Skip to main content
Top
Published 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

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

Published in: Cancer and Metastasis Reviews | Issue 1/2024

Login to get access

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

Literature
2.
go back to reference 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.
go back to reference 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.
go back to reference 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.
46.
go back to reference 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.
go back to reference 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.
go back to reference 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.
82.
99.
103.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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
140.
155.
156.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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
193.
go back to reference 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.
199.
200.
go back to reference 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.
221.
229.
go back to reference 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
237.
238.
go back to reference 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.
264.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
272.
go back to reference 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.
go back to reference 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.
go back to reference 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.
284.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
300.
go back to reference 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
Metadata
Title
Dynamic interplay of nuclear receptors in tumor cell plasticity and drug resistance: Shifting gears in malignant transformations and applications in cancer therapeutics
Authors
Bandari BharathwajChetty
Anjana Sajeev
Ravichandran Vishwa
Babu Santha Aswani
Mohammed S. Alqahtani
Mohamed Abbas
Ajaikumar B. Kunnumakkara
Publication date
22-03-2024
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 1/2024
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-024-10171-0

Other articles of this Issue 1/2024

Cancer and Metastasis Reviews 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine