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

16.01.2024 | REVIEW

Harnessing function of EMT in cancer drug resistance: a metastasis regulator determines chemotherapy response

verfasst von: Nasim Ebrahimi, Mahdokht Sadat Manavi, Ferdos Faghihkhorasani, Siavash Seifollahy Fakhr, Fatemeh Jafari Baei, Fereshteh Faghih Khorasani, Mohammad Mehdi Zare, Nazanin Pazhouhesh Far, Fatemeh Rezaei-Tazangi, Jun Ren, Russel J. Reiter, Noushin Nabavi, Amir Reza Aref, Chu Chen, Yavuz Nuri Ertas, Qi Lu

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

Einloggen, um Zugang zu erhalten

Abstract

Epithelial-mesenchymal transition (EMT) is a complicated molecular process that governs cellular shape and function changes throughout tissue development and embryogenesis. In addition, EMT contributes to the development and spread of tumors. Expanding and degrading the surrounding microenvironment, cells undergoing EMT move away from the main location. On the basis of the expression of fibroblast-specific protein-1 (FSP1), fibroblast growth factor (FGF), collagen, and smooth muscle actin (-SMA), the mesenchymal phenotype exhibited in fibroblasts is crucial for promoting EMT. While EMT is not entirely reliant on its regulators like ZEB1/2, Twist, and Snail proteins, investigation of upstream signaling (like EGF, TGF-β, Wnt) is required to get a more thorough understanding of tumor EMT. Throughout numerous cancers, connections between tumor epithelial and fibroblast cells that influence tumor growth have been found. The significance of cellular crosstalk stems from the fact that these events affect therapeutic response and disease prognosis. This study examines how classical EMT signals emanating from various cancer cells interfere to tumor metastasis, treatment resistance, and tumor recurrence.
Literatur
2.
Zurück zum Zitat Yaray, K., Norbakhsh, A., Rashidzadeh, H., Mohammadi, A., Mozafari, F., Ghaffarlou, M., Mousazadeh, N., Ghaderzadeh, R., Ghorbani, Y., Nasehi, L., et al. (2023). Chemoradiation therapy of 4T1 cancer cells with methotrexate conjugated platinum nanoparticles under X-Ray irradiation. Inorganic Chemistry Communications, 150, 110457. https://doi.org/10.1016/j.inoche.2023.110457CrossRef Yaray, K., Norbakhsh, A., Rashidzadeh, H., Mohammadi, A., Mozafari, F., Ghaffarlou, M., Mousazadeh, N., Ghaderzadeh, R., Ghorbani, Y., Nasehi, L., et al. (2023). Chemoradiation therapy of 4T1 cancer cells with methotrexate conjugated platinum nanoparticles under X-Ray irradiation. Inorganic Chemistry Communications, 150, 110457. https://​doi.​org/​10.​1016/​j.​inoche.​2023.​110457CrossRef
4.
Zurück zum Zitat Ashrafizadeh, M., Mirzaei, S., Hashemi, F., Zarrabi, A., Zabolian, A., Saleki, H., Sharifzadeh, S. O., Soleymani, L., Daneshi, S., & Hushmandi, K. (2021). New insight towards development of paclitaxel and docetaxel resistance in cancer cells: EMT as a novel molecular mechanism and therapeutic possibilities. Biomedicine & Pharmacotherapy, 141, 111824.CrossRef Ashrafizadeh, M., Mirzaei, S., Hashemi, F., Zarrabi, A., Zabolian, A., Saleki, H., Sharifzadeh, S. O., Soleymani, L., Daneshi, S., & Hushmandi, K. (2021). New insight towards development of paclitaxel and docetaxel resistance in cancer cells: EMT as a novel molecular mechanism and therapeutic possibilities. Biomedicine & Pharmacotherapy, 141, 111824.CrossRef
5.
Zurück zum Zitat Ashrafizadeh, M., Zarrabi, A., Hushmandi, K., Kalantari, M., Mohammadinejad, R., Javaheri, T., & Sethi, G. (2020). Association of the epithelial–mesenchymal transition (EMT) with cisplatin resistance. International journal of Molecular Sciences, 21, 4002.CrossRefPubMedPubMedCentral Ashrafizadeh, M., Zarrabi, A., Hushmandi, K., Kalantari, M., Mohammadinejad, R., Javaheri, T., & Sethi, G. (2020). Association of the epithelial–mesenchymal transition (EMT) with cisplatin resistance. International journal of Molecular Sciences, 21, 4002.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Mirzaei, S., Abadi, A. J., Gholami, M. H., Hashemi, F., Zabolian, A., Hushmandi, K., Zarrabi, A., Entezari, M., Aref, A. R., & Khan, H. (2021). The involvement of epithelial-to-mesenchymal transition in doxorubicin resistance: Possible molecular targets. European Journal of Pharmacology, 908, 174344.CrossRefPubMed Mirzaei, S., Abadi, A. J., Gholami, M. H., Hashemi, F., Zabolian, A., Hushmandi, K., Zarrabi, A., Entezari, M., Aref, A. R., & Khan, H. (2021). The involvement of epithelial-to-mesenchymal transition in doxorubicin resistance: Possible molecular targets. European Journal of Pharmacology, 908, 174344.CrossRefPubMed
10.
Zurück zum Zitat Ashrafizadeh, M., Hushmandi, K., Hashemi, M., Akbari, M. E., Kubatka, P., Raei, M., Koklesova, L., Shahinozzaman, M., Mohammadinejad, R., & Najafi, M. (2020). Role of microRNA/epithelial-to-mesenchymal transition axis in the metastasis of bladder cancer. Biomolecules, 10, 1159.CrossRefPubMedPubMedCentral Ashrafizadeh, M., Hushmandi, K., Hashemi, M., Akbari, M. E., Kubatka, P., Raei, M., Koklesova, L., Shahinozzaman, M., Mohammadinejad, R., & Najafi, M. (2020). Role of microRNA/epithelial-to-mesenchymal transition axis in the metastasis of bladder cancer. Biomolecules, 10, 1159.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Mirzaei, S., Gholami, M. H., Aghdaei, H. A., Hashemi, M., Parivar, K., Karamian, A., Zarrabi, A., Ashrafizadeh, M., & Lu, J. (2023). Exosome-mediated miR-200a delivery into TGF-β-treated AGS cells abolished epithelial-mesenchymal transition with normalization of ZEB1, vimentin and Snail1 expression. Environmental Research, 231, 116115.CrossRefPubMed Mirzaei, S., Gholami, M. H., Aghdaei, H. A., Hashemi, M., Parivar, K., Karamian, A., Zarrabi, A., Ashrafizadeh, M., & Lu, J. (2023). Exosome-mediated miR-200a delivery into TGF-β-treated AGS cells abolished epithelial-mesenchymal transition with normalization of ZEB1, vimentin and Snail1 expression. Environmental Research, 231, 116115.CrossRefPubMed
31.
Zurück zum Zitat Sommers, C. L., Heckford, S. E., Skerker, J. M., Worland, P., Torri, J. A., Thompson, E. W., Byers, S. W., & Gelmann, E. P. (1992). Loss of epithelial markers and acquisition of vimentin expression in adriamycin- and vinblastine-resistant human breast cancer cell lines. Cancer Research, 52, 5190–5197.PubMed Sommers, C. L., Heckford, S. E., Skerker, J. M., Worland, P., Torri, J. A., Thompson, E. W., Byers, S. W., & Gelmann, E. P. (1992). Loss of epithelial markers and acquisition of vimentin expression in adriamycin- and vinblastine-resistant human breast cancer cell lines. Cancer Research, 52, 5190–5197.PubMed
32.
Zurück zum Zitat Della Corte, C. M., Bellevicine, C., Vicidomini, G., Vitagliano, D., Malapelle, U., Accardo, M., Fabozzi, A., Fiorelli, A., Fasano, M., Papaccio, F., et al. (2015). SMO gene amplification and activation of the hedgehog pathway as novel mechanisms of resistance to anti-epidermal growth factor receptor drugs in human lung cancer. Clinical Cancer Research, 21, 4686–4697. https://doi.org/10.1158/1078-0432.Ccr-14-3319CrossRefPubMed Della Corte, C. M., Bellevicine, C., Vicidomini, G., Vitagliano, D., Malapelle, U., Accardo, M., Fabozzi, A., Fiorelli, A., Fasano, M., Papaccio, F., et al. (2015). SMO gene amplification and activation of the hedgehog pathway as novel mechanisms of resistance to anti-epidermal growth factor receptor drugs in human lung cancer. Clinical Cancer Research, 21, 4686–4697. https://​doi.​org/​10.​1158/​1078-0432.​Ccr-14-3319CrossRefPubMed
36.
Zurück zum Zitat Katsuno, Y., Meyer, D. S., Zhang, Z., Shokat, K. M., Akhurst, R. J., Miyazono, K., & Derynck, R. (2019). Chronic TGF-β exposure drives stabilized EMT, tumor stemness, and cancer drug resistance with vulnerability to bitopic mTOR inhibition. Science Signaling, 12, 570. https://doi.org/10.1126/scisignal.aau8544 Katsuno, Y., Meyer, D. S., Zhang, Z., Shokat, K. M., Akhurst, R. J., Miyazono, K., & Derynck, R. (2019). Chronic TGF-β exposure drives stabilized EMT, tumor stemness, and cancer drug resistance with vulnerability to bitopic mTOR inhibition. Science Signaling, 12, 570. https://​doi.​org/​10.​1126/​scisignal.​aau8544
40.
44.
Zurück zum Zitat Li, R. N., Liu, B., Li, X. M., Hou, L. S., Mu, X. L., Wang, H., & Linghu, H. (2017). DACT1 Overexpression in type I ovarian cancer inhibits malignant expansion and cis-platinum resistance by modulating canonical Wnt signalling and autophagy. Science and Reports, 7, 9285. https://doi.org/10.1038/s41598-017-08249-7CrossRef Li, R. N., Liu, B., Li, X. M., Hou, L. S., Mu, X. L., Wang, H., & Linghu, H. (2017). DACT1 Overexpression in type I ovarian cancer inhibits malignant expansion and cis-platinum resistance by modulating canonical Wnt signalling and autophagy. Science and Reports, 7, 9285. https://​doi.​org/​10.​1038/​s41598-017-08249-7CrossRef
47.
52.
Zurück zum Zitat Yang, M. H., Hsu, D. S., Wang, H. W., Wang, H. J., Lan, H. Y., Yang, W. H., Huang, C. H., Kao, S. Y., Tzeng, C. H., Tai, S. K., et al. (2010). Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nature Cell Biology, 12, 982–992. https://doi.org/10.1038/ncb2099CrossRefPubMed Yang, M. H., Hsu, D. S., Wang, H. W., Wang, H. J., Lan, H. Y., Yang, W. H., Huang, C. H., Kao, S. Y., Tzeng, C. H., Tai, S. K., et al. (2010). Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nature Cell Biology, 12, 982–992. https://​doi.​org/​10.​1038/​ncb2099CrossRefPubMed
60.
Zurück zum Zitat Bao, Z., Zeng, W., Zhang, D., Wang, L., Deng, X., Lai, J., Li, J., Gong, J., & Xiang, G. (2022). SNAIL induces EMT and lung metastasis of tumours secreting CXCL2 to promote the invasion of M2-type immunosuppressed macrophages in colorectal cancer. International Journal of Biological Sciences, 18, 2867.CrossRefPubMedPubMedCentral Bao, Z., Zeng, W., Zhang, D., Wang, L., Deng, X., Lai, J., Li, J., Gong, J., & Xiang, G. (2022). SNAIL induces EMT and lung metastasis of tumours secreting CXCL2 to promote the invasion of M2-type immunosuppressed macrophages in colorectal cancer. International Journal of Biological Sciences, 18, 2867.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Wang, W.-D., Shang, Y., Li, Y., & Chen, S.-Z. (2019). Honokiol inhibits breast cancer cell metastasis by blocking EMT through modulation of Snail/Slug protein translation. Acta Pharmacologica Sinica, 40, 1219–1227.CrossRefPubMedPubMedCentral Wang, W.-D., Shang, Y., Li, Y., & Chen, S.-Z. (2019). Honokiol inhibits breast cancer cell metastasis by blocking EMT through modulation of Snail/Slug protein translation. Acta Pharmacologica Sinica, 40, 1219–1227.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Kao, S. H., Wang, W. L., Chen, C. Y., Chang, Y. L., Wu, Y. Y., Wang, Y. T., Wang, S. P., Nesvizhskii, A. I., Chen, Y. J., Hong, T. M., et al. (2014). GSK3β controls epithelial-mesenchymal transition and tumor metastasis by CHIP-mediated degradation of Slug. Oncogene, 33, 3172–3182. https://doi.org/10.1038/onc.2013.279CrossRefPubMed Kao, S. H., Wang, W. L., Chen, C. Y., Chang, Y. L., Wu, Y. Y., Wang, Y. T., Wang, S. P., Nesvizhskii, A. I., Chen, Y. J., Hong, T. M., et al. (2014). GSK3β controls epithelial-mesenchymal transition and tumor metastasis by CHIP-mediated degradation of Slug. Oncogene, 33, 3172–3182. https://​doi.​org/​10.​1038/​onc.​2013.​279CrossRefPubMed
65.
67.
Zurück zum Zitat Wang, S. P., Wang, W. L., Chang, Y. L., Wu, C. T., Chao, Y. C., Kao, S. H., Yuan, A., Lin, C. W., Yang, S. C., Chan, W. K., et al. (2009). p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nature Cell Biology, 11, 694–704. https://doi.org/10.1038/ncb1875CrossRefPubMed Wang, S. P., Wang, W. L., Chang, Y. L., Wu, C. T., Chao, Y. C., Kao, S. H., Yuan, A., Lin, C. W., Yang, S. C., Chan, W. K., et al. (2009). p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nature Cell Biology, 11, 694–704. https://​doi.​org/​10.​1038/​ncb1875CrossRefPubMed
72.
Zurück zum Zitat Madhry, D., Pandey, K. K., Kaur, J., Rawat, Y., Sapra, L., Ravikumar, Y. S., Srivastava, R. K., Bhattacharyya, S., & Verma, B. (2021). Role of non-coding RNAs in Dengue virus-host interaction. Frontiers in Bioscience-Scholar, 13(44), 55. Madhry, D., Pandey, K. K., Kaur, J., Rawat, Y., Sapra, L., Ravikumar, Y. S., Srivastava, R. K., Bhattacharyya, S., & Verma, B. (2021). Role of non-coding RNAs in Dengue virus-host interaction. Frontiers in Bioscience-Scholar, 13(44), 55.
76.
78.
81.
Zurück zum Zitat Yang, Z., Qu, C. B., Zhang, Y., Zhang, W. F., Wang, D. D., Gao, C. C., Ma, L., Chen, J. S., Liu, K. L., Zheng, B., et al. (2019). Dysregulation of p53-RBM25-mediated circAMOTL1L biogenesis contributes to prostate cancer progression through the circAMOTL1L-miR-193a-5p-Pcdha pathway. Oncogene, 38, 2516–2532. https://doi.org/10.1038/s41388-018-0602-8CrossRefPubMed Yang, Z., Qu, C. B., Zhang, Y., Zhang, W. F., Wang, D. D., Gao, C. C., Ma, L., Chen, J. S., Liu, K. L., Zheng, B., et al. (2019). Dysregulation of p53-RBM25-mediated circAMOTL1L biogenesis contributes to prostate cancer progression through the circAMOTL1L-miR-193a-5p-Pcdha pathway. Oncogene, 38, 2516–2532. https://​doi.​org/​10.​1038/​s41388-018-0602-8CrossRefPubMed
83.
Zurück zum Zitat Khanbabaei, H., Ebrahimi, S., García-Rodríguez, J. L., Ghasemi, Z., Pourghadamyari, H., Mohammadi, M., & Kristensen, L. S. (2022). Non-coding RNAs and epithelial mesenchymal transition in cancer: Molecular mechanisms and clinical implications. Journal of Experimental & Clinical Cancer Research, 41, 278. https://doi.org/10.1186/s13046-022-02488-xCrossRef Khanbabaei, H., Ebrahimi, S., García-Rodríguez, J. L., Ghasemi, Z., Pourghadamyari, H., Mohammadi, M., & Kristensen, L. S. (2022). Non-coding RNAs and epithelial mesenchymal transition in cancer: Molecular mechanisms and clinical implications. Journal of Experimental & Clinical Cancer Research, 41, 278. https://​doi.​org/​10.​1186/​s13046-022-02488-xCrossRef
85.
Zurück zum Zitat Lu, H. Y., Zu, Y. X., Jiang, X. W., Sun, X. T., Liu, T. Y., Li, R. L., Wu, Q., Zhang, Y. S., & Zhao, Q. C. (2019). Novel ADAM-17 inhibitor ZLDI-8 inhibits the proliferation and metastasis of chemo-resistant non-small-cell lung cancer by reversing Notch and epithelial mesenchymal transition in vitro and in vivo. Pharmacological Research, 148, 104406. https://doi.org/10.1016/j.phrs.2019.104406CrossRefPubMed Lu, H. Y., Zu, Y. X., Jiang, X. W., Sun, X. T., Liu, T. Y., Li, R. L., Wu, Q., Zhang, Y. S., & Zhao, Q. C. (2019). Novel ADAM-17 inhibitor ZLDI-8 inhibits the proliferation and metastasis of chemo-resistant non-small-cell lung cancer by reversing Notch and epithelial mesenchymal transition in vitro and in vivo. Pharmacological Research, 148, 104406. https://​doi.​org/​10.​1016/​j.​phrs.​2019.​104406CrossRefPubMed
86.
Zurück zum Zitat Blazquez, R., Rietkötter, E., Wenske, B., Wlochowitz, D., Sparrer, D., Vollmer, E., Müller, G., Seegerer, J., Sun, X., Dettmer, K., et al. (2020). LEF1 supports metastatic brain colonization by regulating glutathione metabolism and increasing ROS resistance in breast cancer. International Journal of Cancer, 146, 3170–3183. https://doi.org/10.1002/ijc.32742CrossRefPubMed Blazquez, R., Rietkötter, E., Wenske, B., Wlochowitz, D., Sparrer, D., Vollmer, E., Müller, G., Seegerer, J., Sun, X., Dettmer, K., et al. (2020). LEF1 supports metastatic brain colonization by regulating glutathione metabolism and increasing ROS resistance in breast cancer. International Journal of Cancer, 146, 3170–3183. https://​doi.​org/​10.​1002/​ijc.​32742CrossRefPubMed
91.
95.
Zurück zum Zitat Zhong, C., Tao, B., Chen, Y., Guo, Z., Yang, X., Peng, L., Xia, X., & Chen, L. (2020). B7–H3 regulates glioma growth and cell invasion through a JAK2/STAT3/Slug-dependent signaling pathway. OncoTargets and therapy, 13, 2215.CrossRefPubMedPubMedCentral Zhong, C., Tao, B., Chen, Y., Guo, Z., Yang, X., Peng, L., Xia, X., & Chen, L. (2020). B7–H3 regulates glioma growth and cell invasion through a JAK2/STAT3/Slug-dependent signaling pathway. OncoTargets and therapy, 13, 2215.CrossRefPubMedPubMedCentral
100.
Zurück zum Zitat Sandberg, C. J., Altschuler, G., Jeong, J., Strømme, K. K., Stangeland, B., Murrell, W., Grasmo-Wendler, U. H., Myklebost, O., Helseth, E., Vik-Mo, E. O., et al. (2013). Comparison of glioma stem cells to neural stem cells from the adult human brain identifies dysregulated Wnt- signaling and a fingerprint associated with clinical outcome. Experimental Cell Research, 319, 2230–2243. https://doi.org/10.1016/j.yexcr.2013.06.004CrossRefPubMed Sandberg, C. J., Altschuler, G., Jeong, J., Strømme, K. K., Stangeland, B., Murrell, W., Grasmo-Wendler, U. H., Myklebost, O., Helseth, E., Vik-Mo, E. O., et al. (2013). Comparison of glioma stem cells to neural stem cells from the adult human brain identifies dysregulated Wnt- signaling and a fingerprint associated with clinical outcome. Experimental Cell Research, 319, 2230–2243. https://​doi.​org/​10.​1016/​j.​yexcr.​2013.​06.​004CrossRefPubMed
104.
Zurück zum Zitat Ebrahimi, N., Adelian, S., Shakerian, S., Afshinpour, M., Chaleshtori, S. R., Rostami, N., Rezaei-Tazangi, F., Beiranvand, S., Hamblin, M. R., & Aref, A. R. (2022). Crosstalk between ferroptosis and the epithelial-mesenchymal transition: Implications for inflammation and cancer therapy. Cytokine & Growth Factor Reviews, 64, 33–45. https://doi.org/10.1016/j.cytogfr.2022.01.006CrossRef Ebrahimi, N., Adelian, S., Shakerian, S., Afshinpour, M., Chaleshtori, S. R., Rostami, N., Rezaei-Tazangi, F., Beiranvand, S., Hamblin, M. R., & Aref, A. R. (2022). Crosstalk between ferroptosis and the epithelial-mesenchymal transition: Implications for inflammation and cancer therapy. Cytokine & Growth Factor Reviews, 64, 33–45. https://​doi.​org/​10.​1016/​j.​cytogfr.​2022.​01.​006CrossRef
106.
Zurück zum Zitat Fan, X., Khaki, L., Zhu, T. S., Soules, M. E., Talsma, C. E., Gul, N., Koh, C., Zhang, J., Li, Y. M., Maciaczyk, J., et al. (2010). NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells, 28, 5–16. https://doi.org/10.1002/stem.254CrossRefPubMed Fan, X., Khaki, L., Zhu, T. S., Soules, M. E., Talsma, C. E., Gul, N., Koh, C., Zhang, J., Li, Y. M., Maciaczyk, J., et al. (2010). NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells, 28, 5–16. https://​doi.​org/​10.​1002/​stem.​254CrossRefPubMed
107.
Zurück zum Zitat Kristoffersen, K., Villingshøj, M., Poulsen, H. S., & Stockhausen, M. T. (2013). Level of Notch activation determines the effect on growth and stem cell-like features in glioblastoma multiforme neurosphere cultures. Cancer Biology & Therapy, 14, 625–637. https://doi.org/10.4161/cbt.24595CrossRef Kristoffersen, K., Villingshøj, M., Poulsen, H. S., & Stockhausen, M. T. (2013). Level of Notch activation determines the effect on growth and stem cell-like features in glioblastoma multiforme neurosphere cultures. Cancer Biology & Therapy, 14, 625–637. https://​doi.​org/​10.​4161/​cbt.​24595CrossRef
110.
Zurück zum Zitat Xu, F., Shangguan, X., Pan, J., Yue, Z., Shen, K., Ji, Y., Zhang, W., Zhu, Y., Sha, J., Wang, Y., et al. (2021). HOXD13 suppresses prostate cancer metastasis and BMP4-induced epithelial-mesenchymal transition by inhibiting SMAD1. International Journal of Cancer, 148, 3060–3070. https://doi.org/10.1002/ijc.33494CrossRefPubMed Xu, F., Shangguan, X., Pan, J., Yue, Z., Shen, K., Ji, Y., Zhang, W., Zhu, Y., Sha, J., Wang, Y., et al. (2021). HOXD13 suppresses prostate cancer metastasis and BMP4-induced epithelial-mesenchymal transition by inhibiting SMAD1. International Journal of Cancer, 148, 3060–3070. https://​doi.​org/​10.​1002/​ijc.​33494CrossRefPubMed
112.
Zurück zum Zitat Xie, S. Y., Shi, D. B., Ouyang, Y., Lin, F., Chen, X. Y., Jiang, T. C., Xia, W., Guo, L., & Lin, H. X. (2022). SHMT2 promotes tumor growth through VEGF and MAPK signaling pathway in breast cancer. American Journal of Cancer Research, 12, 3405–3421.PubMedPubMedCentral Xie, S. Y., Shi, D. B., Ouyang, Y., Lin, F., Chen, X. Y., Jiang, T. C., Xia, W., Guo, L., & Lin, H. X. (2022). SHMT2 promotes tumor growth through VEGF and MAPK signaling pathway in breast cancer. American Journal of Cancer Research, 12, 3405–3421.PubMedPubMedCentral
116.
Zurück zum Zitat Chen, B., Liu, C., Bai, G., Zhu, Y., & Xu, H. (2021). BTBD7 accelerates the epithelial-mesenchymal transition, proliferation and invasion of prostate cancer cells. Journal of B.U.ON., 26, 2111–2116.PubMed Chen, B., Liu, C., Bai, G., Zhu, Y., & Xu, H. (2021). BTBD7 accelerates the epithelial-mesenchymal transition, proliferation and invasion of prostate cancer cells. Journal of B.U.ON., 26, 2111–2116.PubMed
124.
Zurück zum Zitat Choi, J. D., Kim, T. J., Jeong, B. C., Jeon, H. G., Jeon, S. S., Kang, M. Y., Yeom, S. Y., & Seo, S. I. (2021). ISL1 promotes enzalutamide resistance in castration-resistant prostate cancer (CRPC) through epithelial to mesenchymal transition (EMT). Science and Reports, 11, 21984. https://doi.org/10.1038/s41598-021-01003-0CrossRef Choi, J. D., Kim, T. J., Jeong, B. C., Jeon, H. G., Jeon, S. S., Kang, M. Y., Yeom, S. Y., & Seo, S. I. (2021). ISL1 promotes enzalutamide resistance in castration-resistant prostate cancer (CRPC) through epithelial to mesenchymal transition (EMT). Science and Reports, 11, 21984. https://​doi.​org/​10.​1038/​s41598-021-01003-0CrossRef
125.
Zurück zum Zitat Marín-Aguilera, M., Codony-Servat, J., Reig, Ò., Lozano, J. J., Fernández, P. L., Pereira, M. V., Jiménez, N., Donovan, M., Puig, P., Mengual, L., et al. (2014). Epithelial-to-mesenchymal transition mediates docetaxel resistance and high risk of relapse in prostate cancer. Molecular Cancer Therapeutics, 13, 1270–1284. https://doi.org/10.1158/1535-7163.Mct-13-0775CrossRefPubMed Marín-Aguilera, M., Codony-Servat, J., Reig, Ò., Lozano, J. J., Fernández, P. L., Pereira, M. V., Jiménez, N., Donovan, M., Puig, P., Mengual, L., et al. (2014). Epithelial-to-mesenchymal transition mediates docetaxel resistance and high risk of relapse in prostate cancer. Molecular Cancer Therapeutics, 13, 1270–1284. https://​doi.​org/​10.​1158/​1535-7163.​Mct-13-0775CrossRefPubMed
126.
127.
Zurück zum Zitat Shiota, M., Itsumi, M., Takeuchi, A., Imada, K., Yokomizo, A., Kuruma, H., Inokuchi, J., Tatsugami, K., Uchiumi, T., Oda, Y., et al. (2015). Crosstalk between epithelial-mesenchymal transition and castration resistance mediated by Twist1/AR signaling in prostate cancer. Endocrine-Related Cancer, 22, 889–900. https://doi.org/10.1530/erc-15-0225CrossRefPubMed Shiota, M., Itsumi, M., Takeuchi, A., Imada, K., Yokomizo, A., Kuruma, H., Inokuchi, J., Tatsugami, K., Uchiumi, T., Oda, Y., et al. (2015). Crosstalk between epithelial-mesenchymal transition and castration resistance mediated by Twist1/AR signaling in prostate cancer. Endocrine-Related Cancer, 22, 889–900. https://​doi.​org/​10.​1530/​erc-15-0225CrossRefPubMed
132.
Zurück zum Zitat Yadav, S., Kowolik, C. M., Lin, M., Zuro, D., Hui, S. K., Riggs, A. D., & Horne, D. A. (2019). SMC1A is associated with radioresistance in prostate cancer and acts by regulating epithelial-mesenchymal transition and cancer stem-like properties. Molecular Carcinogenesis, 58, 113–125. https://doi.org/10.1002/mc.22913CrossRefPubMed Yadav, S., Kowolik, C. M., Lin, M., Zuro, D., Hui, S. K., Riggs, A. D., & Horne, D. A. (2019). SMC1A is associated with radioresistance in prostate cancer and acts by regulating epithelial-mesenchymal transition and cancer stem-like properties. Molecular Carcinogenesis, 58, 113–125. https://​doi.​org/​10.​1002/​mc.​22913CrossRefPubMed
134.
135.
Zurück zum Zitat Zhang, H., Wei, X., Lu, S., Lin, X., Huang, J., Chen, L., Huang, X., Jiang, L., Li, Y., Qin, L., et al. (2019). Protective effect of DMDD, isolated from the root of Averrhoa carambola L., on high glucose induced EMT in HK-2 cells by inhibiting the TLR4-BAMBI-Smad2/3 signaling pathway. Biomedicine & Pharmacotherapy, 113, 108705. https://doi.org/10.1016/j.biopha.2019.108705CrossRef Zhang, H., Wei, X., Lu, S., Lin, X., Huang, J., Chen, L., Huang, X., Jiang, L., Li, Y., Qin, L., et al. (2019). Protective effect of DMDD, isolated from the root of Averrhoa carambola L., on high glucose induced EMT in HK-2 cells by inhibiting the TLR4-BAMBI-Smad2/3 signaling pathway. Biomedicine & Pharmacotherapy, 113, 108705. https://​doi.​org/​10.​1016/​j.​biopha.​2019.​108705CrossRef
138.
Zurück zum Zitat Wang, Y., Wang, G., Zhang, X., Zhou, X., Liu, Z., Huang, L., Liu, R., Lang, B., Xu, X., Liu, W., et al. (2015). γ-Secretase inhibitor inhibits bladder cancer cell drug resistance and invasion by reducing epithelial-mesenchymal transition. Molecular Medicine Reports, 12, 2821–2827. https://doi.org/10.3892/mmr.2015.3750CrossRefPubMed Wang, Y., Wang, G., Zhang, X., Zhou, X., Liu, Z., Huang, L., Liu, R., Lang, B., Xu, X., Liu, W., et al. (2015). γ-Secretase inhibitor inhibits bladder cancer cell drug resistance and invasion by reducing epithelial-mesenchymal transition. Molecular Medicine Reports, 12, 2821–2827. https://​doi.​org/​10.​3892/​mmr.​2015.​3750CrossRefPubMed
141.
Zurück zum Zitat Tsai, T. F., Chang, A. C., Chen, P. C., Ho, C. Y., Chen, H. E., Chou, K. Y., & Hwang, T. I. (2022). Autophagy blockade potentiates cancer-associated immunosuppression through programmed death ligand-1 upregulation in bladder cancer. Journal of Cellular Physiology, 237, 3587–3597. https://doi.org/10.1002/jcp.30817CrossRefPubMed Tsai, T. F., Chang, A. C., Chen, P. C., Ho, C. Y., Chen, H. E., Chou, K. Y., & Hwang, T. I. (2022). Autophagy blockade potentiates cancer-associated immunosuppression through programmed death ligand-1 upregulation in bladder cancer. Journal of Cellular Physiology, 237, 3587–3597. https://​doi.​org/​10.​1002/​jcp.​30817CrossRefPubMed
146.
Zurück zum Zitat Zhu, H., Jiang, W., Zhu, H., Hu, J., Tang, B., Zhou, Z., & He, X. (2022). Elevation of ADAM12 facilitates tumor progression by enhancing metastasis and immune infiltration in gastric cancer. International Journal of Oncology, 60, 1–13. https://doi.org/10.3892/ijo.2022.5341 Zhu, H., Jiang, W., Zhu, H., Hu, J., Tang, B., Zhou, Z., & He, X. (2022). Elevation of ADAM12 facilitates tumor progression by enhancing metastasis and immune infiltration in gastric cancer. International Journal of Oncology, 60, 1–13. https://​doi.​org/​10.​3892/​ijo.​2022.​5341
164.
Zurück zum Zitat Ohashi, S., Natsuizaka, M., Wong, G. S., Michaylira, C. Z., Grugan, K. D., Stairs, D. B., Kalabis, J., Vega, M. E., Kalman, R. A., Nakagawa, M., et al. (2010). Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Research, 70, 4174–4184. https://doi.org/10.1158/0008-5472.Can-09-4614CrossRefPubMedPubMedCentral Ohashi, S., Natsuizaka, M., Wong, G. S., Michaylira, C. Z., Grugan, K. D., Stairs, D. B., Kalabis, J., Vega, M. E., Kalman, R. A., Nakagawa, M., et al. (2010). Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Research, 70, 4174–4184. https://​doi.​org/​10.​1158/​0008-5472.​Can-09-4614CrossRefPubMedPubMedCentral
166.
168.
Zurück zum Zitat Voon, D. C., Wang, H., Koo, J. K., Nguyen, T. A., Hor, Y. T., Chu, Y. S., Ito, K., Fukamachi, H., Chan, S. L., Thiery, J. P., et al. (2012). Runx3 protects gastric epithelial cells against epithelial-mesenchymal transition-induced cellular plasticity and tumorigenicity. Stem Cells, 30, 2088–2099. https://doi.org/10.1002/stem.1183CrossRefPubMed Voon, D. C., Wang, H., Koo, J. K., Nguyen, T. A., Hor, Y. T., Chu, Y. S., Ito, K., Fukamachi, H., Chan, S. L., Thiery, J. P., et al. (2012). Runx3 protects gastric epithelial cells against epithelial-mesenchymal transition-induced cellular plasticity and tumorigenicity. Stem Cells, 30, 2088–2099. https://​doi.​org/​10.​1002/​stem.​1183CrossRefPubMed
170.
173.
Zurück zum Zitat Zhou, H., Lin, C., Zhang, Y., Zhang, X., Zhang, C., Zhang, P., Xie, X., & Ren, Z. (2017). miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression. Cell Proliferation, 50, e12341. https://doi.org/10.1111/cpr.12341 Zhou, H., Lin, C., Zhang, Y., Zhang, X., Zhang, C., Zhang, P., Xie, X., & Ren, Z. (2017). miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression. Cell Proliferation, 50, e12341. https://​doi.​org/​10.​1111/​cpr.​12341
175.
176.
Zurück zum Zitat Cozzolino, A. M., Alonzi, T., Santangelo, L., Mancone, C., Conti, B., Steindler, C., Musone, M., Cicchini, C., Tripodi, M., & Marchetti, A. (2013). TGFβ overrides HNF4α tumor suppressing activity through GSK3β inactivation: Implication for hepatocellular carcinoma gene therapy. Journal of Hepatology, 58, 65–72. https://doi.org/10.1016/j.jhep.2012.08.023CrossRefPubMed Cozzolino, A. M., Alonzi, T., Santangelo, L., Mancone, C., Conti, B., Steindler, C., Musone, M., Cicchini, C., Tripodi, M., & Marchetti, A. (2013). TGFβ overrides HNF4α tumor suppressing activity through GSK3β inactivation: Implication for hepatocellular carcinoma gene therapy. Journal of Hepatology, 58, 65–72. https://​doi.​org/​10.​1016/​j.​jhep.​2012.​08.​023CrossRefPubMed
183.
Zurück zum Zitat Davidowitz, R. A., Selfors, L. M., Iwanicki, M. P., Elias, K. M., Karst, A., Piao, H., Ince, T. A., Drage, M. G., Dering, J., Konecny, G. E., et al. (2014). Mesenchymal gene program-expressing ovarian cancer spheroids exhibit enhanced mesothelial clearance. The Journal of Clinical Investigation, 124, 2611–2625. https://doi.org/10.1172/jci69815CrossRefPubMedPubMedCentral Davidowitz, R. A., Selfors, L. M., Iwanicki, M. P., Elias, K. M., Karst, A., Piao, H., Ince, T. A., Drage, M. G., Dering, J., Konecny, G. E., et al. (2014). Mesenchymal gene program-expressing ovarian cancer spheroids exhibit enhanced mesothelial clearance. The Journal of Clinical Investigation, 124, 2611–2625. https://​doi.​org/​10.​1172/​jci69815CrossRefPubMedPubMedCentral
184.
Zurück zum Zitat Rodrigues, I. S., Lavorato-Rocha, A. M., de, M. M. B., Stiepcich, M. M., de Carvalho, F. M., Baiocchi, G., Soares, F. A., Rocha, R. M. (2013). Epithelial-mesenchymal transition-like events in vulvar cancer and its relation with HPV. British Journal of Cancer, 109: 184–194, https://doi.org/10.1038/bjc.2013.273. Rodrigues, I. S., Lavorato-Rocha, A. M., de, M. M. B., Stiepcich, M. M., de Carvalho, F. M., Baiocchi, G., Soares, F. A., Rocha, R. M. (2013). Epithelial-mesenchymal transition-like events in vulvar cancer and its relation with HPV. British Journal of Cancer, 109: 184–194, https://​doi.​org/​10.​1038/​bjc.​2013.​273.
185.
Zurück zum Zitat Dong, P., Kaneuchi, M., Xiong, Y., Cao, L., Cai, M., Liu, X., Guo, S. W., Ju, J., Jia, N., Konno, Y., et al. (2014). Identification of KLF17 as a novel epithelial to mesenchymal transition inducer via direct activation of TWIST1 in endometrioid endometrial cancer. Carcinogenesis, 35, 760–768. https://doi.org/10.1093/carcin/bgt369CrossRefPubMed Dong, P., Kaneuchi, M., Xiong, Y., Cao, L., Cai, M., Liu, X., Guo, S. W., Ju, J., Jia, N., Konno, Y., et al. (2014). Identification of KLF17 as a novel epithelial to mesenchymal transition inducer via direct activation of TWIST1 in endometrioid endometrial cancer. Carcinogenesis, 35, 760–768. https://​doi.​org/​10.​1093/​carcin/​bgt369CrossRefPubMed
190.
192.
196.
197.
198.
201.
Zurück zum Zitat Wei, C. R., Liu, J., & Yu, X. J. (2015). Targeting SLUG sensitizes leukemia cells to ADR-induced apoptosis. International Journal of Clinical and Experimental Medicine, 8, 22139–22148.PubMedPubMedCentral Wei, C. R., Liu, J., & Yu, X. J. (2015). Targeting SLUG sensitizes leukemia cells to ADR-induced apoptosis. International Journal of Clinical and Experimental Medicine, 8, 22139–22148.PubMedPubMedCentral
203.
Zurück zum Zitat Pérez-Mancera, P. A., Pérez-Caro, M., González-Herrero, I., Flores, T., Orfao, A., de Herreros, A. G., Gutiérrez-Adán, A., Pintado, B., Sagrera, A., Sánchez-Martín, M., et al. (2005). Cancer development induced by graded expression of Snail in mice. Human Molecular Genetics, 14, 3449–3461. https://doi.org/10.1093/hmg/ddi373CrossRefPubMed Pérez-Mancera, P. A., Pérez-Caro, M., González-Herrero, I., Flores, T., Orfao, A., de Herreros, A. G., Gutiérrez-Adán, A., Pintado, B., Sagrera, A., Sánchez-Martín, M., et al. (2005). Cancer development induced by graded expression of Snail in mice. Human Molecular Genetics, 14, 3449–3461. https://​doi.​org/​10.​1093/​hmg/​ddi373CrossRefPubMed
204.
Zurück zum Zitat Li, H., Mar, B. G., Zhang, H., Puram, R. V., Vazquez, F., Weir, B. A., Hahn, W. C., Ebert, B., & Pellman, D. (2017). The EMT regulator ZEB2 is a novel dependency of human and murine acute myeloid leukemia. Blood, the Journal of the American Society of Hematology, 129, 497–508. Li, H., Mar, B. G., Zhang, H., Puram, R. V., Vazquez, F., Weir, B. A., Hahn, W. C., Ebert, B., & Pellman, D. (2017). The EMT regulator ZEB2 is a novel dependency of human and murine acute myeloid leukemia. Blood, the Journal of the American Society of Hematology, 129, 497–508.
209.
Zurück zum Zitat Shahin, S. A., Wang, R., Simargi, S. I., Contreras, A., Parra Echavarria, L., Qu, L., Wen, W., Dellinger, T., Unternaehrer, J., Tamanoi, F., et al. (2018). Hyaluronic acid conjugated nanoparticle delivery of siRNA against TWIST reduces tumor burden and enhances sensitivity to cisplatin in ovarian cancer. Nanomedicine, 14, 1381–1394. https://doi.org/10.1016/j.nano.2018.04.008CrossRefPubMed Shahin, S. A., Wang, R., Simargi, S. I., Contreras, A., Parra Echavarria, L., Qu, L., Wen, W., Dellinger, T., Unternaehrer, J., Tamanoi, F., et al. (2018). Hyaluronic acid conjugated nanoparticle delivery of siRNA against TWIST reduces tumor burden and enhances sensitivity to cisplatin in ovarian cancer. Nanomedicine, 14, 1381–1394. https://​doi.​org/​10.​1016/​j.​nano.​2018.​04.​008CrossRefPubMed
217.
Zurück zum Zitat Mattijssen, S., Arimbasseri, A. G., Iben, J. R., Gaidamakov, S., Lee, J., Hafner, M., & Maraia, R. J. (2017). LARP4 mRNA codon-tRNA match contributes to LARP4 activity for ribosomal protein mRNA poly(A) tail length protection. Elife, 6, e28889. https://doi.org/10.7554/eLife.28889 Mattijssen, S., Arimbasseri, A. G., Iben, J. R., Gaidamakov, S., Lee, J., Hafner, M., & Maraia, R. J. (2017). LARP4 mRNA codon-tRNA match contributes to LARP4 activity for ribosomal protein mRNA poly(A) tail length protection. Elife, 6, e28889. https://​doi.​org/​10.​7554/​eLife.​28889
Metadaten
Titel
Harnessing function of EMT in cancer drug resistance: a metastasis regulator determines chemotherapy response
verfasst von
Nasim Ebrahimi
Mahdokht Sadat Manavi
Ferdos Faghihkhorasani
Siavash Seifollahy Fakhr
Fatemeh Jafari Baei
Fereshteh Faghih Khorasani
Mohammad Mehdi Zare
Nazanin Pazhouhesh Far
Fatemeh Rezaei-Tazangi
Jun Ren
Russel J. Reiter
Noushin Nabavi
Amir Reza Aref
Chu Chen
Yavuz Nuri Ertas
Qi Lu
Publikationsdatum
16.01.2024
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/s10555-023-10162-7

Weitere Artikel der Ausgabe 1/2024

Cancer and Metastasis Reviews 1/2024 Zur Ausgabe

EditorialNotes

Preface

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.