khan, A.U. Awareness and current knowledge of breast cancer. Biol. Res. 2017, 50, 33. [Google Scholar] [CrossRef]
- Gong, L.; Zhang, Y.; Liu, C.; Zhang, M.; Han, S. Application of Radiosensitizers in Cancer Radiotherapy. Int. J. Nanomed. 2021, 16, 1083–1102. [Google Scholar] [CrossRef]
- He, L.; Lv, Y.; Song, Y.; Zhang, B. The prognosis comparison of different molecular subtypes of breast tumors after radiotherapy and the intrinsic reasons for their distinct radiosensitivity. Cancer Manag. Res. 2019, 11, 5765–5775. [Google Scholar] [CrossRef] [PubMed]
- Costa, M.S.; Gonçalves, Y.G.; Borges, B.C.; Silva, M.J.B.; Amstalden, M.K.; Costa, T.R.; Antunes, L.M.G.; Rodrigues, R.S.; Rodrigues, V.M.; de Faria Franca, E.; et al. Ruthenium (II) complex cis-[RuII(ŋ2-O2CC7H7O2)(dppm)2]PF6-hmxbato induces ROS-mediated apoptosis in lung tumor cells producing selective cytotoxicity. Sci. Rep. 2020, 10, 15410. [Google Scholar] [CrossRef] [PubMed]
- Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef]
- McDonald, E.S.; Clark, A.S.; Tchou, J.; Zhang, P.; Freedman, G.M. Clinical Diagnosis and Management of Breast Cancer. J. Nucl. Med. 2016, 57 (Suppl. 1), 9s–16s. [Google Scholar] [CrossRef]
- Lim, E.; Winer, E.P. Adjuvant chemotherapy in luminal breast cancers. Breast 2011, 20 (Suppl. 3), S128–S131. [Google Scholar] [CrossRef]
- Goncalves, A.; Finetti, P.; Birnbaum, D.; Bertucci, F. The CINSARC signature predicts the clinical outcome in patients with Luminal B breast cancer. npj Breast Cancer 2021, 7, 48. [Google Scholar] [CrossRef]
- Uchida, N.; Suda, T.; Ishiguro, K. Effect of chemotherapy for luminal a breast cancer. Yonago Acta Med. 2013, 56, 51–56. [Google Scholar] [PubMed]
- de Gregorio, A.; Janni, W.; Friedl, T.W.P.; Nitz, U.; Rack, B.; Schneeweiss, A.; Kates, R.; Fehm, T.; Kreipe, H.; Christgen, M.; et al. The impact of anthracyclines in intermediate and high-risk HER2-negative early breast cancer-a pooled analysis of the randomised clinical trials PlanB and SUCCESS C. Br. J. Cancer 2022, 126, 1715–1724. [Google Scholar] [CrossRef] [PubMed]
- Denduluri, N.; Somerfield, M.R.; Eisen, A.; Holloway, J.N.; Hurria, A.; King, T.A.; Lyman, G.H.; Partridge, A.H.; Telli, M.L.; Trudeau, M.E.; et al. Selection of Optimal Adjuvant Chemotherapy Regimens for Human Epidermal Growth Factor Receptor 2 (HER2) -Negative and Adjuvant Targeted Therapy for HER2-Positive Breast Cancers: An American Society of Clinical Oncology Guideline Adaptation of the Cancer Care Ontario Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 2416–2427. [Google Scholar] [CrossRef] [PubMed]
- Burstein, H.J. Systemic Therapy for Estrogen Receptor-Positive, HER2-Negative Breast Cancer. N. Engl. J. Med. 2020, 383, 2557–2570. [Google Scholar] [CrossRef]
- Awan, A.; Esfahani, K. Endocrine therapy for breast cancer in the primary care setting. Curr. Oncol. 2018, 25, 285–291. [Google Scholar] [CrossRef]
- Liu, C.Y.; Wu, C.Y.; Petrossian, K.; Huang, T.T.; Tseng, L.M.; Chen, S. Treatment for the endocrine resistant breast cancer: Current options and future perspectives. J. Steroid Biochem. Mol. Biol. 2017, 172, 166–175. [Google Scholar] [CrossRef]
- Liu, S.; Han, S.J.; Smith, C.L. Cooperative activation of gene expression by agonists and antagonists mediated by estrogen receptor heteroligand dimer complexes. Mol. Pharmacol. 2013, 83, 1066–1077. [Google Scholar] [CrossRef]
- Meade, E.; Dowling, M. Early breast cancer: Diagnosis, treatment and survivorship. Br. J. Nurs. 2012, 21, S4–S7. [Google Scholar] [CrossRef]
- Reinbolt, R.E.; Mangini, N.; Hill, J.L.; Levine, L.B.; Dempsey, J.L.; Singaravelu, J.; Koehler, K.A.; Talley, A.; Lustberg, M.B. Endocrine therapy in breast cancer: The neoadjuvant, adjuvant, and metastatic approach. Semin. Oncol. Nurs. 2015, 31, 146–155. [Google Scholar] [CrossRef]
- Cuzick, J.; Sestak, I.; Cawthorn, S.; Hamed, H.; Holli, K.; Howell, A.; Forbes, J.F. Tamoxifen for prevention of breast cancer: Extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet. Oncol. 2015, 16, 67–75. [Google Scholar] [CrossRef]
- Rimawi, M.; Ferrero, J.M.; de la Haba-Rodriguez, J.; Poole, C.; De Placido, S.; Osborne, C.K.; Hegg, R.; Easton, V.; Wohlfarth, C.; Arpino, G. First-Line Trastuzumab Plus an Aromatase Inhibitor, With or Without Pertuzumab, in Human Epidermal Growth Factor Receptor 2-Positive and Hormone Receptor-Positive Metastatic or Locally Advanced Breast Cancer (PERTAIN): A Randomized, Open-Label Phase II Trial. J. Clin. Oncol. 2018, 36, 2826–2835. [Google Scholar] [CrossRef] [PubMed]
- Emons, G.; Mustea, A.; Tempfer, C. Tamoxifen and Endometrial Cancer: A Janus-Headed Drug. Cancers 2020, 12, 2535. [Google Scholar] [CrossRef] [PubMed]
- Belachew, E.B.; Sewasew, D.T. Molecular Mechanisms of Endocrine Resistance in Estrogen-Positive Breast Cancer. Front. Endocrinol. 2021, 12, 599586. [Google Scholar] [CrossRef]
- Ferraldeschi, R.; Newman, W.G. The Impact of CYP2D6 Genotyping on Tamoxifen Treatment. Pharmaceuticals 2010, 3, 1122–1138. [Google Scholar] [CrossRef]
- Reid, J.M.; Goetz, M.P.; Buhrow, S.A.; Walden, C.; Safgren, S.L.; Kuffel, M.J.; Reinicke, K.E.; Suman, V.; Haluska, P.; Hou, X.; et al. Pharmacokinetics of endoxifen and tamoxifen in female mice: Implications for comparative in vivo activity studies. Cancer Chemother. Pharmacol. 2014, 74, 1271–1278. [Google Scholar] [CrossRef]
- Jones, C.J.; Subramaniam, M.; Emch, M.J.; Bruinsma, E.S.; Ingle, J.N.; Goetz, M.P.; Hawse, J.R. Development and Characterization of Novel Endoxifen-Resistant Breast Cancer Cell Lines Highlight Numerous Differences from Tamoxifen-Resistant Models. Mol. Cancer Res. 2021, 19, 1026–1039. [Google Scholar] [CrossRef]
- Davies, C.; Pan, H.; Godwin, J.; Gray, R.; Arriagada, R.; Raina, V.; Abraham, M.; Medeiros Alencar, V.H.; Badran, A.; Bonfill, X.; et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013, 381, 805–816. [Google Scholar] [CrossRef] [PubMed]
- Lumachi, F.; Santeufemia, D.A.; Basso, S.M. Current medical treatment of estrogen receptor-positive breast cancer. World J. Biol. Chem. 2015, 6, 231–239. [Google Scholar] [CrossRef] [PubMed]
- Datta, J.; Willingham, N.; Manouchehri, J.M.; Schnell, P.; Sheth, M.; David, J.J.; Kassem, M.; Wilson, T.A.; Radomska, H.S.; Coss, C.C.; et al. Activity of Estrogen Receptor β Agonists in Therapy-Resistant Estrogen Receptor-Positive Breast Cancer. Front. Oncol. 2022, 12, 857590. [Google Scholar] [CrossRef]
- Shagufta; Ahmad, I.; Mathew, S.; Rahman, S. Recent progress in selective estrogen receptor downregulators (SERDs) for the treatment of breast cancer. RSC Med. Chem. 2020, 11, 438–454. [Google Scholar] [CrossRef]
- Osborne, C.K.; Wakeling, A.; Nicholson, R.I. Fulvestrant: An oestrogen receptor antagonist with a novel mechanism of action. Br. J. Cancer 2004, 90 (Suppl. 1), S2–S6. [Google Scholar] [CrossRef] [PubMed]
- Mottamal, M.; Kang, B.; Peng, X.; Wang, G. From Pure Antagonists to Pure Degraders of the Estrogen Receptor: Evolving Strategies for the Same Target. ACS Omega 2021, 6, 9334–9343. [Google Scholar] [CrossRef] [PubMed]
- Rocca, A.; Maltoni, R.; Bravaccini, S.; Donati, C.; Andreis, D. Clinical utility of fulvestrant in the treatment of breast cancer: A report on the emerging clinical evidence. Cancer Manag. Res. 2018, 10, 3083–3099. [Google Scholar] [CrossRef]
- Ikeda, H.; Taira, N.; Nogami, T.; Shien, K.; Okada, M.; Shien, T.; Doihara, H.; Miyoshi, S. Combination treatment with fulvestrant and various cytotoxic agents (doxorubicin, paclitaxel, docetaxel, vinorelbine, and 5-fluorouracil) has a synergistic effect in estrogen receptor-positive breast cancer. Cancer Sci. 2011, 102, 2038–2042. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Cai, L.; Song, Y.; Sun, T.; Tong, Z.; Teng, Y.; Li, H.; Ouyang, Q.; Chen, Q.; Cui, S.; et al. Clinical efficacy of fulvestrant versus exemestane as first-line therapies for Chinese postmenopausal oestrogen-receptor positive/human epidermal growth factor receptor 2 -advanced breast cancer (FRIEND study). Eur. J. Cancer 2023, 184, 73–82. [Google Scholar] [CrossRef]
- Santen, R.J.; Brodie, H.; Simpson, E.R.; Siiteri, P.K.; Brodie, A. History of aromatase: Saga of an important biological mediator and therapeutic target. Endocr. Rev. 2009, 30, 343–375. [Google Scholar] [CrossRef]
- Clarke, R.; Tyson, J.J.; Dixon, J.M. Endocrine resistance in breast cancer—An overview and update. Mol. Cell. Endocrinol. 2015, 418 Pt 3, 220–234. [Google Scholar] [CrossRef]
- Chumsri, S. Clinical utilities of aromatase inhibitors in breast cancer. Int. J. Women’s Health 2015, 7, 493–499. [Google Scholar] [CrossRef]
- Walker, G.; Xenophontos, M.; Chen, L.; Cheung, K. Long-term efficacy and safety of exemestane in the treatment of breast cancer. Patient Prefer. Adherence 2013, 7, 245–258. [Google Scholar] [CrossRef]
- Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Aromatase inhibitors versus tamoxifen in early breast cancer: Patient-level meta-analysis of the randomised trials. Lancet 2015, 386, 1341–1352. [Google Scholar] [CrossRef]
- Gnant, M.; Fitzal, F.; Rinnerthaler, G.; Steger, G.G.; Greil-Ressler, S.; Balic, M.; Heck, D.; Jakesz, R.; Thaler, J.; Egle, D.; et al. Duration of Adjuvant Aromatase-Inhibitor Therapy in Postmenopausal Breast Cancer. N. Engl. J. Med. 2021, 385, 395–405. [Google Scholar] [CrossRef] [PubMed]
- Hyder, T.; Marino, C.C.; Ahmad, S.; Nasrazadani, A.; Brufsky, A.M. Aromatase Inhibitor-Associated Musculoskeletal Syndrome: Understanding Mechanisms and Management. Front. Endocrinol. 2021, 12, 713700. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Pan, W.; Tang, X.; Wu, S.; Sun, X. A meta-analysis of randomized controlled trials comparing the efficacy and safety of anastrozole versus tamoxifen for breast cancer. Oncotarget 2017, 8, 48362–48374. [Google Scholar] [CrossRef] [PubMed]
- Huerta-Reyes, M.; Maya-Núñez, G.; Pérez-Solis, M.A.; López-Muñoz, E.; Guillén, N.; Olivo-Marin, J.C.; Aguilar-Rojas, A. Treatment of Breast Cancer with Gonadotropin-Releasing Hormone Analogs. Front. Oncol. 2019, 9, 943. [Google Scholar] [CrossRef]
- Dos Santos, B.S.; Bordignon, C.; Rosa, D.D. Managing Common Estrogen Deprivation Side Effects in HR+ Breast Cancer: An Evidence-Based Review. Curr. Oncol. Rep. 2021, 23, 63. [Google Scholar] [CrossRef]
- Prieto-Callejero, B.; Rivera, F.; Fagundo-Rivera, J.; Romero, A.; Romero-Martín, M.; Gómez-Salgado, J.; Ruiz-Frutos, C. Relationship between chemotherapy-induced adverse reactions and health-related quality of life in patients with breast cancer. Medicine 2020, 99, e21695. [Google Scholar] [CrossRef]
- Aiello Bowles, E.J.; Boudreau, D.M.; Chubak, J.; Yu, O.; Fujii, M.; Chestnut, J.; Buist, D.S. Patient-reported discontinuation of endocrine therapy and related adverse effects among women with early-stage breast cancer. J. Oncol. Pract. 2012, 8, e149–e157. [Google Scholar] [CrossRef]
- Riemsma, R.; Forbes, C.A.; Kessels, A.; Lykopoulos, K.; Amonkar, M.M.; Rea, D.W.; Kleijnen, J. Systematic review of aromatase inhibitors in the first-line treatment for hormone sensitive advanced or metastatic breast cancer. Breast Cancer Res. Treat. 2010, 123, 9–24. [Google Scholar] [CrossRef]
- Gibson, L.; Lawrence, D.; Dawson, C.; Bliss, J. Aromatase inhibitors for treatment of advanced breast cancer in postmenopausal women. Cochrane Database Syst. Rev. 2009, 2009, CD003370. [Google Scholar] [CrossRef]
- Fontes-Sousa, M.; Amorim, M.; Salta, S.; Palma De Sousa, S.; Henrique, R.; Jerónimo, C. Predicting resistance to endocrine therapy in breast cancer: It’s time for epigenetic biomarkers (Review). Oncol. Rep. 2019, 41, 1431–1438. [Google Scholar] [CrossRef]
- Krauss, K.; Stickeler, E. Endocrine Therapy in Early Breast Cancer. Breast Care 2020, 15, 337–346. [Google Scholar] [CrossRef] [PubMed]
- Hernando, C.; Ortega-Morillo, B.; Tapia, M.; Moragón, S.; Martínez, M.T.; Eroles, P.; Garrido-Cano, I.; Adam-Artigues, A.; Lluch, A.; Bermejo, B.; et al. Oral Selective Estrogen Receptor Degraders (SERDs) as a Novel Breast Cancer Therapy: Present and Future from a Clinical Perspective. Int. J. Mol. Sci. 2021, 22, 7812. [Google Scholar] [CrossRef] [PubMed]
- Ferraro, E.; Walsh, E.M.; Tao, J.J.; Chandarlapaty, S.; Jhaveri, K. Accelerating drug development in breast cancer: New frontiers for ER inhibition. Cancer Treat. Rev. 2022, 109, 102432. [Google Scholar] [CrossRef] [PubMed]
- Hartkopf, A.D.; Grischke, E.M.; Brucker, S.Y. Endocrine-Resistant Breast Cancer: Mechanisms and Treatment. Breast Care 2020, 15, 347–354. [Google Scholar] [CrossRef] [PubMed]
- Cottu, P.; D’Hondt, V.; Dureau, S.; Lerebours, F.; Desmoulins, I.; Heudel, P.E.; Duhoux, F.P.; Levy, C.; Mouret-Reynier, M.A.; Dalenc, F.; et al. Letrozole and palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer. Ann. Oncol. 2018, 29, 2334–2340. [Google Scholar] [CrossRef]
- Goss, P.E.; Ingle, J.N.; Pritchard, K.I.; Robert, N.J.; Muss, H.; Gralow, J.; Gelmon, K.; Whelan, T.; Strasser-Weippl, K.; Rubin, S.; et al. Extending Aromatase-Inhibitor Adjuvant Therapy to 10 Years. N. Engl. J. Med. 2016, 375, 209–219. [Google Scholar] [CrossRef]
- Schiff, R.; Massarweh, S.A.; Shou, J.; Bharwani, L.; Mohsin, S.K.; Osborne, C.K. Cross-talk between estrogen receptor and growth factor pathways as a molecular target for overcoming endocrine resistance. Clin. Cancer Res. 2004, 10, 331s–336s. [Google Scholar] [CrossRef]
- Murphy, C.G.; Dickler, M.N. Endocrine resistance in hormone-responsive breast cancer: Mechanisms and therapeutic strategies. Endocr.-Relat. Cancer 2016, 23, R337–R352. [Google Scholar] [CrossRef]
- García-Becerra, R.; Santos, N.; Díaz, L.; Camacho, J. Mechanisms of resistance to endocrine therapy in breast cancer: Focus on signaling pathways, miRNAs and genetically based resistance. Int. J. Mol. Sci. 2012, 14, 108–145. [Google Scholar] [CrossRef]
- Fan, W.; Chang, J.; Fu, P. Endocrine therapy resistance in breast cancer: Current status, possible mechanisms and overcoming strategies. Future Med. Chem. 2015, 7, 1511–1519. [Google Scholar] [CrossRef]
- Cardoso, F.; Paluch-Shimon, S.; Senkus, E.; Curigliano, G.; Aapro, M.S.; André, F.; Barrios, C.H.; Bergh, J.; Bhattacharyya, G.S.; Biganzoli, L.; et al. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann. Oncol. 2020, 31, 1623–1649. [Google Scholar] [CrossRef]
- Chandarlapaty, S.; Chen, D.; He, W.; Sung, P.; Samoila, A.; You, D.; Bhatt, T.; Patel, P.; Voi, M.; Gnant, M.; et al. Prevalence of ESR1 Mutations in Cell-Free DNA and Outcomes in Metastatic Breast Cancer: A Secondary Analysis of the BOLERO-2 Clinical Trial. JAMA Oncol. 2016, 2, 1310–1315. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.; Lee, J.; Choi, J.; Sim, S.H.; Kim, J.E.; Kim, M.H.; Park, Y.H.; Kim, J.H.; Koh, S.J.; Park, K.H.; et al. Genomic analysis of plasma circulating tumor DNA in patients with heavily pretreated HER2+ metastatic breast cancer. Sci. Rep. 2023, 13, 9928. [Google Scholar] [CrossRef] [PubMed]
- De Santo, I.; McCartney, A.; Migliaccio, I.; Di Leo, A.; Malorni, L. The Emerging Role of ESR1 Mutations in Luminal Breast Cancer as a Prognostic and Predictive Biomarker of Response to Endocrine Therapy. Cancers 2019, 11, 1894. [Google Scholar] [CrossRef] [PubMed]
- Rasha, F.; Sharma, M.; Pruitt, K. Mechanisms of endocrine therapy resistance in breast cancer. Mol. Cell. Endocrinol. 2021, 532, 111322. [Google Scholar] [CrossRef]
- Bostner, J.; Alayev, A.; Berman, A.Y.; Fornander, T.; Nordenskjöld, B.; Holz, M.K.; Stål, O. Raptor localization predicts prognosis and tamoxifen response in estrogen receptor-positive breast cancer. Breast Cancer Res. Treat. 2018, 168, 17–27. [Google Scholar] [CrossRef]
- Kastrati, I.; Semina, S.; Gordon, B.; Smart, E. Insights into how phosphorylation of estrogen receptor at serine 305 modulates tamoxifen activity in breast cancer. Mol. Cell. Endocrinol. 2019, 483, 97–101. [Google Scholar] [CrossRef]
- Barone, I.; Iacopetta, D.; Covington, K.R.; Cui, Y.; Tsimelzon, A.; Beyer, A.; Andò, S.; Fuqua, S.A. Phosphorylation of the mutant K303R estrogen receptor alpha at serine 305 affects aromatase inhibitor sensitivity. Oncogene 2010, 29, 2404–2414. [Google Scholar] [CrossRef]
- Karami Fath, M.; Azargoonjahromi, A.; Kiani, A.; Jalalifar, F.; Osati, P.; Akbari Oryani, M.; Shakeri, F.; Nasirzadeh, F.; Khalesi, B.; Nabi-Afjadi, M.; et al. The role of epigenetic modifications in drug resistance and treatment of breast cancer. Cell. Mol. Biol. Lett. 2022, 27, 52. [Google Scholar] [CrossRef]
- Ma, C.X.; Reinert, T.; Chmielewska, I.; Ellis, M.J. Mechanisms of aromatase inhibitor resistance. Nat. Rev. Cancer 2015, 15, 261–275. [Google Scholar] [CrossRef]
- Fribbens, C.; O’Leary, B.; Kilburn, L.; Hrebien, S.; Garcia-Murillas, I.; Beaney, M.; Cristofanilli, M.; Andre, F.; Loi, S.; Loibl, S.; et al. Plasma ESR1 Mutations and the Treatment of Estrogen Receptor-Positive Advanced Breast Cancer. J. Clin. Oncol. 2016, 34, 2961–2968. [Google Scholar] [CrossRef] [PubMed]
- Hopp, T.A.; Weiss, H.L.; Parra, I.S.; Cui, Y.; Osborne, C.K.; Fuqua, S.A. Low levels of estrogen receptor beta protein predict resistance to tamoxifen therapy in breast cancer. Clin. Cancer Res. 2004, 10, 7490–7499. [Google Scholar] [CrossRef] [PubMed]
- Choi, Y. Estrogen Receptor β Expression and Its Clinical Implication in Breast Cancers: Favorable or Unfavorable? J. Breast Cancer 2022, 25, 75–93. [Google Scholar] [CrossRef] [PubMed]
- Murphy, L.C.; Leygue, E.; Niu, Y.; Snell, L.; Ho, S.M.; Watson, P.H. Relationship of coregulator and oestrogen receptor isoform expression to de novo tamoxifen resistance in human breast cancer. Br. J. Cancer 2002, 87, 1411–1416. [Google Scholar] [CrossRef] [PubMed]
- Hamilton-Burke, W.; Coleman, L.; Cummings, M.; Green, C.A.; Holliday, D.L.; Horgan, K.; Maraqa, L.; Peter, M.B.; Pollock, S.; Shaaban, A.M.; et al. Phosphorylation of estrogen receptor beta at serine 105 is associated with good prognosis in breast cancer. Am. J. Pathol. 2010, 177, 1079–1086. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Deng, C.; Lu, W.; Xiao, J.; Ma, D.; Guo, M.; Recker, R.R.; Gatalica, Z.; Wang, Z.; Xiao, G.G. let-7 microRNAs induce tamoxifen sensitivity by downregulation of estrogen receptor α signaling in breast cancer. Mol. Med. 2011, 17, 1233–1241. [Google Scholar] [CrossRef]
- Szostakowska, M.; Trębińska-Stryjewska, A.; Grzybowska, E.A.; Fabisiewicz, A. Resistance to endocrine therapy in breast cancer: Molecular mechanisms and future goals. Breast Cancer Res. Treat. 2019, 173, 489–497. [Google Scholar] [CrossRef]
- Butti, R.; Das, S.; Gunasekaran, V.P.; Yadav, A.S.; Kumar, D.; Kundu, G.C. Receptor tyrosine kinases (RTKs) in breast cancer: Signaling, therapeutic implications and challenges. Mol. Cancer 2018, 17, 34. [Google Scholar] [CrossRef]
- Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef]
- Lee, J.S.; Tocheny, C.E.; Shaw, L.M. The Insulin-like Growth Factor Signaling Pathway in Breast Cancer: An Elusive Therapeutic Target. Life 2022, 12, 1992. [Google Scholar] [CrossRef]
- Sutherland, R.L. Endocrine resistance in breast cancer: New roles for ErbB3 and ErbB4. Breast Cancer Res. 2011, 13, 106. [Google Scholar] [CrossRef] [PubMed]
- Rani, A.; Stebbing, J.; Giamas, G.; Murphy, J. Endocrine Resistance in Hormone Receptor Positive Breast Cancer-From Mechanism to Therapy. Front. Endocrinol. 2019, 10, 245. [Google Scholar] [CrossRef] [PubMed]
- Ortega, M.A.; Fraile-Martínez, O.; Asúnsolo, Á.; Buján, J.; García-Honduvilla, N.; Coca, S. Signal Transduction Pathways in Breast Cancer: The Important Role of PI3K/Akt/mTOR. J. Oncol. 2020, 2020, 9258396. [Google Scholar] [CrossRef]
- Dong, C.; Wu, J.; Chen, Y.; Nie, J.; Chen, C. Activation of PI3K/AKT/mTOR Pathway Causes Drug Resistance in Breast Cancer. Front. Pharmacol. 2021, 12, 628690. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Prever, L.; Hirsch, E.; Gulluni, F. Targeting PI3K/AKT/mTOR Signaling Pathway in Breast Cancer. Cancers 2021, 13, 3517. [Google Scholar] [CrossRef]
- Fusco, N.; Malapelle, U.; Fassan, M.; Marchiò, C.; Buglioni, S.; Zupo, S.; Criscitiello, C.; Vigneri, P.; Dei Tos, A.P.; Maiorano, E.; et al. PIK3CA Mutations as a Molecular Target for Hormone Receptor-Positive, HER2-Negative Metastatic Breast Cancer. Front. Oncol. 2021, 11, 644737. [Google Scholar] [CrossRef]
- Hare, S.H.; Harvey, A.J. mTOR function and therapeutic targeting in breast cancer. Am. J. Cancer Res. 2017, 7, 383–404. [Google Scholar]
- Ngeow, J.; Sesock, K.; Eng, C. Breast cancer risk and clinical implications for germline PTEN mutation carriers. Breast Cancer Res. Treat. 2017, 165, 1–8. [Google Scholar] [CrossRef]
- Kilker, R.L.; Planas-Silva, M.D. Cyclin D1 is necessary for tamoxifen-induced cell cycle progression in human breast cancer cells. Cancer Res. 2006, 66, 11478–11484. [Google Scholar] [CrossRef]
- Wilcken, N.R.; Prall, O.W.; Musgrove, E.A.; Sutherland, R.L. Inducible overexpression of cyclin D1 in breast cancer cells reverses the growth-inhibitory effects of antiestrogens. Clin. Cancer Res. 1997, 3, 849–854. [Google Scholar]
- Brufsky, A.M.; Dickler, M.N. Estrogen Receptor-Positive Breast Cancer: Exploiting Signaling Pathways Implicated in Endocrine Resistance. Oncologist 2018, 23, 528–539. [Google Scholar] [CrossRef]
- Kenny, F.S.; Hui, R.; Musgrove, E.A.; Gee, J.M.; Blamey, R.W.; Nicholson, R.I.; Sutherland, R.L.; Robertson, J.F. Overexpression of cyclin D1 messenger RNA predicts for poor prognosis in estrogen receptor-positive breast cancer. Clin. Cancer Res. 1999, 5, 2069–2076. [Google Scholar] [PubMed]
- Thangavel, C.; Dean, J.L.; Ertel, A.; Knudsen, K.E.; Aldaz, C.M.; Witkiewicz, A.K.; Clarke, R.; Knudsen, E.S. Therapeutically activating RB: Reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr.-Relat. Cancer 2011, 18, 333–345. [Google Scholar] [CrossRef] [PubMed]
- Abraham, J.; Coleman, R.; Elias, A.; Holmes, F.A.; Kalinsky, K.; Kittaneh, M.; Lower, E.; Mahtani, R.; Terry Mamounas, E.; Pegram, M.; et al. Use of cyclin-dependent kinase (CDK) 4/6 inhibitors for hormone receptor-positive, human epidermal growth factor receptor 2-negative, metastatic breast cancer: A roundtable discussion by The Breast Cancer Therapy Expert Group (BCTEG). Breast Cancer Res. Treat. 2018, 171, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Lian, B.; Chen, X.; Shen, K. Inhibition of histone deacetylases attenuates tumor progression and improves immunotherapy in breast cancer. Front. Immunol. 2023, 14, 1164514. [Google Scholar] [CrossRef]
- Slamon, D.J.; Neven, P.; Chia, S.; Jerusalem, G.; De Laurentiis, M.; Im, S.; Petrakova, K.; Valeria Bianchi, G.; Martín, M.; Nusch, A.; et al. Ribociclib plus fulvestrant for postmenopausal women with hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer in the phase III randomized MONALEESA-3 trial: Updated overall survival. Ann. Oncol. 2021, 32, 1015–1024. [Google Scholar] [CrossRef]
- Raha, P.; Thomas, S.; Thurn, K.T.; Park, J.; Munster, P.N. Combined histone deacetylase inhibition and tamoxifen induces apoptosis in tamoxifen-resistant breast cancer models, by reversing Bcl-2 overexpression. Breast Cancer Res. 2015, 17, 26. [Google Scholar] [CrossRef]
- Baselga, J.; Campone, M.; Piccart, M.; Burris, H.A., 3rd; Rugo, H.S.; Sahmoud, T.; Noguchi, S.; Gnant, M.; Pritchard, K.I.; Lebrun, F.; et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N. Engl. J. Med. 2012, 366, 520–529. [Google Scholar] [CrossRef]
- Prajapati, K.S.; Gupta, S.; Kumar, S. Targeting Breast Cancer-Derived Stem Cells by Dietary Phytochemicals: A Strategy for Cancer Prevention and Treatment. Cancers 2022, 14, 2864. [Google Scholar] [CrossRef]
- Kushwaha, P.P.; Vardhan, P.S.; Kapewangolo, P.; Shuaib, M.; Prajapati, S.K.; Singh, A.K.; Kumar, S. Bulbine frutescens phytochemical inhibits notch signaling pathway and induces apoptosis in triple negative and luminal breast cancer cells. Life Sci. 2019, 234, 116783. [Google Scholar] [CrossRef]
- Moudgil, K.D.; Venkatesha, S.H. The Anti-Inflammatory and Immunomodulatory Activities of Natural Products to Control Autoimmune Inflammation. Int. J. Mol. Sci. 2022, 24, 95. [Google Scholar] [CrossRef] [PubMed]
- Zhong, L.; Luo, N.; Zhong, X.; Xu, T.; Hao, P. The immunoregulatory effects of natural products on psoriasis via its action on Th17 cells versus regulatory T cells balance. Int. Immunopharmacol. 2022, 110, 109032. [Google Scholar] [CrossRef] [PubMed]
- Liu, Q.P.; Chen, Y.Y.; An, P.; Rahman, K.; Luan, X.; Zhang, H. Natural products targeting macrophages in tumor microenvironment are a source of potential antitumor agents. Phytomedicine 2023, 109, 154612. [Google Scholar] [CrossRef] [PubMed]
- Wurtzel, E.T.; Kutchan, T.M. Plant metabolism, the diverse chemistry set of the future. Science 2016, 353, 1232–1236. [Google Scholar] [CrossRef]
- Khan, M.I.; Bouyahya, A.; Hachlafi, N.E.L.; Menyiy, N.E.; Akram, M.; Sultana, S.; Zengin, G.; Ponomareva, L.; Shariati, M.A.; Ojo, O.A.; et al. Anticancer properties of medicinal plants and their bioactive compounds against breast cancer: A review on recent investigations. Environ. Sci. Pollut. Res. Int. 2022, 29, 24411–24444. [Google Scholar] [CrossRef]
- Tanwar, A.K.; Dhiman, N.; Kumar, A.; Jaitak, V. Engagement of phytoestrogens in breast cancer suppression: Structural classification and mechanistic approach. Eur. J. Med. Chem. 2021, 213, 113037. [Google Scholar] [CrossRef]
- Dewi, C.; Fristiohady, A.; Amalia, R.; Ikram, N.K.K.; Ibrahim, S.; Muchtaridi, M. Signaling Pathways and Natural Compounds in Triple-Negative Breast Cancer Cell Line. Molecules 2022, 27, 3661. [Google Scholar] [CrossRef]
- Malayil, A.S.; Poothaadammal, A.K.; Narayanankutty, A. Natural Bioactive Compounds as Emerging Therapeutic Molecules Against Breast Cancer: Emphasis on the Role of Phytoestrogens. Curr. Drug Targets 2021, 22, 1255–1271. [Google Scholar] [CrossRef]
- Yuan, Y.; Long, H.; Zhou, Z.; Fu, Y.; Jiang, B. PI3K-AKT-Targeting Breast Cancer Treatments: Natural Products and Synthetic Compounds. Biomolecules 2023, 13, 93. [Google Scholar] [CrossRef]
- Madureira, M.B.; Concato, V.M.; Cruz, E.M.S.; Bitencourt de Morais, J.M.; Inoue, F.S.R.; Concimo Santos, N.; Gonçalves, M.D.; Cremer de Souza, M.; Basso Scandolara, T.; Fontana Mezoni, M.; et al. Naringenin and Hesperidin as Promising Alternatives for Prevention and Co-Adjuvant Therapy for Breast Cancer. Antioxidants 2023, 12, 586. [Google Scholar] [CrossRef]
- Choi, E.J. Hesperetin induced G1-phase cell cycle arrest in human breast cancer MCF-7 cells: Involvement of CDK4 and p21. Nutr. Cancer 2007, 59, 115–119. [Google Scholar] [CrossRef] [PubMed]
- Fasoulakis, Z.; Koutras, A.; Syllaios, A.; Schizas, D.; Garmpis, N.; Diakosavvas, M.; Angelou, K.; Tsatsaris, G.; Pagkalos, A.; Ntounis, T.; et al. Breast Cancer Apoptosis and the Therapeutic Role of Luteolin. Chirurgia 2021, 116, 170–177. [Google Scholar] [CrossRef] [PubMed]
- Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef] [PubMed]
- Cao, X.; Liu, B.; Cao, W.; Zhang, W.; Zhang, F.; Zhao, H.; Meng, R.; Zhang, L.; Niu, R.; Hao, X.; et al. Autophagy inhibition enhances apigenin-induced apoptosis in human breast cancer cells. Chin. J. Cancer Res. 2013, 25, 212–222. [Google Scholar] [CrossRef] [PubMed]
- Vrhovac Madunić, I.; Madunić, J.; Antunović, M.; Paradžik, M.; Garaj-Vrhovac, V.; Breljak, D.; Marijanović, I.; Gajski, G. Apigenin, a dietary flavonoid, induces apoptosis, DNA damage, and oxidative stress in human breast cancer MCF-7 and MDA MB-231 cells. Naunyn-Schmiedebergs Arch. Pharmacol. 2018, 391, 537–550. [Google Scholar] [CrossRef] [PubMed]
- Hwang, K.A.; Choi, K.C. Anticarcinogenic Effects of Dietary Phytoestrogens and Their Chemopreventive Mechanisms. Nutr. Cancer 2015, 67, 796–803. [Google Scholar] [CrossRef] [PubMed]
- Zafar, A.; Singh, S.; Naseem, I. Cytotoxic activity of soy phytoestrogen coumestrol against human breast cancer MCF-7 cells: Insights into the molecular mechanism. Food Chem. Toxicol. 2017, 99, 149–161. [Google Scholar] [CrossRef]
- Zava, D.T.; Dollbaum, C.M.; Blen, M. Estrogen and progestin bioactivity of foods, herbs, and spices. Proc. Soc. Exp. Biol. Med. Soc. Exp. Biol. Med. 1998, 217, 369–378. [Google Scholar] [CrossRef]
- Chen, H.H.; Chen, S.P.; Zheng, Q.L.; Nie, S.P.; Li, W.J.; Hu, X.J.; Xie, M.Y. Genistein Promotes Proliferation of Human Cervical Cancer Cells Through Estrogen Receptor-Mediated PI3K/Akt-NF-κB Pathway. J. Cancer 2018, 9, 288–295. [Google Scholar] [CrossRef]
- Li, Y.; Upadhyay, S.; Bhuiyan, M.; Sarkar, F.H. Induction of apoptosis in breast cancer cells MDA-MB-231 by genistein. Oncogene 1999, 18, 3166–3172. [Google Scholar] [CrossRef]
- Gong, L.; Li, Y.; Nedeljkovic-Kurepa, A.; Sarkar, F.H. Inactivation of NF-kappaB by genistein is mediated via Akt signaling pathway in breast cancer cells. Oncogene 2003, 22, 4702–4709. [Google Scholar] [CrossRef] [PubMed]
- Uifălean, A.; Schneider, S.; Gierok, P.; Ionescu, C.; Iuga, C.A.; Lalk, M. The Impact of Soy Isoflavones on MCF-7 and MDA-MB-231 Breast Cancer Cells Using a Global Metabolomic Approach. Int. J. Mol. Sci. 2016, 17, 1443. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Ge, B.; Wang, Y.; Ye, Y.; Zeng, S.; Huang, Z. Biochanin A promotes proliferation that involves a feedback loop of microRNA-375 and estrogen receptor alpha in breast cancer cells. Cell. Physiol. Biochem. 2015, 35, 639–646. [Google Scholar] [CrossRef] [PubMed]
- Sirotkin, A.V.; Harrath, A.H. Phytoestrogens and their effects. Eur. J. Pharmacol. 2014, 741, 230–236. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.X.; Zhang, H.; Peng, C. Puerarin: A review of pharmacological effects. Phytother. Res. 2014, 28, 961–975. [Google Scholar] [CrossRef]
- Tian, J.; Duan, Y.X.; Bei, C.Y.; Chen, J. Calycosin induces apoptosis by upregulation of RASD1 in human breast cancer cells MCF-7. Horm. Metab. Res. 2013, 45, 593–598. [Google Scholar] [CrossRef]
- Do, M.T.; Kim, H.G.; Choi, J.H.; Khanal, T.; Park, B.H.; Tran, T.P.; Jeong, T.C.; Jeong, H.G. Antitumor efficacy of piperine in the treatment of human HER2-overexpressing breast cancer cells. Food Chem. 2013, 141, 2591–2599. [Google Scholar] [CrossRef]
- Monteiro, C.B.A.d.G. EGCG do Chá Verde—Um Agente Natural Contra o Cancro de Mama. Master’s Thesis, Universidade de Coimbra, Coimbra, Portugal, 2018. [Google Scholar]
- López-Biedma, A.; Sánchez-Quesada, C.; Beltrán, G.; Delgado-Rodríguez, M.; Gaforio, J.J. Phytoestrogen (+)-pinoresinol exerts antitumor activity in breast cancer cells with different oestrogen receptor statuses. BMC Complement. Altern. Med. 2016, 16, 350. [Google Scholar] [CrossRef]
- He, Y.; Fan, Q.; Cai, T.; Huang, W.; Xie, X.; Wen, Y.; Shi, Z. Molecular mechanisms of the action of Arctigenin in cancer. Biomed. Pharmacother. 2018, 108, 403–407. [Google Scholar] [CrossRef]
- Hsieh, C.J.; Kuo, P.L.; Hsu, Y.C.; Huang, Y.F.; Tsai, E.M.; Hsu, Y.L. Arctigenin, a dietary phytoestrogen, induces apoptosis of estrogen receptor-negative breast cancer cells through the ROS/p38 MAPK pathway and epigenetic regulation. Free Radic. Biol. Med. 2014, 67, 159–170. [Google Scholar] [CrossRef]
- Xiong, X.Y.; Hu, X.J.; Li, Y.; Liu, C.M. Inhibitory Effects of Enterolactone on Growth and Metastasis in Human Breast Cancer. Nutr. Cancer 2015, 67, 1324–1332. [Google Scholar] [CrossRef] [PubMed]
- Abarzua, S.; Serikawa, T.; Szewczyk, M.; Richter, D.U.; Piechulla, B.; Briese, V. Antiproliferative activity of lignans against the breast carcinoma cell lines MCF 7 and BT 20. Arch. Gynecol. Obstet. 2012, 285, 1145–1151. [Google Scholar] [CrossRef] [PubMed]
- Reuben, S.C.; Gopalan, A.; Petit, D.M.; Bishayee, A. Modulation of angiogenesis by dietary phytoconstituents in the prevention and intervention of breast cancer. Mol. Nutr. Food Res. 2012, 56, 14–29. [Google Scholar] [CrossRef] [PubMed]
- Truan, J.S.; Chen, J.M.; Thompson, L.U. Comparative effects of sesame seed lignan and flaxseed lignan in reducing the growth of human breast tumors (MCF-7) at high levels of circulating estrogen in athymic mice. Nutr. Cancer 2012, 64, 65–71. [Google Scholar] [CrossRef]
- Bowers, L.W.; Lineberger, C.G.; Ford, N.A.; Rossi, E.L.; Punjala, A.; Camp, K.K.; Kimler, B.K.; Fabian, C.J.; Hursting, S.D. The flaxseed lignan secoisolariciresinol diglucoside decreases local inflammation, suppresses NFκB signaling, and inhibits mammary tumor growth. Breast Cancer Res. Treat. 2019, 173, 545–557. [Google Scholar] [CrossRef]
- Saggar, J.K.; Chen, J.; Corey, P.; Thompson, L.U. The effect of secoisolariciresinol diglucoside and flaxseed oil, alone and in combination, on MCF-7 tumor growth and signaling pathways. Nutr. Cancer 2010, 62, 533–542. [Google Scholar] [CrossRef]
- Lee, Y.H.; Yuk, H.J.; Park, K.H.; Bae, Y.S. Coumestrol induces senescence through protein kinase CKII inhibition-mediated reactive oxygen species production in human breast cancer and colon cancer cells. Food Chem. 2013, 141, 381–388. [Google Scholar] [CrossRef]
- Liu, S.; Hsieh, D.; Yang, Y.L.; Xu, Z.; Peto, C.; Jablons, D.M.; You, L. Coumestrol from the national cancer Institute’s natural product library is a novel inhibitor of protein kinase CK2. BMC Pharmacol. Toxicol. 2013, 14, 36. [Google Scholar] [CrossRef]
- Mikstacka, R.; Przybylska, D.; Rimando, A.M.; Baer-Dubowska, W. Inhibition of human recombinant cytochromes P450 CYP1A1 and CYP1B1 by trans-resveratrol methyl ethers. Mol. Nutr. Food Res. 2007, 51, 517–524. [Google Scholar] [CrossRef]
- Ma, Z.; Molavi, O.; Haddadi, A.; Lai, R.; Gossage, R.A.; Lavasanifar, A. Resveratrol analog trans 3,4,5,4′-tetramethoxystilbene (DMU-212) mediates anti-tumor effects via mechanism different from that of resveratrol. Cancer Chemother. Pharmacol. 2008, 63, 27–35. [Google Scholar] [CrossRef]
- Wang, Y.; Ding, L.; Wang, X.; Zhang, J.; Han, W.; Feng, L.; Sun, J.; Jin, H.; Wang, X.J. Pterostilbene simultaneously induces apoptosis, cell cycle arrest and cyto-protective autophagy in breast cancer cells. Am. J. Transl. Res. 2012, 4, 44–51. [Google Scholar] [PubMed]
- Woo, C.C.; Loo, S.Y.; Gee, V.; Yap, C.W.; Sethi, G.; Kumar, A.P.; Tan, K.H. Anticancer activity of thymoquinone in breast cancer cells: Possible involvement of PPAR-γ pathway. Biochem. Pharmacol. 2011, 82, 464–475. [Google Scholar] [CrossRef] [PubMed]
- Arafa el, S.A.; Zhu, Q.; Shah, Z.I.; Wani, G.; Barakat, B.M.; Racoma, I.; El-Mahdy, M.A.; Wani, A.A. Thymoquinone up-regulates PTEN expression and induces apoptosis in doxorubicin-resistant human breast cancer cells. Mutat. Res. 2011, 706, 28–35. [Google Scholar] [CrossRef] [PubMed]
- Charalambous, C.; Pitta, C.A.; Constantinou, A.I. Equol enhances tamoxifen’s anti-tumor activity by induction of caspase-mediated apoptosis in MCF-7 breast cancer cells. BMC Cancer 2013, 13, 238. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H. Ginsenoside Rh2 reverses P-glycoprotein-mediated multidrug resistance of MCF-7/ADM cells. Tumor 2007, 12, 365–369. [Google Scholar]
- Huynh, D.T.N.; Jin, Y.; Myung, C.S.; Heo, K.-S. Ginsenoside Rh1 Induces MCF-7 Cell Apoptosis and Autophagic Cell Death through ROS-Mediated Akt Signaling. Cancers 2021, 13, 1892. [Google Scholar] [CrossRef]
- Selvakumar, P.; Badgeley, A.; Murphy, P.; Anwar, H.; Sharma, U.; Lawrence, K.; Lakshmikuttyamma, A. Flavonoids and Other Polyphenols Act as Epigenetic Modifiers in Breast Cancer. Nutrients 2020, 12, 761. [Google Scholar] [CrossRef]
- Park, M.Y.; Kim, Y.; Ha, S.E.; Kim, H.H.; Bhosale, P.B.; Abusaliya, A.; Jeong, S.H.; Kim, G.S. Function and Application of Flavonoids in the Breast Cancer. Int. J. Mol. Sci. 2022, 23, 7732. [Google Scholar] [CrossRef]
- Kabała-Dzik, A.; Rzepecka-Stojko, A.; Kubina, R.; Iriti, M.; Wojtyczka, R.D.; Buszman, E.; Stojko, J. Flavonoids, bioactive components of propolis, exhibit cytotoxic activity and induce cell cycle arrest and apoptosis in human breast cancer cells MDA-MB-231 and MCF-7—A comparative study. Cell. Mol. Biol. 2018, 64, 1–10. [Google Scholar] [CrossRef]
- Pyrzynska, K. Hesperidin: A Review on Extraction Methods, Stability and Biological Activities. Nutrients 2022, 14, 2387. [Google Scholar] [CrossRef]
- Khamis, A.A.A.; Ali, E.M.M.; El-Moneim, M.A.A.; Abd-Alhaseeb, M.M.; El-Magd, M.A.; Salim, E.I. Hesperidin, piperine and bee venom synergistically potentiate the anticancer effect of tamoxifen against breast cancer cells. Biomed. Pharmacother. 2018, 105, 1335–1343. [Google Scholar] [CrossRef] [PubMed]
- Hsu, P.H.; Chen, W.H.; Juan-Lu, C.; Hsieh, S.C.; Lin, S.C.; Mai, R.-T.; Chen, S.-Y. Hesperidin and Chlorogenic Acid Synergistically Inhibit the Growth of Breast Cancer Cells via Estrogen Receptor/Mitochondrial Pathway. Life 2021, 11, 950. [Google Scholar] [CrossRef]
- Yap, K.M.; Sekar, M.; Wu, Y.S.; Gan, S.H.; Rani, N.; Seow, L.J.; Subramaniyan, V.; Fuloria, N.K.; Fuloria, S.; Lum, P.T. Hesperidin and its aglycone hesperetin in breast cancer therapy: A review of recent developments and future prospects. Saudi J. Biol. Sci. 2021, 28, 6730–6747. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Shi, R.; Wang, X.; Shen, H.M. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr. Cancer Drug Targets 2008, 8, 634–646. [Google Scholar] [CrossRef] [PubMed]
- Tsai, K.J.; Tsai, H.Y.; Tsai, C.C.; Chen, T.Y.; Hsieh, T.H.; Chen, C.-L.; Mbuyisa, L.; Huang, Y.-B.; Lin, M.-W. Luteolin Inhibits Breast Cancer Stemness and Enhances Chemosensitivity through the Nrf2-Mediated Pathway. Molecules 2021, 26, 6452. [Google Scholar] [CrossRef] [PubMed]
- Markaverich, B.M.; Shoulars, K.; Rodriguez, M.A. Luteolin Regulation of Estrogen Signaling and Cell Cycle Pathway Genes in MCF-7 Human Breast Cancer Cells. Int. J. Biomed. Sci. 2011, 7, 101–111. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Zhang, L.; Dai, Q.; Si, H.; Zhang, L.; Eltom, S.E.; Si, H. Combined Luteolin and Indole-3-Carbinol Synergistically Constrains ERα-Positive Breast Cancer by Dual Inhibiting Estrogen Receptor Alpha and Cyclin-Dependent Kinase 4/6 Pathway in Cultured Cells and Xenograft Mice. Cancers 2021, 13, 2116. [Google Scholar] [CrossRef] [PubMed]
- Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef]
- Imran, M.; Aslam Gondal, T.; Atif, M.; Shahbaz, M.; Batool Qaisarani, T.; Hanif Mughal, M.; Salehi, B.; Martorell, M.; Sharifi-Rad, J. Apigenin as an anticancer agent. Phytother. Res. 2020, 34, 1812–1828. [Google Scholar] [CrossRef]
- Singh, D.; Gupta, M.; Sarwat, M.; Siddique, H.R. Apigenin in cancer prevention and therapy: A systematic review and meta-analysis of animal models. Crit. Rev. Oncol./Hematol. 2022, 176, 103751. [Google Scholar] [CrossRef]
- Yao, L.; Fan, Z.; Han, S.; Sun, N.; Che, H. Apigenin acts as a partial agonist action at estrogen receptors in vivo. Eur. J. Pharmacol. 2021, 906, 174175. [Google Scholar] [CrossRef] [PubMed]
- Adel, M.; Zahmatkeshan, M.; Akbarzadeh, A.; Rabiee, N.; Ahmadi, S.; Keyhanvar, P.; Rezayat, S.M.; Seifalian, A.M. Chemotherapeutic effects of Apigenin in breast cancer: Preclinical evidence and molecular mechanisms; enhanced bioavailability by nanoparticles. Biotechnol. Rep. 2022, 34, e00730. [Google Scholar] [CrossRef] [PubMed]
- Boutas, I.; Kontogeorgi, A.; Dimitrakakis, C.; Kalantaridou, S.N. Soy Isoflavones and Breast Cancer Risk: A Meta-analysis. In Vivo 2022, 36, 556–562. [Google Scholar] [CrossRef] [PubMed]
- Křížová, L.; Dadáková, K.; Kašparovská, J.; Kašparovský, T. Isoflavones. Molecules 2019, 24, 1076. [Google Scholar] [CrossRef]
- Alshehri, M.M.; Sharifi-Rad, J.; Herrera-Bravo, J.; Jara, E.L.; Salazar, L.A.; Kregiel, D.; Uprety, Y.; Akram, M.; Iqbal, M.; Martorell, M.; et al. Therapeutic Potential of Isoflavones with an Emphasis on Daidzein. Oxid. Med. Cell. Longev. 2021, 2021, 6331630. [Google Scholar] [CrossRef]
- Vitale, D.C.; Piazza, C.; Melilli, B.; Drago, F.; Salomone, S. Isoflavones: Estrogenic activity, biological effect and bioavailability. Eur. J. Drug Metab. Pharmacokinet. 2013, 38, 15–25. [Google Scholar] [CrossRef]
- Feng, X.L.; Ho, S.C.; Zhan, X.X.; Zuo, L.S.; Mo, X.F.; Zhang, X.; Abulimiti, A.; Huang, C.Y.; Zhang, C.X. Serum isoflavones and lignans and odds of breast cancer in pre- and postmenopausal Chinese women. Menopause 2021, 28, 413–422. [Google Scholar] [CrossRef]
- Jin, S.; Zhang, Q.Y.; Kang, X.M.; Wang, J.X.; Zhao, W.H. Daidzein induces MCF-7 breast cancer cell apoptosis via the mitochondrial pathway. Ann. Oncol. 2010, 21, 263–268. [Google Scholar] [CrossRef]
- Phase I Clinical Study of Soy Isoflavones in Healthy, Post-Menopausal Women. Identifier NCT00491595. University of North Carolina. 2007. Available online: https://www.clinicaltrials.gov/study/NCT00491595 (accessed on 23 September 2023).
- Yu, L.; Rios, E.; Castro, L.; Liu, J.; Yan, Y.; Dixon, D. Genistein: Dual Role in Women’s Health. Nutrients 2021, 13, 3048. [Google Scholar] [CrossRef]
- Kim, I.S. Current Perspectives on the Beneficial Effects of Soybean Isoflavones and Their Metabolites for Humans. Antioxidants 2021, 10, 1064. [Google Scholar] [CrossRef]
- Marik, R.; Allu, M.; Anchoori, R.; Stearns, V.; Umbricht, C.B.; Khan, S. Potent genistein derivatives as inhibitors of estrogen receptor alpha-positive breast cancer. Cancer Biol. Ther. 2011, 11, 883–892. [Google Scholar] [CrossRef]
- Obiorah, I.E.; Fan, P.; Jordan, V.C. Breast cancer cell apoptosis with phytoestrogens is dependent on an estrogen-deprived state. Cancer Prev. Res. 2014, 7, 939–949. [Google Scholar] [CrossRef]
- de Lemos, M.L. Effects of soy phytoestrogens genistein and daidzein on breast cancer growth. Ann. Pharmacother. 2001, 35, 1118–1121. [Google Scholar] [CrossRef] [PubMed]
- Messina, M.; McCaskill-Stevens, W.; Lampe, J.W. Addressing the soy and breast cancer relationship: Review, commentary, and workshop proceedings. J. Natl. Cancer Inst. 2006, 98, 1275–1284. [Google Scholar] [CrossRef] [PubMed]
- Ramachandran, V.; V, I.K.; Hr, K.K.; Tiwari, R.; Tiwari, G. Biochanin-A: A Bioactive Natural Product with Versatile Therapeutic Perspectives. Curr. Drug Res. Rev. 2022, 14, 225–238. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Xu, J.; Sun, Y.; Fu, R.; Ye, D. An Insight on Synergistic Anti-cancer Efficacy of Biochanin A and Sulforaphane Combination Against Breast Cancer. Appl. Biochem. Biotechnol. 2023. [Google Scholar] [CrossRef]
- An, G.; Morris, M.E. The sulfated conjugate of biochanin A is a substrate of breast cancer resistant protein (ABCG2). Biopharm. Drug Dispos. 2011, 32, 446–457. [Google Scholar] [CrossRef]
- Moon, Y.J.; Shin, B.S.; An, G.; Morris, M.E. Biochanin A inhibits breast cancer tumor growth in a murine xenograft model. Pharm. Res. 2008, 25, 2158–2163. [Google Scholar] [CrossRef]
- Mahmoud, M.; Abdollah, M.R.A.; Elsesy, M.E.; Abou El Ella, D.A.; Zada, S.K.; Tolba, M.F. The natural isoflavone Biochanin-A synergizes 5-fluorouracil anticancer activity in vitro and in vivo in Ehrlich solid-phase carcinoma model. Phytother. Res. 2022, 36, 1310–1325. [Google Scholar] [CrossRef]
- Zhou, Q.; Zhang, W.; Li, T.; Tang, R.; Li, C.; Yuan, S.; Fan, D. Formononetin Enhances the Tumoricidal Effect of Everolimus in Breast Cancer MDA-MB-468 Cells by Suppressing the mTOR Pathway. Evid.-Based Complement. Altern. Med. 2019, 2019, 9610629. [Google Scholar] [CrossRef]
- Li, T.; Zhang, S.; Chen, F.; Hu, J.; Yuan, S.; Li, C.; Wang, X.; Zhang, W.; Tang, R. Formononetin ameliorates the drug resistance of Taxol resistant triple negative breast cancer by inhibiting autophagy. Am. J. Transl. Res. 2021, 13, 497–514. [Google Scholar]
- Chen, J.; Zeng, J.; Xin, M.; Huang, W.; Chen, X. Formononetin induces cell cycle arrest of human breast cancer cells via IGF1/PI3K/Akt pathways in vitro and in vivo. Horm. Metab. Res. 2011, 43, 681–686. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.Y.; Xu, H.; Wu, Z.F.; Chen, C.; Liu, J.Y.; Wu, G.N.; Yao, X.Q.; Liu, F.K.; Li, G.; Shen, L. Formononetin, a novel FGFR2 inhibitor, potently inhibits angiogenesis and tumor growth in preclinical models. Oncotarget 2015, 6, 44563–44578. [Google Scholar] [CrossRef]
- Ong, S.K.L.; Shanmugam, M.K.; Fan, L.; Fraser, S.E.; Arfuso, F.; Ahn, K.S.; Sethi, G.; Bishayee, A. Focus on Formononetin: Anticancer Potential and Molecular Targets. Cancers 2019, 11, 611. [Google Scholar] [CrossRef] [PubMed]
- Ye, X.; Jiang, F.; Li, Y.; Mu, J.; Si, L.; Wang, X.; Ning, S.; Li, Z. Glabridin attenuates the migratory and invasive capacity of breast cancer cells by activating microRNA-200c. Cancer Sci. 2014, 105, 875–882. [Google Scholar] [CrossRef] [PubMed]
- Cui, X.; Cui, M. Glabridin induces paraptosis-like cell death via ER stress in breast cancer cells. Heliyon 2022, 8, e10607. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, A.; Ghosh, D.; Mukerjee, N.; Maitra, S.; Das, P.; Dey, A.; Sharkawi, S.M.Z.; Zouganelis, G.D.; Alexiou, A.; Chaudhari, S.Y.; et al. The Efficient Activity of Glabridin and its Derivatives Against EGFR-mediated Inhibition of Breast Cancer. Curr. Med. Chem. 2023, 31, 573–594. [Google Scholar] [CrossRef]
- Tamir, S.; Eizenberg, M.; Somjen, D.; Izrael, S.; Vaya, J. Estrogen-like activity of glabrene and other constituents isolated from licorice root. J. Steroid Biochem. Mol. Biol. 2001, 78, 291–298. [Google Scholar] [CrossRef]
- Jamwal, A.; Chand, J.; Dash, A.; Bhatt, S.; Dhiman, S.; Wazir, P.; Singh, B.; Goswami, A.; Nandi, U. Glabridin plays dual action to intensify anti-metastatic potential of paclitaxel via impeding CYP2C8 in liver and CYP2J2/EETs in tumor of an orthotopic mouse model of breast cancer. Chem.-Biol. Interact. 2023, 382, 110605. [Google Scholar] [CrossRef]
- Somjen, D.; Katzburg, S.; Vaya, J.; Kaye, A.M.; Hendel, D.; Posner, G.H.; Tamir, S. Estrogenic activity of glabridin and glabrene from licorice roots on human osteoblasts and prepubertal rat skeletal tissues. J. Steroid Biochem. Mol. Biol. 2004, 91, 241–246. [Google Scholar] [CrossRef]
- Li, Z.; Xu, W.; Ren, X.; Xu, J.; Chen, J. Puerarin promotes DUSP1 expression by regulating miR-133a-3p in breast cancer. Mol. Med. Rep. 2019, 19, 205–212. [Google Scholar] [CrossRef]
- Hien, T.T.; Kim, H.G.; Han, E.H.; Kang, K.W.; Jeong, H.G. Molecular mechanism of suppression of MDR1 by puerarin from Pueraria lobata via NF-kappaB pathway and cAMP-responsive element transcriptional activity-dependent up-regulation of AMP-activated protein kinase in breast cancer MCF-7/adr cells. Mol. Nutr. Food Res. 2010, 54, 918–928. [Google Scholar] [CrossRef]
- Hou, N.; Huang, Y.; Cai, S.A.; Yuan, W.C.; Li, L.R.; Liu, X.W.; Zhao, G.J.; Qiu, X.X.; Li, A.Q.; Cheng, C.F.; et al. Puerarin ameliorated pressure overload-induced cardiac hypertrophy in ovariectomized rats through activation of the PPARα/PGC-1 pathway. Acta Pharmacol. Sin. 2021, 42, 55–67. [Google Scholar] [CrossRef]
- Michihara, S.; Tanaka, T.; Uzawa, Y.; Moriyama, T.; Kawamura, Y. Puerarin exerted anti-osteoporotic action independent of estrogen receptor-mediated pathway. J. Nutr. Sci. Vitaminol. 2012, 58, 202–209. [Google Scholar] [CrossRef]
- Zhang, Z.; Lin, M.; Wang, J.; Yang, F.; Yang, P.; Liu, Y.; Chen, Z.; Zheng, Y. Calycosin inhibits breast cancer cell migration and invasion by suppressing EMT via BATF/TGF-β1. Aging 2021, 13, 16009–16023. [Google Scholar] [CrossRef]
- Tian, J.; Wang, Y.; Zhang, X.; Ren, Q.; Li, R.; Huang, Y.; Lu, H.; Chen, J. Calycosin inhibits the in vitro and in vivo growth of breast cancer cells through WDR7-7-GPR30 Signaling. J. Exp. Clin. Cancer Res. 2017, 36, 153. [Google Scholar] [CrossRef]
- Chen, J.; Zhao, X.; Ye, Y.; Wang, Y.; Tian, J. Estrogen receptor beta-mediated proliferative inhibition and apoptosis in human breast cancer by calycosin and formononetin. Cell. Physiol. Biochem. 2013, 32, 1790–1797. [Google Scholar] [CrossRef]
- Minatoya, M.; Kutomi, G.; Asakura, S.; Otokozawa, S.; Sugiyama, Y.; Nagata, Y.; Mori, M.; Hirata, K. Equol, adiponectin, insulin levels and risk of breast cancer. Asian Pac. J. Cancer Prev. 2013, 14, 2191–2199. [Google Scholar] [CrossRef]
- Lampe, J.W. Emerging research on equol and cancer. J. Nutr. 2010, 140, 1369s–1372s. [Google Scholar] [CrossRef]
- Atkinson, C.; Ray, R.M.; Li, W.; Lin, M.G.; Gao, D.L.; Shannon, J.; Stalsberg, H.; Porter, P.L.; Frankenfeld, C.L.; Wähälä, K.; et al. Plasma equol concentration is not associated with breast cancer and fibrocystic breast conditions among women in Shanghai, China. Nutr. Res. 2016, 36, 863–871. [Google Scholar] [CrossRef]
- de la Parra, C.; Otero-Franqui, E.; Martinez-Montemayor, M.; Dharmawardhane, S. The soy isoflavone equol may increase cancer malignancy via up-regulation of eukaryotic protein synthesis initiation factor eIF4G. J. Biol. Chem. 2012, 287, 41640–41650. [Google Scholar] [CrossRef] [PubMed]
- Rampogu, S.; Balasubramaniyam, T.; Lee, J.H. Phytotherapeutic applications of alkaloids in treating breast cancer. Biomed. Pharmacother. 2022, 155, 113760. [Google Scholar] [CrossRef] [PubMed]
- Hu, Z.; Pan, J.; Wang, J.; Pei, Y.; Zhou, R. Current research status of alkaloids against breast cancer. Chin. J. Physiol. 2022, 65, 12–20. [Google Scholar] [CrossRef] [PubMed]
- Rather, R.A.; Bhagat, M. Cancer Chemoprevention and Piperine: Molecular Mechanisms and Therapeutic Opportunities. Front. Cell Dev. Biol. 2018, 6, 10. [Google Scholar] [CrossRef]
- Quijia, C.R.; Chorilli, M. Piperine for treating breast cancer: A review of molecular mechanisms, combination with anticancer drugs, and nanosystems. Phytother. Res. 2022, 36, 147–163. [Google Scholar] [CrossRef]
- Mokbel, K.; Mokbel, K. Chemoprevention of Breast Cancer With Vitamins and Micronutrients: A Concise Review. In Vivo 2019, 33, 983–997. [Google Scholar] [CrossRef]
- Lamarão, R.d.C.; Fialho, E. Aspectos funcionais das catequinas do chá verde no metabolismo celular e sua relação com a redução da gordura corporal. Rev. Nutr. 2009, 22, 257–269. [Google Scholar] [CrossRef]
- Senger, A.E.V.; Schwanke, C.H.A.; Gottlieb, M.G.V. Chá verde (Camellia sinensis) e suas propriedades funcionais nas doenças crônicas não transmissíveis. Sci. Med. 2010, 20, 292–300. [Google Scholar]
- Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial Properties of Green Tea Catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef]
- Gianfredi, V.; Vannini, S.; Moretti, M.; Villarini, M.; Bragazzi, N.L.; Izzotti, A.; Nucci, D. Sulforaphane and Epigallocatechin Gallate Restore Estrogen Receptor Expression by Modulating Epigenetic Events in the Breast Cancer Cell Line MDA-MB-231: A Systematic Review and Meta-Analysis. J. Nutrigenet. Nutrigenom. 2017, 10, 126–135. [Google Scholar] [CrossRef]
- Alam, M.; Ali, S.; Ashraf, G.M.; Bilgrami, A.L.; Yadav, D.K.; Hassan, M.I. Epigallocatechin 3-gallate: From green tea to cancer therapeutics. Food Chem. 2022, 379, 132135. [Google Scholar] [CrossRef] [PubMed]
- Romano, A.; Martel, F. The Role of EGCG in Breast Cancer Prevention and Therapy. Mini Rev. Med. Chem. 2021, 21, 883–898. [Google Scholar] [CrossRef] [PubMed]
- Study of Topically Applied Green Tea Extract for Radio Dermatitis and Radiation Mucositis. Identifier NCT01481818. Shandong Cancer Hospital and Institute. 2020. Available online: https://clinicaltrials.gov/study/NCT01481818 (accessed on 23 September 2023).
- Touillaud, M.S.; Thiébaut, A.C.; Fournier, A.; Niravong, M.; Boutron-Ruault, M.C.; Clavel-Chapelon, F. Dietary lignan intake and postmenopausal breast cancer risk by estrogen and progesterone receptor status. J. Natl. Cancer Inst. 2007, 99, 475–486. [Google Scholar] [CrossRef] [PubMed]
- Jang, W.Y.; Kim, M.Y.; Cho, J.Y. Antioxidant, Anti-Inflammatory, Anti-Menopausal, and Anti-Cancer Effects of Lignans and Their Metabolites. Int. J. Mol. Sci. 2022, 23, 15482. [Google Scholar] [CrossRef] [PubMed]
- Ávila-Gálvez, M.; González-Sarrías, A.; Martínez-Díaz, F.; Abellán, B.; Martínez-Torrano, A.J.; Fernández-López, A.J.; Giménez-Bastida, J.A.; Espín, J.C. Disposition of Dietary Polyphenols in Breast Cancer Patients’ Tumors, and Their Associated Anticancer Activity: The Particular Case of Curcumin. Mol. Nutr. Food Res. 2021, 65, e2100163. [Google Scholar] [CrossRef] [PubMed]
- Moral, R.; Escrich, E. Influence of Olive Oil and Its Components on Breast Cancer: Molecular Mechanisms. Molecules 2022, 27, 477. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Li, M.C.; Yang, J.; Yang, D.; Su, Y.F.; Fan, G.W.; Zhu, Y.; Gao, X.M.; Paoletti, R. Estrogenic properties of six compounds derived from Eucommia ulmoides Oliv. and their differing biological activity through estrogen receptors α and β. Food Chem. 2011, 129, 408–416. [Google Scholar] [CrossRef]
- Shi, H.; Zhao, L.; Guo, X.; Fang, R.; Zhang, H.; Dong, G.; Fu, J.; Yan, F.; Zhang, J.; Ning, Z.; et al. Arctigenin Attenuates Breast Cancer Progression through Decreasing GM-CSF/TSLP/STAT3/β-Catenin Signaling. Int. J. Mol. Sci. 2020, 21, 6357. [Google Scholar] [CrossRef]
- Luo, W.; Wang, F.; Luo, H.; Liu, H. Arctigenin inhibits human breast cancer cell proliferation, migratory and invasive abilities and epithelial to mesenchymal transition by targeting 4EBP1. Exp. Ther. Med. 2021, 21, 547. [Google Scholar] [CrossRef]
- Zhu, L.; Shen, X.B.; Yuan, P.C.; Shao, T.L.; Wang, G.D.; Liu, X.P. Arctigenin inhibits proliferation of ER-positive breast cancer cells through cell cycle arrest mediated by GSK3-dependent cyclin D1 degradation. Life Sci. 2020, 256, 117983. [Google Scholar] [CrossRef]
- Carreau, C.; Flouriot, G.; Bennetau-Pelissero, C.; Potier, M. Enterodiol and enterolactone, two major diet-derived polyphenol metabolites have different impact on ERalpha transcriptional activation in human breast cancer cells. J. Steroid Biochem. Mol. Biol. 2008, 110, 176–185. [Google Scholar] [CrossRef] [PubMed]
- Sonestedt, E.; Wirfält, E. Enterolactone and breast cancer: Methodological issues may contribute to conflicting results in observational studies. Nutr. Res. 2010, 30, 667–677. [Google Scholar] [CrossRef] [PubMed]
- Mali, A.V.; Wagh, U.V.; Hegde, M.V.; Chandorkar, S.S.; Surve, S.V.; Patole, M.V. In vitro anti-metastatic activity of enterolactone, a mammalian lignan derived from flax lignan, and down-regulation of matrix metalloproteinases in MCF-7 and MDA MB 231 cell lines. Indian J. Cancer 2012, 49, 181–187. [Google Scholar] [CrossRef] [PubMed]
- Schröder, L.; Richter, D.U.; Piechulla, B.; Chrobak, M.; Kuhn, C.; Schulze, S.; Abarzua, S.; Jeschke, U.; Weissenbacher, T. Effects of Phytoestrogen Extracts Isolated from Elder Flower on Hormone Production and Receptor Expression of Trophoblast Tumor Cells JEG-3 and BeWo, as well as MCF7 Breast Cancer Cells. Nutrients 2016, 8, 616. [Google Scholar] [CrossRef]
- Bergman Jungeström, M.; Thompson, L.U.; Dabrosin, C. Flaxseed and its lignans inhibit estradiol-induced growth, angiogenesis, and secretion of vascular endothelial growth factor in human breast cancer xenografts in vivo. Clin. Cancer Res. 2007, 13, 1061–1067. [Google Scholar] [CrossRef]
- Mali, A.V.; Padhye, S.B.; Anant, S.; Hegde, M.V.; Kadam, S.S. Anticancer and antimetastatic potential of enterolactone: Clinical, preclinical and mechanistic perspectives. Eur. J. Pharmacol. 2019, 852, 107–124. [Google Scholar] [CrossRef]
- Choi, S.W.; Park, K.I.; Yeon, J.T.; Ryu, B.J.; Kim, K.J.; Kim, S.H. Anti-osteoclastogenic activity of matairesinol via suppression of p38/ERK-NFATc1 signaling axis. BMC Complement. Altern. Med. 2014, 14, 35. [Google Scholar] [CrossRef]
- Yokota, T.; Matsuzaki, Y.; Koyama, M.; Hitomi, T.; Kawanaka, M.; Enoki-Konishi, M.; Okuyama, Y.; Takayasu, J.; Nishino, H.; Nishikawa, A.; et al. Sesamin, a lignan of sesame, down-regulates cyclin D1 protein expression in human tumor cells. Cancer Sci. 2007, 98, 1447–1453. [Google Scholar] [CrossRef]
- Sohel, M.; Islam, M.N.; Hossain, M.A.; Sultana, T.; Dutta, A.; Rahman, S.; Aktar, S.; Islam, K.; Al Mamun, A. Pharmacological Properties to Pharmacological Insight of Sesamin in Breast Cancer Treatment: A Literature-Based Review Study. Int. J. Breast Cancer 2022, 2022, 2599689. [Google Scholar] [CrossRef]
- Kongtawelert, P.; Wudtiwai, B.; Shwe, T.H.; Pothacharoen, P.; Phitak, T. Inhibition of programmed death ligand 1 (PD-L1) expression in breast cancer cells by sesamin. Int. Immunopharmacol. 2020, 86, 106759. [Google Scholar] [CrossRef]
- Fabian, C.J.; Khan, S.A.; Garber, J.E.; Dooley, W.C.; Yee, L.D.; Klemp, J.R.; Nydegger, J.L.; Powers, K.R.; Kreutzjans, A.L.; Zalles, C.M.; et al. Randomized Phase IIB Trial of the Lignan Secoisolariciresinol Diglucoside in Premenopausal Women at Increased Risk for Development of Breast Cancer. Cancer Prev. Res. 2020, 13, 623–634. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.M.; Huang, Y.; Wang, J. Developing phytoestrogens for breast cancer prevention. Anti-Cancer Agents Med. Chem. 2012, 12, 1306–1313. [Google Scholar] [CrossRef] [PubMed]
- Peeters, P.H.; Keinan-Boker, L.; van der Schouw, Y.T.; Grobbee, D.E. Phytoestrogens and breast cancer risk. Review of the epidemiological evidence. Breast Cancer Res. Treat. 2003, 77, 171–183. [Google Scholar] [CrossRef] [PubMed]
- Duncan, A.M.; Phipps, W.R.; Kurzer, M.S. Phyto-oestrogens. Best Pract. Res. Clin. Endocrinol. Metab. 2003, 17, 253–271. [Google Scholar] [CrossRef]
- Nehybová, T.; Šmarda, J.; Beneš, P. Plant coumestans: Recent advances and future perspectives in cancer therapy. Anti-Cancer Agents Med. Chem. 2014, 14, 1351–1362. [Google Scholar] [CrossRef]
- Esteves, D.M.B. Fitoestrogénios Alimentares: Saúde e Prevenção da Doença; Universidade do Porto: Porto, Portugal, 2004. [Google Scholar]
- Barakat, R.; Park, C.J.; Pérez, P.A.; Chiu, K.; Ko, C. Female antiestrogens. In Encyclopedia of Reproduction; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar]
- Al-Thamiree Mezban, S.; Fox, S.W. Genistein and coumestrol reduce MCF-7 breast cancer cell viability and inhibit markers of preferential metastasis, bone matrix attachment and tumor-induced osteoclastogenesis. Arch. Biochem. Biophys. 2023, 740, 109583. [Google Scholar] [CrossRef]
- Mazzuco, C.C. A Romã (Punica granatum L.) como Perspectiva Terapêutica para o Câncer de Mama; Universidade de São Paulo: São Paulo, Brazil, 2018. [Google Scholar]
- Diel, P.; Olff, S.; Schmidt, S.; Michna, H. Effects of the environmental estrogens bisphenol A, o,p′-DDT, p-tert-octylphenol and coumestrol on apoptosis induction, cell proliferation and the expression of estrogen sensitive molecular parameters in the human breast cancer cell line MCF-7. J. Steroid Biochem. Mol. Biol. 2002, 80, 61–70. [Google Scholar] [CrossRef]
- Lu, R.; Serrero, G. Resveratrol, a natural product derived from grape, exhibits antiestrogenic activity and inhibits the growth of human breast cancer cells. J. Cell. Physiol. 1999, 179, 297–304. [Google Scholar] [CrossRef]
- Lephart, E.D. Phytoestrogens (Resveratrol and Equol) for Estrogen-Deficient Skin-Controversies/Misinformation versus Anti-Aging In Vitro and Clinical Evidence via Nutraceutical-Cosmetics. Int. J. Mol. Sci. 2021, 22, 11218. [Google Scholar] [CrossRef]
- Behroozaghdam, M.; Dehghani, M.; Zabolian, A.; Kamali, D.; Javanshir, S.; Hasani Sadi, F.; Hashemi, M.; Tabari, T.; Rashidi, M.; Mirzaei, S.; et al. Resveratrol in breast cancer treatment: From cellular effects to molecular mechanisms of action. Cell. Mol. Life Sci. 2022, 79, 539. [Google Scholar] [CrossRef]
- Amini, P.; Nodooshan, S.J.; Ashrafizadeh, M.; Eftekhari, S.M.; Aryafar, T.; Khalafi, L.; Musa, A.E.; Mahdavi, S.R.; Najafi, M.; Farhood, B. Resveratrol Induces Apoptosis and Attenuates Proliferation of MCF-7 Cells in Combination with Radiation and Hyperthermia. Curr. Mol. Med. 2021, 21, 142–150. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Chen, L.; Zhu, F.; Han, X.; Sun, L.; Chen, K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins 2019, 11, 731. [Google Scholar] [CrossRef] [PubMed]
- Leon-Galicia, I.; Diaz-Chavez, J.; Albino-Sanchez, M.E.; Garcia-Villa, E.; Bermudez-Cruz, R.; Garcia-Mena, J.; Herrera, L.A.; García-Carrancá, A.; Gariglio, P. Resveratrol decreases Rad51 expression and sensitizes cisplatin-resistant MCF-7 breast cancer cells. Oncol. Rep. 2018, 39, 3025–3033. [Google Scholar] [CrossRef] [PubMed]
- Ganguly, S.; Arora, I.; Tollefsbol, T.O. Impact of Stilbenes as Epigenetic Modulators of Breast Cancer Risk and Associated Biomarkers. Int. J. Mol. Sci. 2021, 22, 10033. [Google Scholar] [CrossRef] [PubMed]
- Poschner, S.; Maier-Salamon, A.; Thalhammer, T.; Jäger, W. Resveratrol and other dietary polyphenols are inhibitors of estrogen metabolism in human breast cancer cells. J. Steroid Biochem. Mol. Biol. 2019, 190, 11–18. [Google Scholar] [CrossRef] [PubMed]
- Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef]
- Kala, R.; Tollefsbol, T.O. A Novel Combinatorial Epigenetic Therapy Using Resveratrol and Pterostilbene for Restoring Estrogen Receptor-α (ERα) Expression in ERα-Negative Breast Cancer Cells. PLoS ONE 2016, 11, e0155057. [Google Scholar] [CrossRef]
- Pan, C.; Hu, Y.; Li, J.; Wang, Z.; Huang, J.; Zhang, S.; Ding, L. Estrogen receptor-α36 is involved in pterostilbene-induced apoptosis and anti-proliferation in in vitro and in vivo breast cancer. PLoS ONE 2014, 9, e104459. [Google Scholar] [CrossRef]
- Chakraborty, A.; Bodipati, N.; Demonacos, M.K.; Peddinti, R.; Ghosh, K.; Roy, P. Long term induction by pterostilbene results in autophagy and cellular differentiation in MCF-7 cells via ROS dependent pathway. Mol. Cell. Endocrinol. 2012, 355, 25–40. [Google Scholar] [CrossRef]
- Nikhil, K.; Sharan, S.; Chakraborty, A.; Bodipati, N.; Krishna Peddinti, R.; Roy, P. Role of isothiocyanate conjugate of pterostilbene on the inhibition of MCF-7 cell proliferation and tumor growth in Ehrlich ascitic cell induced tumor bearing mice. Exp. Cell Res. 2014, 320, 311–328. [Google Scholar] [CrossRef]
- Moon, D.; McCormack, D.; McDonald, D.; McFadden, D. Pterostilbene induces mitochondrially derived apoptosis in breast cancer cells in vitro. J. Surg. Res. 2013, 180, 208–215. [Google Scholar] [CrossRef] [PubMed]
- Motaghed, M.; Al-Hassan, F.M.; Hamid, S.S. Cellular responses with thymoquinone treatment in human breast cancer cell line MCF-7. Pharmacogn. Res. 2013, 5, 200–206. [Google Scholar] [CrossRef]
- Motaghed, M.; Al-Hassan, F.M.; Hamid, S.S. Thymoquinone regulates gene expression levels in the estrogen metabolic and interferon pathways in MCF7 breast cancer cells. Int. J. Mol. Med. 2014, 33, 8–16. [Google Scholar] [CrossRef]
- Vanduchova, A.; Anzenbacher, P.; Anzenbacherova, E. Isothiocyanate from Broccoli, Sulforaphane, and Its Properties. J. Med. Food 2019, 22, 121–126. [Google Scholar] [CrossRef] [PubMed]
- Ramirez, M.C.; Singletary, K. Regulation of estrogen receptor alpha expression in human breast cancer cells by sulforaphane. J. Nutr. Biochem. 2009, 20, 195–201. [Google Scholar] [CrossRef] [PubMed]
- Su, X.; Jiang, X.; Meng, L.; Dong, X.; Shen, Y.; Xin, Y. Anticancer Activity of Sulforaphane: The Epigenetic Mechanisms and the Nrf2 Signaling Pathway. Oxid. Med. Cell. Longev. 2018, 2018, 5438179. [Google Scholar] [CrossRef]
- Cao, W.; Lu, X.; Zhong, C.; Wu, J. Sulforaphane Suppresses MCF-7 Breast Cancer Cells Growth via miR-19/PTEN Axis to Antagonize the Effect of Butyl Benzyl Phthalate. Nutr. Cancer 2023, 75, 980–991. [Google Scholar] [CrossRef]
- Study to Evaluate the Effect of Sulforaphane in Broccoli Sprout Extract on Breast Tissue. Identifier NCT00982319. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 2018. Available online: https://clinicaltrials.gov/study/NCT00982319 (accessed on 23 September 2023).
- Ha, Y.W.; Ahn, K.S.; Lee, J.C.; Kim, S.H.; Chung, B.C.; Choi, M.H. Validated quantification for selective cellular uptake of ginsenosides on MCF-7 human breast cancer cells by liquid chromatography-mass spectrometry. Anal. Bioanal. Chem. 2010, 396, 3017–3025. [Google Scholar] [CrossRef]
- Jin, J.; Shahi, S.; Kang, H.K.; van Veen, H.W.; Fan, T.P. Metabolites of ginsenosides as novel BCRP inhibitors. Biochem. Biophys. Res. Commun. 2006, 345, 1308–1314. [Google Scholar] [CrossRef]
- Yao, W.; Guan, Y. Ginsenosides in cancer: A focus on the regulation of cell metabolism. Biomed. Pharmacother. 2022, 156, 113756. [Google Scholar] [CrossRef]
- Li, Y.; Li, S.; Meng, X.; Gan, R.Y.; Zhang, J.J.; Li, H.B. Dietary Natural Products for Prevention and Treatment of Breast Cancer. Nutrients 2017, 9, 728. [Google Scholar] [CrossRef] [PubMed]
- Caesar, L.K.; Cech, N.B. Synergy and antagonism in natural product extracts: When 1 + 1 does not equal 2. Nat. Prod. Rep. 2019, 36, 869–888. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Li, H.; Zhang, J.; Zhao, C.; Lu, S.; Qiao, J.; Han, M. The combinatory effects of natural products and chemotherapy drugs and their mechanisms in breast cancer treatment. Phytochem. Rev. 2020, 19, 1179–1197. [Google Scholar] [CrossRef]
- Srinivas, N.R. Recent trends in preclinical drug–drug interaction studies of flavonoids—Review of case studies, issues and perspectives. Phytother. Res. 2015, 29, 1679–1691. [Google Scholar] [CrossRef] [PubMed]
- Pan, M.H.; Lin, C.L.; Tsai, J.H.; Ho, C.T.; Chen, W.J. 3,5,3′,4′,5′-pentamethoxystilbene (MR-5), a synthetically methoxylated analogue of resveratrol, inhibits growth and induces G1 cell cycle arrest of human breast carcinoma MCF-7 cells. J. Agric. Food Chem. 2010, 58, 226–234. [Google Scholar] [CrossRef]
- Gadag, S.; Narayan, R.; Nayak, A.S.; Catalina Ardila, D.; Sant, S.; Nayak, Y.; Garg, S.; Nayak, U.Y. Development and preclinical evaluation of microneedle-assisted resveratrol loaded nanostructured lipid carriers for localized delivery to breast cancer therapy. Int. J. Pharm. 2021, 606, 120877. [Google Scholar] [CrossRef]
- Ubaid, M.; Shadani, M.A.; Kawish, S.M.; Albratty, M.; Makeen, H.A.; Alhazmi, H.A.; Najmi, A.; Zoghebi, K.; Halawi, M.A.; Ali, A.; et al. Daidzein from Dietary Supplement to a Drug Candidate: An Evaluation of Potential. ACS Omega 2023, 8, 32271–32293. [Google Scholar] [CrossRef]
- Tay, K.C.; Tan, L.T.; Chan, C.K.; Hong, S.L.; Chan, K.G.; Yap, W.H.; Pusparajah, P.; Lee, L.H.; Goh, B.H. Formononetin: A Review of Its Anticancer Potentials and Mechanisms. Front. Pharmacol. 2019, 10, 820. [Google Scholar] [CrossRef]
- Chen, J.; Hou, R.; Zhang, X.; Ye, Y.; Wang, Y.; Tian, J. Calycosin suppresses breast cancer cell growth via ERβ-dependent regulation of IGF-1R, p38 MAPK and PI3K/Akt pathways. PLoS ONE 2014, 9, e91245. [Google Scholar] [CrossRef]
- Liu, Z.; Fei, Y.J.; Cao, X.H.; Xu, D.; Tang, W.J.; Yang, K.; Xu, W.X.; Tang, J.H. Lignans intake and enterolactone concentration and prognosis of breast cancer: A systematic review and meta-analysis. J. Cancer 2021, 12, 2787–2796. [Google Scholar] [CrossRef]
- Pietinen, P.; Stumpf, K.; Männistö, S.; Kataja, V.; Uusitupa, M.; Adlercreutz, H. Serum enterolactone and risk of breast cancer: A case-control study in eastern Finland. Cancer Epidemiol. Biomark. Prev. 2001, 10, 339–344. [Google Scholar]
- Bishayee, A. Cancer prevention and treatment with resveratrol: From rodent studies to clinical trials. Cancer Prev. Res. 2009, 2, 409–418. [Google Scholar] [CrossRef] [PubMed]
- Berman, A.Y.; Motechin, R.A.; Wiesenfeld, M.Y.; Holz, M.K. The therapeutic potential of resveratrol: A review of clinical trials. npj Precis. Oncol. 2017, 1, 35. [Google Scholar] [CrossRef] [PubMed]
- Kuran, D.; Pogorzelska, A.; Wiktorska, K. Breast Cancer Prevention-Is there a Future for Sulforaphane and Its Analogs? Nutrients 2020, 12, 1559. [Google Scholar] [CrossRef] [PubMed]
Class | Compound | Main Sources | Cell Lineage | Mechanism of Action | References |
---|---|---|---|---|---|
Flavonoid | Hesperidin | Orange (Citrus × sinensis) | MCF 7 | Induction of cell-cycle arrest in the G1 phase Inhibition of cell proliferation Induction of apoptosis | [213] |
Hesperetin | Orange (Citrus × sinensis) | MCF 7 | Induction of cell-cycle arrest in the G1 phase Inhibition of cell proliferation Induction of apoptosis | [214] | |
Luteolin | Algaroba (Prosopis juliflora) | MCF 7 | Inhibition of IGF-1 stimulation by the PI3K-Akt signal transduction pathway | [215,216] | |
Apigenin | Chamomile (Matricaria recutita L.) | MCF 7 | Phosphotransferase inhibition | [217,218] | |
Isoflavonoid | Daidzein | Soy (Glycine max (L.) Merril) | MCF 7 | Inhibition of CYP1 Induction of apoptosis Inhibition of topoisomerase Inhibition of cell-cycle arrest in G1 and G2 Phosphotransferase inhibition Activation via PI3K/Akt Inhibition of hTERT expression Increased CDKI protein expression Decreased protein expression in cyclins A, B, E, CDK1, CDK2, CDK4, CDK6, p21, p57, and p27 | [219,220,221] |
Genistein | Soy (Glycine max (L.) Merril) | MCF 7 | Inhibition of CYP1 Inhibition of DNMT1 Induction of apoptosis Inhibition of NF-κB activation Inhibition of telomerase and topoisomerase Phosphotransferase inhibition Activation via PI3K/Akt Inhibition of hTERT expression Increased p53 protein expression | [222,223,224] | |
Glycitein | Soy (Glycine max (L.) Merril) | MCF 7 | Inhibition of cell-cycle arrest in G1 and G2, and decrease in glucose uptake | [221,225] | |
Biochanin A | Soy (Glycine max) | MCF 7 | Cell-cycle arrest induced by upregulation of Bcl-2 expression Inhibition of cell-cycle arrest in G1 and G2 | [221,226] | |
Formononetin | Red propolis | MCF 7 | Induction of cell-cycle arrest by the IGF-1/IGF-1R, MAPK, and PI3K/Akt signaling pathways Inhibition of hTERT expression Decreased mRNA and protein expression of D1 cyclins | [227] | |
Glabridin | Licorice (Glycyrrhiza inflata) | MCF 7 | CK activation in estrogen-responsive tissues | [221] | |
Glabrene | Licorice (Glycyrrhiza glabra) | MCF 7 | CK activation in estrogen-responsive tissues | [221] | |
Puerarin | Kudzu (Pueraria montana) | MCF 7 | Activation via PI3K/Akt | [228] | |
Calycosin | Red propolis | MCF 7 | Induction of apoptosis | [229] | |
Equol | Soy (Glycine max) | MCF 7 | Induction of apoptosis | ||
Alkaloid | Piperine | Black pepper (Piper nigrum L.) | MCF 7 | Antiproliferative effect Induction of apoptosis Activation of caspase-3 and PARP cleavage Inhibited expression of the HER2 gene at the transcriptional level Blocked ERK1/2 signaling to reduce SREBP-1 expression Inhibition of AP-1 activation | [230] |
Catechin | Epigallocatechin | Green tea (Camellia sinensis) | MCF 7 | Induction of apoptosis | [231] |
Lignan | Pinoresinol | Indian tea (Camellia sinensis) | MCF 7 | HER2 protein proteasomal degradation Induction of apoptosis | [232] |
Arctigenin | Burdock (Arctium lappa L.) | MCF 7 | Downregulation of cyclin D1 protein expression | [233,234] | |
Enterolactone | Linseed (Linum usitatissimum L.) | MCF 7 | Downregulation of FAK/paxillin pathway phosphorylation | [235] | |
Matairesinol | Linum (Linum sp) | MCF 7 | Downregulation of the ER-β receptor, cutting off the G0 and G1 mitotic phase | [236] | |
Enterodiol | Linseed (Linum usitatissimum L.) | MCF 7 | Inhibition of VEGF secretion | [237] | |
Sesamin | Sesame (Sesamum indicum L.) | MCF 7 | Negative activation of EGFR and MAPK expression | [238] | |
Secoisolariciresinol | Burdock (Arctium lappa L.) | MCF 7 | Inhibition of NF-kB Phosphotransferase inhibition Inhibition of CYP1 Activation via PI3K/Akt Decreased expression of CDK6 | [239,240] | |
Coumestan | Coumestrol | Alfalfa (Medicago sativa L.) | MCF 7 | Inhibition of CK-2 phosphotransferase activity Inhibition of hTERT expression Decreased protein expression in cyclin E and CK-2 | [220,241] |
4-Methoxycoumestrol | Soy (Glycine max) | MCF 7 | Downregulation of CK-2-specific Akt phosphorylation | [242] | |
Stilbenoid | Resveratrol | Blueberry (Vaccinium spp.) and Blackberry (Morus spp.) | MCF 7 | Inhibition of CYP-1A1/1A2/1B1 and 2E1 Decreased protein expression of cyclin D1 | [243,244] |
Pterostilbene | Blueberry (Vaccinium spp.) | MCF 7 | Induction of apoptosis Decreased mRNA and protein expression of D1 cyclins | [243,245] | |
Monoterpene | Thymoquinone | Black cumin (Nigella sativa L.) | MCF 7 e T-47D | Activation of caspases 8, 9, and 7 Increased PPAR-γ activity and Bcl-2/Bcl-xL expression Inhibition of the PI3K/Akt pathway and induction of p53 and p21 protein expression | [246,247] |
Isotiocianate | Sulforaphane | Broccoli (Brassica oleracea) | MCF 7 e T-47D | Interruption of proliferation and mitosis Inhibition of ER-α protein expression B1 cyclin elevation Decreased EGFR, HER2, and hTERT mRNA expression | [248] |
Saponin | Ginsenoside Rh1 | Ginseng (Panax ginseng) | MCF 7 | Induction of apoptosis | [249] |
Tab | Ginsenoside Rh2 | Ginseng (Panax ginseng) | MCF 7 e ADM | Reverses P-gp-mediated drug resistance of MCF 7/ADM cells | [250] |
Natural Compound | Potential Targets and Mechanisms of Action in the Context of Estrogen Signaling | Preclinical and Clinical Evaluation in the Context of BC | Challenges for Its Use in Clinical Practice | Reference |
---|---|---|---|---|
Hesperetin (2) | - Regulates estrogen metabolism, and induces both extrinsic and intrinsic apoptotic pathways - Suppresses aromatase enzyme activity and cyclin D1, CDK4, Bcl-xL, and pS2 expression | - Reduced the tumor growth in female athymic mice with BC | - Possibly toxic to the liver - Recommended long-term animal and clinical studies to understand its therapeutic advantages in cancer | [257] |
Daidzein (5) | - Regulates estrogen and estrogen receptor complex-binding affinity - At high concentrations, exhibits anticancer capacity | - Phase I multiple-dose clinical investigation to test the safety and effects in healthy postmenopausal women (ClinicalTrials.gov Identifier: NCT00491595) | - The mechanisms of action are still not completely known, and its poor bioavailability restricts its clinical application - Possibly causes unwanted side effects | [273,377] |
Formononetin (9) | - Induces cell-cycle arrest in BC cells via IGF1/PI3K/Akt pathways | - Showed growth-inhibitory activity associated with inhibition of tumor angiogenesis in xenograft models of BC | - There is still insufficient evidence to delineate the exact anticancer mechanisms | [287,288,378] |
Calycosin (13) | - Inhibits growth and induces apoptosis in ER-positive BC cells via ERβ-dependent regulation of the IGF-1R, p38 MAPK, and PI3K/Akt pathways | - Inhibited tumor growth in mice bearing MCF 7 or SKBR3 xenografts | - Recommended long-term animal and clinical studies to better understand its toxicity and therapeutic advantages in cancer | [301,379] |
Epigallocatechin (16) | - Can exert cytotoxic effects in MCF 7 cells, possible through the EGFR, STAT3, ERK, ERK1/2, NF-κB, and Akt pathways | - Oral treatments in mice resulted in a reduction in tumor growth and antiangiogenic effects in xenograft and allograft models of BC - Evaluated in phase I clinical trials for the prevention and treatment of radiodermatitis in patients with BC (ClinicalTrials.gov Identifier: NCT01481818) | - Low oral bioavailability is a problem for its therapeutic application - There is still insufficient evidence about the molecular mechanisms involved in its protective effects against mammary carcinogenesis - More in vivo studies are necessary to determine its potential toxicity | [316,317,318] |
Enterolactone (19) | - Indicated anti-estrogenic effects and affected VEGF production in ER-positive breast cancer | - Showed some benefit to BC patients’ prognosis when it was found at higher concentrations in the serum - Its lower concentration in the serum was associated with an increased risk of developing BC | - Long-term studies are needed to understand its potential benefits or harms to BC patients | [380,381] |
Secoisolariciresinol (23) | - Alters the expression of ER1, ER2, EGF, BCL2 747, and IGF1R | - Phase II clinical studies were conducted in premenopausal women at risk of developing breast cancer, but they did not indicate significant results in Ki-67 expression compared to the placebo-treated group - The trials demonstrated that its use is tolerable and safe | - More clinical trials are necessary to determine its real potential for treating BC patients | [240,337] |
Resveratrol (26) | - Reduces the expression of certain breast-cancer-related genes (e.g., RASSF-1α) via epigenetic mechanisms | - Inhibited the growth of Erβ-positive tumor explants, increased apoptosis, and decreased angiogenesis in nude mice - Clinical trials showed that resveratrol was safe and well tolerated, in addition to its action as a chemopreventive agent for BC patients | - The major obstacle presented in the clinical trials was its poor bioavailability | [382,383] |
Sulforaphane (29) | - Can inhibit the expression of ERα protein in MCF 7 cells, affecting its mRNA levels or mediating the degradation of the receptor by the proteasome complex - Suppresses MCF 7 cell growth via the miR-19/PTEN axis | - A phase II clinical study examined whether this compound in a broccoli sprout preparation could increase the levels of protective enzymes in BC tissues (ClinicalTrials.gov Identifier: NCT00982319) | - Available on the market as a food supplement - More efforts are necessary to determine its therapeutic properties in BC patients | [364,366,367,384] |
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. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
da Silva, F.C.; Brandão, D.C.; Ferreira, E.A.; Siqueira, R.P.; Ferreira, H.S.V.; Da Silva Filho, A.A.; Araújo, T.G. Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer. Pharmaceuticals 2023, 16, 1466. https://doi.org/10.3390/ph16101466
da Silva FC, Brandão DC, Ferreira EA, Siqueira RP, Ferreira HSV, Da Silva Filho AA, Araújo TG. Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer. Pharmaceuticals. 2023; 16(10):1466. https://doi.org/10.3390/ph16101466
Chicago/Turabian Styleda Silva, Fernanda Cardoso, Douglas Cardoso Brandão, Everton Allan Ferreira, Raoni Pais Siqueira, Helen Soares Valença Ferreira, Ademar Alves Da Silva Filho, and Thaise Gonçalves Araújo. 2023. "Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer" Pharmaceuticals 16, no. 10: 1466. https://doi.org/10.3390/ph16101466
APA Styleda Silva, F. C., Brandão, D. C., Ferreira, E. A., Siqueira, R. P., Ferreira, H. S. V., Da Silva Filho, A. A., & Araújo, T. G. (2023). Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer. Pharmaceuticals, 16(10), 1466. https://doi.org/10.3390/ph16101466