Skip to main content
Erschienen in: Cancer and Metastasis Reviews 1/2024

22.03.2024 | REVIEW

Dynamic interplay of nuclear receptors in tumor cell plasticity and drug resistance: Shifting gears in malignant transformations and applications in cancer therapeutics

verfasst von: Bandari BharathwajChetty, Anjana Sajeev, Ravichandran Vishwa, Babu Santha Aswani, Mohammed S. Alqahtani, Mohamed Abbas, Ajaikumar B. Kunnumakkara

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 1/2024

Einloggen, um Zugang zu erhalten

Abstract

Recent advances have brought forth the complex interplay between tumor cell plasticity and its consequential impact on drug resistance and tumor recurrence, both of which are critical determinants of neoplastic progression and therapeutic efficacy. Various forms of tumor cell plasticity, instrumental in facilitating neoplastic cells to develop drug resistance, include epithelial-mesenchymal transition (EMT) alternatively termed epithelial-mesenchymal plasticity, the acquisition of cancer stem cell (CSC) attributes, and transdifferentiation into diverse cell lineages. Nuclear receptors (NRs) are a superfamily of transcription factors (TFs) that play an essential role in regulating a multitude of cellular processes, including cell proliferation, differentiation, and apoptosis. NRs have been implicated to play a critical role in modulating gene expression associated with tumor cell plasticity and drug resistance. This review aims to provide a comprehensive overview of the current understanding of how NRs regulate these key aspects of cancer biology. We discuss the diverse mechanisms through which NRs influence tumor cell plasticity, including EMT, stemness, and metastasis. Further, we explore the intricate relationship between NRs and drug resistance, highlighting the impact of NR signaling on chemotherapy, radiotherapy and targeted therapies. We also discuss the emerging therapeutic strategies targeting NRs to overcome tumor cell plasticity and drug resistance. This review also provides valuable insights into the current clinical trials that involve agonists or antagonists of NRs modulating various aspects of tumor cell plasticity, thereby delineating the potential of NRs as therapeutic targets for improved cancer treatment outcomes.

Graphical Abstract

Literatur
2.
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.21660CrossRefPubMed 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.​21660CrossRefPubMed
21.
Zurück zum Zitat Dawood, S., Austin, L., & Cristofanilli, M. (2014). Cancer stem cells: implications for cancer therapy. Oncology (Williston Park, N.Y.), 28(12), 1101–1110. Dawood, S., Austin, L., & Cristofanilli, M. (2014). Cancer stem cells: implications for cancer therapy. Oncology (Williston Park, N.Y.), 28(12), 1101–1110.
36.
Zurück zum Zitat Kurrey, N. K., Jalgaonkar, S. P., Joglekar, A. V., Ghanate, A. D., Chaskar, P. D., Doiphode, R. Y., et al. (2009). Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells, 27(9), 2059–2068. https://doi.org/10.1002/stem.154CrossRefPubMed Kurrey, N. K., Jalgaonkar, S. P., Joglekar, A. V., Ghanate, A. D., Chaskar, P. D., Doiphode, R. Y., et al. (2009). Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells, 27(9), 2059–2068. https://​doi.​org/​10.​1002/​stem.​154CrossRefPubMed
38.
Zurück zum Zitat Battula, V. L., Evans, K. W., Hollier, B. G., Shi, Y., Marini, F. C., Ayyanan, A., et al. (2010). Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem Cells, 28(8), 1435–1445. https://doi.org/10.1002/stem.467CrossRefPubMed Battula, V. L., Evans, K. W., Hollier, B. G., Shi, Y., Marini, F. C., Ayyanan, A., et al. (2010). Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem Cells, 28(8), 1435–1445. https://​doi.​org/​10.​1002/​stem.​467CrossRefPubMed
46.
Zurück zum Zitat Kunnumakkara, A. B., Bordoloi, D., Harsha, C., Banik, K., Gupta, S. C., & Aggarwal, B. B. (2017). Curcumin mediates anticancer effects by modulating multiple cell signaling pathways. Clinical Science (London, England), 131(15), 1781–1799. https://doi.org/10.1042/CS20160935CrossRef Kunnumakkara, A. B., Bordoloi, D., Harsha, C., Banik, K., Gupta, S. C., & Aggarwal, B. B. (2017). Curcumin mediates anticancer effects by modulating multiple cell signaling pathways. Clinical Science (London, England), 131(15), 1781–1799. https://​doi.​org/​10.​1042/​CS20160935CrossRef
47.
Zurück zum Zitat Verma, E., Kumar, A., Daimary, U. D., Parama, D., Girisa, S., Sethi, G., et al. (2021). Potential of baicalein in the prevention and treatment of cancer: A scientometric analyses based review. Journal of Functional Foods, 86, 104660.CrossRef Verma, E., Kumar, A., Daimary, U. D., Parama, D., Girisa, S., Sethi, G., et al. (2021). Potential of baicalein in the prevention and treatment of cancer: A scientometric analyses based review. Journal of Functional Foods, 86, 104660.CrossRef
51.
Zurück zum Zitat Wang, L., Nanayakkara, G., Yang, Q., Tan, H., Drummer, C., Sun, Y., et al. (2017). A comprehensive data mining study shows that most nuclear receptors act as newly proposed homeostasis-associated molecular pattern receptors. Journal of Hematology & Oncology, 10(1), 168. https://doi.org/10.1186/s13045-017-0526-8CrossRef Wang, L., Nanayakkara, G., Yang, Q., Tan, H., Drummer, C., Sun, Y., et al. (2017). A comprehensive data mining study shows that most nuclear receptors act as newly proposed homeostasis-associated molecular pattern receptors. Journal of Hematology & Oncology, 10(1), 168. https://​doi.​org/​10.​1186/​s13045-017-0526-8CrossRef
65.
Zurück zum Zitat Manickasamy, M. K., Sajeev, A., Bharathwaj Chetty, B., Alqahtani, M. S., Abbas, M., Hegde, M., Aswani, B. S., Shakibaei, M., Sethi, G., & Kunnumakkara, A. B. (2024). Exploring the nexus of nuclear receptors in hematological malignancies. Cellular and Molecular Life Sciences: CMLS, 81(1), 78. https://doi.org/10.1007/s00018-023-05085-zCrossRefPubMed Manickasamy, M. K., Sajeev, A., Bharathwaj Chetty, B., Alqahtani, M. S., Abbas, M., Hegde, M., Aswani, B. S., Shakibaei, M., Sethi, G., & Kunnumakkara, A. B. (2024). Exploring the nexus of nuclear receptors in hematological malignancies. Cellular and Molecular Life Sciences: CMLS, 81(1), 78. https://​doi.​org/​10.​1007/​s00018-023-05085-zCrossRefPubMed
82.
Zurück zum Zitat Cano, A., Perez-Moreno, M. A., Rodrigo, I., Locascio, A., Blanco, M. J., del Barrio, M. G., et al. (2000). The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nature Cell Biology, 2(2), 76–83. https://doi.org/10.1038/35000025CrossRefPubMed Cano, A., Perez-Moreno, M. A., Rodrigo, I., Locascio, A., Blanco, M. J., del Barrio, M. G., et al. (2000). The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nature Cell Biology, 2(2), 76–83. https://​doi.​org/​10.​1038/​35000025CrossRefPubMed
99.
Zurück zum Zitat Chua, H. L., Bhat-Nakshatri, P., Clare, S. E., Morimiya, A., Badve, S., & Nakshatri, H. (2007). NF-kappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: Potential involvement of ZEB-1 and ZEB-2. Oncogene, 26(5), 711–724. https://doi.org/10.1038/sj.onc.1209808CrossRefPubMed Chua, H. L., Bhat-Nakshatri, P., Clare, S. E., Morimiya, A., Badve, S., & Nakshatri, H. (2007). NF-kappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: Potential involvement of ZEB-1 and ZEB-2. Oncogene, 26(5), 711–724. https://​doi.​org/​10.​1038/​sj.​onc.​1209808CrossRefPubMed
103.
Zurück zum Zitat Upadhyay, S. K., Verone, A., Shoemaker, S., Qin, M., Liu, S., Campbell, M., et al. (2013). 1,25-Dihydroxyvitamin D3 (1,25(OH)2D3) Signaling capacity and the epithelial-mesenchymal transition in non-small cell lung cancer (NSCLC): Implications for use of 1,25(OH)2D3 in NSCLC treatment. Cancers (Basel), 5(4), 1504–1521. https://doi.org/10.3390/cancers5041504CrossRefPubMed Upadhyay, S. K., Verone, A., Shoemaker, S., Qin, M., Liu, S., Campbell, M., et al. (2013). 1,25-Dihydroxyvitamin D3 (1,25(OH)2D3) Signaling capacity and the epithelial-mesenchymal transition in non-small cell lung cancer (NSCLC): Implications for use of 1,25(OH)2D3 in NSCLC treatment. Cancers (Basel), 5(4), 1504–1521. https://​doi.​org/​10.​3390/​cancers5041504CrossRefPubMed
106.
Zurück zum Zitat Palmer, H. G., Larriba, M. J., Garcia, J. M., Ordonez-Moran, P., Pena, C., Peiro, S., et al. (2004). The transcription factor SNAIL represses vitamin D receptor expression and responsiveness in human colon cancer. Nature Medicine, 10(9), 917–919. https://doi.org/10.1038/nm1095CrossRefPubMed Palmer, H. G., Larriba, M. J., Garcia, J. M., Ordonez-Moran, P., Pena, C., Peiro, S., et al. (2004). The transcription factor SNAIL represses vitamin D receptor expression and responsiveness in human colon cancer. Nature Medicine, 10(9), 917–919. https://​doi.​org/​10.​1038/​nm1095CrossRefPubMed
108.
119.
Zurück zum Zitat Langevin, S. M., Stone, R. A., Bunker, C. H., Lyons-Weiler, M. A., LaFramboise, W. A., Kelly, L., et al. (2011). MicroRNA-137 promoter methylation is associated with poorer overall survival in patients with squamous cell carcinoma of the head and neck. Cancer, 117(7), 1454–1462. https://doi.org/10.1002/cncr.25689CrossRefPubMed Langevin, S. M., Stone, R. A., Bunker, C. H., Lyons-Weiler, M. A., LaFramboise, W. A., Kelly, L., et al. (2011). MicroRNA-137 promoter methylation is associated with poorer overall survival in patients with squamous cell carcinoma of the head and neck. Cancer, 117(7), 1454–1462. https://​doi.​org/​10.​1002/​cncr.​25689CrossRefPubMed
126.
Zurück zum Zitat Fernández, N. B., Sosa, S. M., Roberts, J. T., Recouvreux, M. S., Rocha-Viegas, L., Christenson, J. L., Spoelstra, N. S., Couto, F. L., Raimondi, A. R., Richer, J. K., & Rubinstein, N. (2023). RUNX1 is regulated by androgen receptor to promote cancer stem markers and chemotherapy resistance in triple negative breast cancer. Cells, 12(3), 444. https://doi.org/10.3390/cells12030444CrossRefPubMedPubMedCentral Fernández, N. B., Sosa, S. M., Roberts, J. T., Recouvreux, M. S., Rocha-Viegas, L., Christenson, J. L., Spoelstra, N. S., Couto, F. L., Raimondi, A. R., Richer, J. K., & Rubinstein, N. (2023). RUNX1 is regulated by androgen receptor to promote cancer stem markers and chemotherapy resistance in triple negative breast cancer. Cells, 12(3), 444. https://​doi.​org/​10.​3390/​cells12030444CrossRefPubMedPubMedCentral
130.
140.
Zurück zum Zitat Lettlova, S., Brynychova, V., Blecha, J., Vrana, D., Vondrusova, M., Soucek, P., et al. (2018). MiR-301a-3p suppresses estrogen signaling by directly inhibiting ESR1 in ERalpha positive breast cancer. Cellular Physiology and Biochemistry, 46(6), 2601–2615. https://doi.org/10.1159/000489687CrossRefPubMed Lettlova, S., Brynychova, V., Blecha, J., Vrana, D., Vondrusova, M., Soucek, P., et al. (2018). MiR-301a-3p suppresses estrogen signaling by directly inhibiting ESR1 in ERalpha positive breast cancer. Cellular Physiology and Biochemistry, 46(6), 2601–2615. https://​doi.​org/​10.​1159/​000489687CrossRefPubMed
141.
155.
156.
Zurück zum Zitat Huang, J. W., Guan, B. Z., Yin, L. H., Liu, F. N., Hu, B., Zheng, Q. Y., et al. (2014). Effects of estrogen-related receptor alpha (ERRalpha) on proliferation and metastasis of human lung cancer A549 cells. Journal of Huazhong University of Science and Technology. Medical Sciences, 34(6), 875–881. https://doi.org/10.1007/s11596-014-1367-0CrossRef Huang, J. W., Guan, B. Z., Yin, L. H., Liu, F. N., Hu, B., Zheng, Q. Y., et al. (2014). Effects of estrogen-related receptor alpha (ERRalpha) on proliferation and metastasis of human lung cancer A549 cells. Journal of Huazhong University of Science and Technology. Medical Sciences, 34(6), 875–881. https://​doi.​org/​10.​1007/​s11596-014-1367-0CrossRef
159.
Zurück zum Zitat Singh, T. D., Jeong, S. Y., Lee, S. W., Ha, J. H., Lee, I. K., Kim, S. H., et al. (2015). Inverse agonist of estrogen-related receptor gamma enhances sodium iodide symporter function through mitogen-activated protein kinase signaling in anaplastic thyroid cancer cells. Journal of Nuclear Medicine, 56(11), 1690–1696. https://doi.org/10.2967/jnumed.115.160366CrossRefPubMed Singh, T. D., Jeong, S. Y., Lee, S. W., Ha, J. H., Lee, I. K., Kim, S. H., et al. (2015). Inverse agonist of estrogen-related receptor gamma enhances sodium iodide symporter function through mitogen-activated protein kinase signaling in anaplastic thyroid cancer cells. Journal of Nuclear Medicine, 56(11), 1690–1696. https://​doi.​org/​10.​2967/​jnumed.​115.​160366CrossRefPubMed
163.
Zurück zum Zitat Han, G. H., Yun, H., Kim, J., Chung, J. Y., Kim, J. H., & Cho, H. (2022). Overexpression of glucocorticoid receptor promotes the poor progression and induces cisplatin resistance through p38 MAP kinase in cervical cancer patients. American Journal of Cancer Research, 12(7), 3437–3454.PubMedPubMedCentral Han, G. H., Yun, H., Kim, J., Chung, J. Y., Kim, J. H., & Cho, H. (2022). Overexpression of glucocorticoid receptor promotes the poor progression and induces cisplatin resistance through p38 MAP kinase in cervical cancer patients. American Journal of Cancer Research, 12(7), 3437–3454.PubMedPubMedCentral
165.
Zurück zum Zitat Suzuki, S., Yamamoto, M., Sanomachi, T., Togashi, K., Sugai, A., Seino, S., Yoshioka, T., Okada, M., & Kitanaka, C. (2021). Dexamethasone sensitizes cancer stem cells to gemcitabine and 5-Fluorouracil by increasing reactive oxygen species production through NRF2 Reduction. Life (Basel, Switzerland), 11(9), 885. https://doi.org/10.3390/life11090885CrossRefPubMed Suzuki, S., Yamamoto, M., Sanomachi, T., Togashi, K., Sugai, A., Seino, S., Yoshioka, T., Okada, M., & Kitanaka, C. (2021). Dexamethasone sensitizes cancer stem cells to gemcitabine and 5-Fluorouracil by increasing reactive oxygen species production through NRF2 Reduction. Life (Basel, Switzerland), 11(9), 885. https://​doi.​org/​10.​3390/​life11090885CrossRefPubMed
166.
Zurück zum Zitat Martinez, S. R., Elix, C. C., Ochoa, P. T., Sanchez-Hernandez, E. S., Alkashgari, H. R., Ortiz-Hernandez, G. L., Zhang, L., & Casiano, C. A. (2023). Glucocorticoid receptor and β-Catenin interact in prostate cancer cells and their co-inhibition attenuates tumorsphere formation, stemness, and docetaxel resistance. International Journal of Molecular Sciences, 24(8), 7130. https://doi.org/10.3390/ijms24087130CrossRefPubMedPubMedCentral Martinez, S. R., Elix, C. C., Ochoa, P. T., Sanchez-Hernandez, E. S., Alkashgari, H. R., Ortiz-Hernandez, G. L., Zhang, L., & Casiano, C. A. (2023). Glucocorticoid receptor and β-Catenin interact in prostate cancer cells and their co-inhibition attenuates tumorsphere formation, stemness, and docetaxel resistance. International Journal of Molecular Sciences, 24(8), 7130. https://​doi.​org/​10.​3390/​ijms24087130CrossRefPubMedPubMedCentral
174.
Zurück zum Zitat Haynes, H. R., Scott, H. L., Killick-Cole, C. L., Shaw, G., Brend, T., Hares, K. M., et al. (2019). shRNA-mediated PPARalpha knockdown in human glioma stem cells reduces in vitro proliferation and inhibits orthotopic xenograft tumour growth. The Journal of Pathology, 247(4), 422–434. https://doi.org/10.1002/path.5201CrossRefPubMed Haynes, H. R., Scott, H. L., Killick-Cole, C. L., Shaw, G., Brend, T., Hares, K. M., et al. (2019). shRNA-mediated PPARalpha knockdown in human glioma stem cells reduces in vitro proliferation and inhibits orthotopic xenograft tumour growth. The Journal of Pathology, 247(4), 422–434. https://​doi.​org/​10.​1002/​path.​5201CrossRefPubMed
177.
Zurück zum Zitat Dwyer, A. R., Truong, T. H., Kerkvliet, C. P., Paul, K. V., Kabos, P., Sartorius, C. A., et al. (2021). Insulin receptor substrate-1 (IRS-1) mediates progesterone receptor-driven stemness and endocrine resistance in oestrogen receptor+ breast cancer. British Journal of Cancer, 124(1), 217–227. https://doi.org/10.1038/s41416-020-01094-yCrossRefPubMed Dwyer, A. R., Truong, T. H., Kerkvliet, C. P., Paul, K. V., Kabos, P., Sartorius, C. A., et al. (2021). Insulin receptor substrate-1 (IRS-1) mediates progesterone receptor-driven stemness and endocrine resistance in oestrogen receptor+ breast cancer. British Journal of Cancer, 124(1), 217–227. https://​doi.​org/​10.​1038/​s41416-020-01094-yCrossRefPubMed
178.
Zurück zum Zitat Finlay-Schultz, J., Cittelly, D. M., Hendricks, P., Patel, P., Kabos, P., Jacobsen, B. M., et al. (2015). Progesterone downregulation of miR-141 contributes to expansion of stem-like breast cancer cells through maintenance of progesterone receptor and Stat5a. Oncogene, 34(28), 3676–3687. https://doi.org/10.1038/onc.2014.298CrossRefPubMed Finlay-Schultz, J., Cittelly, D. M., Hendricks, P., Patel, P., Kabos, P., Jacobsen, B. M., et al. (2015). Progesterone downregulation of miR-141 contributes to expansion of stem-like breast cancer cells through maintenance of progesterone receptor and Stat5a. Oncogene, 34(28), 3676–3687. https://​doi.​org/​10.​1038/​onc.​2014.​298CrossRefPubMed
186.
Zurück zum Zitat Singh, D. K., Carcamo, S., Farias, E. F., Hasson, D., Zheng, W., Sun, D., Huang, X., Cheung, J., Nobre, A. R., Kale, N., Sosa, M. S., Bernstein, E., & Aguirre-Ghiso, J. A. (2023). 5-Azacytidine- and retinoic-acid-induced reprogramming of DCCs into dormancy suppresses metastasis via restored TGF-β-SMAD4 signaling. Cell reports, 42(6), 112560. https://doi.org/10.1016/j.celrep.2023.112560CrossRefPubMed Singh, D. K., Carcamo, S., Farias, E. F., Hasson, D., Zheng, W., Sun, D., Huang, X., Cheung, J., Nobre, A. R., Kale, N., Sosa, M. S., Bernstein, E., & Aguirre-Ghiso, J. A. (2023). 5-Azacytidine- and retinoic-acid-induced reprogramming of DCCs into dormancy suppresses metastasis via restored TGF-β-SMAD4 signaling. Cell reports, 42(6), 112560. https://​doi.​org/​10.​1016/​j.​celrep.​2023.​112560CrossRefPubMed
192.
193.
Zurück zum Zitat Chavali, P. L., Saini, R. K., Zhai, Q., Vizlin-Hodzic, D., Venkatabalasubramanian, S., Hayashi, A., et al. (2014). TLX activates MMP-2, promotes self-renewal of tumor spheres in neuroblastoma and correlates with poor patient survival. Cell Death & Disease, 5(10), e1502. https://doi.org/10.1038/cddis.2014.449CrossRef Chavali, P. L., Saini, R. K., Zhai, Q., Vizlin-Hodzic, D., Venkatabalasubramanian, S., Hayashi, A., et al. (2014). TLX activates MMP-2, promotes self-renewal of tumor spheres in neuroblastoma and correlates with poor patient survival. Cell Death & Disease, 5(10), e1502. https://​doi.​org/​10.​1038/​cddis.​2014.​449CrossRef
194.
Zurück zum Zitat Yang, D. R., Ding, X. F., Luo, J., Shan, Y. X., Wang, R., Lin, S. J., et al. (2013). Increased chemosensitivity via targeting testicular nuclear receptor 4 (TR4)-Oct4-interleukin 1 receptor antagonist (IL1Ra) axis in prostate cancer CD133+ stem/progenitor cells to battle prostate cancer. Journal of Biological Chemistry, 288(23), 16476–16483. https://doi.org/10.1074/jbc.M112.448142CrossRefPubMedPubMedCentral Yang, D. R., Ding, X. F., Luo, J., Shan, Y. X., Wang, R., Lin, S. J., et al. (2013). Increased chemosensitivity via targeting testicular nuclear receptor 4 (TR4)-Oct4-interleukin 1 receptor antagonist (IL1Ra) axis in prostate cancer CD133+ stem/progenitor cells to battle prostate cancer. Journal of Biological Chemistry, 288(23), 16476–16483. https://​doi.​org/​10.​1074/​jbc.​M112.​448142CrossRefPubMedPubMedCentral
199.
200.
Zurück zum Zitat Davey, R. A., & Grossmann, M. (2016). Androgen Receptor Structure, Function and Biology: From Bench to Bedside. Clinical Biochemist Reviews, 37(1), 3–15.PubMedPubMedCentral Davey, R. A., & Grossmann, M. (2016). Androgen Receptor Structure, Function and Biology: From Bench to Bedside. Clinical Biochemist Reviews, 37(1), 3–15.PubMedPubMedCentral
205.
Zurück zum Zitat Schreiber, S. N., Knutti, D., Brogli, K., Uhlmann, T., & Kralli, A. (2003). The transcriptional coactivator PGC-1 regulates the expression and activity of the orphan nuclear receptor estrogen-related receptor alpha (ERRalpha). Journal of Biological Chemistry, 278(11), 9013–9018. https://doi.org/10.1074/jbc.M212923200CrossRefPubMed Schreiber, S. N., Knutti, D., Brogli, K., Uhlmann, T., & Kralli, A. (2003). The transcriptional coactivator PGC-1 regulates the expression and activity of the orphan nuclear receptor estrogen-related receptor alpha (ERRalpha). Journal of Biological Chemistry, 278(11), 9013–9018. https://​doi.​org/​10.​1074/​jbc.​M212923200CrossRefPubMed
221.
Zurück zum Zitat Neschen, S., Morino, K., Dong, J., Wang-Fischer, Y., Cline, G. W., Romanelli, A. J., et al. (2007). n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome proliferator-activated receptor-alpha-dependent manner. Diabetes, 56(4), 1034–1041. https://doi.org/10.2337/db06-1206CrossRefPubMed Neschen, S., Morino, K., Dong, J., Wang-Fischer, Y., Cline, G. W., Romanelli, A. J., et al. (2007). n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome proliferator-activated receptor-alpha-dependent manner. Diabetes, 56(4), 1034–1041. https://​doi.​org/​10.​2337/​db06-1206CrossRefPubMed
229.
Zurück zum Zitat Steven, A., Kliewer John, T., Laura, Moore, Wade Jeff, L., Staudinger Michael, A., Watson Stacey, A., Jones David, D., McKee Beverly, B., Oliver Timothy, M., Willson Rolf, H., Thomas, Zetterström, Perlmann Jürgen, M., & Lehmann,. (1998). An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell, 92(1), 73–82. https://doi.org/10.1016/S0092-8674(00)80900-9CrossRef Steven, A., Kliewer John, T., Laura, Moore, Wade Jeff, L., Staudinger Michael, A., Watson Stacey, A., Jones David, D., McKee Beverly, B., Oliver Timothy, M., Willson Rolf, H., Thomas, Zetterström, Perlmann Jürgen, M., & Lehmann,. (1998). An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell, 92(1), 73–82. https://​doi.​org/​10.​1016/​S0092-8674(00)80900-9CrossRef
230.
237.
Zurück zum Zitat Delacroix, L., Moutier, E., Altobelli, G., Legras, S., Poch, O., Choukrallah, M. A., et al. (2010). Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Molecular and Cellular Biology, 30(1), 231–244. https://doi.org/10.1128/MCB.00756-09CrossRefPubMed Delacroix, L., Moutier, E., Altobelli, G., Legras, S., Poch, O., Choukrallah, M. A., et al. (2010). Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Molecular and Cellular Biology, 30(1), 231–244. https://​doi.​org/​10.​1128/​MCB.​00756-09CrossRefPubMed
238.
Zurück zum Zitat Eifert, C., Sangster-Guity, N., Yu, L. M., Chittur, S. V., Perez, A. V., Tine, J. A., et al. (2006). Global gene expression profiles associated with retinoic acid-induced differentiation of embryonal carcinoma cells. Molecular Reproduction and Development, 73(7), 796–824. https://doi.org/10.1002/mrd.20444CrossRefPubMed Eifert, C., Sangster-Guity, N., Yu, L. M., Chittur, S. V., Perez, A. V., Tine, J. A., et al. (2006). Global gene expression profiles associated with retinoic acid-induced differentiation of embryonal carcinoma cells. Molecular Reproduction and Development, 73(7), 796–824. https://​doi.​org/​10.​1002/​mrd.​20444CrossRefPubMed
253.
Zurück zum Zitat Giguere, V., Tini, M., Flock, G., Ong, E., Evans, R. M., & Otulakowski, G. (1994). Isoform-specific amino-terminal domains dictate DNA-binding properties of ROR alpha, a novel family of orphan hormone nuclear receptors. Genes & Development, 8(5), 538–553. https://doi.org/10.1101/gad.8.5.538CrossRef Giguere, V., Tini, M., Flock, G., Ong, E., Evans, R. M., & Otulakowski, G. (1994). Isoform-specific amino-terminal domains dictate DNA-binding properties of ROR alpha, a novel family of orphan hormone nuclear receptors. Genes & Development, 8(5), 538–553. https://​doi.​org/​10.​1101/​gad.​8.​5.​538CrossRef
264.
Zurück zum Zitat Sweeney, C. J., Martin, A. J., Stockler, M. R., Begbie, S., Cheung, L., Chi, K. N., et al. (2023). Testosterone suppression plus enzalutamide versus testosterone suppression plus standard antiandrogen therapy for metastatic hormone-sensitive prostate cancer (ENZAMET): An international, open-label, randomised, phase 3 trial. The lancet Oncology, 24(4), 323–334. https://doi.org/10.1016/S1470-2045(23)00063-3CrossRefPubMed Sweeney, C. J., Martin, A. J., Stockler, M. R., Begbie, S., Cheung, L., Chi, K. N., et al. (2023). Testosterone suppression plus enzalutamide versus testosterone suppression plus standard antiandrogen therapy for metastatic hormone-sensitive prostate cancer (ENZAMET): An international, open-label, randomised, phase 3 trial. The lancet Oncology, 24(4), 323–334. https://​doi.​org/​10.​1016/​S1470-2045(23)00063-3CrossRefPubMed
265.
Zurück zum Zitat Merseburger, A. S., Attard, G., Astrom, L., Matveev, V. B., Bracarda, S., Esen, A., et al. (2022). Continuous enzalutamide after progression of metastatic castration-resistant prostate cancer treated with docetaxel (PRESIDE): An international, randomised, phase 3b study. The lancet Oncology, 23(11), 1398–1408. https://doi.org/10.1016/S1470-2045(22)00560-5CrossRefPubMed Merseburger, A. S., Attard, G., Astrom, L., Matveev, V. B., Bracarda, S., Esen, A., et al. (2022). Continuous enzalutamide after progression of metastatic castration-resistant prostate cancer treated with docetaxel (PRESIDE): An international, randomised, phase 3b study. The lancet Oncology, 23(11), 1398–1408. https://​doi.​org/​10.​1016/​S1470-2045(22)00560-5CrossRefPubMed
268.
Zurück zum Zitat Gu, W., Han, W., Luo, H., Zhou, F., He, D., Ma, L., et al. (2022). Rezvilutamide versus bicalutamide in combination with androgen-deprivation therapy in patients with high-volume, metastatic, hormone-sensitive prostate cancer (CHART): A randomised, open-label, phase 3 trial. The lancet Oncology, 23(10), 1249–1260. https://doi.org/10.1016/S1470-2045(22)00507-1CrossRefPubMed Gu, W., Han, W., Luo, H., Zhou, F., He, D., Ma, L., et al. (2022). Rezvilutamide versus bicalutamide in combination with androgen-deprivation therapy in patients with high-volume, metastatic, hormone-sensitive prostate cancer (CHART): A randomised, open-label, phase 3 trial. The lancet Oncology, 23(10), 1249–1260. https://​doi.​org/​10.​1016/​S1470-2045(22)00507-1CrossRefPubMed
269.
Zurück zum Zitat Penson, D. F., Armstrong, A. J., Concepcion, R. S., Agarwal, N., Olsson, C. A., Karsh, L. I., et al. (2022). Enzalutamide versus bicalutamide in patients with nonmetastatic castration-resistant prostate cancer: A prespecified subgroup analysis of the STRIVE trial. Prostate Cancer and Prostatic Diseases, 25(2), 363–365. https://doi.org/10.1038/s41391-021-00465-7CrossRefPubMed Penson, D. F., Armstrong, A. J., Concepcion, R. S., Agarwal, N., Olsson, C. A., Karsh, L. I., et al. (2022). Enzalutamide versus bicalutamide in patients with nonmetastatic castration-resistant prostate cancer: A prespecified subgroup analysis of the STRIVE trial. Prostate Cancer and Prostatic Diseases, 25(2), 363–365. https://​doi.​org/​10.​1038/​s41391-021-00465-7CrossRefPubMed
270.
Zurück zum Zitat Yuan, Y., Lee, J. S., Yost, S. E., Frankel, P. H., Ruel, C., Egelston, C. A., et al. (2021). A phase II clinical trial of pembrolizumab and enobosarm in patients with androgen receptor-positive metastatic triple-negative breast cancer. The Oncologist, 26(2), 99-e217. https://doi.org/10.1002/onco.13583CrossRefPubMed Yuan, Y., Lee, J. S., Yost, S. E., Frankel, P. H., Ruel, C., Egelston, C. A., et al. (2021). A phase II clinical trial of pembrolizumab and enobosarm in patients with androgen receptor-positive metastatic triple-negative breast cancer. The Oncologist, 26(2), 99-e217. https://​doi.​org/​10.​1002/​onco.​13583CrossRefPubMed
272.
Zurück zum Zitat Cmero, M., Kurganovs, N. J., Stuchbery, R., McCoy, P., Grima, C., Ngyuen, A., Chow, K., Mangiola, S., Macintyre, G., Howard, N., Kerger, M., Dundee, P., Ruljancich, P., Clarke, D., Grummet, J., Peters, J. S., Costello, A. J., Norden, S., Ryan, A., … Corcoran, N. M. (2021). Loss of SNAI2 in prostate cancer correlates with clinical response to androgen deprivation therapy. JCO Precision Oncology, 5, PO.20.00337. https://doi.org/10.1200/PO.20.00337CrossRefPubMedPubMedCentral Cmero, M., Kurganovs, N. J., Stuchbery, R., McCoy, P., Grima, C., Ngyuen, A., Chow, K., Mangiola, S., Macintyre, G., Howard, N., Kerger, M., Dundee, P., Ruljancich, P., Clarke, D., Grummet, J., Peters, J. S., Costello, A. J., Norden, S., Ryan, A., … Corcoran, N. M. (2021). Loss of SNAI2 in prostate cancer correlates with clinical response to androgen deprivation therapy. JCO Precision Oncology, 5, PO.20.00337. https://​doi.​org/​10.​1200/​PO.​20.​00337CrossRefPubMedPubMedCentral
275.
Zurück zum Zitat Baek, S. Y., Noh, W. C., Ahn, S. H., Kim, H. A., Ryu, J. M., Kim, S. I., et al. (2023). Adding ovarian suppression to tamoxifen for premenopausal women with hormone receptor-positive breast cancer after chemotherapy: An 8-year follow-up of the ASTRRA trial. Journal of Clinical Oncology, 41(31), 4864–4871. https://doi.org/10.1200/JCO.23.00557CrossRefPubMed Baek, S. Y., Noh, W. C., Ahn, S. H., Kim, H. A., Ryu, J. M., Kim, S. I., et al. (2023). Adding ovarian suppression to tamoxifen for premenopausal women with hormone receptor-positive breast cancer after chemotherapy: An 8-year follow-up of the ASTRRA trial. Journal of Clinical Oncology, 41(31), 4864–4871. https://​doi.​org/​10.​1200/​JCO.​23.​00557CrossRefPubMed
278.
Zurück zum Zitat Howell, S. J., Casbard, A., Carucci, M., Ingarfield, K., Butler, R., Morgan, S., et al. (2022). Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive, HER2-negative breast cancer (FAKTION): Overall survival, updated progression-free survival, and expanded biomarker analysis from a randomised, phase 2 trial. The lancet Oncology, 23(7), 851–864. https://doi.org/10.1016/S1470-2045(22)00284-4CrossRefPubMedPubMedCentral Howell, S. J., Casbard, A., Carucci, M., Ingarfield, K., Butler, R., Morgan, S., et al. (2022). Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive, HER2-negative breast cancer (FAKTION): Overall survival, updated progression-free survival, and expanded biomarker analysis from a randomised, phase 2 trial. The lancet Oncology, 23(7), 851–864. https://​doi.​org/​10.​1016/​S1470-2045(22)00284-4CrossRefPubMedPubMedCentral
280.
282.
Zurück zum Zitat Campbell, R. A., Bhat-Nakshatri, P., Patel, N. M., Constantinidou, D., Ali, S., & Nakshatri, H. (2001). Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: A new model for anti-estrogen resistance. Journal of Biological Chemistry, 276(13), 9817–9824. https://doi.org/10.1074/jbc.M010840200CrossRefPubMed Campbell, R. A., Bhat-Nakshatri, P., Patel, N. M., Constantinidou, D., Ali, S., & Nakshatri, H. (2001). Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: A new model for anti-estrogen resistance. Journal of Biological Chemistry, 276(13), 9817–9824. https://​doi.​org/​10.​1074/​jbc.​M010840200CrossRefPubMed
284.
Zurück zum Zitat Sonneveld, P., Chanan-Khan, A., Weisel, K., Nooka, A. K., Masszi, T., Beksac, M., et al. (2023). Overall survival with daratumumab, bortezomib, and dexamethasone in previously treated multiple myeloma (CASTOR): A randomized, open-label, phase III trial. Journal of Clinical Oncology, 41(8), 1600–1609. https://doi.org/10.1200/JCO.21.02734CrossRefPubMed Sonneveld, P., Chanan-Khan, A., Weisel, K., Nooka, A. K., Masszi, T., Beksac, M., et al. (2023). Overall survival with daratumumab, bortezomib, and dexamethasone in previously treated multiple myeloma (CASTOR): A randomized, open-label, phase III trial. Journal of Clinical Oncology, 41(8), 1600–1609. https://​doi.​org/​10.​1200/​JCO.​21.​02734CrossRefPubMed
285.
Zurück zum Zitat Dimopoulos, M. A., Dytfeld, D., Grosicki, S., Moreau, P., Takezako, N., Hori, M., et al. (2023). Elotuzumab plus pomalidomide and dexamethasone for relapsed/refractory multiple myeloma: Final overall survival analysis from the randomized phase II ELOQUENT-3 Trial. Journal of Clinical Oncology, 41(3), 568–578. https://doi.org/10.1200/JCO.21.02815CrossRefPubMed Dimopoulos, M. A., Dytfeld, D., Grosicki, S., Moreau, P., Takezako, N., Hori, M., et al. (2023). Elotuzumab plus pomalidomide and dexamethasone for relapsed/refractory multiple myeloma: Final overall survival analysis from the randomized phase II ELOQUENT-3 Trial. Journal of Clinical Oncology, 41(3), 568–578. https://​doi.​org/​10.​1200/​JCO.​21.​02815CrossRefPubMed
287.
288.
Zurück zum Zitat Costa, L. J., Chhabra, S., Medvedova, E., Dholaria, B. R., Schmidt, T. M., Godby, K. N., et al. (2022). Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma. Journal of Clinical Oncology, 40(25), 2901–2912. https://doi.org/10.1200/JCO.21.01935CrossRefPubMed Costa, L. J., Chhabra, S., Medvedova, E., Dholaria, B. R., Schmidt, T. M., Godby, K. N., et al. (2022). Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma. Journal of Clinical Oncology, 40(25), 2901–2912. https://​doi.​org/​10.​1200/​JCO.​21.​01935CrossRefPubMed
290.
Zurück zum Zitat Akutsu, T., Okada, S., Hirooka, S., Ikegami, M., Ohdaira, H., Suzuki, Y., et al. (2020). Effect of vitamin D on relapse-free survival in a subgroup of patients with p53 protein-positive digestive tract cancer: A post hoc analysis of the AMATERASU trial. Cancer Epidemiology, Biomarkers & Prevention, 29(2), 406–413. https://doi.org/10.1158/1055-9965.EPI-19-0986CrossRef Akutsu, T., Okada, S., Hirooka, S., Ikegami, M., Ohdaira, H., Suzuki, Y., et al. (2020). Effect of vitamin D on relapse-free survival in a subgroup of patients with p53 protein-positive digestive tract cancer: A post hoc analysis of the AMATERASU trial. Cancer Epidemiology, Biomarkers & Prevention, 29(2), 406–413. https://​doi.​org/​10.​1158/​1055-9965.​EPI-19-0986CrossRef
293.
Zurück zum Zitat Haidari, F., Abiri, B., Iravani, M., Ahmadi-Angali, K., & Vafa, M. (2020). Effects of vitamin D and omega-3 fatty acids co-supplementation on inflammatory factors and tumor marker cea in colorectal cancer patients undergoing chemotherapy: A randomized, double-blind, placebo-controlled clinical trial. Nutrition and Cancer, 72(6), 948–958. https://doi.org/10.1080/01635581.2019.1659380CrossRefPubMed Haidari, F., Abiri, B., Iravani, M., Ahmadi-Angali, K., & Vafa, M. (2020). Effects of vitamin D and omega-3 fatty acids co-supplementation on inflammatory factors and tumor marker cea in colorectal cancer patients undergoing chemotherapy: A randomized, double-blind, placebo-controlled clinical trial. Nutrition and Cancer, 72(6), 948–958. https://​doi.​org/​10.​1080/​01635581.​2019.​1659380CrossRefPubMed
296.
Zurück zum Zitat Kutny, M. A., Alonzo, T. A., Abla, O., Rajpurkar, M., Gerbing, R. B., Wang, Y. C., et al. (2022). Assessment of arsenic trioxide and all-trans retinoic acid for the treatment of pediatric acute promyelocytic leukemia: A report from the Children’s Oncology group AAML1331 Trial. JAMA Oncology, 8(1), 79–87. https://doi.org/10.1001/jamaoncol.2021.5206CrossRefPubMed Kutny, M. A., Alonzo, T. A., Abla, O., Rajpurkar, M., Gerbing, R. B., Wang, Y. C., et al. (2022). Assessment of arsenic trioxide and all-trans retinoic acid for the treatment of pediatric acute promyelocytic leukemia: A report from the Children’s Oncology group AAML1331 Trial. JAMA Oncology, 8(1), 79–87. https://​doi.​org/​10.​1001/​jamaoncol.​2021.​5206CrossRefPubMed
300.
Zurück zum Zitat Smith, M. R., Manola, J., Kaufman, D. S., George, D., Oh, W. K., Mueller, E., et al. (2004). Rosiglitazone versus placebo for men with prostate carcinoma and a rising serum prostate-specific antigen level after radical prostatectomy and/or radiation therapy. Cancer, 101(7), 1569–1574. https://doi.org/10.1002/cncr.20493CrossRefPubMed Smith, M. R., Manola, J., Kaufman, D. S., George, D., Oh, W. K., Mueller, E., et al. (2004). Rosiglitazone versus placebo for men with prostate carcinoma and a rising serum prostate-specific antigen level after radical prostatectomy and/or radiation therapy. Cancer, 101(7), 1569–1574. https://​doi.​org/​10.​1002/​cncr.​20493CrossRefPubMed
Metadaten
Titel
Dynamic interplay of nuclear receptors in tumor cell plasticity and drug resistance: Shifting gears in malignant transformations and applications in cancer therapeutics
verfasst von
Bandari BharathwajChetty
Anjana Sajeev
Ravichandran Vishwa
Babu Santha Aswani
Mohammed S. Alqahtani
Mohamed Abbas
Ajaikumar B. Kunnumakkara
Publikationsdatum
22.03.2024
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 1/2024
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-024-10171-0

Weitere Artikel der Ausgabe 1/2024

Cancer and Metastasis Reviews 1/2024 Zur Ausgabe

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

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.

Update Onkologie

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