Skip to main content
Erschienen in: Cancer and Metastasis Reviews 3/2023

08.12.2022

Natural compounds targeting nuclear receptors for effective cancer therapy

verfasst von: Mangala Hegde, Sosmitha Girisa, Nikunj Naliyadhara, Aviral Kumar, Mohammed S. Alqahtani, Mohamed Abbas, Chakrabhavi Dhananjaya Mohan, Sudha Warrier, Kam Man Hui, Kanchugarakoppal S. Rangappa, Gautam Sethi, Ajaikumar B. Kunnumakkara

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 3/2023

Einloggen, um Zugang zu erhalten

Abstract

Human nuclear receptors (NRs) are a family of forty-eight transcription factors that modulate gene expression both spatially and temporally. Numerous biochemical, physiological, and pathological processes including cell survival, proliferation, differentiation, metabolism, immune modulation, development, reproduction, and aging are extensively orchestrated by different NRs. The involvement of dysregulated NRs and NR-mediated signaling pathways in driving cancer cell hallmarks has been thoroughly investigated. Targeting NRs has been one of the major focuses of drug development strategies for cancer interventions. Interestingly, rapid progress in molecular biology and drug screening reveals that the naturally occurring compounds are promising modern oncology drugs which are free of potentially inevitable repercussions that are associated with synthetic compounds. Therefore, the purpose of this review is to draw our attention to the potential therapeutic effects of various classes of natural compounds that target NRs such as phytochemicals, dietary components, venom constituents, royal jelly–derived compounds, and microbial derivatives in the establishment of novel and safe medications for cancer treatment. This review also emphasizes molecular mechanisms and signaling pathways that are leveraged to promote the anti-cancer effects of these natural compounds. We have also critically reviewed and assessed the advantages and limitations of current preclinical and clinical studies on this subject for cancer prophylaxis. This might subsequently pave the way for new paradigms in the discovery of drugs that target specific cancer types.
Literatur
1.
Zurück zum Zitat Sung, H., Ferlay, J., Siegel, R. L., Laversanne, M., Soerjomataram, I., Jemal, A., et al. (2021). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians, 71(3), 209–249, https://doi.org/10.3322/caac.21660. Sung, H., Ferlay, J., Siegel, R. L., Laversanne, M., Soerjomataram, I., Jemal, A., et al. (2021). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians, 71(3), 209–249, https://​doi.​org/​10.​3322/​caac.​21660.
6.
Zurück zum Zitat Garg, M., Shanmugam, M. K., Bhardwaj, V., Goel, A., Gupta, R., Sharma, A., et al. (2021). The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Medicinal Research Reviews, 41(3), 1291–1336. https://doi.org/10.1002/med.21761CrossRef Garg, M., Shanmugam, M. K., Bhardwaj, V., Goel, A., Gupta, R., Sharma, A., et al. (2021). The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Medicinal Research Reviews, 41(3), 1291–1336. https://​doi.​org/​10.​1002/​med.​21761CrossRef
9.
Zurück zum Zitat Jayaprakash, S., Hegde, M., Girisa, S., Alqahtani, M. S., Abbas, M., Lee, E. H. C., et al. (2022). Demystifying the functional role of nuclear receptors in esophageal cancer. International Journal of Molecular Sciences, 23(18), 10952. https://doi.org/10.3390/ijms231810952. Jayaprakash, S., Hegde, M., Girisa, S., Alqahtani, M. S., Abbas, M., Lee, E. H. C., et al. (2022). Demystifying the functional role of nuclear receptors in esophageal cancer. International Journal of Molecular Sciences, 23(18), 10952. https://​doi.​org/​10.​3390/​ijms231810952.
11.
17.
Zurück zum Zitat Zhao, L., Zhou, S., & Gustafsson, J. -Å. (2019). Nuclear receptors: Recent drug discovery for cancer therapies. Endocrine Reviews, 40(5), 1207–1249.PubMed Zhao, L., Zhou, S., & Gustafsson, J. -Å. (2019). Nuclear receptors: Recent drug discovery for cancer therapies. Endocrine Reviews, 40(5), 1207–1249.PubMed
20.
Zurück zum Zitat Knapp, P., Gardner, P. H., Raynor, D. K., Woolf, E., & McMillan, B. (2010). Perceived risk of tamoxifen side effects: A study of the use of absolute frequencies or frequency bands, with or without verbal descriptors. Patient Education and Counseling, 79(2), 267–271.PubMedCrossRef Knapp, P., Gardner, P. H., Raynor, D. K., Woolf, E., & McMillan, B. (2010). Perceived risk of tamoxifen side effects: A study of the use of absolute frequencies or frequency bands, with or without verbal descriptors. Patient Education and Counseling, 79(2), 267–271.PubMedCrossRef
21.
Zurück zum Zitat Pratheeshkumar, P., Sreekala, C., Zhang, Z., Budhraja, A., Ding, S., Son, Y.-O., et al. (2012). Cancer prevention with promising natural products: Mechanisms of action and molecular targets. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents), 12(10), 1159–1184. Pratheeshkumar, P., Sreekala, C., Zhang, Z., Budhraja, A., Ding, S., Son, Y.-O., et al. (2012). Cancer prevention with promising natural products: Mechanisms of action and molecular targets. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents), 12(10), 1159–1184.
22.
Zurück zum Zitat Harsha, C., Banik, K., Bordoloi, D., & Kunnumakkara, A. B. (2017). Antiulcer properties of fruits and vegetables: A mechanism based perspective. Food and Chemical Toxicology, 108, 104–119.PubMedCrossRef Harsha, C., Banik, K., Bordoloi, D., & Kunnumakkara, A. B. (2017). Antiulcer properties of fruits and vegetables: A mechanism based perspective. Food and Chemical Toxicology, 108, 104–119.PubMedCrossRef
23.
Zurück zum Zitat Elkordy, A. A., Haj-Ahmad, R. R., Awaad, A. S., & Zaki, R. M. (2021). An overview on natural product drug formulations from conventional medicines to nanomedicines: Past, present and future. Journal of Drug Delivery Science and Technology, 63, 102459.CrossRef Elkordy, A. A., Haj-Ahmad, R. R., Awaad, A. S., & Zaki, R. M. (2021). An overview on natural product drug formulations from conventional medicines to nanomedicines: Past, present and future. Journal of Drug Delivery Science and Technology, 63, 102459.CrossRef
24.
Zurück zum Zitat Kunnumakkara, A. B., Nair, A. S., Ahn, K. S., Pandey, M. K., Yi, Z., Liu, M., et al. (2007). Gossypin, a pentahydroxy glucosyl flavone, inhibits the transforming growth factor beta-activated kinase-1-mediated NF-kappaB activation pathway, leading to potentiation of apoptosis, suppression of invasion, and abrogation of osteoclastogenesis. Blood, 109(12), 5112–5121. https://doi.org/10.1182/blood-2007-01-067256CrossRefPubMedPubMedCentral Kunnumakkara, A. B., Nair, A. S., Ahn, K. S., Pandey, M. K., Yi, Z., Liu, M., et al. (2007). Gossypin, a pentahydroxy glucosyl flavone, inhibits the transforming growth factor beta-activated kinase-1-mediated NF-kappaB activation pathway, leading to potentiation of apoptosis, suppression of invasion, and abrogation of osteoclastogenesis. Blood, 109(12), 5112–5121. https://​doi.​org/​10.​1182/​blood-2007-01-067256CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kunnumakkara, A. B., Sung, B., Ravindran, J., Diagaradjane, P., Deorukhkar, A., Dey, S., et al. (2012). Zyflamend suppresses growth and sensitizes human pancreatic tumors to gemcitabine in an orthotopic mouse model through modulation of multiple targets. International Journal of Cancer, 131(3), E292-303. https://doi.org/10.1002/ijc.26442CrossRefPubMed Kunnumakkara, A. B., Sung, B., Ravindran, J., Diagaradjane, P., Deorukhkar, A., Dey, S., et al. (2012). Zyflamend suppresses growth and sensitizes human pancreatic tumors to gemcitabine in an orthotopic mouse model through modulation of multiple targets. International Journal of Cancer, 131(3), E292-303. https://​doi.​org/​10.​1002/​ijc.​26442CrossRefPubMed
27.
Zurück zum Zitat Huang, M., Lu, J.-J., & Ding, J. (2021). Natural products in cancer therapy: Past, present and future. Natural Products and Bioprospecting, 11(1), 5–13.PubMedPubMedCentralCrossRef Huang, M., Lu, J.-J., & Ding, J. (2021). Natural products in cancer therapy: Past, present and future. Natural Products and Bioprospecting, 11(1), 5–13.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Huang, M.-Y., Zhang, L.-L., Ding, J., & Lu, J.-J. (2018). Anticancer drug discovery from Chinese medicinal herbs. Chinese Medicine, 13(1), 1–9.CrossRef Huang, M.-Y., Zhang, L.-L., Ding, J., & Lu, J.-J. (2018). Anticancer drug discovery from Chinese medicinal herbs. Chinese Medicine, 13(1), 1–9.CrossRef
29.
Zurück zum Zitat Newman, D. J., & Cragg, G. M. (2020). Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. Journal of Natural Products, 83(3), 770–803.PubMedCrossRef Newman, D. J., & Cragg, G. M. (2020). Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. Journal of Natural Products, 83(3), 770–803.PubMedCrossRef
30.
Zurück zum Zitat Liu, C., Ho, P.C.-L., Wong, F. C., Sethi, G., Wang, L. Z., & Goh, B. C. (2015). Garcinol: Current status of its anti-oxidative, anti-inflammatory and anti-cancer effects. Cancer Letters, 362(1), 8–14.PubMedCrossRef Liu, C., Ho, P.C.-L., Wong, F. C., Sethi, G., Wang, L. Z., & Goh, B. C. (2015). Garcinol: Current status of its anti-oxidative, anti-inflammatory and anti-cancer effects. Cancer Letters, 362(1), 8–14.PubMedCrossRef
31.
Zurück zum Zitat Kirtonia, A., Gala, K., Fernandes, S. G., Pandya, G., Pandey, A. K., Sethi, G., et al. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. In Seminars in Cancer Biology, 2021 (Vol. 68, pp. 258–278): Elsevier Kirtonia, A., Gala, K., Fernandes, S. G., Pandya, G., Pandey, A. K., Sethi, G., et al. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. In Seminars in Cancer Biology, 2021 (Vol. 68, pp. 258–278): Elsevier
39.
Zurück zum Zitat Moore, L. B., Parks, D. J., Jones, S. A., Bledsoe, R. K., Consler, T. G., Stimmel, J. B., et al. (2000). Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands. Journal of Biological Chemistry, 275(20), 15122–15127. https://doi.org/10.1074/jbc.M001215200CrossRefPubMed Moore, L. B., Parks, D. J., Jones, S. A., Bledsoe, R. K., Consler, T. G., Stimmel, J. B., et al. (2000). Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands. Journal of Biological Chemistry, 275(20), 15122–15127. https://​doi.​org/​10.​1074/​jbc.​M001215200CrossRefPubMed
41.
Zurück zum Zitat Orth, D. N., Kovacs, W., & DeBold, C. R. (1998). Williams textbook of endocrinology. In Williams Textbook of Endocrinology. WB Saunders Co. Orth, D. N., Kovacs, W., & DeBold, C. R. (1998). Williams textbook of endocrinology. In Williams Textbook of Endocrinology. WB Saunders Co.
42.
Zurück zum Zitat Sporn, M. B., Roberts, A. B., & Goodman, D. S. (1994). The retinoids: Biology, chemistry, and medicine (pp. 319). Lippincott Williams & Wilkins. Sporn, M. B., Roberts, A. B., & Goodman, D. S. (1994). The retinoids: Biology, chemistry, and medicine (pp. 319). Lippincott Williams & Wilkins.
46.
Zurück zum Zitat Germain, P., Staels, B., Dacquet, C., Spedding, M., & Laudet, V. (2006). Overview of nomenclature of nuclear receptors. Pharmacological Reviews, 58(4), 685–704.PubMedCrossRef Germain, P., Staels, B., Dacquet, C., Spedding, M., & Laudet, V. (2006). Overview of nomenclature of nuclear receptors. Pharmacological Reviews, 58(4), 685–704.PubMedCrossRef
48.
Zurück zum Zitat Weikum, E. R., Liu, X., & Ortlund, E. A. (2018). The nuclear receptor superfamily: A structural perspective. Protein Science, 27(11), 1876–1892.PubMedPubMedCentralCrossRef Weikum, E. R., Liu, X., & Ortlund, E. A. (2018). The nuclear receptor superfamily: A structural perspective. Protein Science, 27(11), 1876–1892.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Zhang, J., Chalmers, M. J., Stayrook, K. R., Burris, L. L., Wang, Y., Busby, S. A., et al. (2011). DNA binding alters coactivator interaction surfaces of the intact VDR-RXR complex. Nature Structural & Molecular Biology, 18(5), 556–563. https://doi.org/10.1038/nsmb.2046CrossRef Zhang, J., Chalmers, M. J., Stayrook, K. R., Burris, L. L., Wang, Y., Busby, S. A., et al. (2011). DNA binding alters coactivator interaction surfaces of the intact VDR-RXR complex. Nature Structural & Molecular Biology, 18(5), 556–563. https://​doi.​org/​10.​1038/​nsmb.​2046CrossRef
51.
Zurück zum Zitat Yu, X., Yi, P., Hamilton, R. A., Shen, H., Chen, M., Foulds, C. E., et al. (2020). Structural insights of transcriptionally active, full-length androgen receptor coactivator complexes. Molecular Cell, 79(5), 812–823. e814. Yu, X., Yi, P., Hamilton, R. A., Shen, H., Chen, M., Foulds, C. E., et al. (2020). Structural insights of transcriptionally active, full-length androgen receptor coactivator complexes. Molecular Cell, 79(5), 812–823. e814.
53.
Zurück zum Zitat Cotnoir-White, D., Laperrière, D., & Mader, S. (2011). Evolution of the repertoire of nuclear receptor binding sites in genomes. Molecular and Cellular Endocrinology, 334(1–2), 76–82.PubMedCrossRef Cotnoir-White, D., Laperrière, D., & Mader, S. (2011). Evolution of the repertoire of nuclear receptor binding sites in genomes. Molecular and Cellular Endocrinology, 334(1–2), 76–82.PubMedCrossRef
54.
Zurück zum Zitat Penvose, A., Keenan, J. L., Bray, D., Ramlall, V., & Siggers, T. (2019). Comprehensive study of nuclear receptor DNA binding provides a revised framework for understanding receptor specificity. Nature Communications, 10(1), 1–15.CrossRef Penvose, A., Keenan, J. L., Bray, D., Ramlall, V., & Siggers, T. (2019). Comprehensive study of nuclear receptor DNA binding provides a revised framework for understanding receptor specificity. Nature Communications, 10(1), 1–15.CrossRef
55.
Zurück zum Zitat Hong, H., Kohli, K., Trivedi, A., Johnson, D. L., & Stallcup, M. R. (1996). GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors. Proceedings of the National Academy of Sciences, 93(10), 4948–4952.CrossRef Hong, H., Kohli, K., Trivedi, A., Johnson, D. L., & Stallcup, M. R. (1996). GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors. Proceedings of the National Academy of Sciences, 93(10), 4948–4952.CrossRef
57.
Zurück zum Zitat Kamei, Y., Xu, L., Heinzel, T., Torchia, J., Kurokawa, R., Gloss, B., et al. (1996). A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors. Cell, 85(3), 403–414.PubMedCrossRef Kamei, Y., Xu, L., Heinzel, T., Torchia, J., Kurokawa, R., Gloss, B., et al. (1996). A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors. Cell, 85(3), 403–414.PubMedCrossRef
58.
Zurück zum Zitat Takeshita, A., Yen, P. M., Misiti, S., Cardona, G. R., Liu, Y., & Chin, W. W. (1996). Molecular cloning and properties of a full-length putative thyroid hormone receptor coactivator. Endocrinology, 137(8), 3594–3597.PubMedCrossRef Takeshita, A., Yen, P. M., Misiti, S., Cardona, G. R., Liu, Y., & Chin, W. W. (1996). Molecular cloning and properties of a full-length putative thyroid hormone receptor coactivator. Endocrinology, 137(8), 3594–3597.PubMedCrossRef
59.
Zurück zum Zitat Voegel, J. J., Heine, M., Zechel, C., Chambon, P., & Gronemeyer, H. (1996). TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors. The EMBO Journal, 15(14), 3667–3675.PubMedPubMedCentralCrossRef Voegel, J. J., Heine, M., Zechel, C., Chambon, P., & Gronemeyer, H. (1996). TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors. The EMBO Journal, 15(14), 3667–3675.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Anzick, S. L., Kononen, J., Walker, R. L., Azorsa, D. O., Tanner, M. M., Guan, X.-Y., et al. (1997). AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science, 277(5328), 965–968.PubMedCrossRef Anzick, S. L., Kononen, J., Walker, R. L., Azorsa, D. O., Tanner, M. M., Guan, X.-Y., et al. (1997). AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science, 277(5328), 965–968.PubMedCrossRef
61.
Zurück zum Zitat Torchia, J., Rose, D. W., Inostroza, J., Kamei, Y., Westin, S., Glass, C. K., et al. (1997). The transcriptional co-activator p/CIP binds CBP and mediates nuclear-receptor function. Nature, 387(6634), 677–684.PubMedCrossRef Torchia, J., Rose, D. W., Inostroza, J., Kamei, Y., Westin, S., Glass, C. K., et al. (1997). The transcriptional co-activator p/CIP binds CBP and mediates nuclear-receptor function. Nature, 387(6634), 677–684.PubMedCrossRef
62.
Zurück zum Zitat McKenna, N. J., & O’Malley, B. W. (2002). Combinatorial control of gene expression by nuclear receptors and coregulators. Cell, 108(4), 465–474.PubMedCrossRef McKenna, N. J., & O’Malley, B. W. (2002). Combinatorial control of gene expression by nuclear receptors and coregulators. Cell, 108(4), 465–474.PubMedCrossRef
63.
Zurück zum Zitat Chen, H., Lin, R. J., Schiltz, R. L., Chakravarti, D., Nash, A., Nagy, L., et al. (1997). Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300. Cell, 90(3), 569–580.PubMedCrossRef Chen, H., Lin, R. J., Schiltz, R. L., Chakravarti, D., Nash, A., Nagy, L., et al. (1997). Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300. Cell, 90(3), 569–580.PubMedCrossRef
64.
Zurück zum Zitat Yoshinaga, S. K., Peterson, C. L., Herskowitz, I., & Yamamoto, K. R. (1992). Roles of SWI1, SWI2, and SWI3 proteins for transcriptional enhancement by steroid receptors. Science, 258(5088), 1598–1604.PubMedCrossRef Yoshinaga, S. K., Peterson, C. L., Herskowitz, I., & Yamamoto, K. R. (1992). Roles of SWI1, SWI2, and SWI3 proteins for transcriptional enhancement by steroid receptors. Science, 258(5088), 1598–1604.PubMedCrossRef
65.
Zurück zum Zitat Chakravarti, D. (1996). LaMorte VJ, Nelson MC, Nakajima T, Schulman IG, Juguilon H, Montminy M, Evans RM. Role of CBP/P300 in nuclear receptor signalling. Nature, 383, 99–103.PubMedCrossRef Chakravarti, D. (1996). LaMorte VJ, Nelson MC, Nakajima T, Schulman IG, Juguilon H, Montminy M, Evans RM. Role of CBP/P300 in nuclear receptor signalling. Nature, 383, 99–103.PubMedCrossRef
67.
Zurück zum Zitat Fryer, C. J., & Archer, T. K. (1998). Chromatin remodelling by the glucocorticoid receptor requires the BRG1 complex. Nature, 393(6680), 88–91.PubMedCrossRef Fryer, C. J., & Archer, T. K. (1998). Chromatin remodelling by the glucocorticoid receptor requires the BRG1 complex. Nature, 393(6680), 88–91.PubMedCrossRef
68.
Zurück zum Zitat Rachez, C., Suldan, Z., Ward, J., Chang, C.-P.B., Burakov, D., Erdjument-Bromage, H., et al. (1998). A novel protein complex that interacts with the vitamin D3 receptor in a ligand-dependent manner and enhances VDR transactivation in a cell-free system. Genes & Development, 12(12), 1787–1800.CrossRef Rachez, C., Suldan, Z., Ward, J., Chang, C.-P.B., Burakov, D., Erdjument-Bromage, H., et al. (1998). A novel protein complex that interacts with the vitamin D3 receptor in a ligand-dependent manner and enhances VDR transactivation in a cell-free system. Genes & Development, 12(12), 1787–1800.CrossRef
72.
Zurück zum Zitat Näär, A. M., Lemon, B. D., & Tjian, R. (2001). Transcriptional coactivator complexes. Annual Review of Biochemistry, 70(1), 475–501.PubMedCrossRef Näär, A. M., Lemon, B. D., & Tjian, R. (2001). Transcriptional coactivator complexes. Annual Review of Biochemistry, 70(1), 475–501.PubMedCrossRef
82.
Zurück zum Zitat Windahl, S. H., Treuter, E., Ford, J., Zilliacus, J., Gustafsson, J. A., & McEwan, I. J. (1999). The nuclear-receptor interacting protein (RIP) 140 binds to the human glucocorticoid receptor and modulates hormone-dependent transactivation. Journal of Steroid Biochemistry and Molecular Biology, 71(3–4), 93–102. https://doi.org/10.1016/S0960-0760(99)00128-4CrossRefPubMed Windahl, S. H., Treuter, E., Ford, J., Zilliacus, J., Gustafsson, J. A., & McEwan, I. J. (1999). The nuclear-receptor interacting protein (RIP) 140 binds to the human glucocorticoid receptor and modulates hormone-dependent transactivation. Journal of Steroid Biochemistry and Molecular Biology, 71(3–4), 93–102. https://​doi.​org/​10.​1016/​S0960-0760(99)00128-4CrossRefPubMed
84.
Zurück zum Zitat Ong, P. S., Wang, L. Z., Dai, X., Tseng, S. H., Loo, S. J., & Sethi, G. (2016). Judicious toggling of mTOR activity to combat insulin resistance and cancer: Current evidence and perspectives. Frontiers in Pharmacology, 7, 395.PubMedPubMedCentralCrossRef Ong, P. S., Wang, L. Z., Dai, X., Tseng, S. H., Loo, S. J., & Sethi, G. (2016). Judicious toggling of mTOR activity to combat insulin resistance and cancer: Current evidence and perspectives. Frontiers in Pharmacology, 7, 395.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Ramachandran, L., Manu, K. A., Shanmugam, M. K., Li, F., Siveen, K. S., Vali, S., et al. (2012). Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor γ activation pathway in gastric cancer. Journal of Biological Chemistry, 287(45), 38028–38040.PubMedPubMedCentralCrossRef Ramachandran, L., Manu, K. A., Shanmugam, M. K., Li, F., Siveen, K. S., Vali, S., et al. (2012). Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor γ activation pathway in gastric cancer. Journal of Biological Chemistry, 287(45), 38028–38040.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Sikka, S., Chen, L., Sethi, G., & Kumar, A. P. (2012). Targeting PPARγ signaling cascade for the prevention and treatment of prostate cancer. PPAR Research, 2012, 968040.PubMedPubMedCentralCrossRef Sikka, S., Chen, L., Sethi, G., & Kumar, A. P. (2012). Targeting PPARγ signaling cascade for the prevention and treatment of prostate cancer. PPAR Research, 2012, 968040.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Zhang, J., Ahn, K. S., Kim, C., Shanmugam, M. K., Siveen, K. S., Arfuso, F., et al. (2016). Nimbolide-induced oxidative stress abrogates STAT3 signaling cascade and inhibits tumor growth in transgenic adenocarcinoma of mouse prostate model. Antioxidants & Redox Signaling, 24(11), 575–589.CrossRef Zhang, J., Ahn, K. S., Kim, C., Shanmugam, M. K., Siveen, K. S., Arfuso, F., et al. (2016). Nimbolide-induced oxidative stress abrogates STAT3 signaling cascade and inhibits tumor growth in transgenic adenocarcinoma of mouse prostate model. Antioxidants & Redox Signaling, 24(11), 575–589.CrossRef
89.
Zurück zum Zitat Raghunath, A., Sundarraj, K., Arfuso, F., Sethi, G., & Perumal, E. (2018). Dysregulation of Nrf2 in hepatocellular carcinoma: Role in cancer progression and chemoresistance. Cancers, 10(12), 481.PubMedPubMedCentralCrossRef Raghunath, A., Sundarraj, K., Arfuso, F., Sethi, G., & Perumal, E. (2018). Dysregulation of Nrf2 in hepatocellular carcinoma: Role in cancer progression and chemoresistance. Cancers, 10(12), 481.PubMedPubMedCentralCrossRef
90.
91.
92.
Zurück zum Zitat Whyte-Allman, S.-K., Hoque, M. T., Jenabian, M.-A., Routy, J.-P., & Bendayan, R. (2017). Xenobiotic nuclear receptors pregnane X receptor and constitutive androstane receptor regulate antiretroviral drug efflux transporters at the blood-testis barrier. Journal of Pharmacology and Experimental Therapeutics, 363(3), 324–335.PubMedCrossRef Whyte-Allman, S.-K., Hoque, M. T., Jenabian, M.-A., Routy, J.-P., & Bendayan, R. (2017). Xenobiotic nuclear receptors pregnane X receptor and constitutive androstane receptor regulate antiretroviral drug efflux transporters at the blood-testis barrier. Journal of Pharmacology and Experimental Therapeutics, 363(3), 324–335.PubMedCrossRef
93.
Zurück zum Zitat Lemmen, J., Tozakidis, I. E., Bele, P., & Galla, H.-J. (2013). Constitutive androstane receptor upregulates Abcb1 and Abcg2 at the blood–brain barrier after CITCO activation. Brain Research, 1501, 68–80.PubMedCrossRef Lemmen, J., Tozakidis, I. E., Bele, P., & Galla, H.-J. (2013). Constitutive androstane receptor upregulates Abcb1 and Abcg2 at the blood–brain barrier after CITCO activation. Brain Research, 1501, 68–80.PubMedCrossRef
94.
Zurück zum Zitat Jigorel, E., Le Vee, M., Boursier-Neyret, C., Parmentier, Y., & Fardel, O. (2006). Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drug-sensing receptors in primary human hepatocytes. Drug Metabolism and Disposition, 34(10), 1756–1763.PubMedCrossRef Jigorel, E., Le Vee, M., Boursier-Neyret, C., Parmentier, Y., & Fardel, O. (2006). Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drug-sensing receptors in primary human hepatocytes. Drug Metabolism and Disposition, 34(10), 1756–1763.PubMedCrossRef
95.
Zurück zum Zitat Chisaki, I., Kobayashi, M., Itagaki, S., Hirano, T., & Iseki, K. (2009). Liver X receptor regulates expression of MRP2 but not that of MDR1 and BCRP in the liver. Biochimica et Biophysica Acta (BBA)-Biomembranes, 1788(11), 2396–2403. Chisaki, I., Kobayashi, M., Itagaki, S., Hirano, T., & Iseki, K. (2009). Liver X receptor regulates expression of MRP2 but not that of MDR1 and BCRP in the liver. Biochimica et Biophysica Acta (BBA)-Biomembranes, 1788(11), 2396–2403.
98.
Zurück zum Zitat Cerveny, L., Svecova, L., Anzenbacherova, E., Vrzal, R., Staud, F., Dvorak, Z., et al. (2007). Valproic acid induces CYP3A4 and MDR1 gene expression by activation of constitutive androstane receptor and pregnane X receptor pathways. Drug Metabolism and Disposition, 35(7), 1032–1041.PubMedCrossRef Cerveny, L., Svecova, L., Anzenbacherova, E., Vrzal, R., Staud, F., Dvorak, Z., et al. (2007). Valproic acid induces CYP3A4 and MDR1 gene expression by activation of constitutive androstane receptor and pregnane X receptor pathways. Drug Metabolism and Disposition, 35(7), 1032–1041.PubMedCrossRef
99.
Zurück zum Zitat Ee, P. L. R., Kamalakaran, S., Tonetti, D., He, X., Ross, D. D., & Beck, W. T. (2004). Identification of a novel estrogen response element in the breast cancer resistance protein (ABCG2) gene. Cancer research, 64(4), 1247–1251.PubMedCrossRef Ee, P. L. R., Kamalakaran, S., Tonetti, D., He, X., Ross, D. D., & Beck, W. T. (2004). Identification of a novel estrogen response element in the breast cancer resistance protein (ABCG2) gene. Cancer research, 64(4), 1247–1251.PubMedCrossRef
100.
Zurück zum Zitat Szatmari, I., Vámosi, G. r., Brazda, P., Balint, B. L., Benko, S., Széles, L., et al. (2006). Peroxisome proliferator-activated receptor γ-regulated ABCG2 expression confers cytoprotection to human dendritic cells. Journal of Biological Chemistry, 281(33), 23812-23823. Szatmari, I., Vámosi, G. r., Brazda, P., Balint, B. L., Benko, S., Széles, L., et al. (2006). Peroxisome proliferator-activated receptor γ-regulated ABCG2 expression confers cytoprotection to human dendritic cells. Journal of Biological Chemistry, 281(33), 23812-23823.
103.
Zurück zum Zitat Hu, L., Sun, Y., Luo, J., He, X., Ye, M., Li, G., et al. (2020). Targeting TR4 nuclear receptor with antagonist bexarotene increases docetaxel sensitivity to better suppress the metastatic castration-resistant prostate cancer progression. Oncogene, 39(9), 1891–1903.PubMedCrossRef Hu, L., Sun, Y., Luo, J., He, X., Ye, M., Li, G., et al. (2020). Targeting TR4 nuclear receptor with antagonist bexarotene increases docetaxel sensitivity to better suppress the metastatic castration-resistant prostate cancer progression. Oncogene, 39(9), 1891–1903.PubMedCrossRef
104.
Zurück zum Zitat Kai, L., & Levenson, A. S. (2011). Combination of resveratrol and antiandrogen flutamide has synergistic effect on androgen receptor inhibition in prostate cancer cells. Anticancer Research, 31(10), 3323–3330.PubMed Kai, L., & Levenson, A. S. (2011). Combination of resveratrol and antiandrogen flutamide has synergistic effect on androgen receptor inhibition in prostate cancer cells. Anticancer Research, 31(10), 3323–3330.PubMed
106.
Zurück zum Zitat de Bono, J. S., Chowdhury, S., Feyerabend, S., Elliott, T., Grande, E., Melhem-Bertrandt, A., et al. (2018). Antitumour activity and safety of enzalutamide in patients with metastatic castration-resistant prostate cancer previously treated with abiraterone acetate plus prednisone for >= 24 weeks in Europe. European Urology, 74(1), 37–45. https://doi.org/10.1016/j.eururo.2017.07.035CrossRefPubMed de Bono, J. S., Chowdhury, S., Feyerabend, S., Elliott, T., Grande, E., Melhem-Bertrandt, A., et al. (2018). Antitumour activity and safety of enzalutamide in patients with metastatic castration-resistant prostate cancer previously treated with abiraterone acetate plus prednisone for >= 24 weeks in Europe. European Urology, 74(1), 37–45. https://​doi.​org/​10.​1016/​j.​eururo.​2017.​07.​035CrossRefPubMed
107.
Zurück zum Zitat Lee, H. Y., Chen, H. L., Teoh, J. Y. C., Chen, T. C., Hao, S. Y., Tsai, H. Y., et al. (2021). Abiraterone and enzalutamide had different adverse effects on the cardiovascular system: A systematic review with pairwise and network meta-analyses. Prostate Cancer and Prostatic Diseases, 24(1), 244–252. https://doi.org/10.1038/s41391-020-00275-3CrossRefPubMed Lee, H. Y., Chen, H. L., Teoh, J. Y. C., Chen, T. C., Hao, S. Y., Tsai, H. Y., et al. (2021). Abiraterone and enzalutamide had different adverse effects on the cardiovascular system: A systematic review with pairwise and network meta-analyses. Prostate Cancer and Prostatic Diseases, 24(1), 244–252. https://​doi.​org/​10.​1038/​s41391-020-00275-3CrossRefPubMed
108.
Zurück zum Zitat Johnson, D. B., & Sonthalia, S. (2022). Flutamide. In StatPearls. StatPearls Publishing. Johnson, D. B., & Sonthalia, S. (2022). Flutamide. In StatPearls. StatPearls Publishing.
111.
Zurück zum Zitat Culig, Z. (2014). Targeting the androgen receptor in prostate cancer. Expert Opinion on Pharmacotherapy, 15(10), 1427–1437.PubMedCrossRef Culig, Z. (2014). Targeting the androgen receptor in prostate cancer. Expert Opinion on Pharmacotherapy, 15(10), 1427–1437.PubMedCrossRef
113.
Zurück zum Zitat Facciola, A., Venanzi Rullo, E., Ceccarelli, M., D'Aleo, F., Di Rosa, M., Pinzone, M. R., et al. (2017). Kaposi’s sarcoma in HIV-infected patients in the era of new antiretrovirals.European Review for Medical Pharmacological Sciences, 21(24), 5868–5869, https://doi.org/10.26355/eurrev_201712_14036. Facciola, A., Venanzi Rullo, E., Ceccarelli, M., D'Aleo, F., Di Rosa, M., Pinzone, M. R., et al. (2017). Kaposi’s sarcoma in HIV-infected patients in the era of new antiretrovirals.European Review for Medical Pharmacological Sciences, 21(24), 5868–5869, https://​doi.​org/​10.​26355/​eurrev_​201712_​14036.
116.
117.
Zurück zum Zitat Nathan, M. R., & Schmid, P. (2017). A review of fulvestrant in breast cancer. Oncology and Therapy, 5(1), 17–29.PubMedCrossRef Nathan, M. R., & Schmid, P. (2017). A review of fulvestrant in breast cancer. Oncology and Therapy, 5(1), 17–29.PubMedCrossRef
120.
Zurück zum Zitat Rathkopf, D. E., Smith, M., Ryan, C., Berry, W., Shore, N., Liu, G., et al. (2017). Androgen receptor mutations in patients with castration-resistant prostate cancer treated with apalutamide. Annals of Oncology, 28(9), 2264–2271.PubMedPubMedCentralCrossRef Rathkopf, D. E., Smith, M., Ryan, C., Berry, W., Shore, N., Liu, G., et al. (2017). Androgen receptor mutations in patients with castration-resistant prostate cancer treated with apalutamide. Annals of Oncology, 28(9), 2264–2271.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Smith, M. R., Antonarakis, E. S., Ryan, C. J., Berry, W. R., Shore, N. D., Liu, G., et al. (2016). Phase 2 study of the safety and antitumor activity of apalutamide (ARN-509), a potent androgen receptor antagonist, in the high-risk nonmetastatic castration-resistant prostate cancer cohort. European Urology, 70(6), 963–970.PubMedPubMedCentralCrossRef Smith, M. R., Antonarakis, E. S., Ryan, C. J., Berry, W. R., Shore, N. D., Liu, G., et al. (2016). Phase 2 study of the safety and antitumor activity of apalutamide (ARN-509), a potent androgen receptor antagonist, in the high-risk nonmetastatic castration-resistant prostate cancer cohort. European Urology, 70(6), 963–970.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Smith, M. R., Saad, F., Chowdhury, S., Oudard, S., Hadaschik, B. A., Graff, J. N., et al. (2018). Apalutamide treatment and metastasis-free survival in prostate cancer. New England Journal of Medicine, 378(15), 1408–1418.PubMedCrossRef Smith, M. R., Saad, F., Chowdhury, S., Oudard, S., Hadaschik, B. A., Graff, J. N., et al. (2018). Apalutamide treatment and metastasis-free survival in prostate cancer. New England Journal of Medicine, 378(15), 1408–1418.PubMedCrossRef
128.
Zurück zum Zitat Wu, P. A., & Stern, R. S. (2012). Topical tretinoin, another failure in the pursuit of practical chemoprevention for non-melanoma skin cancer. Journal of Investigative Dermatology, 132(6), 1532–1535.PubMedCrossRef Wu, P. A., & Stern, R. S. (2012). Topical tretinoin, another failure in the pursuit of practical chemoprevention for non-melanoma skin cancer. Journal of Investigative Dermatology, 132(6), 1532–1535.PubMedCrossRef
129.
132.
Zurück zum Zitat Babu, B. H., Jayram, H. N., Nair, M. G., Ajaikumar, K. B., & Padikkala, J. (2003). Free radical scavenging, antitumor and anticarcinogenic activity of gossypin. Journal of Experimental & Clinical Cancer Research, 22(4), 581–589. Babu, B. H., Jayram, H. N., Nair, M. G., Ajaikumar, K. B., & Padikkala, J. (2003). Free radical scavenging, antitumor and anticarcinogenic activity of gossypin. Journal of Experimental & Clinical Cancer Research, 22(4), 581–589.
135.
137.
139.
Zurück zum Zitat Delfosse, V., Maire, A. I., Balaguer, P., & Bourguet, W. (2015). A structural perspective on nuclear receptors as targets of environmental compounds. Acta Pharmacologica Sinica, 36(1), 88–101.PubMedCrossRef Delfosse, V., Maire, A. I., Balaguer, P., & Bourguet, W. (2015). A structural perspective on nuclear receptors as targets of environmental compounds. Acta Pharmacologica Sinica, 36(1), 88–101.PubMedCrossRef
140.
Zurück zum Zitat Manu, K. A., Shanmugam, M. K., Li, F., Chen, L., Siveen, K. S., Ahn, K. S., et al. (2014). Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. Journal of Molecular Medicine, 92(3), 267–276.PubMedCrossRef Manu, K. A., Shanmugam, M. K., Li, F., Chen, L., Siveen, K. S., Ahn, K. S., et al. (2014). Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. Journal of Molecular Medicine, 92(3), 267–276.PubMedCrossRef
142.
Zurück zum Zitat Patel, S. M., Venkata, K. C. N., Bhattacharyya, P., Sethi, G., & Bishayee, A. Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic diseases. In Seminars in Cancer Biology, 2016 (Vol. 40, pp. 100–115): Elsevier Patel, S. M., Venkata, K. C. N., Bhattacharyya, P., Sethi, G., & Bishayee, A. Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic diseases. In Seminars in Cancer Biology, 2016 (Vol. 40, pp. 100–115): Elsevier
143.
Zurück zum Zitat Kim, C., Lee, S.-G., Yang, W. M., Arfuso, F., Um, J.-Y., Kumar, A. P., et al. (2018). Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Letters, 431, 123–141.PubMedCrossRef Kim, C., Lee, S.-G., Yang, W. M., Arfuso, F., Um, J.-Y., Kumar, A. P., et al. (2018). Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Letters, 431, 123–141.PubMedCrossRef
147.
Zurück zum Zitat Li, W., Shao, Y., Hu, L., Zhang, X., Chen, Y., Tong, L., et al. (2007). BM6, a new semi-synthetic vinca alkaloid, exhibits its potent in vivo anti-tumor activities via its high binding affinity for tubulin and improved pharmacokinetic profiles. Cancer Biology & Therapy, 6(5), 787–794. https://doi.org/10.4161/cbt.6.5.4006CrossRef Li, W., Shao, Y., Hu, L., Zhang, X., Chen, Y., Tong, L., et al. (2007). BM6, a new semi-synthetic vinca alkaloid, exhibits its potent in vivo anti-tumor activities via its high binding affinity for tubulin and improved pharmacokinetic profiles. Cancer Biology & Therapy, 6(5), 787–794. https://​doi.​org/​10.​4161/​cbt.​6.​5.​4006CrossRef
150.
Zurück zum Zitat Cancer, I. A. f. R. o. (1991). Coffee, tea, mate, methylxanthines, and methyglyoxal. IARC Monograph on the Evaluation of Carcinogenic Risk of Chemicals to Humans, 51, 1–513. Cancer, I. A. f. R. o. (1991). Coffee, tea, mate, methylxanthines, and methyglyoxal. IARC Monograph on the Evaluation of Carcinogenic Risk of Chemicals to Humans, 51, 1–513.
151.
Zurück zum Zitat Tolmach, L. J., Jones, R. W., & Busse, P. M. (1977). The action of caffeine on X-irradiated HeLa cells. I. Delayed inhibition of DNA synthesis. Radiation Research, 71(3), 653–665. Tolmach, L. J., Jones, R. W., & Busse, P. M. (1977). The action of caffeine on X-irradiated HeLa cells. I. Delayed inhibition of DNA synthesis. Radiation Research, 71(3), 653–665.
152.
Zurück zum Zitat Busse, P. M., Bose, S. K., Jones, R. W., & Tolmach, L. J. (1978). The action of caffeine on X-irradiated HeLa cells. III. Enhancement of X-ray-induced killing during G2 arrest. Radiation Research, 76(2), 292–307. Busse, P. M., Bose, S. K., Jones, R. W., & Tolmach, L. J. (1978). The action of caffeine on X-irradiated HeLa cells. III. Enhancement of X-ray-induced killing during G2 arrest. Radiation Research, 76(2), 292–307.
155.
157.
Zurück zum Zitat Faudone, G., Kilu, W., Ni, X., Chaikuad, A., Sreeramulu, S., Heitel, P., et al. (2021). The transcriptional repressor orphan nuclear receptor TLX is responsive to xanthines. ACS Pharmacology & Translational Science, 4(6), 1794–1807.CrossRef Faudone, G., Kilu, W., Ni, X., Chaikuad, A., Sreeramulu, S., Heitel, P., et al. (2021). The transcriptional repressor orphan nuclear receptor TLX is responsive to xanthines. ACS Pharmacology & Translational Science, 4(6), 1794–1807.CrossRef
158.
Zurück zum Zitat Clark, R., & Lee, S. H. (2016). Anticancer properties of capsaicin against human cancer. Anticancer Research, 36(3), 837–843.PubMed Clark, R., & Lee, S. H. (2016). Anticancer properties of capsaicin against human cancer. Anticancer Research, 36(3), 837–843.PubMed
159.
Zurück zum Zitat Kim, C. S., Park, W. H., Park, J. Y., Kang, J. H., Kim, M. O., Kawada, T., et al. (2004). Capsaicin, a spicy component of hot pepper, induces apoptosis by activation of the peroxisome proliferator-activated receptor gamma in HT-29 human colon cancer cells. Journal of Medicinal Food, 7(3), 267–273. https://doi.org/10.1089/1096620041938713CrossRefPubMed Kim, C. S., Park, W. H., Park, J. Y., Kang, J. H., Kim, M. O., Kawada, T., et al. (2004). Capsaicin, a spicy component of hot pepper, induces apoptosis by activation of the peroxisome proliferator-activated receptor gamma in HT-29 human colon cancer cells. Journal of Medicinal Food, 7(3), 267–273. https://​doi.​org/​10.​1089/​1096620041938713​CrossRefPubMed
160.
Zurück zum Zitat Bort, A., Sanchez, B. G., Mateos-Gomez, P. A., Diaz-Laviada, I., & Rodriguez-Henche, N. (2019). Capsaicin targets lipogenesis in HepG2 cells through AMPK activation, AKT inhibition and PPARs regulation. International Journal of Molecular Sciences, 20(7), ARTN 1660. https://doi.org/10.3390/ijms20071660. Bort, A., Sanchez, B. G., Mateos-Gomez, P. A., Diaz-Laviada, I., & Rodriguez-Henche, N. (2019). Capsaicin targets lipogenesis in HepG2 cells through AMPK activation, AKT inhibition and PPARs regulation. International Journal of Molecular Sciences, 20(7), ARTN 1660. https://​doi.​org/​10.​3390/​ijms20071660.
161.
Zurück zum Zitat Kumar, S., Kamboj, J., & Sharma, S. (2011). Overview for various aspects of the health benefits of Piper longum linn. fruit. Journal of Acupuncture and Meridian Studies, 4(2), 134–140. Kumar, S., Kamboj, J., & Sharma, S. (2011). Overview for various aspects of the health benefits of Piper longum linn. fruit. Journal of Acupuncture and Meridian Studies, 4(2), 134–140.
163.
164.
Zurück zum Zitat Vershinin, A. (1999). Biological functions of carotenoids - Diversity and evolution. BioFactors, 10(2–3), 99–104.PubMedCrossRef Vershinin, A. (1999). Biological functions of carotenoids - Diversity and evolution. BioFactors, 10(2–3), 99–104.PubMedCrossRef
166.
Zurück zum Zitat Zare, M., Norouzi Roshan, Z., Assadpour, E., & Jafari, S. M. (2021). Improving the cancer prevention/treatment role of carotenoids through various nano-delivery systems. Critical Reviews in Food Science and Nutrition, 61(3), 522–534.PubMedCrossRef Zare, M., Norouzi Roshan, Z., Assadpour, E., & Jafari, S. M. (2021). Improving the cancer prevention/treatment role of carotenoids through various nano-delivery systems. Critical Reviews in Food Science and Nutrition, 61(3), 522–534.PubMedCrossRef
167.
Zurück zum Zitat Zhang, X., Zhao, W.-E., Hu, L., Zhao, L., & Huang, J. (2011). Carotenoids inhibit proliferation and regulate expression of peroxisome proliferators-activated receptor gamma (PPARγ) in K562 cancer cells. Archives of Biochemistry and Biophysics, 512(1), 96–106.PubMedCrossRef Zhang, X., Zhao, W.-E., Hu, L., Zhao, L., & Huang, J. (2011). Carotenoids inhibit proliferation and regulate expression of peroxisome proliferators-activated receptor gamma (PPARγ) in K562 cancer cells. Archives of Biochemistry and Biophysics, 512(1), 96–106.PubMedCrossRef
168.
Zurück zum Zitat Liu, C.-L., Lim, Y.-P., & Hu, M.-L. (2012). Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Marine Drugs, 10(1), 242–257.PubMedPubMedCentralCrossRef Liu, C.-L., Lim, Y.-P., & Hu, M.-L. (2012). Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Marine Drugs, 10(1), 242–257.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Hosokawa, M., Kudo, M., Maeda, H., Kohno, H., Tanaka, T., & Miyashita, K. (2004). Fucoxanthin induces apoptosis and enhances the antiproliferative effect of the PPARγ ligand, troglitazone, on colon cancer cells. Biochimica et Biophysica Acta (BBA)-General Subjects, 1675(1–3), 113–119. Hosokawa, M., Kudo, M., Maeda, H., Kohno, H., Tanaka, T., & Miyashita, K. (2004). Fucoxanthin induces apoptosis and enhances the antiproliferative effect of the PPARγ ligand, troglitazone, on colon cancer cells. Biochimica et Biophysica Acta (BBA)-General Subjects, 1675(1–3), 113–119.
170.
Zurück zum Zitat Lian, F., Hu, K. Q., Russell, R. M., & Wang, X. D. (2006). β-Cryptoxanthin suppresses the growth of immortalized human bronchial epithelial cells and non-small-cell lung cancer cells and up-regulates retinoic acid receptor β expression. International Journal of Cancer, 119(9), 2084–2089.PubMedCrossRef Lian, F., Hu, K. Q., Russell, R. M., & Wang, X. D. (2006). β-Cryptoxanthin suppresses the growth of immortalized human bronchial epithelial cells and non-small-cell lung cancer cells and up-regulates retinoic acid receptor β expression. International Journal of Cancer, 119(9), 2084–2089.PubMedCrossRef
171.
Zurück zum Zitat Iskandar, A. R., Liu, C., Smith, D. E., Hu, K.-Q., Choi, S.-W., Ausman, L. M., et al. (2013). β-Cryptoxanthin restores nicotine-reduced lung SIRT1 to normal levels and inhibits nicotine-promoted lung tumorigenesis and emphysema in A/J mice. Cancer Prevention Research, 6(4), 309–320.PubMedCrossRef Iskandar, A. R., Liu, C., Smith, D. E., Hu, K.-Q., Choi, S.-W., Ausman, L. M., et al. (2013). β-Cryptoxanthin restores nicotine-reduced lung SIRT1 to normal levels and inhibits nicotine-promoted lung tumorigenesis and emphysema in A/J mice. Cancer Prevention Research, 6(4), 309–320.PubMedCrossRef
172.
Zurück zum Zitat Cheng, J., Miao, B., Hu, K.-Q., Fu, X., & Wang, X.-D. (2018). Apo-10’-lycopenoic acid inhibits cancer cell migration and angiogenesis and induces peroxisome proliferator-activated receptor γ. The Journal of Nutritional Biochemistry, 56, 26–34.PubMedCrossRef Cheng, J., Miao, B., Hu, K.-Q., Fu, X., & Wang, X.-D. (2018). Apo-10’-lycopenoic acid inhibits cancer cell migration and angiogenesis and induces peroxisome proliferator-activated receptor γ. The Journal of Nutritional Biochemistry, 56, 26–34.PubMedCrossRef
173.
Zurück zum Zitat Manach, C., Scalbert, A., Morand, C., Rémésy, C., & Jiménez, L. (2004). Polyphenols: Food sources and bioavailability. The American journal of Clinical Nutrition, 79(5), 727–747.PubMedCrossRef Manach, C., Scalbert, A., Morand, C., Rémésy, C., & Jiménez, L. (2004). Polyphenols: Food sources and bioavailability. The American journal of Clinical Nutrition, 79(5), 727–747.PubMedCrossRef
175.
Zurück zum Zitat Zhou, Y., Zheng, J., Li, Y., Xu, D.-P., Li, S., Chen, Y.-M., et al. (2016). Natural polyphenols for prevention and treatment of cancer. Nutrients, 8(8), 515.PubMedPubMedCentralCrossRef Zhou, Y., Zheng, J., Li, Y., Xu, D.-P., Li, S., Chen, Y.-M., et al. (2016). Natural polyphenols for prevention and treatment of cancer. Nutrients, 8(8), 515.PubMedPubMedCentralCrossRef
186.
189.
Zurück zum Zitat Lu, Z.-H., Peng, J.-H., Zhang, R.-X., Wang, F., Sun, H.-P., Fang, Y.-J., et al. (2018). Dihydroartemisinin inhibits colon cancer cell viability by inducing apoptosis through up-regulation of PPARγ expression. Saudi Journal of Biological Sciences, 25(2), 372–376.PubMedCrossRef Lu, Z.-H., Peng, J.-H., Zhang, R.-X., Wang, F., Sun, H.-P., Fang, Y.-J., et al. (2018). Dihydroartemisinin inhibits colon cancer cell viability by inducing apoptosis through up-regulation of PPARγ expression. Saudi Journal of Biological Sciences, 25(2), 372–376.PubMedCrossRef
192.
Zurück zum Zitat Takahashi, N., Kang, M.-S., Kuroyanagi, K., Goto, T., Hirai, S., Ohyama, K., et al. (2008). Auraptene, a citrus fruit compound, regulates gene expression as a PPARα agonist in HepG2 hepatocytes. BioFactors, 33(1), 25–32.PubMedCrossRef Takahashi, N., Kang, M.-S., Kuroyanagi, K., Goto, T., Hirai, S., Ohyama, K., et al. (2008). Auraptene, a citrus fruit compound, regulates gene expression as a PPARα agonist in HepG2 hepatocytes. BioFactors, 33(1), 25–32.PubMedCrossRef
194.
Zurück zum Zitat Charmforoshan, E., Karimi, E., Oskoueian, E., Es-Haghi, A., & Iranshahi, M. (2019). Inhibition of human breast cancer cells (MCF-7 cell line) growth via cell proliferation, migration, and angiogenesis by auraptene of Ferula szowitsiana root extract. Journal of Food Measurement and Characterization, 13(4), 2644–2653. https://doi.org/10.1007/s11694-019-00185-6CrossRef Charmforoshan, E., Karimi, E., Oskoueian, E., Es-Haghi, A., & Iranshahi, M. (2019). Inhibition of human breast cancer cells (MCF-7 cell line) growth via cell proliferation, migration, and angiogenesis by auraptene of Ferula szowitsiana root extract. Journal of Food Measurement and Characterization, 13(4), 2644–2653. https://​doi.​org/​10.​1007/​s11694-019-00185-6CrossRef
196.
204.
208.
Zurück zum Zitat Lim, H. Y., Ong, P. S., Wang, L., Goel, A., Ding, L., Wong, A.L.-A., et al. (2021). Celastrol in cancer therapy: Recent developments, challenges and prospects. Cancer Letters, 521, 252–267.PubMedCrossRef Lim, H. Y., Ong, P. S., Wang, L., Goel, A., Ding, L., Wong, A.L.-A., et al. (2021). Celastrol in cancer therapy: Recent developments, challenges and prospects. Cancer Letters, 521, 252–267.PubMedCrossRef
209.
213.
Zurück zum Zitat Chan, K., Chui, S., Wong, D., Ha, W., Chan, C., & Wong, R. (2004). Protective effects of Danshensu from the aqueous extract of Salvia miltiorrhiza (Danshen) against homocysteine-induced endothelial dysfunction. Life Sciences, 75(26), 3157–3171.PubMedCrossRef Chan, K., Chui, S., Wong, D., Ha, W., Chan, C., & Wong, R. (2004). Protective effects of Danshensu from the aqueous extract of Salvia miltiorrhiza (Danshen) against homocysteine-induced endothelial dysfunction. Life Sciences, 75(26), 3157–3171.PubMedCrossRef
217.
Zurück zum Zitat Moballegh Nasery, M., Abadi, B., Poormoghadam, D., Zarrabi, A., Keyhanvar, P., Khanbabaei, H., et al. (2020). Curcumin delivery mediated by bio-based nanoparticles: A review. Molecules, 25(3), 689.PubMedPubMedCentralCrossRef Moballegh Nasery, M., Abadi, B., Poormoghadam, D., Zarrabi, A., Keyhanvar, P., Khanbabaei, H., et al. (2020). Curcumin delivery mediated by bio-based nanoparticles: A review. Molecules, 25(3), 689.PubMedPubMedCentralCrossRef
218.
Zurück zum Zitat Abadi, A. J., Mirzaei, S., Mahabady, M. K., Hashemi, F., Zabolian, A., Hashemi, F., et al. (2022). Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytotherapy Research, 36(1), 189–213.PubMedCrossRef Abadi, A. J., Mirzaei, S., Mahabady, M. K., Hashemi, F., Zabolian, A., Hashemi, F., et al. (2022). Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytotherapy Research, 36(1), 189–213.PubMedCrossRef
226.
Zurück zum Zitat Jiang, A., Wang, X., Shan, X., Li, Y., Wang, P., Jiang, P., et al. (2015). Curcumin reactivates silenced tumor suppressor gene RARβ by reducing DNA methylation. Phytotherapy Research, 29(8), 1237–1245.PubMedCrossRef Jiang, A., Wang, X., Shan, X., Li, Y., Wang, P., Jiang, P., et al. (2015). Curcumin reactivates silenced tumor suppressor gene RARβ by reducing DNA methylation. Phytotherapy Research, 29(8), 1237–1245.PubMedCrossRef
227.
Zurück zum Zitat Lee, Y. K., Lee, W. S., Hwang, J. T., Kwon, D. Y., Surh, Y. J., & Park, O. J. (2009). Curcumin exerts antidifferentiation effect through AMPKα-PPAR-γ in 3T3-L1 adipocytes and antiproliferatory effect through AMPKα-COX-2 in cancer cells. Journal of Agricultural and Food Chemistry, 57(1), 305–310.PubMedCrossRef Lee, Y. K., Lee, W. S., Hwang, J. T., Kwon, D. Y., Surh, Y. J., & Park, O. J. (2009). Curcumin exerts antidifferentiation effect through AMPKα-PPAR-γ in 3T3-L1 adipocytes and antiproliferatory effect through AMPKα-COX-2 in cancer cells. Journal of Agricultural and Food Chemistry, 57(1), 305–310.PubMedCrossRef
230.
Zurück zum Zitat Chen, A. P., & Xu, J. Y. (2005). Activation of PPAR gamma by curcumin inhibits Moser cell growth and mediates suppression of gene expression of cyclin D1 and EGFR. American Journal of Physiology-Gastrointestinal and Liver Physiology, 288(3), G447-G456, https://doi.org/10.1152/ajpgi.00209.2004. Chen, A. P., & Xu, J. Y. (2005). Activation of PPAR gamma by curcumin inhibits Moser cell growth and mediates suppression of gene expression of cyclin D1 and EGFR. American Journal of Physiology-Gastrointestinal and Liver Physiology, 288(3), G447-G456, https://​doi.​org/​10.​1152/​ajpgi.​00209.​2004.
233.
Zurück zum Zitat Altenburg, J. D., Bieberich, A. A., Terry, C., Harvey, K. A., VanHorn, J. F., Xu, Z., et al. (2011). A synergistic antiproliferation effect of curcumin and docosahexaenoic acid in SK-BR-3 breast cancer cells: Unique signaling not explained by the effects of either compound alone. BMC Cancer, 11(1), 1–16.CrossRef Altenburg, J. D., Bieberich, A. A., Terry, C., Harvey, K. A., VanHorn, J. F., Xu, Z., et al. (2011). A synergistic antiproliferation effect of curcumin and docosahexaenoic acid in SK-BR-3 breast cancer cells: Unique signaling not explained by the effects of either compound alone. BMC Cancer, 11(1), 1–16.CrossRef
236.
Zurück zum Zitat Kumar, K. N., Raja, S. B., Vidhya, N., & Devaraj, S. N. (2012). Ellagic acid modulates antioxidant status, ornithine decarboxylase expression, and aberrant crypt foci progression in 1,2-dimethylhydrazine-instigated colon preneoplastic lesions in rats. Journal of Agriculture and Food Chemistry, 60(14), 3665–3672. https://doi.org/10.1021/jf204128zCrossRef Kumar, K. N., Raja, S. B., Vidhya, N., & Devaraj, S. N. (2012). Ellagic acid modulates antioxidant status, ornithine decarboxylase expression, and aberrant crypt foci progression in 1,2-dimethylhydrazine-instigated colon preneoplastic lesions in rats. Journal of Agriculture and Food Chemistry, 60(14), 3665–3672. https://​doi.​org/​10.​1021/​jf204128zCrossRef
238.
Zurück zum Zitat Anitha, P., Priyadarsini, R. V., Kavitha, K., Thiyagarajan, P., & Nagini, S. (2013). Ellagic acid coordinately attenuates Wnt/beta-catenin and NF-kappaB signaling pathways to induce intrinsic apoptosis in an animal model of oral oncogenesis. European Journal of Nutrition, 52(1), 75–84. https://doi.org/10.1007/s00394-011-0288-yCrossRefPubMed Anitha, P., Priyadarsini, R. V., Kavitha, K., Thiyagarajan, P., & Nagini, S. (2013). Ellagic acid coordinately attenuates Wnt/beta-catenin and NF-kappaB signaling pathways to induce intrinsic apoptosis in an animal model of oral oncogenesis. European Journal of Nutrition, 52(1), 75–84. https://​doi.​org/​10.​1007/​s00394-011-0288-yCrossRefPubMed
241.
Zurück zum Zitat Kim, S. W., Kim, S. M., Bae, H., Nam, D., Lee, J. H., Lee, S. G., et al. (2013). Embelin inhibits growth and induces apoptosis through the suppression of Akt/mTOR/S6K1 signaling cascades. The Prostate, 73(3), 296–305.PubMedCrossRef Kim, S. W., Kim, S. M., Bae, H., Nam, D., Lee, J. H., Lee, S. G., et al. (2013). Embelin inhibits growth and induces apoptosis through the suppression of Akt/mTOR/S6K1 signaling cascades. The Prostate, 73(3), 296–305.PubMedCrossRef
243.
Zurück zum Zitat Manu, K. A., Shanmugam, M. K., Ong, T. H., Subramaniam, A., Siveen, K. S., Perumal, E., et al. (2013). Emodin suppresses migration and invasion through the modulation of CXCR4 expression in an orthotopic model of human hepatocellular carcinoma. PLoS One, 8(3), e57015.PubMedPubMedCentralCrossRef Manu, K. A., Shanmugam, M. K., Ong, T. H., Subramaniam, A., Siveen, K. S., Perumal, E., et al. (2013). Emodin suppresses migration and invasion through the modulation of CXCR4 expression in an orthotopic model of human hepatocellular carcinoma. PLoS One, 8(3), e57015.PubMedPubMedCentralCrossRef
244.
Zurück zum Zitat Subramaniam, A., Loo, S. Y., Rajendran, P., Manu, K. A., Perumal, E., Li, F., et al. (2013). An anthraquinone derivative, emodin sensitizes hepatocellular carcinoma cells to TRAIL induced apoptosis through the induction of death receptors and downregulation of cell survival proteins. Apoptosis, 18(10), 1175–1187.PubMedCrossRef Subramaniam, A., Loo, S. Y., Rajendran, P., Manu, K. A., Perumal, E., Li, F., et al. (2013). An anthraquinone derivative, emodin sensitizes hepatocellular carcinoma cells to TRAIL induced apoptosis through the induction of death receptors and downregulation of cell survival proteins. Apoptosis, 18(10), 1175–1187.PubMedCrossRef
248.
Zurück zum Zitat Bektic, J., Berger, A. P., Pfeil, K., Dobler, G., Bartsch, G., & Klocker, H. (2004). Androgen receptor regulation by physiological concentrations of the isoflavonoid genistein in androgen-dependent LNCaP cells is mediated by estrogen receptor β. European Urology, 45(2), 245–251.PubMedCrossRef Bektic, J., Berger, A. P., Pfeil, K., Dobler, G., Bartsch, G., & Klocker, H. (2004). Androgen receptor regulation by physiological concentrations of the isoflavonoid genistein in androgen-dependent LNCaP cells is mediated by estrogen receptor β. European Urology, 45(2), 245–251.PubMedCrossRef
251.
Zurück zum Zitat Oh, H. Y., Leem, J., Yoon, S. J., Yoon, S., & Hong, S. J. (2010). Lipid raft cholesterol and genistein inhibit the cell viability of prostate cancer cells via the partial contribution of EGFR-Akt/p70S6k pathway and down-regulation of androgen receptor. Biochemical and Biophysical Research Communications, 393(2), 319–324. https://doi.org/10.1016/j.bbrc.2010.01.133CrossRefPubMed Oh, H. Y., Leem, J., Yoon, S. J., Yoon, S., & Hong, S. J. (2010). Lipid raft cholesterol and genistein inhibit the cell viability of prostate cancer cells via the partial contribution of EGFR-Akt/p70S6k pathway and down-regulation of androgen receptor. Biochemical and Biophysical Research Communications, 393(2), 319–324. https://​doi.​org/​10.​1016/​j.​bbrc.​2010.​01.​133CrossRefPubMed
253.
Zurück zum Zitat Zhang, T., Wang, F., Xu, H. X., Yi, L., Qin, Y., Chang, H., et al. (2013). Activation of nuclear factor erythroid 2-related factor 2 and PPARgamma plays a role in the genistein-mediated attenuation of oxidative stress-induced endothelial cell injury. British Journal of Nutrition, 109(2), 223–235. https://doi.org/10.1017/S0007114512001110CrossRefPubMed Zhang, T., Wang, F., Xu, H. X., Yi, L., Qin, Y., Chang, H., et al. (2013). Activation of nuclear factor erythroid 2-related factor 2 and PPARgamma plays a role in the genistein-mediated attenuation of oxidative stress-induced endothelial cell injury. British Journal of Nutrition, 109(2), 223–235. https://​doi.​org/​10.​1017/​S000711451200111​0CrossRefPubMed
255.
256.
Zurück zum Zitat Ahn, K. S., Sethi, G., Sung, B., Goel, A., Ralhan, R., & Aggarwal, B. B. (2008). Guggulsterone, a farnesoid X receptor antagonist, inhibits constitutive and inducible STAT3 activation through induction of a protein tyrosine phosphatase SHP-1 (Publication with Expression of Concern. See vol. 78, pg. 5184, 2018). Cancer Research, 68(11), 4406–4415, https://doi.org/10.1158/0008-5472.Can-07-6696. Ahn, K. S., Sethi, G., Sung, B., Goel, A., Ralhan, R., & Aggarwal, B. B. (2008). Guggulsterone, a farnesoid X receptor antagonist, inhibits constitutive and inducible STAT3 activation through induction of a protein tyrosine phosphatase SHP-1 (Publication with Expression of Concern. See vol. 78, pg. 5184, 2018). Cancer Research, 68(11), 4406–4415, https://​doi.​org/​10.​1158/​0008-5472.​Can-07-6696.
257.
Zurück zum Zitat Shishodia, S., Harikumar, K. B., Dass, S., Ramawat, K. G., & Aggarwal, B. B. (2008). The guggul for chronic diseases: Ancient medicine, modern targets. Anticancer Research, 28(6a), 3647–3664.PubMed Shishodia, S., Harikumar, K. B., Dass, S., Ramawat, K. G., & Aggarwal, B. B. (2008). The guggul for chronic diseases: Ancient medicine, modern targets. Anticancer Research, 28(6a), 3647–3664.PubMed
258.
Zurück zum Zitat Girisa, S., Parama, D., Harsha, C., Banik, K., & Kunnumakkara, A. B. (2020). Potential of guggulsterone, a farnesoid X receptor antagonist, in the prevention and treatment of cancer. Exploration of Targeted Anti-tumor Theraphy, 1(5), 313–342, https://doi.org/10.37349/etat.2020.00019. Girisa, S., Parama, D., Harsha, C., Banik, K., & Kunnumakkara, A. B. (2020). Potential of guggulsterone, a farnesoid X receptor antagonist, in the prevention and treatment of cancer. Exploration of Targeted Anti-tumor Theraphy, 1(5), 313–342, https://​doi.​org/​10.​37349/​etat.​2020.​00019.
262.
Zurück zum Zitat Yu, J.-H., Zheng, J.-B., Qi, J., Yang, K., Wu, Y.-H., Wang, K., et al. (2019). Bile acids promote gastric intestinal metaplasia by upregulating CDX2 and MUC2 expression via the FXR/NF-κB signalling pathway. International Journal of Oncology, 54(3), 879–892.PubMedPubMedCentral Yu, J.-H., Zheng, J.-B., Qi, J., Yang, K., Wu, Y.-H., Wang, K., et al. (2019). Bile acids promote gastric intestinal metaplasia by upregulating CDX2 and MUC2 expression via the FXR/NF-κB signalling pathway. International Journal of Oncology, 54(3), 879–892.PubMedPubMedCentral
264.
Zurück zum Zitat Lee, J., Lee, K., Lee, J., Lee, K., Jang, K., Heo, J., et al. (2011). Farnesoid X receptor, overexpressed in pancreatic cancer with lymph node metastasis promotes cell migration and invasion. British Journal of Cancer, 104(6), 1027–1037.PubMedPubMedCentralCrossRef Lee, J., Lee, K., Lee, J., Lee, K., Jang, K., Heo, J., et al. (2011). Farnesoid X receptor, overexpressed in pancreatic cancer with lymph node metastasis promotes cell migration and invasion. British Journal of Cancer, 104(6), 1027–1037.PubMedPubMedCentralCrossRef
265.
268.
Zurück zum Zitat Wang, J., & Zhao, J. (1993). The effect of preventing recurrence of cancer metastasis on jiaogulan soup in clinical study. Zhejiang Zhong Yi Za Zhii, 28(20), 529–530. Wang, J., & Zhao, J. (1993). The effect of preventing recurrence of cancer metastasis on jiaogulan soup in clinical study. Zhejiang Zhong Yi Za Zhii, 28(20), 529–530.
272.
275.
Zurück zum Zitat Ahmadi, A., & Shadboorestan, A. (2016). Oxidative stress and cancer; the role of hesperidin, a citrus natural bioflavonoid, as a cancer chemoprotective agent. Nutrition and Cancer, 68(1), 29–39.PubMedCrossRef Ahmadi, A., & Shadboorestan, A. (2016). Oxidative stress and cancer; the role of hesperidin, a citrus natural bioflavonoid, as a cancer chemoprotective agent. Nutrition and Cancer, 68(1), 29–39.PubMedCrossRef
276.
Zurück zum Zitat Saiprasad, G., Chitra, P., Manikandan, R., & Sudhandiran, G. (2014). Hesperidin induces apoptosis and triggers autophagic markers through inhibition of Aurora-A mediated phosphoinositide-3-kinase/Akt/mammalian target of rapamycin and glycogen synthase kinase-3 beta signalling cascades in experimental colon carcinogenesis. European Journal of Cancer, 50(14), 2489–2507. https://doi.org/10.1016/j.ejca.2014.06.013CrossRefPubMed Saiprasad, G., Chitra, P., Manikandan, R., & Sudhandiran, G. (2014). Hesperidin induces apoptosis and triggers autophagic markers through inhibition of Aurora-A mediated phosphoinositide-3-kinase/Akt/mammalian target of rapamycin and glycogen synthase kinase-3 beta signalling cascades in experimental colon carcinogenesis. European Journal of Cancer, 50(14), 2489–2507. https://​doi.​org/​10.​1016/​j.​ejca.​2014.​06.​013CrossRefPubMed
278.
279.
Zurück zum Zitat Hsu, P. H., Chen, W. H., Chen, J. L., Hsieh, S. C., Lin, S. C., Mai, R. T., et al. (2021). Hesperidin and chlorogenic acid synergistically inhibit the growth of breast cancer cells via estrogen receptor/mitochondrial pathway. Life-Basel, 11(9), ARTN 950. https://doi.org/10.3390/life11090950. Hsu, P. H., Chen, W. H., Chen, J. L., Hsieh, S. C., Lin, S. C., Mai, R. T., et al. (2021). Hesperidin and chlorogenic acid synergistically inhibit the growth of breast cancer cells via estrogen receptor/mitochondrial pathway. Life-Basel, 11(9), ARTN 950. https://​doi.​org/​10.​3390/​life11090950.
280.
Zurück zum Zitat Banik, K., Ranaware, A. M., Deshpande, V., Nalawade, S. P., Padmavathi, G., Bordoloi, D., et al. (2019). Honokiol for cancer therapeutics: A traditional medicine that can modulate multiple oncogenic targets. Pharmacological Research, 144, 192–209.PubMedCrossRef Banik, K., Ranaware, A. M., Deshpande, V., Nalawade, S. P., Padmavathi, G., Bordoloi, D., et al. (2019). Honokiol for cancer therapeutics: A traditional medicine that can modulate multiple oncogenic targets. Pharmacological Research, 144, 192–209.PubMedCrossRef
284.
Zurück zum Zitat Lim, S. L., Park, S. Y., Kang, S., Park, D., Kim, S. H., Um, J. Y., et al. (2015). Morusin induces cell death through inactivating STAT3 signaling in prostate cancer cells. American Journal of Cancer Research, 5(1), 289-U482.PubMed Lim, S. L., Park, S. Y., Kang, S., Park, D., Kim, S. H., Um, J. Y., et al. (2015). Morusin induces cell death through inactivating STAT3 signaling in prostate cancer cells. American Journal of Cancer Research, 5(1), 289-U482.PubMed
286.
Zurück zum Zitat Dat, N. T., Binh, P. T. X., Van Minh, C., Huong, H. T., & Lee, J. J. (2010). Cytotoxic prenylated flavonoids from Morus alba. Fitoterapia, 81(8), 1224–1227.PubMedCrossRef Dat, N. T., Binh, P. T. X., Van Minh, C., Huong, H. T., & Lee, J. J. (2010). Cytotoxic prenylated flavonoids from Morus alba. Fitoterapia, 81(8), 1224–1227.PubMedCrossRef
288.
Zurück zum Zitat Lee, J.-C., Won, S.-J., Chao, C.-L., Wu, F.-L., Liu, H.-S., Ling, P., et al. (2008). Morusin induces apoptosis and suppresses NF-κB activity in human colorectal cancer HT-29 cells. Biochemical and Biophysical Research Communications, 372(1), 236–242.PubMedCrossRef Lee, J.-C., Won, S.-J., Chao, C.-L., Wu, F.-L., Liu, H.-S., Ling, P., et al. (2008). Morusin induces apoptosis and suppresses NF-κB activity in human colorectal cancer HT-29 cells. Biochemical and Biophysical Research Communications, 372(1), 236–242.PubMedCrossRef
289.
Zurück zum Zitat Li, H., Wang, Q., Dong, L., Liu, C., Sun, Z., Gao, L., et al. (2015). Morusin suppresses breast cancer cell growth in vitro and in vivo through C/EBPβ and PPARγ mediated lipoapoptosis. Journal of Experimental & Clinical Cancer Research, 34(1), 1–12.CrossRef Li, H., Wang, Q., Dong, L., Liu, C., Sun, Z., Gao, L., et al. (2015). Morusin suppresses breast cancer cell growth in vitro and in vivo through C/EBPβ and PPARγ mediated lipoapoptosis. Journal of Experimental & Clinical Cancer Research, 34(1), 1–12.CrossRef
290.
Zurück zum Zitat Baek, S. H., Ko, J.-H., Lee, H., Jung, J., Kong, M., Lee, J.-W., et al. (2016). Resveratrol inhibits STAT3 signaling pathway through the induction of SOCS-1: Role in apoptosis induction and radiosensitization in head and neck tumor cells. Phytomedicine, 23(5), 566–577.PubMedCrossRef Baek, S. H., Ko, J.-H., Lee, H., Jung, J., Kong, M., Lee, J.-W., et al. (2016). Resveratrol inhibits STAT3 signaling pathway through the induction of SOCS-1: Role in apoptosis induction and radiosensitization in head and neck tumor cells. Phytomedicine, 23(5), 566–577.PubMedCrossRef
292.
Zurück zum Zitat Nonomura, S., Kanagawa, H., & Makimoto, A. (1963). Chemical Constituents of Polygonaceous Plants. I. Studies on the Components of Ko-J O-Kon. (Polygonum Cuspidatum Sieb. Et Zucc.). Yakugaku Zasshi, 83, 988–990.PubMedCrossRef Nonomura, S., Kanagawa, H., & Makimoto, A. (1963). Chemical Constituents of Polygonaceous Plants. I. Studies on the Components of Ko-J O-Kon. (Polygonum Cuspidatum Sieb. Et Zucc.). Yakugaku Zasshi, 83, 988–990.PubMedCrossRef
295.
Zurück zum Zitat Li, Y. T., Tian, X. T., Wu, M. L., Zheng, X., Kong, Q. Y., Cheng, X. X., et al. (2018). Resveratrol suppresses the growth and enhances retinoic acid sensitivity of anaplastic thyroid cancer cells. International Journal of Molecular Sciences, 19(4), ARTN 1030. https://doi.org/10.3390/ijms19041030. Li, Y. T., Tian, X. T., Wu, M. L., Zheng, X., Kong, Q. Y., Cheng, X. X., et al. (2018). Resveratrol suppresses the growth and enhances retinoic acid sensitivity of anaplastic thyroid cancer cells. International Journal of Molecular Sciences, 19(4), ARTN 1030. https://​doi.​org/​10.​3390/​ijms19041030.
298.
Zurück zum Zitat Shiragannavar, V. D., Gowda, N. G. S., Kumar, D. P., Mirshahi, F., & Santhekadur, P. K. (2021). Withaferin A acts as a novel regulator of liver X receptor-α in HCC. Frontiers in Oncology, 3124. Shiragannavar, V. D., Gowda, N. G. S., Kumar, D. P., Mirshahi, F., & Santhekadur, P. K. (2021). Withaferin A acts as a novel regulator of liver X receptor-α in HCC. Frontiers in Oncology, 3124.
299.
Zurück zum Zitat Lim, Y. P., Cheng, C. H., Chen, W. C., Chang, S. Y., Hung, D. Z., Chen, J. J., et al. (2015). Allyl isothiocyanate (AITC) inhibits pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation and protects against acetaminophen- and amiodarone-induced cytotoxicity. Archives of Toxicology, 89(1), 57–72. https://doi.org/10.1007/s00204-014-1230-xCrossRefPubMed Lim, Y. P., Cheng, C. H., Chen, W. C., Chang, S. Y., Hung, D. Z., Chen, J. J., et al. (2015). Allyl isothiocyanate (AITC) inhibits pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation and protects against acetaminophen- and amiodarone-induced cytotoxicity. Archives of Toxicology, 89(1), 57–72. https://​doi.​org/​10.​1007/​s00204-014-1230-xCrossRefPubMed
300.
Zurück zum Zitat Oh, H., Park, S.-H., Kang, M.-K., Kim, Y.-H., Lee, E.-J., Kim, D. Y., et al. (2020). Asaronic acid inhibited glucose-triggered M2-phenotype shift through disrupting the formation of coordinated signaling of IL-4Rα-Tyk2-STAT6 and GLUT1-Akt-mTOR-AMPK. Nutrients, 12(7), 2006.PubMedPubMedCentralCrossRef Oh, H., Park, S.-H., Kang, M.-K., Kim, Y.-H., Lee, E.-J., Kim, D. Y., et al. (2020). Asaronic acid inhibited glucose-triggered M2-phenotype shift through disrupting the formation of coordinated signaling of IL-4Rα-Tyk2-STAT6 and GLUT1-Akt-mTOR-AMPK. Nutrients, 12(7), 2006.PubMedPubMedCentralCrossRef
303.
Zurück zum Zitat Papaioannou, M., Schleich, S., Roell, D., Schubert, U., Tanner, T., Claessens, F., et al. (2010). NBBS isolated from Pygeum africanum bark exhibits androgen antagonistic activity, inhibits AR nuclear translocation and prostate cancer cell growth. Investigational New Drugs, 28(6), 729–743. https://doi.org/10.1007/s10637-009-9304-yCrossRefPubMed Papaioannou, M., Schleich, S., Roell, D., Schubert, U., Tanner, T., Claessens, F., et al. (2010). NBBS isolated from Pygeum africanum bark exhibits androgen antagonistic activity, inhibits AR nuclear translocation and prostate cancer cell growth. Investigational New Drugs, 28(6), 729–743. https://​doi.​org/​10.​1007/​s10637-009-9304-yCrossRefPubMed
307.
308.
Zurück zum Zitat Weng, J.-R., Bai, L.-Y., Chiu, C.-F., Hu, J.-L., Chiu, S.-J., & Wu, C.-Y. (2013). Cucurbitane triterpenoid from Momordica charantia induces apoptosis and autophagy in breast cancer cells, in part, through peroxisome proliferator-activated receptor γ activation. Evidence-Based Complementary and Alternative Medicine, 2013, 93675. https://doi.org/10.1155/2013/935675CrossRef Weng, J.-R., Bai, L.-Y., Chiu, C.-F., Hu, J.-L., Chiu, S.-J., & Wu, C.-Y. (2013). Cucurbitane triterpenoid from Momordica charantia induces apoptosis and autophagy in breast cancer cells, in part, through peroxisome proliferator-activated receptor γ activation. Evidence-Based Complementary and Alternative Medicine, 2013, 93675. https://​doi.​org/​10.​1155/​2013/​935675CrossRef
309.
Zurück zum Zitat Somjen, D., Grafi-Cohen, M., Weisinger, G., Izkhakov, E., Sharon, O., Kraiem, Z., et al. (2012). Growth inhibition of human thyroid carcinoma and goiter cells in vitro by the isoflavone derivative 7-(O)-carboxymethyl daidzein conjugated to Nt-boc-hexylenediamine. Thyroid, 22(8), 809–813.PubMedCrossRef Somjen, D., Grafi-Cohen, M., Weisinger, G., Izkhakov, E., Sharon, O., Kraiem, Z., et al. (2012). Growth inhibition of human thyroid carcinoma and goiter cells in vitro by the isoflavone derivative 7-(O)-carboxymethyl daidzein conjugated to Nt-boc-hexylenediamine. Thyroid, 22(8), 809–813.PubMedCrossRef
310.
Zurück zum Zitat Ichikawa, H., Nair, M. S., Takada, Y., Sheeja, D. A., Kumar, M. S., Oommen, O. V., et al. (2006). Isodeoxyelephantopin, a novel sesquiterpene lactone, potentiates apoptosis, inhibits invasion, and abolishes osteoclastogenesis through suppression of nuclear factor-κB (NF-κB) activation and NF-κB-regulated gene expression. Clinical Cancer Research, 12(19), 5910–5918.PubMedCrossRef Ichikawa, H., Nair, M. S., Takada, Y., Sheeja, D. A., Kumar, M. S., Oommen, O. V., et al. (2006). Isodeoxyelephantopin, a novel sesquiterpene lactone, potentiates apoptosis, inhibits invasion, and abolishes osteoclastogenesis through suppression of nuclear factor-κB (NF-κB) activation and NF-κB-regulated gene expression. Clinical Cancer Research, 12(19), 5910–5918.PubMedCrossRef
311.
Zurück zum Zitat Wolo, M. T., Cowherd, C. M., & Lee, K. H. (1975). Antitumor agents XV: Deoxyelephantopin, an antitumor principle from Elephantopus carolinianus Willd. Journal of Pharmaceutical Sciences, 64(9), 1572–1573.PubMedCrossRef Wolo, M. T., Cowherd, C. M., & Lee, K. H. (1975). Antitumor agents XV: Deoxyelephantopin, an antitumor principle from Elephantopus carolinianus Willd. Journal of Pharmaceutical Sciences, 64(9), 1572–1573.PubMedCrossRef
312.
Zurück zum Zitat Zou, G., Gao, Z., Wang, J., Zhang, Y., Ding, H., Huang, J., et al. (2008). Deoxyelephantopin inhibits cancer cell proliferation and functions as a selective partial agonist against PPARγ. Biochemical Pharmacology, 75(6), 1381–1392. Zou, G., Gao, Z., Wang, J., Zhang, Y., Ding, H., Huang, J., et al. (2008). Deoxyelephantopin inhibits cancer cell proliferation and functions as a selective partial agonist against PPARγ. Biochemical Pharmacology, 75(6), 1381–1392.
321.
Zurück zum Zitat Takahashi, N., Kawada, T., Goto, T., Yamamoto, T., Taimatsu, A., Matsui, N., et al. (2002). Dual action of isoprenols from herbal medicines on both PPARγ and PPARα in 3T3-L1 adipocytes and HepG2 hepatocytes. FEBS Letters, 514(2–3), 315–322.PubMedCrossRef Takahashi, N., Kawada, T., Goto, T., Yamamoto, T., Taimatsu, A., Matsui, N., et al. (2002). Dual action of isoprenols from herbal medicines on both PPARγ and PPARα in 3T3-L1 adipocytes and HepG2 hepatocytes. FEBS Letters, 514(2–3), 315–322.PubMedCrossRef
323.
Zurück zum Zitat Marconett, C. N., Sundar, S. N., Poindexter, K. M., Stueve, T. R., Bjeldanes, L. F., & Firestone, G. L. (2010). Indole-3-carbinol triggers aryl hydrocarbon receptor-dependent estrogen receptor (ER)alpha protein degradation in breast cancer cells disrupting an ERalpha-GATA3 transcriptional cross-regulatory loop. Molecular Biology of the Cell, 21(7), 1166–1177. https://doi.org/10.1091/mbc.E09-08-0689CrossRefPubMedPubMedCentral Marconett, C. N., Sundar, S. N., Poindexter, K. M., Stueve, T. R., Bjeldanes, L. F., & Firestone, G. L. (2010). Indole-3-carbinol triggers aryl hydrocarbon receptor-dependent estrogen receptor (ER)alpha protein degradation in breast cancer cells disrupting an ERalpha-GATA3 transcriptional cross-regulatory loop. Molecular Biology of the Cell, 21(7), 1166–1177. https://​doi.​org/​10.​1091/​mbc.​E09-08-0689CrossRefPubMedPubMedCentral
324.
Zurück zum Zitat Hsu, J. C., Zhang, J., Dev, A., Wing, A., Bjeldanes, L. F., & Firestone, G. L. (2005). Indole-3-carbinol inhibition of androgen receptor expression and downregulation of androgen responsiveness in human prostate cancer cells. Carcinogenesis, 26(11), 1896–1904.PubMedCrossRef Hsu, J. C., Zhang, J., Dev, A., Wing, A., Bjeldanes, L. F., & Firestone, G. L. (2005). Indole-3-carbinol inhibition of androgen receptor expression and downregulation of androgen responsiveness in human prostate cancer cells. Carcinogenesis, 26(11), 1896–1904.PubMedCrossRef
326.
Zurück zum Zitat Boakye, C. H. A., Doddapaneni, R., Shah, P. P., Patel, A. R., Godugu, C., Safe, S., et al. (2013). Chemoprevention of skin cancer with 1,1-bis (3'-indolyl)-1-(aromatic) methane analog through induction of the orphan nuclear receptor, NR4A2 (Nurr1). Plos One, 8(8), ARTN e69519. https://doi.org/10.1371/journal.pone.0069519. Boakye, C. H. A., Doddapaneni, R., Shah, P. P., Patel, A. R., Godugu, C., Safe, S., et al. (2013). Chemoprevention of skin cancer with 1,1-bis (3'-indolyl)-1-(aromatic) methane analog through induction of the orphan nuclear receptor, NR4A2 (Nurr1). Plos One, 8(8), ARTN e69519. https://​doi.​org/​10.​1371/​journal.​pone.​0069519.
329.
Zurück zum Zitat Hossain, S., Liu, Z. H., & Wood, R. J. (2020). Histone deacetylase activity and vitamin D-dependent gene expressions in relation to sulforaphane in human breast cancer cells. Journal of Food Biochemistry, 44(2), ARTN e13114. https://doi.org/10.1111/jfbc.13114. Hossain, S., Liu, Z. H., & Wood, R. J. (2020). Histone deacetylase activity and vitamin D-dependent gene expressions in relation to sulforaphane in human breast cancer cells. Journal of Food Biochemistry, 44(2), ARTN e13114. https://​doi.​org/​10.​1111/​jfbc.​13114.
330.
Zurück zum Zitat Hossain, S., Liu, Z. H., & Wood, R. J. (2021). Association between histone deacetylase activity and vitamin D-dependent gene expressions in relation to sulforaphane in human colorectal cancer cells. Journal of the Science of Food and Agriculture, 101(5), 1833–1843. https://doi.org/10.1002/jsfa.10797CrossRefPubMed Hossain, S., Liu, Z. H., & Wood, R. J. (2021). Association between histone deacetylase activity and vitamin D-dependent gene expressions in relation to sulforaphane in human colorectal cancer cells. Journal of the Science of Food and Agriculture, 101(5), 1833–1843. https://​doi.​org/​10.​1002/​jsfa.​10797CrossRefPubMed
334.
Zurück zum Zitat Ranaware, A. M., Banik, K., Deshpande, V., Padmavathi, G., Roy, N. K., Sethi, G., et al. (2018). Magnolol: A neolignan from the magnolia family for the prevention and treatment of cancer. International Journal of Molecular Sciences, 19(8), https://doi.org/10.3390/ijms19082362. Ranaware, A. M., Banik, K., Deshpande, V., Padmavathi, G., Roy, N. K., Sethi, G., et al. (2018). Magnolol: A neolignan from the magnolia family for the prevention and treatment of cancer. International Journal of Molecular Sciences, 19(8), https://​doi.​org/​10.​3390/​ijms19082362.
339.
Zurück zum Zitat Goto, T., Takahashi, N., Kato, S., Egawa, K., Ebisu, S., Moriyama, T., et al. (2005). Phytol directly activates peroxisome proliferator-activated receptor alpha (PPARalpha) and regulates gene expression involved in lipid metabolism in PPARalpha-expressing HepG2 hepatocytes. Biochemical and Biophysical Research Communications, 337(2), 440–445. https://doi.org/10.1016/j.bbrc.2005.09.077CrossRefPubMed Goto, T., Takahashi, N., Kato, S., Egawa, K., Ebisu, S., Moriyama, T., et al. (2005). Phytol directly activates peroxisome proliferator-activated receptor alpha (PPARalpha) and regulates gene expression involved in lipid metabolism in PPARalpha-expressing HepG2 hepatocytes. Biochemical and Biophysical Research Communications, 337(2), 440–445. https://​doi.​org/​10.​1016/​j.​bbrc.​2005.​09.​077CrossRefPubMed
341.
Zurück zum Zitat He, T., Wu, K., Lv, Y., Gong, X., Chen, G. G., & Liang, N. (2009). Effect of 5F from Pteris semipinnata on expression of Nr1d1 in HO-8910PM cell line. Zhongguo Zhong Yao Za Zhi, 34(10), 1268–1271.PubMed He, T., Wu, K., Lv, Y., Gong, X., Chen, G. G., & Liang, N. (2009). Effect of 5F from Pteris semipinnata on expression of Nr1d1 in HO-8910PM cell line. Zhongguo Zhong Yao Za Zhi, 34(10), 1268–1271.PubMed
343.
346.
Zurück zum Zitat Shanmugam, M. K., Arfuso, F., Kumar, A. P., Wang, L., Goh, B. C., Ahn, K. S., et al. (2018). Modulation of diverse oncogenic transcription factors by thymoquinone, an essential oil compound isolated from the seeds of Nigella sativa Linn. Pharmacological Research, 129, 357–364.PubMedCrossRef Shanmugam, M. K., Arfuso, F., Kumar, A. P., Wang, L., Goh, B. C., Ahn, K. S., et al. (2018). Modulation of diverse oncogenic transcription factors by thymoquinone, an essential oil compound isolated from the seeds of Nigella sativa Linn. Pharmacological Research, 129, 357–364.PubMedCrossRef
347.
Zurück zum Zitat Woo, C. C., Loo, S. Y., Gee, V., Yap, C. W., Sethi, G., Kumar, A. P., et al. (2011). Anticancer activity of thymoquinone in breast cancer cells: Possible involvement of PPAR-γ pathway. Biochemical Pharmacology, 82(5), 464–475.PubMedCrossRef Woo, C. C., Loo, S. Y., Gee, V., Yap, C. W., Sethi, G., Kumar, A. P., et al. (2011). Anticancer activity of thymoquinone in breast cancer cells: Possible involvement of PPAR-γ pathway. Biochemical Pharmacology, 82(5), 464–475.PubMedCrossRef
350.
Zurück zum Zitat Li, L. H., Stanton, J. D., Tolson, A. H., Luo, Y., & Wang, H. B. (2009). Bioactive terpenoids and flavonoids from ginkgo biloba extract induce the expression of hepatic drug-metabolizing enzymes through pregnane X receptor, constitutive androstane receptor, and aryl hydrocarbon receptor-mediated pathways. Pharmaceutical Research, 26(4), 872–882. https://doi.org/10.1007/s11095-008-9788-8CrossRefPubMed Li, L. H., Stanton, J. D., Tolson, A. H., Luo, Y., & Wang, H. B. (2009). Bioactive terpenoids and flavonoids from ginkgo biloba extract induce the expression of hepatic drug-metabolizing enzymes through pregnane X receptor, constitutive androstane receptor, and aryl hydrocarbon receptor-mediated pathways. Pharmaceutical Research, 26(4), 872–882. https://​doi.​org/​10.​1007/​s11095-008-9788-8CrossRefPubMed
352.
Zurück zum Zitat Jia, Y., Hoang, M. H., Jun, H. J., Lee, J. H., & Lee, S. J. (2013). Cyanidin, a natural flavonoid, is an agonistic ligand for liver X receptor alpha and beta and reduces cellular lipid accumulation in macrophages and hepatocytes. Bioorganic & Medicinal Chemistry Letters, 23(14), 4185–4190. https://doi.org/10.1016/j.bmcl.2013.05.030CrossRef Jia, Y., Hoang, M. H., Jun, H. J., Lee, J. H., & Lee, S. J. (2013). Cyanidin, a natural flavonoid, is an agonistic ligand for liver X receptor alpha and beta and reduces cellular lipid accumulation in macrophages and hepatocytes. Bioorganic & Medicinal Chemistry Letters, 23(14), 4185–4190. https://​doi.​org/​10.​1016/​j.​bmcl.​2013.​05.​030CrossRef
355.
Zurück zum Zitat Hoang, M. H., Jia, Y., Jun, H. J., Lee, J. H., Lee, B. Y., & Lee, S. J. (2012). Fucosterol is a selective liver X receptor modulator that regulates the expression of key genes in cholesterol homeostasis in macrophages, hepatocytes, and intestinal cells. Journal of Agricultural and Food Chemistry, 60(46), 11567–11575. https://doi.org/10.1021/jf3019084CrossRefPubMed Hoang, M. H., Jia, Y., Jun, H. J., Lee, J. H., Lee, B. Y., & Lee, S. J. (2012). Fucosterol is a selective liver X receptor modulator that regulates the expression of key genes in cholesterol homeostasis in macrophages, hepatocytes, and intestinal cells. Journal of Agricultural and Food Chemistry, 60(46), 11567–11575. https://​doi.​org/​10.​1021/​jf3019084CrossRefPubMed
356.
Zurück zum Zitat Hoang, M. H., Jia, Y., Jun, H. J., Lee, J. H., Lee, D. H., Hwang, B. Y., et al. (2012). Ethyl 2,4,6-trihydroxybenzoate is an agonistic ligand for liver X receptor that induces cholesterol efflux from macrophages without affecting lipid accumulation in HepG2 cells. Bioorganic & Medicinal Chemistry Letters, 22(12), 4094–4099. https://doi.org/10.1016/j.bmcl.2012.04.071CrossRef Hoang, M. H., Jia, Y., Jun, H. J., Lee, J. H., Lee, D. H., Hwang, B. Y., et al. (2012). Ethyl 2,4,6-trihydroxybenzoate is an agonistic ligand for liver X receptor that induces cholesterol efflux from macrophages without affecting lipid accumulation in HepG2 cells. Bioorganic & Medicinal Chemistry Letters, 22(12), 4094–4099. https://​doi.​org/​10.​1016/​j.​bmcl.​2012.​04.​071CrossRef
357.
Zurück zum Zitat Jun, H. J., Hoang, M. H., Lee, J. W., Yaoyao, J., Lee, J. H., Lee, D. H., et al. (2012). Iristectorigenin B isolated from Belamcanda chinensis is a liver X receptor modulator that increases ABCA1 and ABCG1 expression in macrophage RAW 264.7 cells. Biotechnology Letters, 34(12), 2213–2221, https://doi.org/10.1007/s10529-012-1036-y. Jun, H. J., Hoang, M. H., Lee, J. W., Yaoyao, J., Lee, J. H., Lee, D. H., et al. (2012). Iristectorigenin B isolated from Belamcanda chinensis is a liver X receptor modulator that increases ABCA1 and ABCG1 expression in macrophage RAW 264.7 cells. Biotechnology Letters, 34(12), 2213–2221, https://​doi.​org/​10.​1007/​s10529-012-1036-y.
359.
Zurück zum Zitat Kma, L. (2014). Plant extracts and plant-derived compounds: Promising players in countermeasure strategy against radiological exposure: A review. Asian Pacific Journal of Cancer Prevention, 15(6), 2405–2425.PubMedCrossRef Kma, L. (2014). Plant extracts and plant-derived compounds: Promising players in countermeasure strategy against radiological exposure: A review. Asian Pacific Journal of Cancer Prevention, 15(6), 2405–2425.PubMedCrossRef
361.
Zurück zum Zitat Yang, Y., Ikezoe, T., Takeuchi, T., Adachi, Y., Ohtsuki, Y., Koeffler, H. P., et al. (2006). Zanthoxyli Fructus induces growth arrest and apoptosis of LNCaP human prostate cancer cells in vitro and in vivo in association with blockade of the AKT and AR signal pathways. Oncology Reports, 15(6), 1581–1590.PubMed Yang, Y., Ikezoe, T., Takeuchi, T., Adachi, Y., Ohtsuki, Y., Koeffler, H. P., et al. (2006). Zanthoxyli Fructus induces growth arrest and apoptosis of LNCaP human prostate cancer cells in vitro and in vivo in association with blockade of the AKT and AR signal pathways. Oncology Reports, 15(6), 1581–1590.PubMed
362.
Zurück zum Zitat Yang, Y., Ikezoe, T., Zheng, Z., Taguchi, H., Koeffler, H. P., & Zhu, W. G. (2007). Saw Palmetto induces growth arrest and apoptosis of androgen-dependent prostate cancer LNCaP cells via inactivation of STAT 3 and androgen receptor signaling. International Journal of Oncology, 31(3), 593–600.PubMed Yang, Y., Ikezoe, T., Zheng, Z., Taguchi, H., Koeffler, H. P., & Zhu, W. G. (2007). Saw Palmetto induces growth arrest and apoptosis of androgen-dependent prostate cancer LNCaP cells via inactivation of STAT 3 and androgen receptor signaling. International Journal of Oncology, 31(3), 593–600.PubMed
363.
Zurück zum Zitat Chen, K. C., Peng, C. C., Chiu, W. T., Cheng, Y. T., Huang, G. T., Hsieh, C. L., et al. (2010). Action mechanism and signal pathways of Psidium guajava L. aqueous extract in killing prostate cancer LNCaP cells. Nutrition and Cancer, 62(2), 260–270, https://doi.org/10.1080/01635580903407130. Chen, K. C., Peng, C. C., Chiu, W. T., Cheng, Y. T., Huang, G. T., Hsieh, C. L., et al. (2010). Action mechanism and signal pathways of Psidium guajava L. aqueous extract in killing prostate cancer LNCaP cells. Nutrition and Cancer, 62(2), 260–270, https://​doi.​org/​10.​1080/​0163558090340713​0.
366.
Zurück zum Zitat Chen, J., Power, K. A., Mann, J., Cheng, A., & Thompson, L. U. (2007). Dietary flaxseed interaction with tamoxifen induced tumor regression in athymic mice with MCF-7 xenografts by downregulating the expression of estrogen related gene products and signal transduction pathways. Nutrition and Cancer, 58(2), 162–170. https://doi.org/10.1080/01635580701328271CrossRefPubMed Chen, J., Power, K. A., Mann, J., Cheng, A., & Thompson, L. U. (2007). Dietary flaxseed interaction with tamoxifen induced tumor regression in athymic mice with MCF-7 xenografts by downregulating the expression of estrogen related gene products and signal transduction pathways. Nutrition and Cancer, 58(2), 162–170. https://​doi.​org/​10.​1080/​0163558070132827​1CrossRefPubMed
367.
Zurück zum Zitat Jeyabalan, J., Aqil, F., Munagala, R., Annamalai, L., Vadhanam, M. V., & Gupta, R. C. (2014). Chemopreventive and therapeutic activity of dietary blueberry against estrogen-mediated breast cancer. Journal of Agriculture and Food Chemistry, 62(18), 3963–3971. https://doi.org/10.1021/jf403734jCrossRef Jeyabalan, J., Aqil, F., Munagala, R., Annamalai, L., Vadhanam, M. V., & Gupta, R. C. (2014). Chemopreventive and therapeutic activity of dietary blueberry against estrogen-mediated breast cancer. Journal of Agriculture and Food Chemistry, 62(18), 3963–3971. https://​doi.​org/​10.​1021/​jf403734jCrossRef
370.
Zurück zum Zitat Sanaei, M., Kavoosi, F., & Arabloo, M. (2020). Effect of curcumin in comparison with trichostatin A on the reactivation of estrogen receptor alpha gene expression, cell growth inhibition and apoptosis induction in hepatocellular carcinoma Hepa 1–6 cell lline. Asian Pacific Journal of Cancer Prevention: APJCP, 21(4), 1045.PubMedPubMedCentralCrossRef Sanaei, M., Kavoosi, F., & Arabloo, M. (2020). Effect of curcumin in comparison with trichostatin A on the reactivation of estrogen receptor alpha gene expression, cell growth inhibition and apoptosis induction in hepatocellular carcinoma Hepa 1–6 cell lline. Asian Pacific Journal of Cancer Prevention: APJCP, 21(4), 1045.PubMedPubMedCentralCrossRef
372.
Zurück zum Zitat Nakamura, K., Yasunaga, Y., Segawa, T., Ko, D., Moul, J. W., Srivastava, S., et al. (2002). Curcumin down-regulates AR gene expression and activation in prostate cancer cell lines. International Journal of Oncology, 21(4), 825–830.PubMed Nakamura, K., Yasunaga, Y., Segawa, T., Ko, D., Moul, J. W., Srivastava, S., et al. (2002). Curcumin down-regulates AR gene expression and activation in prostate cancer cell lines. International Journal of Oncology, 21(4), 825–830.PubMed
373.
Zurück zum Zitat Choi, H., Lim, J., & Hong, J. (2010). Curcumin interrupts the interaction between the androgen receptor and Wnt/β-catenin signaling pathway in LNCaP prostate cancer cells. Prostate Cancer and Prostatic Diseases, 13(4), 343–349.PubMedCrossRef Choi, H., Lim, J., & Hong, J. (2010). Curcumin interrupts the interaction between the androgen receptor and Wnt/β-catenin signaling pathway in LNCaP prostate cancer cells. Prostate Cancer and Prostatic Diseases, 13(4), 343–349.PubMedCrossRef
375.
Zurück zum Zitat Guo, H., Xu, Y. M., Ye, Z. Q., Yu, J. H., & Hu, X. Y. (2013). Curcumin induces cell cycle arrest and apoptosis of prostate cancer cells by regulating the expression of IkappaBalpha, c-Jun and androgen receptor. Die Pharmazie, 68(6), 431–434.PubMed Guo, H., Xu, Y. M., Ye, Z. Q., Yu, J. H., & Hu, X. Y. (2013). Curcumin induces cell cycle arrest and apoptosis of prostate cancer cells by regulating the expression of IkappaBalpha, c-Jun and androgen receptor. Die Pharmazie, 68(6), 431–434.PubMed
376.
Zurück zum Zitat Dorai, T., Gehani, N., & Katz, A. (2000). Therapeutic potential of curcumin in human prostate cancer - I. curcumin induces apoptosis in both androgen-dependent and androgen-independent prostate cancer cells. Prostate Cancer and Prostatic Diseases, 3(2), 84–93, https://doi.org/10.1038/sj.pcan.4500399. Dorai, T., Gehani, N., & Katz, A. (2000). Therapeutic potential of curcumin in human prostate cancer - I. curcumin induces apoptosis in both androgen-dependent and androgen-independent prostate cancer cells. Prostate Cancer and Prostatic Diseases, 3(2), 84–93, https://​doi.​org/​10.​1038/​sj.​pcan.​4500399.
377.
379.
Zurück zum Zitat Sharma, V., Kumar, L., Mohanty, S. K., Maikhuri, J. P., Rajender, S., & Gupta, G. (2016). Sensitization of androgen refractory prostate cancer cells to anti androgens through re-expression of epigenetically repressed androgen receptor - Synergistic action of quercetin and curcumin. Molecular and Cellular Endocrinology, 431(C), 12–23, https://doi.org/10.1016/j.mce.2016.04.024. Sharma, V., Kumar, L., Mohanty, S. K., Maikhuri, J. P., Rajender, S., & Gupta, G. (2016). Sensitization of androgen refractory prostate cancer cells to anti androgens through re-expression of epigenetically repressed androgen receptor - Synergistic action of quercetin and curcumin. Molecular and Cellular Endocrinology, 431(C), 12–23, https://​doi.​org/​10.​1016/​j.​mce.​2016.​04.​024.
381.
Zurück zum Zitat Pham, N. K., Bui, H. T., Tran, T. H., Hoang, T. N. A., Vu, T. H., Do, D. T., et al. (2021). Dammarane triterpenes and phytosterols from Dysoxylum tpongense Pierre and their anti-inflammatory activity against liver X receptors and NF-κB activation. Steroids, 175, 108902.PubMedCrossRef Pham, N. K., Bui, H. T., Tran, T. H., Hoang, T. N. A., Vu, T. H., Do, D. T., et al. (2021). Dammarane triterpenes and phytosterols from Dysoxylum tpongense Pierre and their anti-inflammatory activity against liver X receptors and NF-κB activation. Steroids, 175, 108902.PubMedCrossRef
382.
Zurück zum Zitat Schleich, S., Papaioannou, M., Baniahmad, A., & Matusch, R. (2006). Activity-guided isolation of an antiandrogenic compound of Pygeum africanum. Planta medica, 72(06), 547–551.PubMedCrossRef Schleich, S., Papaioannou, M., Baniahmad, A., & Matusch, R. (2006). Activity-guided isolation of an antiandrogenic compound of Pygeum africanum. Planta medica, 72(06), 547–551.PubMedCrossRef
383.
Zurück zum Zitat Levenson, A. S., Gehm, B. D., Pearce, S. T., Horiguchi, J., Simons, L. A., Ward, J. E., et al. (2003). Resveratrol acts as an estrogen receptor (ER) agonist in breast cancer cells stably transfected with ER alpha. International Journal of Cancer, 104(5), 587–596. https://doi.org/10.1002/ijc.10992CrossRefPubMed Levenson, A. S., Gehm, B. D., Pearce, S. T., Horiguchi, J., Simons, L. A., Ward, J. E., et al. (2003). Resveratrol acts as an estrogen receptor (ER) agonist in breast cancer cells stably transfected with ER alpha. International Journal of Cancer, 104(5), 587–596. https://​doi.​org/​10.​1002/​ijc.​10992CrossRefPubMed
384.
Zurück zum Zitat Mitchell, S. H., Zhu, W., & Young, C. Y. F. (1999). Resveratrol inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Cancer Research, 59(23), 5892–5895.PubMed Mitchell, S. H., Zhu, W., & Young, C. Y. F. (1999). Resveratrol inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Cancer Research, 59(23), 5892–5895.PubMed
387.
Zurück zum Zitat Mitani, T., Harada, N., Tanimori, S., Nakano, Y., Inui, H., & Yamaji, R. (2014). Resveratrol inhibits hypoxia-inducible factor-1alpha-mediated androgen receptor signaling and represses tumor progression in castration-resistant prostate cancer. Journal of Nutritional Science and Vitaminology (Tokyo), 60(4), 276–282.CrossRef Mitani, T., Harada, N., Tanimori, S., Nakano, Y., Inui, H., & Yamaji, R. (2014). Resveratrol inhibits hypoxia-inducible factor-1alpha-mediated androgen receptor signaling and represses tumor progression in castration-resistant prostate cancer. Journal of Nutritional Science and Vitaminology (Tokyo), 60(4), 276–282.CrossRef
389.
Zurück zum Zitat Qi, H. Y., Jiang, Z. Y., Wang, C. Q., Yang, Y., Li, L., He, H., et al. (2017). Sensitization of tamoxifen-resistant breast cancer cells by Z-ligustilide through inhibiting autophagy and accumulating DNA damages. Oncotarget, 8(17), 29300–29317, https://doi.org/10.18632/oncotarget.16832. Qi, H. Y., Jiang, Z. Y., Wang, C. Q., Yang, Y., Li, L., He, H., et al. (2017). Sensitization of tamoxifen-resistant breast cancer cells by Z-ligustilide through inhibiting autophagy and accumulating DNA damages. Oncotarget, 8(17), 29300–29317, https://​doi.​org/​10.​18632/​oncotarget.​16832.
390.
Zurück zum Zitat Shrestha, R., Mohankumar, K., & Safe, S. (2020). Bis-indole derived nuclear receptor 4A1 (NR4A1) antagonists inhibit TGFβ-induced invasion of embryonal rhabdomyosarcoma cells. American Journal of Cancer Research, 10(8), 2495.PubMedPubMedCentral Shrestha, R., Mohankumar, K., & Safe, S. (2020). Bis-indole derived nuclear receptor 4A1 (NR4A1) antagonists inhibit TGFβ-induced invasion of embryonal rhabdomyosarcoma cells. American Journal of Cancer Research, 10(8), 2495.PubMedPubMedCentral
391.
Zurück zum Zitat Yeh, S.-L., Yeh, C.-L., Chan, S.-T., & Chuang, C.-H. (2011). Plasma rich in quercetin metabolites induces G2/M arrest by upregulating PPAR-γ expression in human A549 lung cancer cells. Planta Medica, 77(10), 992–998.PubMedCrossRef Yeh, S.-L., Yeh, C.-L., Chan, S.-T., & Chuang, C.-H. (2011). Plasma rich in quercetin metabolites induces G2/M arrest by upregulating PPAR-γ expression in human A549 lung cancer cells. Planta Medica, 77(10), 992–998.PubMedCrossRef
393.
Zurück zum Zitat Helleboid, S., Haug, C., Lamottke, K., Zhou, Y., Wei, J., Daix, S., et al. (2014). The identification of naturally occurring neoruscogenin as a bioavailable, potent, and high-affinity agonist of the nuclear receptor RORα (NR1F1). Journal of Biomolecular Screening, 19(3), 399–406.PubMedCrossRef Helleboid, S., Haug, C., Lamottke, K., Zhou, Y., Wei, J., Daix, S., et al. (2014). The identification of naturally occurring neoruscogenin as a bioavailable, potent, and high-affinity agonist of the nuclear receptor RORα (NR1F1). Journal of Biomolecular Screening, 19(3), 399–406.PubMedCrossRef
394.
Zurück zum Zitat Ao, M., Zhang, J., Qian, Y., Li, B., Wang, X., Chen, J., et al. (2022). Design and synthesis of adamantyl-substituted flavonoid derivatives as anti-inflammatory Nur77 modulators: Compound B7 targets Nur77 and improves LPS-induced inflammation in vitro and in vivo. Bioorganic Chemistry, 120, 105645.PubMedCrossRef Ao, M., Zhang, J., Qian, Y., Li, B., Wang, X., Chen, J., et al. (2022). Design and synthesis of adamantyl-substituted flavonoid derivatives as anti-inflammatory Nur77 modulators: Compound B7 targets Nur77 and improves LPS-induced inflammation in vitro and in vivo. Bioorganic Chemistry, 120, 105645.PubMedCrossRef
395.
Zurück zum Zitat Zhao, J., Khan, S. I., Wang, M., Vasquez, Y., Yang, M. H., Avula, B., et al. (2014). Octulosonic acid derivatives from Roman chamomile (Chamaemelum nobile) with activities against inflammation and metabolic disorder. Journal of Natural Products, 77(3), 509–515.PubMedCrossRef Zhao, J., Khan, S. I., Wang, M., Vasquez, Y., Yang, M. H., Avula, B., et al. (2014). Octulosonic acid derivatives from Roman chamomile (Chamaemelum nobile) with activities against inflammation and metabolic disorder. Journal of Natural Products, 77(3), 509–515.PubMedCrossRef
396.
Zurück zum Zitat Tamehiro, N., Sato, Y., Suzuki, T., Hashimoto, T., Asakawa, Y., Yokoyama, S., et al. (2005). Riccardin C: A natural product that functions as a liver X receptor (LXR) α agonist and an LXRβ antagonist. FEBS Letters, 579(24), 5299–5304.PubMedCrossRef Tamehiro, N., Sato, Y., Suzuki, T., Hashimoto, T., Asakawa, Y., Yokoyama, S., et al. (2005). Riccardin C: A natural product that functions as a liver X receptor (LXR) α agonist and an LXRβ antagonist. FEBS Letters, 579(24), 5299–5304.PubMedCrossRef
397.
Zurück zum Zitat Uemura, T., Goto, T., Kang, M. S., Mizoguchi, N., Hirai, S., Lee, J. Y., et al. (2011). Diosgenin, the main aglycon of fenugreek, inhibits LXRα activity in HepG2 cells and decreases plasma and hepatic triglycerides in obese diabetic mice. The Journal of Nutrition, 141(1), 17–23.PubMedCrossRef Uemura, T., Goto, T., Kang, M. S., Mizoguchi, N., Hirai, S., Lee, J. Y., et al. (2011). Diosgenin, the main aglycon of fenugreek, inhibits LXRα activity in HepG2 cells and decreases plasma and hepatic triglycerides in obese diabetic mice. The Journal of Nutrition, 141(1), 17–23.PubMedCrossRef
398.
Zurück zum Zitat Goldwasser, J., Cohen, P. Y., Yang, E., Balaguer, P., Yarmush, M. L., & Nahmias, Y. (2010). Transcriptional regulation of human and rat hepatic lipid metabolism by the grapefruit flavonoid naringenin: Role of PPARα PPARγ and LXRα. PloS One, 5(8), e12399.PubMedPubMedCentralCrossRef Goldwasser, J., Cohen, P. Y., Yang, E., Balaguer, P., Yarmush, M. L., & Nahmias, Y. (2010). Transcriptional regulation of human and rat hepatic lipid metabolism by the grapefruit flavonoid naringenin: Role of PPARα PPARγ and LXRα. PloS One, 5(8), e12399.PubMedPubMedCentralCrossRef
399.
Zurück zum Zitat Sheng, X., Wang, M., Lu, M., Xi, B., Sheng, H., & Zang, Y. Q. (2011). Rhein ameliorates fatty liver disease through negative energy balance, hepatic lipogenic regulation, and immunomodulation in diet-induced obese mice. American Journal of Physiology-Endocrinology and Metabolism, 300(5), E886–E893.PubMedCrossRef Sheng, X., Wang, M., Lu, M., Xi, B., Sheng, H., & Zang, Y. Q. (2011). Rhein ameliorates fatty liver disease through negative energy balance, hepatic lipogenic regulation, and immunomodulation in diet-induced obese mice. American Journal of Physiology-Endocrinology and Metabolism, 300(5), E886–E893.PubMedCrossRef
400.
Zurück zum Zitat Montserrat-de la Paz, S., Fernández-Arche, M., Bermúdez, B., & García-Giménez, M. D. (2015). The sterols isolated from evening primrose oil inhibit human colon adenocarcinoma cell proliferation and induce cell cycle arrest through upregulation of LXR. Journal of Functional Foods, 12, 64–69. Montserrat-de la Paz, S., Fernández-Arche, M., Bermúdez, B., & García-Giménez, M. D. (2015). The sterols isolated from evening primrose oil inhibit human colon adenocarcinoma cell proliferation and induce cell cycle arrest through upregulation of LXR. Journal of Functional Foods, 12, 64–69.
401.
Zurück zum Zitat Kao, T.-C., Shyu, M.-H., & Yen, G.-C. (2010). Glycyrrhizic acid and 18β-glycyrrhetinic acid inhibit inflammation via PI3K/Akt/GSK3β signaling and glucocorticoid receptor activation. Journal of Agricultural and Food Chemistry, 58(15), 8623–8629.PubMedCrossRef Kao, T.-C., Shyu, M.-H., & Yen, G.-C. (2010). Glycyrrhizic acid and 18β-glycyrrhetinic acid inhibit inflammation via PI3K/Akt/GSK3β signaling and glucocorticoid receptor activation. Journal of Agricultural and Food Chemistry, 58(15), 8623–8629.PubMedCrossRef
402.
Zurück zum Zitat Kao, T.-C., Wu, C.-H., & Yen, G.-C. (2013). Glycyrrhizic acid and 18β-glycyrrhetinic acid recover glucocorticoid resistance via PI3K-induced AP1 CRE and NFAT activation. Phytomedicine, 20(3–4), 295–302.PubMedCrossRef Kao, T.-C., Wu, C.-H., & Yen, G.-C. (2013). Glycyrrhizic acid and 18β-glycyrrhetinic acid recover glucocorticoid resistance via PI3K-induced AP1 CRE and NFAT activation. Phytomedicine, 20(3–4), 295–302.PubMedCrossRef
403.
Zurück zum Zitat Chintharlapalli, S., Papineni, S., Jutooru, I., McAlees, A., & Safe, S. (2007). Structure-dependent activity of glycyrrhetinic acid derivatives as peroxisome proliferator–activated receptor γ agonists in colon cancer cells. Molecular Cancer Therapeutics, 6(5), 1588–1598.PubMedCrossRef Chintharlapalli, S., Papineni, S., Jutooru, I., McAlees, A., & Safe, S. (2007). Structure-dependent activity of glycyrrhetinic acid derivatives as peroxisome proliferator–activated receptor γ agonists in colon cancer cells. Molecular Cancer Therapeutics, 6(5), 1588–1598.PubMedCrossRef
404.
Zurück zum Zitat Lee, Y., Chung, E., Lee, K. Y., Lee, Y. H., Huh, B., & Lee, S. K. (1997). Ginsenoside-Rg1, one of the major active molecules from Panax ginseng, is a functional ligand of glucocorticoid receptor. Molecular and Cellular Endocrinology, 133(2), 135–140.PubMedCrossRef Lee, Y., Chung, E., Lee, K. Y., Lee, Y. H., Huh, B., & Lee, S. K. (1997). Ginsenoside-Rg1, one of the major active molecules from Panax ginseng, is a functional ligand of glucocorticoid receptor. Molecular and Cellular Endocrinology, 133(2), 135–140.PubMedCrossRef
405.
Zurück zum Zitat Ferron, P.-J., Hogeveen, K., De Sousa, G., Rahmani, R., Dubreil, E., Fessard, V. r., et al. (2016). Modulation of CYP3A4 activity alters the cytotoxicity of lipophilic phycotoxins in human hepatic HepaRG cells. Toxicology in Vitro, 33, 136-146. Ferron, P.-J., Hogeveen, K., De Sousa, G., Rahmani, R., Dubreil, E., Fessard, V. r., et al. (2016). Modulation of CYP3A4 activity alters the cytotoxicity of lipophilic phycotoxins in human hepatic HepaRG cells. Toxicology in Vitro, 33, 136-146.
406.
407.
Zurück zum Zitat Gasmi, J., & Sanderson, J. T. (2010). Growth inhibitory, antiandrogenic, and pro-apoptotic effects of punicic acid in LNCaP human prostate cancer cells. Journal of Agricultural and Food Chemistry, 58(23), 12149–12156.PubMedCrossRef Gasmi, J., & Sanderson, J. T. (2010). Growth inhibitory, antiandrogenic, and pro-apoptotic effects of punicic acid in LNCaP human prostate cancer cells. Journal of Agricultural and Food Chemistry, 58(23), 12149–12156.PubMedCrossRef
408.
Zurück zum Zitat Cvoro, A., Paruthiyil, S., Jones, J. O., Tzagarakis-Foster, C., Clegg, N. J., Tatomer, D., et al. (2007). Selective activation of estrogen receptor-β transcriptional pathways by an herbal extract. Endocrinology, 148(2), 538–547.PubMedCrossRef Cvoro, A., Paruthiyil, S., Jones, J. O., Tzagarakis-Foster, C., Clegg, N. J., Tatomer, D., et al. (2007). Selective activation of estrogen receptor-β transcriptional pathways by an herbal extract. Endocrinology, 148(2), 538–547.PubMedCrossRef
409.
Zurück zum Zitat Liu, L., Ma, H., Tang, Y., Chen, W., Lu, Y., Guo, J., et al. (2012). Discovery of estrogen receptor α modulators from natural compounds in Si-Wu-Tang series decoctions using estrogen-responsive MCF-7 breast cancer cells. Bioorganic & Medicinal Chemistry Letters, 22(1), 154–163.CrossRef Liu, L., Ma, H., Tang, Y., Chen, W., Lu, Y., Guo, J., et al. (2012). Discovery of estrogen receptor α modulators from natural compounds in Si-Wu-Tang series decoctions using estrogen-responsive MCF-7 breast cancer cells. Bioorganic & Medicinal Chemistry Letters, 22(1), 154–163.CrossRef
410.
Zurück zum Zitat Huang, T.H.-W., Razmovski-Naumovski, V., Salam, N. K., Duke, R. K., Duke, C. C., & Roufogalis, B. D. (2005). A novel LXR-α activator identified from the natural product Gynostemma pentaphyllum. Biochemical Pharmacology, 70(9), 1298–1308.PubMedCrossRef Huang, T.H.-W., Razmovski-Naumovski, V., Salam, N. K., Duke, R. K., Duke, C. C., & Roufogalis, B. D. (2005). A novel LXR-α activator identified from the natural product Gynostemma pentaphyllum. Biochemical Pharmacology, 70(9), 1298–1308.PubMedCrossRef
411.
Zurück zum Zitat Yuan, H.-Q., Kong, F., Wang, X.-L., Young, C. Y., Hu, X.-Y., & Lou, H.-X. (2008). Inhibitory effect of acetyl-11-keto-β-boswellic acid on androgen receptor by interference of Sp1 binding activity in prostate cancer cells. Biochemical Pharmacology, 75(11), 2112–2121.PubMedCrossRef Yuan, H.-Q., Kong, F., Wang, X.-L., Young, C. Y., Hu, X.-Y., & Lou, H.-X. (2008). Inhibitory effect of acetyl-11-keto-β-boswellic acid on androgen receptor by interference of Sp1 binding activity in prostate cancer cells. Biochemical Pharmacology, 75(11), 2112–2121.PubMedCrossRef
412.
Zurück zum Zitat Chung, B. H., Lee, H.-Y., Lee, J. S., & Young, C. Y. (2006). Perillyl alcohol inhibits the expression and function of the androgen receptor in human prostate cancer cells. Cancer Letters, 236(2), 222–228.PubMedCrossRef Chung, B. H., Lee, H.-Y., Lee, J. S., & Young, C. Y. (2006). Perillyl alcohol inhibits the expression and function of the androgen receptor in human prostate cancer cells. Cancer Letters, 236(2), 222–228.PubMedCrossRef
413.
Zurück zum Zitat Pathirana, C., Stein, R. B., Berger, T. S., Fenical, W., Ianiro, T., Mais, D. E., et al. (1995). Nonsteroidal human progesterone receptor modulators from the marine alga Cymopolia barbata. Molecular Pharmacology, 47(3), 630–635.PubMed Pathirana, C., Stein, R. B., Berger, T. S., Fenical, W., Ianiro, T., Mais, D. E., et al. (1995). Nonsteroidal human progesterone receptor modulators from the marine alga Cymopolia barbata. Molecular Pharmacology, 47(3), 630–635.PubMed
414.
Zurück zum Zitat Zhao, Z., Wang, L., James, T., Jung, Y., Kim, I., Tan, R., et al. (2015). Reciprocal regulation of ERα and ERβ stability and activity by diptoindonesin G. Chemistry & Biology, 22(12), 1608–1621.CrossRef Zhao, Z., Wang, L., James, T., Jung, Y., Kim, I., Tan, R., et al. (2015). Reciprocal regulation of ERα and ERβ stability and activity by diptoindonesin G. Chemistry & Biology, 22(12), 1608–1621.CrossRef
415.
Zurück zum Zitat Lin, W., Huang, J., Liao, X., Yuan, Z., Feng, S., Xie, Y., et al. (2016). Neo-tanshinlactone selectively inhibits the proliferation of estrogen receptor positive breast cancer cells through transcriptional down-regulation of estrogen receptor alpha. Pharmacological Research, 111, 849–858.PubMedCrossRef Lin, W., Huang, J., Liao, X., Yuan, Z., Feng, S., Xie, Y., et al. (2016). Neo-tanshinlactone selectively inhibits the proliferation of estrogen receptor positive breast cancer cells through transcriptional down-regulation of estrogen receptor alpha. Pharmacological Research, 111, 849–858.PubMedCrossRef
416.
Zurück zum Zitat Onogi, K., Niwa, K., Tang, L., Yun, W., Mori, H., & Tamaya, T. (2006). Inhibitory effects of Hochu-ekki-to on endometrial carcinogenesis induced by N-methyl-N-nitrosourea and 17β-estradiol in mice. Oncology Reports, 16(6), 1343–1348. Onogi, K., Niwa, K., Tang, L., Yun, W., Mori, H., & Tamaya, T. (2006). Inhibitory effects of Hochu-ekki-to on endometrial carcinogenesis induced by N-methyl-N-nitrosourea and 17β-estradiol in mice. Oncology Reports, 16(6), 1343–1348.
417.
Zurück zum Zitat Marconett, C. N., Morgenstern, T. J., San Roman, A. K., Sundar, S. N., Singhal, A. K., & Firestone, G. L. (2010). BZL101, a phytochemical extract from the Scutellaria barbata plant, disrupts proliferation of human breast and prostate cancer cells through distinct mechanisms dependent on the cancer cell phenotype. Cancer Biology & Therapy, 10(4), 397–405.CrossRef Marconett, C. N., Morgenstern, T. J., San Roman, A. K., Sundar, S. N., Singhal, A. K., & Firestone, G. L. (2010). BZL101, a phytochemical extract from the Scutellaria barbata plant, disrupts proliferation of human breast and prostate cancer cells through distinct mechanisms dependent on the cancer cell phenotype. Cancer Biology & Therapy, 10(4), 397–405.CrossRef
418.
Zurück zum Zitat Mora, F. D., Jones, D. K., Desai, P. V., Patny, A., Avery, M. A., Feller, D. R., et al. (2006). Bioassay for the identification of natural product-based activators of peroxisome proliferator-activated receptor-gamma (PPARgamma): The marine sponge metabolite psammaplin A activates PPARgamma and induces apoptosis in human breast tumor cells. Journal of Natural Products, 69(4), 547–552. https://doi.org/10.1021/np050397qCrossRefPubMed Mora, F. D., Jones, D. K., Desai, P. V., Patny, A., Avery, M. A., Feller, D. R., et al. (2006). Bioassay for the identification of natural product-based activators of peroxisome proliferator-activated receptor-gamma (PPARgamma): The marine sponge metabolite psammaplin A activates PPARgamma and induces apoptosis in human breast tumor cells. Journal of Natural Products, 69(4), 547–552. https://​doi.​org/​10.​1021/​np050397qCrossRefPubMed
419.
Zurück zum Zitat Sadar, M. D., Williams, D. E., Mawji, N. R., Patrick, B. O., Wikanta, T., Chasanah, E., et al. (2008). Sintokamides A to E, chlorinated peptides from the sponge Dysidea sp that inhibit transactivation of the N-terminus of the androgen receptor in prostate cancer cells. Organic Letters, 10(21), 4947–4950. https://doi.org/10.1021/ol802021wCrossRefPubMed Sadar, M. D., Williams, D. E., Mawji, N. R., Patrick, B. O., Wikanta, T., Chasanah, E., et al. (2008). Sintokamides A to E, chlorinated peptides from the sponge Dysidea sp that inhibit transactivation of the N-terminus of the androgen receptor in prostate cancer cells. Organic Letters, 10(21), 4947–4950. https://​doi.​org/​10.​1021/​ol802021wCrossRefPubMed
420.
Zurück zum Zitat Festa, C., De Marino, S., D’Auria, M. V., Bifulco, G., Renga, B., Fiorucci, S., et al. (2011). Solomonsterols A and B from Theonella swinhoei. The first example of C-24 and C-23 sulfated sterols from a marine source endowed with a PXR agonistic activity. Journal of Medicinal Chemistry, 54(1), 401–405, https://doi.org/10.1021/jm100968b. Festa, C., De Marino, S., D’Auria, M. V., Bifulco, G., Renga, B., Fiorucci, S., et al. (2011). Solomonsterols A and B from Theonella swinhoei. The first example of C-24 and C-23 sulfated sterols from a marine source endowed with a PXR agonistic activity. Journal of Medicinal Chemistry, 54(1), 401–405, https://​doi.​org/​10.​1021/​jm100968b.
421.
Zurück zum Zitat Sepe, V., Ummarino, R., D’Auria, M. V., Mencarelli, A., D’Amore, C., Renga, B., et al. (2011). Total synthesis and pharmacological characterization of solomonsterol A, a potent marine pregnane-X-receptor agonist endowed with anti-inflammatory activity. Journal of Medicinal Chemistry, 54(13), 4590–4599. https://doi.org/10.1021/jm200241sCrossRefPubMed Sepe, V., Ummarino, R., D’Auria, M. V., Mencarelli, A., D’Amore, C., Renga, B., et al. (2011). Total synthesis and pharmacological characterization of solomonsterol A, a potent marine pregnane-X-receptor agonist endowed with anti-inflammatory activity. Journal of Medicinal Chemistry, 54(13), 4590–4599. https://​doi.​org/​10.​1021/​jm200241sCrossRefPubMed
422.
Zurück zum Zitat Sepe, V., Ummarino, R., D'Auria, M. V., Lauro, G., Bifulco, G., D'Amore, C., et al. (2012). Modification in the side chain of solomonsterol A: discovery of cholestan disulfate as a potent pregnane-X-receptor agonist. Organic & Biomolecular Chemistry, 10(31), 6350-6362, doi:10.1039/c2ob25800e. Sepe, V., Ummarino, R., D'Auria, M. V., Lauro, G., Bifulco, G., D'Amore, C., et al. (2012). Modification in the side chain of solomonsterol A: discovery of cholestan disulfate as a potent pregnane-X-receptor agonist. Organic & Biomolecular Chemistry, 10(31), 6350-6362, doi:10.1039/c2ob25800e.
423.
Zurück zum Zitat De Marino, S., Ummarino, R., D’Auria, M. V., Chini, M. G., Bifulco, G., Renga, B., et al. (2011). Theonellasterols and conicasterols from Theonella swinhoei. Novel marine natural ligands for human nuclear receptors. Journal of Medicinal Chemistry, 54(8), 3065–3075, https://doi.org/10.1021/jm200169t. De Marino, S., Ummarino, R., D’Auria, M. V., Chini, M. G., Bifulco, G., Renga, B., et al. (2011). Theonellasterols and conicasterols from Theonella swinhoei. Novel marine natural ligands for human nuclear receptors. Journal of Medicinal Chemistry, 54(8), 3065–3075, https://​doi.​org/​10.​1021/​jm200169t.
424.
Zurück zum Zitat Renga, B., Mencarelli, A., D’Amore, C., Cipriani, S., D’Auria, M. V., Sepe, V., et al. (2012). Discovery that theonellasterol a marine sponge sterol is a highly selective FXR antagonist that protects against liver injury in cholestasis. PloS One, 7(1), ARTN e30443. https://doi.org/10.1371/journal.pone.0030443. Renga, B., Mencarelli, A., D’Amore, C., Cipriani, S., D’Auria, M. V., Sepe, V., et al. (2012). Discovery that theonellasterol a marine sponge sterol is a highly selective FXR antagonist that protects against liver injury in cholestasis. PloS One, 7(1), ARTN e30443. https://​doi.​org/​10.​1371/​journal.​pone.​0030443.
426.
Zurück zum Zitat Sepe, V., Ummarino, R., D’Auria, M. V., Chini, M. G., Bifulco, G., Renga, B., et al. (2012). Conicasterol E, a small heterodimer partner sparing farnesoid X receptor modulator endowed with a pregnane X receptor agonistic activity, from the marine sponge Theonella swinhoei. Journal of Medicinal Chemistry, 55(1), 84–93. https://doi.org/10.1021/jm201004pCrossRefPubMed Sepe, V., Ummarino, R., D’Auria, M. V., Chini, M. G., Bifulco, G., Renga, B., et al. (2012). Conicasterol E, a small heterodimer partner sparing farnesoid X receptor modulator endowed with a pregnane X receptor agonistic activity, from the marine sponge Theonella swinhoei. Journal of Medicinal Chemistry, 55(1), 84–93. https://​doi.​org/​10.​1021/​jm201004pCrossRefPubMed
427.
Zurück zum Zitat Chini, M. G., Jones, C. R., Zampella, A., D’Auria, M. V., Renga, B., Fiorucci, S., et al. (2012). Quantitative NMR-derived interproton distances combined with quantum mechanical calculations of 13C chemical shifts in the stereochemical determination of conicasterol F, a nuclear receptor ligand from Theonella swinhoei. Journal of Organic Chemistry, 77(3), 1489–1496. https://doi.org/10.1021/jo2023763CrossRefPubMed Chini, M. G., Jones, C. R., Zampella, A., D’Auria, M. V., Renga, B., Fiorucci, S., et al. (2012). Quantitative NMR-derived interproton distances combined with quantum mechanical calculations of 13C chemical shifts in the stereochemical determination of conicasterol F, a nuclear receptor ligand from Theonella swinhoei. Journal of Organic Chemistry, 77(3), 1489–1496. https://​doi.​org/​10.​1021/​jo2023763CrossRefPubMed
428.
Zurück zum Zitat De Marino, S., Sepe, V., D'Auria, M. V., Bifulco, G., Renga, B., Petek, S., et al. (2011). Towards new ligands of nuclear receptors. Discovery of malaitasterol A, an unique bis-secosterol from marine sponge Theonella swinhoei. Organic & Biomolecular Chemistry, 9(13), 4856–4862, https://doi.org/10.1039/c1ob05378g. De Marino, S., Sepe, V., D'Auria, M. V., Bifulco, G., Renga, B., Petek, S., et al. (2011). Towards new ligands of nuclear receptors. Discovery of malaitasterol A, an unique bis-secosterol from marine sponge Theonella swinhoei. Organic & Biomolecular Chemistry, 9(13), 4856–4862, https://​doi.​org/​10.​1039/​c1ob05378g.
431.
Zurück zum Zitat Festa, C., Lauro, G., De Marino, S., D’Auria, M. V., Monti, M. C., Casapullo, A., et al. (2012). Plakilactones from the marine sponge Plakinastrella mamillaris. Discovery of a new class of marine ligands of peroxisome proliferator-activated receptor γ. Journal of Medicinal Chemistry, 55(19), 8303–8317. Festa, C., Lauro, G., De Marino, S., D’Auria, M. V., Monti, M. C., Casapullo, A., et al. (2012). Plakilactones from the marine sponge Plakinastrella mamillaris. Discovery of a new class of marine ligands of peroxisome proliferator-activated receptor γ. Journal of Medicinal Chemistry, 55(19), 8303–8317.
434.
Zurück zum Zitat Meimetis, L. G., Williams, D. E., Mawji, N. R., Banuelos, C. A., Lal, A. A., Park, J. J., et al. (2012). Niphatenones, glycerol ethers from the sponge Niphates digitalis block androgen receptor transcriptional activity in prostate cancer cells: Structure elucidation, synthesis, and biological activity. Journal of Medicinal Chemistry, 55(1), 503–514. https://doi.org/10.1021/jm2014056CrossRefPubMed Meimetis, L. G., Williams, D. E., Mawji, N. R., Banuelos, C. A., Lal, A. A., Park, J. J., et al. (2012). Niphatenones, glycerol ethers from the sponge Niphates digitalis block androgen receptor transcriptional activity in prostate cancer cells: Structure elucidation, synthesis, and biological activity. Journal of Medicinal Chemistry, 55(1), 503–514. https://​doi.​org/​10.​1021/​jm2014056CrossRefPubMed
435.
Zurück zum Zitat Wang, S. S., Wang, Z., Lin, S. C., Zheng, W. L., Wang, R., Jin, S. K., et al. (2012). Revealing a natural marine product as a novel agonist for retinoic acid receptors with a unique binding mode and inhibitory effects on cancer cells. Biochemical Journal, 446, 79–87. https://doi.org/10.1042/Bj20120726CrossRefPubMed Wang, S. S., Wang, Z., Lin, S. C., Zheng, W. L., Wang, R., Jin, S. K., et al. (2012). Revealing a natural marine product as a novel agonist for retinoic acid receptors with a unique binding mode and inhibitory effects on cancer cells. Biochemical Journal, 446, 79–87. https://​doi.​org/​10.​1042/​Bj20120726CrossRefPubMed
436.
Zurück zum Zitat Di Leva, F. S., Festa, C., D’Amore, C., De Marino, S., Renga, B., D’Auria, M. V., et al. (2013). Binding mechanism of the farnesoid X receptor marine antagonist suvanine reveals a strategy to forestall drug modulation on nuclear receptors. Design, synthesis, and biological evaluation of novel ligands. Journal of Medicinal Chemistry, 56(11), 4701–4717, https://doi.org/10.1021/jm400419e. Di Leva, F. S., Festa, C., D’Amore, C., De Marino, S., Renga, B., D’Auria, M. V., et al. (2013). Binding mechanism of the farnesoid X receptor marine antagonist suvanine reveals a strategy to forestall drug modulation on nuclear receptors. Design, synthesis, and biological evaluation of novel ligands. Journal of Medicinal Chemistry, 56(11), 4701–4717, https://​doi.​org/​10.​1021/​jm400419e.
438.
Zurück zum Zitat Wu, Y., Liao, H., Liu, L.-Y., Sun, F., Chen, H.-F., Jiao, W.-H., et al. (2020). Phakefustatins A-C: Kynurenine-bearing cycloheptapeptides as RXRα modulators from the marine sponge Phakellia fusca. Organic Letters, 22(17), 6703–6708.PubMedCrossRef Wu, Y., Liao, H., Liu, L.-Y., Sun, F., Chen, H.-F., Jiao, W.-H., et al. (2020). Phakefustatins A-C: Kynurenine-bearing cycloheptapeptides as RXRα modulators from the marine sponge Phakellia fusca. Organic Letters, 22(17), 6703–6708.PubMedCrossRef
439.
Zurück zum Zitat Machida, K., Abe, T., Arai, D., Okamoto, M., Shimizu, I., de Voogd, N. J., et al. (2014). Cinanthrenol A, an estrogenic steroid containing phenanthrene nucleus, from a marine sponge Cinachyrella sp. Organic Letters, 16(6), 1539–1541.PubMedCrossRef Machida, K., Abe, T., Arai, D., Okamoto, M., Shimizu, I., de Voogd, N. J., et al. (2014). Cinanthrenol A, an estrogenic steroid containing phenanthrene nucleus, from a marine sponge Cinachyrella sp. Organic Letters, 16(6), 1539–1541.PubMedCrossRef
444.
Zurück zum Zitat Mensah-Osman, E., Lin, H.-L., Reinke, D., Hollenberg, P., & Baker, L. (2005). Ecteinascidin-743 is a potent inhibitor of P450 3A4 enzyme and accumulates cytoplasmic PXR to inhibit transcription of P450 3A4 and MDR1: Implications for the enhancement of cytotoxicity to chemotherapeutic agents in osteosarcoma. Journal of Clinical Oncology, 23(16_suppl), 9026–9026. Mensah-Osman, E., Lin, H.-L., Reinke, D., Hollenberg, P., & Baker, L. (2005). Ecteinascidin-743 is a potent inhibitor of P450 3A4 enzyme and accumulates cytoplasmic PXR to inhibit transcription of P450 3A4 and MDR1: Implications for the enhancement of cytotoxicity to chemotherapeutic agents in osteosarcoma. Journal of Clinical Oncology, 23(16_suppl), 9026–9026.
446.
Zurück zum Zitat Sepe, V., Bifulco, G., Renga, B., D’Amore, C., Fiorucci, S., & Zampella, A. (2011). Discovery of sulfated sterols from marine invertebrates as a new class of marine natural antagonists of farnesoid-X-receptor. Journal of Medicinal Chemistry, 54(5), 1314–1320. https://doi.org/10.1021/jm101336mCrossRefPubMed Sepe, V., Bifulco, G., Renga, B., D’Amore, C., Fiorucci, S., & Zampella, A. (2011). Discovery of sulfated sterols from marine invertebrates as a new class of marine natural antagonists of farnesoid-X-receptor. Journal of Medicinal Chemistry, 54(5), 1314–1320. https://​doi.​org/​10.​1021/​jm101336mCrossRefPubMed
447.
452.
Zurück zum Zitat Shiraki, T., Kamiya, N., Shiki, S., Kodama, T. S., Kakizuka, A., & Jingami, H. (2005). α, β-unsaturated ketone is a core moiety of natural ligands for covalent binding to peroxisome proliferator-activated receptor γ. Journal of Biological Chemistry, 280(14), 14145–14153.PubMedCrossRef Shiraki, T., Kamiya, N., Shiki, S., Kodama, T. S., Kakizuka, A., & Jingami, H. (2005). α, β-unsaturated ketone is a core moiety of natural ligands for covalent binding to peroxisome proliferator-activated receptor γ. Journal of Biological Chemistry, 280(14), 14145–14153.PubMedCrossRef
453.
Zurück zum Zitat Shappell, S. B., Gupta, R. A., Manning, S., Whitehead, R., Boeglin, W. E., Schneider, C., et al. (2001). 15S-Hydroxyeicosatetraenoic acid activates peroxisome proliferator-activated receptor gamma and inhibits proliferation in PC3 prostate carcinoma cells. Cancer Research, 61(2), 497–503.PubMed Shappell, S. B., Gupta, R. A., Manning, S., Whitehead, R., Boeglin, W. E., Schneider, C., et al. (2001). 15S-Hydroxyeicosatetraenoic acid activates peroxisome proliferator-activated receptor gamma and inhibits proliferation in PC3 prostate carcinoma cells. Cancer Research, 61(2), 497–503.PubMed
456.
Zurück zum Zitat Kinjo, J., Tsuchihashi, R., Morito, K., Hirose, T., Aomori, T., Nagao, T., et al. (2004). Interactions of phytoestrogens with estrogen receptors alpha and beta (III). Estrogenic activities of soy isoflavone aglycones and their metabolites isolated from human urine. Biological & Pharmaceutical Bulletin, 27(2), 185–188, https://doi.org/10.1248/bpb.27.185. Kinjo, J., Tsuchihashi, R., Morito, K., Hirose, T., Aomori, T., Nagao, T., et al. (2004). Interactions of phytoestrogens with estrogen receptors alpha and beta (III). Estrogenic activities of soy isoflavone aglycones and their metabolites isolated from human urine. Biological & Pharmaceutical Bulletin, 27(2), 185–188, https://​doi.​org/​10.​1248/​bpb.​27.​185.
457.
Zurück zum Zitat Maggiora, M., Bologna, M., Ceru, M. P., Possati, L., Angelucci, A., Cimini, A., et al. (2004). An overview of the effect of linoleic and conjugated-linoleic acids on the growth of several human tumor cell lines. International Journal of Cancer, 112(6), 909–919. https://doi.org/10.1002/ijc.20519CrossRefPubMed Maggiora, M., Bologna, M., Ceru, M. P., Possati, L., Angelucci, A., Cimini, A., et al. (2004). An overview of the effect of linoleic and conjugated-linoleic acids on the growth of several human tumor cell lines. International Journal of Cancer, 112(6), 909–919. https://​doi.​org/​10.​1002/​ijc.​20519CrossRefPubMed
459.
Zurück zum Zitat Evans, N. P., Misyak, S. A., Schmelz, E. M., Guri, A. J., Hontecillas, R., & Bassaganya-Riera, J. (2010). Conjugated linoleic acid ameliorates inflammation-induced colorectal cancer in mice through activation of PPARγ. The Journal of Nutrition, 140(3), 515–521.PubMedPubMedCentralCrossRef Evans, N. P., Misyak, S. A., Schmelz, E. M., Guri, A. J., Hontecillas, R., & Bassaganya-Riera, J. (2010). Conjugated linoleic acid ameliorates inflammation-induced colorectal cancer in mice through activation of PPARγ. The Journal of Nutrition, 140(3), 515–521.PubMedPubMedCentralCrossRef
467.
Zurück zum Zitat Smolarek, A. K., So, J. Y., Burgess, B., Kong, A.-N.T., Reuhl, K., Lin, Y., et al. (2012). Dietary administration of δ-and γ-tocopherol inhibits tumorigenesis in the animal model of estrogen receptor–positive, but not HER-2 breast cancer. Cancer Prevention Research, 5(11), 1310–1320.PubMedCrossRef Smolarek, A. K., So, J. Y., Burgess, B., Kong, A.-N.T., Reuhl, K., Lin, Y., et al. (2012). Dietary administration of δ-and γ-tocopherol inhibits tumorigenesis in the animal model of estrogen receptor–positive, but not HER-2 breast cancer. Cancer Prevention Research, 5(11), 1310–1320.PubMedCrossRef
468.
Zurück zum Zitat Smolarek, A. K., So, J. Y., Thomas, P. E., Lee, H. J., Paul, S., Dombrowski, A., et al. (2013). Dietary tocopherols inhibit cell proliferation, regulate expression of ERalpha, PPARgamma, and Nrf2, and decrease serum inflammatory markers during the development of mammary hyperplasia. Molecular Carcinogenesis, 52(7), 514–525. https://doi.org/10.1002/mc.21886CrossRefPubMed Smolarek, A. K., So, J. Y., Thomas, P. E., Lee, H. J., Paul, S., Dombrowski, A., et al. (2013). Dietary tocopherols inhibit cell proliferation, regulate expression of ERalpha, PPARgamma, and Nrf2, and decrease serum inflammatory markers during the development of mammary hyperplasia. Molecular Carcinogenesis, 52(7), 514–525. https://​doi.​org/​10.​1002/​mc.​21886CrossRefPubMed
469.
Zurück zum Zitat Yang, L., Yuan, J., Liu, L., Shi, C., Wang, L., Tian, F., et al. (2013). α-linolenic acid inhibits human renal cell carcinoma cell proliferation through PPAR-γ activation and COX-2 inhibition. Oncology Letters, 6(1), 197–202.PubMedPubMedCentralCrossRef Yang, L., Yuan, J., Liu, L., Shi, C., Wang, L., Tian, F., et al. (2013). α-linolenic acid inhibits human renal cell carcinoma cell proliferation through PPAR-γ activation and COX-2 inhibition. Oncology Letters, 6(1), 197–202.PubMedPubMedCentralCrossRef
471.
Zurück zum Zitat Fang, H., Zhang, J., Ao, M., He, F., Chen, W., Qian, Y., et al. (2020). Synthesis and discovery of ω-3 polyunsaturated fatty acid-alkanolamine (PUFA-AA) derivatives as anti-inflammatory agents targeting Nur77. Bioorganic Chemistry, 105, 104456.PubMedCrossRef Fang, H., Zhang, J., Ao, M., He, F., Chen, W., Qian, Y., et al. (2020). Synthesis and discovery of ω-3 polyunsaturated fatty acid-alkanolamine (PUFA-AA) derivatives as anti-inflammatory agents targeting Nur77. Bioorganic Chemistry, 105, 104456.PubMedCrossRef
472.
Zurück zum Zitat Tabata, Y., Iizuka, Y., Kashiwa, J., Masuda, N. T., Shinei, R., Kurihara, K.-I., et al. (2001). Fungal metabolites, PF1092 compounds and their derivatives, are nonsteroidal and selective progesterone receptor modulators. European Journal of Pharmacology, 430(2–3), 159–165.PubMedCrossRef Tabata, Y., Iizuka, Y., Kashiwa, J., Masuda, N. T., Shinei, R., Kurihara, K.-I., et al. (2001). Fungal metabolites, PF1092 compounds and their derivatives, are nonsteroidal and selective progesterone receptor modulators. European Journal of Pharmacology, 430(2–3), 159–165.PubMedCrossRef
473.
Zurück zum Zitat Muthyala, R. S., Ju, Y. H., Sheng, S. B., Williams, L. D., Doerge, D. R., Katzenellenbogen, B. S., et al. (2004). Equol, a natural estrogenic metabolite from soy isoflavones: Convenient preparation and resolution of R- and S-equols and their differing binding and biological activity through estrogen receptors alpha and beta. Bioorganic & Medicinal Chemistry, 12(6), 1559–1567. https://doi.org/10.1016/j.bmc.2003.11.035CrossRef Muthyala, R. S., Ju, Y. H., Sheng, S. B., Williams, L. D., Doerge, D. R., Katzenellenbogen, B. S., et al. (2004). Equol, a natural estrogenic metabolite from soy isoflavones: Convenient preparation and resolution of R- and S-equols and their differing binding and biological activity through estrogen receptors alpha and beta. Bioorganic & Medicinal Chemistry, 12(6), 1559–1567. https://​doi.​org/​10.​1016/​j.​bmc.​2003.​11.​035CrossRef
474.
Zurück zum Zitat Zheng, W., Zhang, Y., Ma, D., Li, G., & Wang, P. (2011). Anti-invasion effects of R- and S-enantiomers of equol on prostate cancer PC3, DU145 cells. Wei Sheng Yan Jiu, 40(4), 423–425, 430. Zheng, W., Zhang, Y., Ma, D., Li, G., & Wang, P. (2011). Anti-invasion effects of R- and S-enantiomers of equol on prostate cancer PC3, DU145 cells. Wei Sheng Yan Jiu, 40(4), 423–425, 430.
475.
Zurück zum Zitat Kelly, D., Campbell, J. I., King, T. P., Grant, G., Jansson, E. A., Coutts, A. G., et al. (2004). Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nature Immunology, 5(1), 104–112. https://doi.org/10.1038/ni1018CrossRefPubMed Kelly, D., Campbell, J. I., King, T. P., Grant, G., Jansson, E. A., Coutts, A. G., et al. (2004). Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nature Immunology, 5(1), 104–112. https://​doi.​org/​10.​1038/​ni1018CrossRefPubMed
476.
Zurück zum Zitat Lee, S. K., Kim, H. J., Chi, S. G., Jang, J. Y., Nam, K. D., Kim, N. H., et al. (2005). Saccharomyces boulardii activates expression of peroxisome proliferator-activated receptor-gamma in HT-29 cells. The Korean Journal of Gastroenterology, 45(5), 328–334.PubMed Lee, S. K., Kim, H. J., Chi, S. G., Jang, J. Y., Nam, K. D., Kim, N. H., et al. (2005). Saccharomyces boulardii activates expression of peroxisome proliferator-activated receptor-gamma in HT-29 cells. The Korean Journal of Gastroenterology, 45(5), 328–334.PubMed
479.
Zurück zum Zitat Moon, H. S., Lim, H., Moon, S., Oh, H. L., Kim, Y. T., Kim, M. K., et al. (2009). Benzyldihydroxyoctenone, a novel anticancer agent, induces apoptosis via mitochondrial-mediated pathway in androgen-sensitive LNCaP prostate cancer cells. Bioorganic & Medicinal Chemistry Letters, 19(3), 742–744. https://doi.org/10.1016/j.bmcl.2008.12.029CrossRef Moon, H. S., Lim, H., Moon, S., Oh, H. L., Kim, Y. T., Kim, M. K., et al. (2009). Benzyldihydroxyoctenone, a novel anticancer agent, induces apoptosis via mitochondrial-mediated pathway in androgen-sensitive LNCaP prostate cancer cells. Bioorganic & Medicinal Chemistry Letters, 19(3), 742–744. https://​doi.​org/​10.​1016/​j.​bmcl.​2008.​12.​029CrossRef
481.
Zurück zum Zitat Meng, J., Fan, Y., Su, M., Chen, C., Ren, T., Wang, J., et al. (2014). WLIP derived from Lasiosphaera fenzlii Reich exhibits anti-tumor activity and induces cell cycle arrest through PPAR-γ associated pathways. International Immunopharmacology, 19(1), 37–44.PubMedCrossRef Meng, J., Fan, Y., Su, M., Chen, C., Ren, T., Wang, J., et al. (2014). WLIP derived from Lasiosphaera fenzlii Reich exhibits anti-tumor activity and induces cell cycle arrest through PPAR-γ associated pathways. International Immunopharmacology, 19(1), 37–44.PubMedCrossRef
484.
Zurück zum Zitat Khan, I., Huang, G., Li, X.-A., Liao, W., Leong, W. K., Xia, W., et al. (2019). Mushroom polysaccharides and jiaogulan saponins exert cancer preventive effects by shaping the gut microbiota and microenvironment in ApcMin/+ mice. Pharmacological Research, 148, 104448.PubMedCrossRef Khan, I., Huang, G., Li, X.-A., Liao, W., Leong, W. K., Xia, W., et al. (2019). Mushroom polysaccharides and jiaogulan saponins exert cancer preventive effects by shaping the gut microbiota and microenvironment in ApcMin/+ mice. Pharmacological Research, 148, 104448.PubMedCrossRef
486.
Zurück zum Zitat Zhang, X., Li, T., Liu, S., Xu, Y., Meng, M., Li, X., et al. (2020). beta-glucan from Lentinus edodes inhibits breast cancer progression via the Nur77/HIF-1alpha axis. Bioscience Reports, 40(12). 10.1042/BSR20201006. Zhang, X., Li, T., Liu, S., Xu, Y., Meng, M., Li, X., et al. (2020). beta-glucan from Lentinus edodes inhibits breast cancer progression via the Nur77/HIF-1alpha axis. Bioscience Reports, 40(12). 10.1042/BSR20201006.
489.
Zurück zum Zitat Meng, L., Feng, B., Tao, H., Yang, T., Meng, Y., Zhu, W., et al. (2011). A novel antioestrogen agent (3R,6R)-bassiatin inhibits cell proliferation and cell cycle progression by repressing cyclin D1 expression in 17beta-oestradiol-treated MCF-7 cells. Cell Biology International, 35(6), 599–605. https://doi.org/10.1042/CBI20100765CrossRefPubMed Meng, L., Feng, B., Tao, H., Yang, T., Meng, Y., Zhu, W., et al. (2011). A novel antioestrogen agent (3R,6R)-bassiatin inhibits cell proliferation and cell cycle progression by repressing cyclin D1 expression in 17beta-oestradiol-treated MCF-7 cells. Cell Biology International, 35(6), 599–605. https://​doi.​org/​10.​1042/​CBI20100765CrossRefPubMed
491.
Zurück zum Zitat Nepelska, M., de Wouters, T., Jacouton, E., Beguet-Crespel, F., Lapaque, N., Dore, J., et al. (2017). Commensal gut bacteria modulate phosphorylation-dependent PPARgamma transcriptional activity in human intestinal epithelial cells. Science and Reports, 7, 43199. https://doi.org/10.1038/srep43199CrossRef Nepelska, M., de Wouters, T., Jacouton, E., Beguet-Crespel, F., Lapaque, N., Dore, J., et al. (2017). Commensal gut bacteria modulate phosphorylation-dependent PPARgamma transcriptional activity in human intestinal epithelial cells. Science and Reports, 7, 43199. https://​doi.​org/​10.​1038/​srep43199CrossRef
495.
Zurück zum Zitat Sari, Z., Miko, E., Kovacs, T., Janko, L., Csonka, T., Lente, G., et al. (2020). Indolepropionic acid, a metabolite of the microbiome, has cytostatic properties in breast cancer by activating AHR and PXR receptors and inducing oxidative stress. Cancers, 12(9), ARTN 2411. https://doi.org/10.3390/cancers12092411. Sari, Z., Miko, E., Kovacs, T., Janko, L., Csonka, T., Lente, G., et al. (2020). Indolepropionic acid, a metabolite of the microbiome, has cytostatic properties in breast cancer by activating AHR and PXR receptors and inducing oxidative stress. Cancers, 12(9), ARTN 2411. https://​doi.​org/​10.​3390/​cancers12092411.
498.
Zurück zum Zitat Liang, Z., Gu, T., Wang, J., She, J., Ye, Y., Cao, W., et al. (2021). Chromene and chromone derivatives as liver X receptors modulators from a marine-derived Pestalotiopsis neglecta fungus. Bioorganic Chemistry, 112, 104927.PubMedCrossRef Liang, Z., Gu, T., Wang, J., She, J., Ye, Y., Cao, W., et al. (2021). Chromene and chromone derivatives as liver X receptors modulators from a marine-derived Pestalotiopsis neglecta fungus. Bioorganic Chemistry, 112, 104927.PubMedCrossRef
509.
Zurück zum Zitat Calcabrini, C., Catanzaro, E., Bishayee, A., Turrini, E., & Fimognari, C. (2017). Marine sponge natural products with anticancer potential: An updated review. Marine Drugs, 15(10), 310.PubMedPubMedCentralCrossRef Calcabrini, C., Catanzaro, E., Bishayee, A., Turrini, E., & Fimognari, C. (2017). Marine sponge natural products with anticancer potential: An updated review. Marine Drugs, 15(10), 310.PubMedPubMedCentralCrossRef
511.
Zurück zum Zitat El Ouar, I., Braicu, C., Naimi, D., Irimie, A., & Berindan-Neagoe, I. (2017). Effect of Helix aspersa extract on TNFα, NF-κB and some tumor suppressor genes in breast cancer cell line Hs578T. Pharmacognosy Magazine, 13(50), 281.PubMedPubMedCentralCrossRef El Ouar, I., Braicu, C., Naimi, D., Irimie, A., & Berindan-Neagoe, I. (2017). Effect of Helix aspersa extract on TNFα, NF-κB and some tumor suppressor genes in breast cancer cell line Hs578T. Pharmacognosy Magazine, 13(50), 281.PubMedPubMedCentralCrossRef
512.
Zurück zum Zitat Jeyamogan, S., Khan, N. A., & Siddiqui, R. (2017). Animals living in polluted environments are a potential source of anti-tumor molecule(s). Cancer Chemotherapy and Pharmacology, 80(5), 919–924.PubMedCrossRef Jeyamogan, S., Khan, N. A., & Siddiqui, R. (2017). Animals living in polluted environments are a potential source of anti-tumor molecule(s). Cancer Chemotherapy and Pharmacology, 80(5), 919–924.PubMedCrossRef
515.
Zurück zum Zitat Ramesh, C., Tulasi, B. R., Raju, M., Thakur, N., & Dufossé, L. (2021). Marine natural products from tunicates and their associated microbes. Marine Drugs, 19(6), 308.PubMedPubMedCentralCrossRef Ramesh, C., Tulasi, B. R., Raju, M., Thakur, N., & Dufossé, L. (2021). Marine natural products from tunicates and their associated microbes. Marine Drugs, 19(6), 308.PubMedPubMedCentralCrossRef
516.
Zurück zum Zitat Waheed, M., Hussain, M. B., Javed, A., Mushtaq, Z., Hassan, S., Shariati, M. A., et al. (2019). Honey and cancer: A mechanistic review. Clinical Nutrition, 38(6), 2499–2503.PubMedCrossRef Waheed, M., Hussain, M. B., Javed, A., Mushtaq, Z., Hassan, S., Shariati, M. A., et al. (2019). Honey and cancer: A mechanistic review. Clinical Nutrition, 38(6), 2499–2503.PubMedCrossRef
517.
Zurück zum Zitat Arung, E. T., Ramadhan, R., Khairunnisa, B., Amen, Y., Matsumoto, M., Nagata, M., et al. (2021). Cytotoxicity effect of honey, bee pollen, and propolis from seven stingless bees in some cancer cell lines. Saudi Journal of Biological Sciences, 28(12), 7182–7189.PubMedPubMedCentralCrossRef Arung, E. T., Ramadhan, R., Khairunnisa, B., Amen, Y., Matsumoto, M., Nagata, M., et al. (2021). Cytotoxicity effect of honey, bee pollen, and propolis from seven stingless bees in some cancer cell lines. Saudi Journal of Biological Sciences, 28(12), 7182–7189.PubMedPubMedCentralCrossRef
518.
Zurück zum Zitat Ding, J., Chua, P.-J., Bay, B.-H., & Gopalakrishnakone, P. (2014). Scorpion venoms as a potential source of novel cancer therapeutic compounds. Experimental Biology and Medicine, 239(4), 387–393.PubMedCrossRef Ding, J., Chua, P.-J., Bay, B.-H., & Gopalakrishnakone, P. (2014). Scorpion venoms as a potential source of novel cancer therapeutic compounds. Experimental Biology and Medicine, 239(4), 387–393.PubMedCrossRef
519.
Zurück zum Zitat Sarfo-Poku, C., Eshun, O., & Lee, K. H. (2016). Medical application of scorpion venom to breast cancer: A mini-review. Toxicon, 122, 109–112.PubMedCrossRef Sarfo-Poku, C., Eshun, O., & Lee, K. H. (2016). Medical application of scorpion venom to breast cancer: A mini-review. Toxicon, 122, 109–112.PubMedCrossRef
520.
Zurück zum Zitat Lee, N., Spears, M. E., Carlisle, A. E., & Kim, D. (2020). Endogenous toxic metabolites and implications in cancer therapy. Oncogene, 39(35), 5709–5720.PubMedPubMedCentralCrossRef Lee, N., Spears, M. E., Carlisle, A. E., & Kim, D. (2020). Endogenous toxic metabolites and implications in cancer therapy. Oncogene, 39(35), 5709–5720.PubMedPubMedCentralCrossRef
522.
Zurück zum Zitat Pham, J. V., Yilma, M. A., Feliz, A., Majid, M. T., Maffetone, N., Walker, J. R., et al. (2019). A review of the microbial production of bioactive natural products and biologics. Frontiers in Microbiology, 10, 1404.PubMedPubMedCentralCrossRef Pham, J. V., Yilma, M. A., Feliz, A., Majid, M. T., Maffetone, N., Walker, J. R., et al. (2019). A review of the microbial production of bioactive natural products and biologics. Frontiers in Microbiology, 10, 1404.PubMedPubMedCentralCrossRef
523.
Zurück zum Zitat Sharma, M., Tuaine, J., McLaren, B., Waters, D. L., Black, K., Jones, L. M., et al. (2016). Chemotherapy agents alter plasma lipids in breast cancer patients and show differential effects on lipid metabolism genes in liver cells. Plos One, 11(1), ARTN e0148049. https://doi.org/10.1371/journal.pone.0148049. Sharma, M., Tuaine, J., McLaren, B., Waters, D. L., Black, K., Jones, L. M., et al. (2016). Chemotherapy agents alter plasma lipids in breast cancer patients and show differential effects on lipid metabolism genes in liver cells. Plos One, 11(1), ARTN e0148049. https://​doi.​org/​10.​1371/​journal.​pone.​0148049.
524.
Zurück zum Zitat Pirouzpanah, S., Asemani, S., Shayanfar, A., Baradaran, B., & Montazeri, V. (2019). The effects of Berberis vulgaris consumption on plasma levels of IGF-1, IGFBPs, PPAR-gamma and the expression of angiogenic genes in women with benign breast disease: A randomized controlled clinical trial. BMC Complementary and Alternative Medicine, 19(1), 324. https://doi.org/10.1186/s12906-019-2715-1CrossRefPubMedPubMedCentral Pirouzpanah, S., Asemani, S., Shayanfar, A., Baradaran, B., & Montazeri, V. (2019). The effects of Berberis vulgaris consumption on plasma levels of IGF-1, IGFBPs, PPAR-gamma and the expression of angiogenic genes in women with benign breast disease: A randomized controlled clinical trial. BMC Complementary and Alternative Medicine, 19(1), 324. https://​doi.​org/​10.​1186/​s12906-019-2715-1CrossRefPubMedPubMedCentral
525.
Zurück zum Zitat Komatsu, Y., Yoshino, T., Yamazaki, K., Yuki, S., Machida, N., Sasaki, T., et al. (2014). Phase 1 study of efatutazone, a novel oral peroxisome proliferator-activated receptor gamma agonist, in combination with FOLFIRI as second-line therapy in patients with metastatic colorectal cancer. Investigational New Drugs, 32(3), 473–480. https://doi.org/10.1007/s10637-013-0056-3CrossRefPubMed Komatsu, Y., Yoshino, T., Yamazaki, K., Yuki, S., Machida, N., Sasaki, T., et al. (2014). Phase 1 study of efatutazone, a novel oral peroxisome proliferator-activated receptor gamma agonist, in combination with FOLFIRI as second-line therapy in patients with metastatic colorectal cancer. Investigational New Drugs, 32(3), 473–480. https://​doi.​org/​10.​1007/​s10637-013-0056-3CrossRefPubMed
526.
Zurück zum Zitat Hamilton-Reeves, J. M., Rebello, S. A., Thomas, W., Slaton, J. W., & Kurzer, M. S. (2007). Isoflavone-rich soy protein isolate suppresses androgen receptor expression without altering estrogen receptor-beta expression or serum hormonal profiles in men at high risk of prostate cancer. Journal of Nutrition, 137(7), 1769–1775. https://doi.org/10.1093/jn/137.7.1769CrossRefPubMed Hamilton-Reeves, J. M., Rebello, S. A., Thomas, W., Slaton, J. W., & Kurzer, M. S. (2007). Isoflavone-rich soy protein isolate suppresses androgen receptor expression without altering estrogen receptor-beta expression or serum hormonal profiles in men at high risk of prostate cancer. Journal of Nutrition, 137(7), 1769–1775. https://​doi.​org/​10.​1093/​jn/​137.​7.​1769CrossRefPubMed
529.
Zurück zum Zitat Kwan, E. M., Spain, L., Anton, A., Gan, C. L., Garrett, L., Chang, D., et al. (2022). Avelumab combined with stereotactic ablative body radiotherapy in metastatic castration-resistant prostate cancer: The phase 2 ICE-PAC clinical trial. European Urology, 81(3), 253–262.PubMedCrossRef Kwan, E. M., Spain, L., Anton, A., Gan, C. L., Garrett, L., Chang, D., et al. (2022). Avelumab combined with stereotactic ablative body radiotherapy in metastatic castration-resistant prostate cancer: The phase 2 ICE-PAC clinical trial. European Urology, 81(3), 253–262.PubMedCrossRef
530.
Zurück zum Zitat Bailly, C. (2019). Irinotecan: 25 years of cancer treatment. Pharmacological Research, 148, 104398.PubMedCrossRef Bailly, C. (2019). Irinotecan: 25 years of cancer treatment. Pharmacological Research, 148, 104398.PubMedCrossRef
531.
Zurück zum Zitat Kciuk, M., Marciniak, B., & Kontek, R. (2020). Irinotecan—Still an important player in cancer chemotherapy: A comprehensive overview. International Journal of Molecular Sciences, 21(14), 4919.PubMedPubMedCentralCrossRef Kciuk, M., Marciniak, B., & Kontek, R. (2020). Irinotecan—Still an important player in cancer chemotherapy: A comprehensive overview. International Journal of Molecular Sciences, 21(14), 4919.PubMedPubMedCentralCrossRef
532.
Zurück zum Zitat Noda, K., Nishiwaki, Y., Kawahara, M., Negoro, S., Sugiura, T., Yokoyama, A., et al. (2002). Irinotecan plus cisplatin compared with etoposide plus cisplatin for extensive small-cell lung cancer. New England Journal of Medicine, 346(2), 85–91.PubMedCrossRef Noda, K., Nishiwaki, Y., Kawahara, M., Negoro, S., Sugiura, T., Yokoyama, A., et al. (2002). Irinotecan plus cisplatin compared with etoposide plus cisplatin for extensive small-cell lung cancer. New England Journal of Medicine, 346(2), 85–91.PubMedCrossRef
533.
Zurück zum Zitat Fuchs, C., Mitchell, E. P., & Hoff, P. M. (2006). Irinotecan in the treatment of colorectal cancer. Cancer Treatment Reviews, 32(7), 491–503.PubMedCrossRef Fuchs, C., Mitchell, E. P., & Hoff, P. M. (2006). Irinotecan in the treatment of colorectal cancer. Cancer Treatment Reviews, 32(7), 491–503.PubMedCrossRef
534.
Zurück zum Zitat Feldman, E., Kalaycio, M., Weiner, G., Frankel, S., Schulman, P., Schwartzberg, L., et al. (2003). Treatment of relapsed or refractory acute myeloid leukemia with humanized anti-CD33 monoclonal antibody HuM195. Leukemia, 17(2), 314–318.PubMedCrossRef Feldman, E., Kalaycio, M., Weiner, G., Frankel, S., Schulman, P., Schwartzberg, L., et al. (2003). Treatment of relapsed or refractory acute myeloid leukemia with humanized anti-CD33 monoclonal antibody HuM195. Leukemia, 17(2), 314–318.PubMedCrossRef
535.
Zurück zum Zitat Grillo-López, A. J., White, C. A., Varns, C., Shen, D., Wei, A., McClure, A., et al. Overview of the clinical development of rituximab: First monoclonal antibody approved for the treatment of lymphoma. In Seminars in Oncology, 1999 (Vol. 26, pp. 66–73, Vol. 5 Suppl 14) Grillo-López, A. J., White, C. A., Varns, C., Shen, D., Wei, A., McClure, A., et al. Overview of the clinical development of rituximab: First monoclonal antibody approved for the treatment of lymphoma. In Seminars in Oncology, 1999 (Vol. 26, pp. 66–73, Vol. 5 Suppl 14)
536.
Zurück zum Zitat Seiden, M., Burris, H., Matulonis, U., Hall, J., Armstrong, D., Speyer, J., et al. (2007). A phase II trial of EMD72000 (matuzumab), a humanized anti-EGFR monoclonal antibody, in patients with platinum-resistant ovarian and primary peritoneal malignancies. Gynecologic Oncology, 104(3), 727–731.PubMedCrossRef Seiden, M., Burris, H., Matulonis, U., Hall, J., Armstrong, D., Speyer, J., et al. (2007). A phase II trial of EMD72000 (matuzumab), a humanized anti-EGFR monoclonal antibody, in patients with platinum-resistant ovarian and primary peritoneal malignancies. Gynecologic Oncology, 104(3), 727–731.PubMedCrossRef
537.
Zurück zum Zitat Smaletz, O., Ismael, G., Estevez-Diz, M. D. P., Nascimento, I. L., de Morais, A. L. G., Cunha-Junior, G. F., et al. (2021). Phase II consolidation trial with anti-Lewis-Y monoclonal antibody (hu3S193) in platinum-sensitive ovarian cancer after a second remission. International Journal of Gynecologic Cancer, 31(4), 62–568. https://doi.org/10.1136/ijgc-2020-002239CrossRef Smaletz, O., Ismael, G., Estevez-Diz, M. D. P., Nascimento, I. L., de Morais, A. L. G., Cunha-Junior, G. F., et al. (2021). Phase II consolidation trial with anti-Lewis-Y monoclonal antibody (hu3S193) in platinum-sensitive ovarian cancer after a second remission. International Journal of Gynecologic Cancer, 31(4), 62–568. https://​doi.​org/​10.​1136/​ijgc-2020-002239CrossRef
539.
Zurück zum Zitat Hor, S., Lee, S., Wong, C., Lim, Y., Lim, R., Wang, L., et al. (2008). PXR, CAR and HNF4α genotypes and their association with pharmacokinetics and pharmacodynamics of docetaxel and doxorubicin in Asian patients. The Pharmacogenomics Journal, 8(2), 139–146.PubMedCrossRef Hor, S., Lee, S., Wong, C., Lim, Y., Lim, R., Wang, L., et al. (2008). PXR, CAR and HNF4α genotypes and their association with pharmacokinetics and pharmacodynamics of docetaxel and doxorubicin in Asian patients. The Pharmacogenomics Journal, 8(2), 139–146.PubMedCrossRef
540.
Zurück zum Zitat Chew, S.-C., Lim, J., Singh, O., Chen, X., Tan, E.-H., Lee, E.-J., et al. (2014). Pharmacogenetic effects of regulatory nuclear receptors (PXR, CAR, RXRα and HNF4α) on docetaxel disposition in Chinese nasopharyngeal cancer patients. European Journal of Clinical Pharmacology, 70(2), 155–166.PubMedCrossRef Chew, S.-C., Lim, J., Singh, O., Chen, X., Tan, E.-H., Lee, E.-J., et al. (2014). Pharmacogenetic effects of regulatory nuclear receptors (PXR, CAR, RXRα and HNF4α) on docetaxel disposition in Chinese nasopharyngeal cancer patients. European Journal of Clinical Pharmacology, 70(2), 155–166.PubMedCrossRef
541.
Zurück zum Zitat Deeken, J. F., Cormier, T., Price, D. K., Sissung, T. M., Steinberg, S. M., Tran, K., et al. (2010). A pharmacogenetic study of docetaxel and thalidomide in patients with castration-resistant prostate cancer using the DMET genotyping platform. Pharmacogenomics Journal, 10(3), 191–199. https://doi.org/10.1038/tpj.2009.57CrossRefPubMed Deeken, J. F., Cormier, T., Price, D. K., Sissung, T. M., Steinberg, S. M., Tran, K., et al. (2010). A pharmacogenetic study of docetaxel and thalidomide in patients with castration-resistant prostate cancer using the DMET genotyping platform. Pharmacogenomics Journal, 10(3), 191–199. https://​doi.​org/​10.​1038/​tpj.​2009.​57CrossRefPubMed
Metadaten
Titel
Natural compounds targeting nuclear receptors for effective cancer therapy
verfasst von
Mangala Hegde
Sosmitha Girisa
Nikunj Naliyadhara
Aviral Kumar
Mohammed S. Alqahtani
Mohamed Abbas
Chakrabhavi Dhananjaya Mohan
Sudha Warrier
Kam Man Hui
Kanchugarakoppal S. Rangappa
Gautam Sethi
Ajaikumar B. Kunnumakkara
Publikationsdatum
08.12.2022
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 3/2023
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-022-10068-w

Weitere Artikel der Ausgabe 3/2023

Cancer and Metastasis Reviews 3/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.