Skip to main content
Top
Published in: Cancer and Metastasis Reviews 3-4/2012

01-12-2012 | NON-THEMATIC REVIEW

The role of FOXP3 in the development and metastatic spread of breast cancer

Authors: Stephen Douglass, Simi Ali, Annette P. Meeson, David Browell, John A. Kirby

Published in: Cancer and Metastasis Reviews | Issue 3-4/2012

Login to get access

Abstract

The transcription factor FOXP3 is widely known for its role in the development and function of immunoregulatory T cells. However, it has been discovered recently that FOXP3 is also expressed in epithelial cells of the normal human breast, ovary and prostate. Aggressive cancer of these epithelial tissues often correlates with abnormal expression of FOXP3, which can be either absent or underexpressed at transcript or protein levels. It is becoming clear that this failure of normal FOXP3 expression can result in dysregulation of the expression of a range of oncogenes which have been implicated in the development and metastasis of cancer. Recent evidence suggests that FOXP3 might also regulate chemokine receptor expression, providing a possible explanation for the chemokine-driven, tissue-specific spread that is characteristic of many cancers. This review first summarises the general structure, function and properties of FOXP3. This is followed by an analysis of the tumour-suppressive properties of this transcription factor, with particular reference to the development and chemokine-mediated spread of human breast cancer. A final section focuses on potential applications of this new knowledge for therapeutic intervention.
Literature
1.
go back to reference Wang, L., et al. (2009). Somatic single hits inactivate the X-linked tumor suppressor FOXP3 in the prostate. Cancer Cell, 16(4), 336–346.PubMedCrossRef Wang, L., et al. (2009). Somatic single hits inactivate the X-linked tumor suppressor FOXP3 in the prostate. Cancer Cell, 16(4), 336–346.PubMedCrossRef
2.
go back to reference Liu, R., et al. (2009). FOXP3 up-regulates p21 expression by site-specific inhibition of histone deacetylase 2/histone deacetylase 4 association to the locus. Cancer Research, 69(6), 2252–2259.PubMedCrossRef Liu, R., et al. (2009). FOXP3 up-regulates p21 expression by site-specific inhibition of histone deacetylase 2/histone deacetylase 4 association to the locus. Cancer Research, 69(6), 2252–2259.PubMedCrossRef
3.
go back to reference Zuo, T., et al. (2007). FOXP3 is a novel transcriptional repressor for the breast cancer oncogene SKP2. The Journal of Clinical Investigation, 117(12), 3765–3773.PubMed Zuo, T., et al. (2007). FOXP3 is a novel transcriptional repressor for the breast cancer oncogene SKP2. The Journal of Clinical Investigation, 117(12), 3765–3773.PubMed
4.
go back to reference Zuo, T., et al. (2007). FOXP3 is an X-linked breast cancer suppressor gene and an important repressor of the HER-2/ErbB2 oncogene. Cell, 129(7), 1275–1286.PubMedCrossRef Zuo, T., et al. (2007). FOXP3 is an X-linked breast cancer suppressor gene and an important repressor of the HER-2/ErbB2 oncogene. Cell, 129(7), 1275–1286.PubMedCrossRef
5.
go back to reference Li, W., et al. (2011). Identification of a tumor suppressor relay between the FOXP3 and the Hippo pathways in breast and prostate cancers. Cancer Research, 71(6), 2162–2171.PubMedCrossRef Li, W., et al. (2011). Identification of a tumor suppressor relay between the FOXP3 and the Hippo pathways in breast and prostate cancers. Cancer Research, 71(6), 2162–2171.PubMedCrossRef
6.
go back to reference Kaestner, K. H., Knochel, W., & Martinez, D. E. (2000). Unified nomenclature for the winged helix/forkhead transcription factors. Genes & Development, 14(2), 142–146. Kaestner, K. H., Knochel, W., & Martinez, D. E. (2000). Unified nomenclature for the winged helix/forkhead transcription factors. Genes & Development, 14(2), 142–146.
7.
go back to reference Clark, K. L., et al. (1993). Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature, 364(6436), 412–420.PubMedCrossRef Clark, K. L., et al. (1993). Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature, 364(6436), 412–420.PubMedCrossRef
8.
go back to reference Lopes, J. E., et al. (2006). Analysis of FOXP3 reveals multiple domains required for its function as a transcriptional repressor. Journal of Immunology, 177(5), 3133–3142. Lopes, J. E., et al. (2006). Analysis of FOXP3 reveals multiple domains required for its function as a transcriptional repressor. Journal of Immunology, 177(5), 3133–3142.
9.
go back to reference Weigel, D., et al. (1989). The homeotic gene fork head encodes a nuclear protein and is expressed in the terminal regions of the Drosophila embryo. Cell, 57(4), 645–658.PubMedCrossRef Weigel, D., et al. (1989). The homeotic gene fork head encodes a nuclear protein and is expressed in the terminal regions of the Drosophila embryo. Cell, 57(4), 645–658.PubMedCrossRef
10.
go back to reference Coffer, P. J., & Burgering, B. M. (2004). Forkhead-box transcription factors and their role in the immune system. Nature Reviews Immunology, 4(11), 889–899.PubMedCrossRef Coffer, P. J., & Burgering, B. M. (2004). Forkhead-box transcription factors and their role in the immune system. Nature Reviews Immunology, 4(11), 889–899.PubMedCrossRef
11.
go back to reference Lin, L., & Peng, S. L. (2006). Coordination of NF-kappaB and NFAT antagonism by the forkhead transcription factor Foxd1. Journal of Immunology, 176(8), 4793–4803. Lin, L., & Peng, S. L. (2006). Coordination of NF-kappaB and NFAT antagonism by the forkhead transcription factor Foxd1. Journal of Immunology, 176(8), 4793–4803.
12.
go back to reference Hu, H., et al. (2006). Foxp1 is an essential transcriptional regulator of B cell development. Nature Immunology, 7(8), 819–826.PubMedCrossRef Hu, H., et al. (2006). Foxp1 is an essential transcriptional regulator of B cell development. Nature Immunology, 7(8), 819–826.PubMedCrossRef
13.
go back to reference Hong, H. K., et al. (2001). The winged helix/forkhead transcription factor Foxq1 regulates differentiation of hair in satin mice. Genesis, 29(4), 163–171.PubMedCrossRef Hong, H. K., et al. (2001). The winged helix/forkhead transcription factor Foxq1 regulates differentiation of hair in satin mice. Genesis, 29(4), 163–171.PubMedCrossRef
14.
go back to reference Hulander, M., et al. (1998). The winged helix transcription factor Fkh10 is required for normal development of the inner ear. Nature Genetics, 20(4), 374–376.PubMedCrossRef Hulander, M., et al. (1998). The winged helix transcription factor Fkh10 is required for normal development of the inner ear. Nature Genetics, 20(4), 374–376.PubMedCrossRef
15.
go back to reference Lai, C. S., et al. (2001). A forkhead-domain gene is mutated in a severe speech and language disorder. Nature, 413(6855), 519–523.PubMedCrossRef Lai, C. S., et al. (2001). A forkhead-domain gene is mutated in a severe speech and language disorder. Nature, 413(6855), 519–523.PubMedCrossRef
16.
go back to reference Chatila, T. A., et al. (2000). JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. The Journal of Clinical Investigation, 106(12), R75–R81.PubMedCrossRef Chatila, T. A., et al. (2000). JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. The Journal of Clinical Investigation, 106(12), R75–R81.PubMedCrossRef
17.
go back to reference Bennett, C. L., et al. (2001). A rare polyadenylation signal mutation of the FOXP3 gene (AAUAAA– > AAUGAA) leads to the IPEX syndrome. Immunogenetics, 53(6), 435–439.PubMedCrossRef Bennett, C. L., et al. (2001). A rare polyadenylation signal mutation of the FOXP3 gene (AAUAAA– > AAUGAA) leads to the IPEX syndrome. Immunogenetics, 53(6), 435–439.PubMedCrossRef
18.
go back to reference Bennett, C. L., et al. (2001). The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nature Genetics, 27(1), 20–21.PubMedCrossRef Bennett, C. L., et al. (2001). The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nature Genetics, 27(1), 20–21.PubMedCrossRef
19.
go back to reference Bennett, C. L., & Ochs, H. D. (2001). IPEX is a unique X-linked syndrome characterized by immune dysfunction, polyendocrinopathy, enteropathy, and a variety of autoimmune phenomena. Current Opinion in Pediatrics, 13(6), 533–538.PubMedCrossRef Bennett, C. L., & Ochs, H. D. (2001). IPEX is a unique X-linked syndrome characterized by immune dysfunction, polyendocrinopathy, enteropathy, and a variety of autoimmune phenomena. Current Opinion in Pediatrics, 13(6), 533–538.PubMedCrossRef
20.
go back to reference Sakaguchi, S. (2000). Regulatory T cells: Key controllers of immunologic self-tolerance. Cell, 101(5), 455–458.PubMedCrossRef Sakaguchi, S. (2000). Regulatory T cells: Key controllers of immunologic self-tolerance. Cell, 101(5), 455–458.PubMedCrossRef
21.
go back to reference Sakaguchi, S. (2005). Naturally arising Foxp3-expressing CD25 + CD4+ regulatory T cells in immunological tolerance to self and non-self. Nature Immunology, 6(4), 345–352.PubMedCrossRef Sakaguchi, S. (2005). Naturally arising Foxp3-expressing CD25 + CD4+ regulatory T cells in immunological tolerance to self and non-self. Nature Immunology, 6(4), 345–352.PubMedCrossRef
22.
go back to reference Chen, G. Y., et al. (2008). Cutting edge: Broad expression of the FoxP3 locus in epithelial cells: A caution against early interpretation of fatal inflammatory diseases following in vivo depletion of FoxP3-expressing cells. Journal of Immunology, 180(8), 5163–5166. Chen, G. Y., et al. (2008). Cutting edge: Broad expression of the FoxP3 locus in epithelial cells: A caution against early interpretation of fatal inflammatory diseases following in vivo depletion of FoxP3-expressing cells. Journal of Immunology, 180(8), 5163–5166.
23.
go back to reference Karanikas, V., et al. (2008). Foxp3 expression in human cancer cells. Journal of Translational Medicine, 6, 19.PubMedCrossRef Karanikas, V., et al. (2008). Foxp3 expression in human cancer cells. Journal of Translational Medicine, 6, 19.PubMedCrossRef
24.
go back to reference Zheng, Y., et al. (2007). Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature, 445(7130), 936–940.PubMedCrossRef Zheng, Y., et al. (2007). Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature, 445(7130), 936–940.PubMedCrossRef
25.
go back to reference Li, B., et al. (2007). FOXP3 interactions with histone acetyltransferase and class II histone deacetylases are required for repression. Proceedings of the National Academy of Sciences of the United States of America, 104(11), 4571–4576.PubMedCrossRef Li, B., et al. (2007). FOXP3 interactions with histone acetyltransferase and class II histone deacetylases are required for repression. Proceedings of the National Academy of Sciences of the United States of America, 104(11), 4571–4576.PubMedCrossRef
26.
go back to reference Bettelli, E., Dastrange, M., & Oukka, M. (2005). Foxp3 interacts with nuclear factor of activated T cells and NF-kappa B to repress cytokine gene expression and effector functions of T helper cells. Proceedings of the National Academy of Sciences of the United States of America, 102(14), 5138–5143.PubMedCrossRef Bettelli, E., Dastrange, M., & Oukka, M. (2005). Foxp3 interacts with nuclear factor of activated T cells and NF-kappa B to repress cytokine gene expression and effector functions of T helper cells. Proceedings of the National Academy of Sciences of the United States of America, 102(14), 5138–5143.PubMedCrossRef
27.
go back to reference Rudensky, A. Y., Gavin, M., & Zheng, Y. (2006). FOXP3 and NFAT: Partners in tolerance. Cell, 126(2), 253–256.PubMedCrossRef Rudensky, A. Y., Gavin, M., & Zheng, Y. (2006). FOXP3 and NFAT: Partners in tolerance. Cell, 126(2), 253–256.PubMedCrossRef
28.
go back to reference Wu, Y., et al. (2006). FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell, 126(2), 375–387.PubMedCrossRef Wu, Y., et al. (2006). FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell, 126(2), 375–387.PubMedCrossRef
29.
30.
go back to reference Bennett, C. L., et al. (2000). X-Linked syndrome of polyendocrinopathy, immune dysfunction, and diarrhea maps to Xp11.23-Xq13.3. American Journal of Human Genetics, 66(2), 461–468.PubMedCrossRef Bennett, C. L., et al. (2000). X-Linked syndrome of polyendocrinopathy, immune dysfunction, and diarrhea maps to Xp11.23-Xq13.3. American Journal of Human Genetics, 66(2), 461–468.PubMedCrossRef
31.
go back to reference Lal, G., & Bromberg, J. S. (2009). Epigenetic mechanisms of regulation of Foxp3 expression. Blood, 114(18), 3727–3735.PubMedCrossRef Lal, G., & Bromberg, J. S. (2009). Epigenetic mechanisms of regulation of Foxp3 expression. Blood, 114(18), 3727–3735.PubMedCrossRef
32.
go back to reference Ebert, L. M., et al. (2008). The regulatory T cell-associated transcription factor FoxP3 is expressed by tumor cells. Cancer Research, 68(8), 3001–3009.PubMedCrossRef Ebert, L. M., et al. (2008). The regulatory T cell-associated transcription factor FoxP3 is expressed by tumor cells. Cancer Research, 68(8), 3001–3009.PubMedCrossRef
33.
go back to reference Lee, S. M., Gao, B., & Fang, D. (2008). FoxP3 maintains Treg unresponsiveness by selectively inhibiting the promoter DNA-binding activity of AP-1. Blood, 111(7), 3599–3606.PubMedCrossRef Lee, S. M., Gao, B., & Fang, D. (2008). FoxP3 maintains Treg unresponsiveness by selectively inhibiting the promoter DNA-binding activity of AP-1. Blood, 111(7), 3599–3606.PubMedCrossRef
34.
go back to reference Allan, S. E., et al. (2005). The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. The Journal of Clinical Investigation, 115(11), 3276–3284.PubMedCrossRef Allan, S. E., et al. (2005). The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. The Journal of Clinical Investigation, 115(11), 3276–3284.PubMedCrossRef
35.
go back to reference Yagi, H., et al. (2004). Crucial role of FOXP3 in the development and function of human CD25 + CD4+ regulatory T cells. International Immunology, 16(11), 1643–1656.PubMedCrossRef Yagi, H., et al. (2004). Crucial role of FOXP3 in the development and function of human CD25 + CD4+ regulatory T cells. International Immunology, 16(11), 1643–1656.PubMedCrossRef
36.
go back to reference Du, J., et al. (2008). Isoform-specific inhibition of ROR alpha-mediated transcriptional activation by human FOXP3. Journal of Immunology, 180(7), 4785–4792. Du, J., et al. (2008). Isoform-specific inhibition of ROR alpha-mediated transcriptional activation by human FOXP3. Journal of Immunology, 180(7), 4785–4792.
37.
go back to reference Hancock, W. W., & Ozkaynak, E. (2009). Three distinct domains contribute to nuclear transport of murine Foxp3. PLoS One, 4(11), e7890.PubMedCrossRef Hancock, W. W., & Ozkaynak, E. (2009). Three distinct domains contribute to nuclear transport of murine Foxp3. PLoS One, 4(11), e7890.PubMedCrossRef
38.
go back to reference Banham, A. H., et al. (2009). Monoclonal antibodies raised to the human FOXP3 protein can be used effectively for detecting Foxp3(+) T cells in other mammalian species. Veterinary Immunology and Immunopathology, 127(3–4), 376–381.PubMedCrossRef Banham, A. H., et al. (2009). Monoclonal antibodies raised to the human FOXP3 protein can be used effectively for detecting Foxp3(+) T cells in other mammalian species. Veterinary Immunology and Immunopathology, 127(3–4), 376–381.PubMedCrossRef
39.
go back to reference Mikami, Y., et al. (2005). The functional region of CENP-H interacts with the Nuf2 complex that localizes to centromere during mitosis. Molecular and Cellular Biology, 25(5), 1958–1970.PubMedCrossRef Mikami, Y., et al. (2005). The functional region of CENP-H interacts with the Nuf2 complex that localizes to centromere during mitosis. Molecular and Cellular Biology, 25(5), 1958–1970.PubMedCrossRef
40.
go back to reference Song, J. H., Waataja, J. J., & Martemyanov, K. A. (2006). Subcellular targeting of RGS9-2 is controlled by multiple molecular determinants on its membrane anchor, R7BP. Journal of Biological Chemistry, 281(22), 15361–15369.PubMedCrossRef Song, J. H., Waataja, J. J., & Martemyanov, K. A. (2006). Subcellular targeting of RGS9-2 is controlled by multiple molecular determinants on its membrane anchor, R7BP. Journal of Biological Chemistry, 281(22), 15361–15369.PubMedCrossRef
41.
go back to reference Qian, X., & Costa, R. H. (1995). Analysis of hepatocyte nuclear factor-3 beta protein domains required for transcriptional activation and nuclear targeting. Nucleic Acids Research, 23(7), 1184–1191.PubMedCrossRef Qian, X., & Costa, R. H. (1995). Analysis of hepatocyte nuclear factor-3 beta protein domains required for transcriptional activation and nuclear targeting. Nucleic Acids Research, 23(7), 1184–1191.PubMedCrossRef
42.
go back to reference Dowsland, M. H., et al. (2003). Chemokines and breast cancer: A gateway to revolutionary targeted cancer treatments? Current Medicinal Chemistry, 10(7), 579–592.PubMedCrossRef Dowsland, M. H., et al. (2003). Chemokines and breast cancer: A gateway to revolutionary targeted cancer treatments? Current Medicinal Chemistry, 10(7), 579–592.PubMedCrossRef
43.
go back to reference Merlo, A., et al. (2009). FOXP3 expression and overall survival in breast cancer. Journal of Clinical Oncology, 27(11), 1746–1752.PubMedCrossRef Merlo, A., et al. (2009). FOXP3 expression and overall survival in breast cancer. Journal of Clinical Oncology, 27(11), 1746–1752.PubMedCrossRef
44.
go back to reference Zhang, H. Y., & Sun, H. (2010). Up-regulation of Foxp3 inhibits cell proliferation, migration and invasion in epithelial ovarian cancer. Cancer Letters, 287(1), 91–97.PubMedCrossRef Zhang, H. Y., & Sun, H. (2010). Up-regulation of Foxp3 inhibits cell proliferation, migration and invasion in epithelial ovarian cancer. Cancer Letters, 287(1), 91–97.PubMedCrossRef
45.
go back to reference Biyikoglu, B., et al. (2009). Gingival crevicular fluid MMP-8 and −13 and TIMP-1 levels in patients with rheumatoid arthritis and inflammatory periodontal disease. Journal of Periodontology, 80(8), 1307–1314.PubMedCrossRef Biyikoglu, B., et al. (2009). Gingival crevicular fluid MMP-8 and −13 and TIMP-1 levels in patients with rheumatoid arthritis and inflammatory periodontal disease. Journal of Periodontology, 80(8), 1307–1314.PubMedCrossRef
46.
go back to reference He, Y., et al. (2007). Interaction between cancer cells and stromal fibroblasts is required for activation of the uPAR-uPA-MMP-2 cascade in pancreatic cancer metastasis. Clinical Cancer Research, 13(11), 3115–3124.PubMedCrossRef He, Y., et al. (2007). Interaction between cancer cells and stromal fibroblasts is required for activation of the uPAR-uPA-MMP-2 cascade in pancreatic cancer metastasis. Clinical Cancer Research, 13(11), 3115–3124.PubMedCrossRef
47.
go back to reference Wu, X., et al. (2001). Generation of a prostate epithelial cell-specific Cre transgenic mouse model for tissue-specific gene ablation. Mechanisms of Development, 101(1–2), 61–69.PubMedCrossRef Wu, X., et al. (2001). Generation of a prostate epithelial cell-specific Cre transgenic mouse model for tissue-specific gene ablation. Mechanisms of Development, 101(1–2), 61–69.PubMedCrossRef
48.
go back to reference Hinz, S., et al. (2007). Foxp3 expression in pancreatic carcinoma cells as a novel mechanism of immune evasion in cancer. Cancer Research, 67(17), 8344–8350.PubMedCrossRef Hinz, S., et al. (2007). Foxp3 expression in pancreatic carcinoma cells as a novel mechanism of immune evasion in cancer. Cancer Research, 67(17), 8344–8350.PubMedCrossRef
49.
go back to reference Boumil, R. M., & Lee, J. T. (2001). Forty years of decoding the silence in X-chromosome inactivation. Human Molecular Genetics, 10(20), 2225–2232.PubMedCrossRef Boumil, R. M., & Lee, J. T. (2001). Forty years of decoding the silence in X-chromosome inactivation. Human Molecular Genetics, 10(20), 2225–2232.PubMedCrossRef
50.
go back to reference Knudson, A. G., Jr. (1971). Mutation and cancer: Statistical study of retinoblastoma. Proceedings of the National Academy of Sciences of the United States of America, 68(4), 820–823.PubMedCrossRef Knudson, A. G., Jr. (1971). Mutation and cancer: Statistical study of retinoblastoma. Proceedings of the National Academy of Sciences of the United States of America, 68(4), 820–823.PubMedCrossRef
51.
go back to reference Woolf, C. M. (1960). An investigation of the familial aspects of carcinoma of the prostate. Cancer, 13, 739–744.PubMedCrossRef Woolf, C. M. (1960). An investigation of the familial aspects of carcinoma of the prostate. Cancer, 13, 739–744.PubMedCrossRef
52.
go back to reference Xu, J., et al. (1998). Evidence for a prostate cancer susceptibility locus on the X chromosome. Nature Genetics, 20(2), 175–179.PubMedCrossRef Xu, J., et al. (1998). Evidence for a prostate cancer susceptibility locus on the X chromosome. Nature Genetics, 20(2), 175–179.PubMedCrossRef
53.
go back to reference Slamon, D. J., et al. (1987). Human breast cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science, 235(4785), 177–182.PubMedCrossRef Slamon, D. J., et al. (1987). Human breast cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science, 235(4785), 177–182.PubMedCrossRef
54.
go back to reference Press, M. F., et al. (1997). HER-2/neu gene amplification characterized by fluorescence in situ hybridization: Poor prognosis in node-negative breast carcinomas. Journal of Clinical Oncology, 15(8), 2894–2904.PubMed Press, M. F., et al. (1997). HER-2/neu gene amplification characterized by fluorescence in situ hybridization: Poor prognosis in node-negative breast carcinomas. Journal of Clinical Oncology, 15(8), 2894–2904.PubMed
55.
go back to reference Roy, V., & Perez, E. A. (2009). Beyond trastuzumab: Small molecule tyrosine kinase inhibitors in HER-2-positive breast cancer. The Oncologist, 14(11), 1061–1069.PubMedCrossRef Roy, V., & Perez, E. A. (2009). Beyond trastuzumab: Small molecule tyrosine kinase inhibitors in HER-2-positive breast cancer. The Oncologist, 14(11), 1061–1069.PubMedCrossRef
56.
go back to reference Nakayama, K. I., & Nakayama, K. (2006). Ubiquitin ligases: Cell-cycle control and cancer. Nature Reviews. Cancer, 6(5), 369–381.PubMedCrossRef Nakayama, K. I., & Nakayama, K. (2006). Ubiquitin ligases: Cell-cycle control and cancer. Nature Reviews. Cancer, 6(5), 369–381.PubMedCrossRef
57.
go back to reference Sonoda, H., et al. (2006). Significance of skp2 expression in primary breast cancer. Clinical Cancer Research, 12(4), 1215–1220.PubMedCrossRef Sonoda, H., et al. (2006). Significance of skp2 expression in primary breast cancer. Clinical Cancer Research, 12(4), 1215–1220.PubMedCrossRef
58.
go back to reference Radke, S., Pirkmaier, A., & Germain, D. (2005). Differential expression of the F-box proteins Skp2 and Skp2B in breast cancer. Oncogene, 24(21), 3448–3458.PubMedCrossRef Radke, S., Pirkmaier, A., & Germain, D. (2005). Differential expression of the F-box proteins Skp2 and Skp2B in breast cancer. Oncogene, 24(21), 3448–3458.PubMedCrossRef
59.
go back to reference Signoretti, S., et al. (2002). Oncogenic role of the ubiquitin ligase subunit Skp2 in human breast cancer. The Journal of Clinical Investigation, 110(5), 633–641.PubMed Signoretti, S., et al. (2002). Oncogenic role of the ubiquitin ligase subunit Skp2 in human breast cancer. The Journal of Clinical Investigation, 110(5), 633–641.PubMed
60.
go back to reference Nakayama, K., et al. (2004). Skp2-mediated degradation of p27 regulates progression into mitosis. Developmental Cell, 6(5), 661–672.PubMedCrossRef Nakayama, K., et al. (2004). Skp2-mediated degradation of p27 regulates progression into mitosis. Developmental Cell, 6(5), 661–672.PubMedCrossRef
61.
go back to reference Grandori, C., et al. (2000). The Myc/Max/Mad network and the transcriptional control of cell behavior. Annual Review of Cell and Developmental Biology, 16, 653–699.PubMedCrossRef Grandori, C., et al. (2000). The Myc/Max/Mad network and the transcriptional control of cell behavior. Annual Review of Cell and Developmental Biology, 16, 653–699.PubMedCrossRef
62.
go back to reference Jung, P., et al. (2008). AP4 encodes a c-MYC-inducible repressor of p21. Proceedings of the National Academy of Sciences of the United States of America, 105(39), 15046–15051.PubMedCrossRef Jung, P., et al. (2008). AP4 encodes a c-MYC-inducible repressor of p21. Proceedings of the National Academy of Sciences of the United States of America, 105(39), 15046–15051.PubMedCrossRef
63.
go back to reference Iavarone, A., & Massague, J. (1997). Repression of the CDK activator Cdc25A and cell-cycle arrest by cytokine TGF-beta in cells lacking the CDK inhibitor p15. Nature, 387(6631), 417–422.PubMedCrossRef Iavarone, A., & Massague, J. (1997). Repression of the CDK activator Cdc25A and cell-cycle arrest by cytokine TGF-beta in cells lacking the CDK inhibitor p15. Nature, 387(6631), 417–422.PubMedCrossRef
64.
go back to reference Pinto, A. E., et al. (2005). Correlations of cell cycle regulators (p53, p21, pRb and mdm2) and c-erbB-2 with biological markers of proliferation and overall survival in breast cancer. Pathology, 37(1), 45–50.PubMedCrossRef Pinto, A. E., et al. (2005). Correlations of cell cycle regulators (p53, p21, pRb and mdm2) and c-erbB-2 with biological markers of proliferation and overall survival in breast cancer. Pathology, 37(1), 45–50.PubMedCrossRef
65.
go back to reference Shiohara, M., et al. (1994). Absence of WAF1 mutations in a variety of human malignancies. Blood, 84(11), 3781–3784.PubMed Shiohara, M., et al. (1994). Absence of WAF1 mutations in a variety of human malignancies. Blood, 84(11), 3781–3784.PubMed
66.
go back to reference el-Deiry, W. S., et al. (1993). WAF1, a potential mediator of p53 tumor suppression. Cell, 75(4), 817–825.PubMedCrossRef el-Deiry, W. S., et al. (1993). WAF1, a potential mediator of p53 tumor suppression. Cell, 75(4), 817–825.PubMedCrossRef
67.
go back to reference Dulic, V., et al. (1994). p53-Dependent inhibition of cyclin-dependent kinase activities in human fibroblasts during radiation-induced G1 arrest. Cell, 76(6), 1013–1023.PubMedCrossRef Dulic, V., et al. (1994). p53-Dependent inhibition of cyclin-dependent kinase activities in human fibroblasts during radiation-induced G1 arrest. Cell, 76(6), 1013–1023.PubMedCrossRef
68.
go back to reference Somasundaram, K., et al. (1997). Arrest of the cell cycle by the tumour-suppressor BRCA1 requires the CDK-inhibitor p21WAF1/CiP1. Nature, 389(6647), 187–190.PubMedCrossRef Somasundaram, K., et al. (1997). Arrest of the cell cycle by the tumour-suppressor BRCA1 requires the CDK-inhibitor p21WAF1/CiP1. Nature, 389(6647), 187–190.PubMedCrossRef
69.
go back to reference Xiong, Y., et al. (1993). p21 is a universal inhibitor of cyclin kinases. Nature, 366(6456), 701–704.PubMedCrossRef Xiong, Y., et al. (1993). p21 is a universal inhibitor of cyclin kinases. Nature, 366(6456), 701–704.PubMedCrossRef
70.
go back to reference Cayrol, C., Knibiehler, M., & Ducommun, B. (1998). p21 binding to PCNA causes G1 and G2 cell cycle arrest in p53-deficient cells. Oncogene, 16(3), 311–320.PubMedCrossRef Cayrol, C., Knibiehler, M., & Ducommun, B. (1998). p21 binding to PCNA causes G1 and G2 cell cycle arrest in p53-deficient cells. Oncogene, 16(3), 311–320.PubMedCrossRef
71.
go back to reference Takahashi, Y., et al. (2005). Down-regulation of LATS1 and LATS2 mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers. Clinical Cancer Research, 11(4), 1380–1385.PubMedCrossRef Takahashi, Y., et al. (2005). Down-regulation of LATS1 and LATS2 mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers. Clinical Cancer Research, 11(4), 1380–1385.PubMedCrossRef
72.
go back to reference Powzaniuk, M., et al. (2004). The LATS2/KPM tumor suppressor is a negative regulator of the androgen receptor. Molecular Endocrinology, 18(8), 2011–2023.PubMedCrossRef Powzaniuk, M., et al. (2004). The LATS2/KPM tumor suppressor is a negative regulator of the androgen receptor. Molecular Endocrinology, 18(8), 2011–2023.PubMedCrossRef
73.
go back to reference Jiang, Z., et al. (2006). Promoter hypermethylation-mediated down-regulation of LATS1 and LATS2 in human astrocytoma. Neuroscience Research, 56(4), 450–458.PubMedCrossRef Jiang, Z., et al. (2006). Promoter hypermethylation-mediated down-regulation of LATS1 and LATS2 in human astrocytoma. Neuroscience Research, 56(4), 450–458.PubMedCrossRef
74.
go back to reference Jimenez-Velasco, A., et al. (2005). Downregulation of the large tumor suppressor 2 (LATS2/KPM) gene is associated with poor prognosis in acute lymphoblastic leukemia. Leukemia, 19(12), 2347–2350.PubMedCrossRef Jimenez-Velasco, A., et al. (2005). Downregulation of the large tumor suppressor 2 (LATS2/KPM) gene is associated with poor prognosis in acute lymphoblastic leukemia. Leukemia, 19(12), 2347–2350.PubMedCrossRef
75.
go back to reference Zhao, B., et al. (2007). Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes & Development, 21(21), 2747–2761.CrossRef Zhao, B., et al. (2007). Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes & Development, 21(21), 2747–2761.CrossRef
76.
go back to reference Steinhardt, A. A., et al. (2008). Expression of Yes-associated protein in common solid tumors. Human Pathology, 39(11), 1582–1589.PubMedCrossRef Steinhardt, A. A., et al. (2008). Expression of Yes-associated protein in common solid tumors. Human Pathology, 39(11), 1582–1589.PubMedCrossRef
77.
go back to reference Bruce, J., Carter, D. C., & Fraser, J. (1970). Patterns of recurrent disease in breast cancer. Lancet, 1(7644), 433–435.PubMedCrossRef Bruce, J., Carter, D. C., & Fraser, J. (1970). Patterns of recurrent disease in breast cancer. Lancet, 1(7644), 433–435.PubMedCrossRef
78.
go back to reference Campbell, J. J., & Butcher, E. C. (2000). Chemokines in tissue-specific and microenvironment-specific lymphocyte homing. Current Opinion in Immunology, 12(3), 336–341.PubMedCrossRef Campbell, J. J., & Butcher, E. C. (2000). Chemokines in tissue-specific and microenvironment-specific lymphocyte homing. Current Opinion in Immunology, 12(3), 336–341.PubMedCrossRef
79.
go back to reference Muller, A., et al. (2001). Involvement of chemokine receptors in breast cancer metastasis. Nature, 410(6824), 50–56.PubMedCrossRef Muller, A., et al. (2001). Involvement of chemokine receptors in breast cancer metastasis. Nature, 410(6824), 50–56.PubMedCrossRef
80.
go back to reference Nasser, M. W., et al. (2011). Crosstalk between chemokine receptor CXCR4 and cannabinoid receptor CB2 in modulating breast cancer growth and invasion. PLoS One, 6(9), e23901.PubMedCrossRef Nasser, M. W., et al. (2011). Crosstalk between chemokine receptor CXCR4 and cannabinoid receptor CB2 in modulating breast cancer growth and invasion. PLoS One, 6(9), e23901.PubMedCrossRef
81.
go back to reference Harvey, J. R., et al. (2007). Inhibition of CXCR4-mediated breast cancer metastasis: A potential role for heparinoids? Clinical Cancer Research, 13(5), 1562–1570.PubMedCrossRef Harvey, J. R., et al. (2007). Inhibition of CXCR4-mediated breast cancer metastasis: A potential role for heparinoids? Clinical Cancer Research, 13(5), 1562–1570.PubMedCrossRef
82.
go back to reference Overbeck-Zubrzycka, D., et al. (2010). FOXP3 regulates metastatic spread of breast cancer via control of expression of CXCR4 chemokine receptor. Cancer Prevention Research, 3(12). Overbeck-Zubrzycka, D., et al. (2010). FOXP3 regulates metastatic spread of breast cancer via control of expression of CXCR4 chemokine receptor. Cancer Prevention Research, 3(12).
83.
go back to reference Li, Y. M., et al. (2004). Upregulation of CXCR4 is essential for HER2-mediated tumor metastasis. Cancer Cell, 6(5), 459–469.PubMedCrossRef Li, Y. M., et al. (2004). Upregulation of CXCR4 is essential for HER2-mediated tumor metastasis. Cancer Cell, 6(5), 459–469.PubMedCrossRef
84.
go back to reference Mellor, P., et al. (2007). Modulatory effects of heparin and short-length oligosaccharides of heparin on the metastasis and growth of LMD MDA-MB 231 breast cancer cells in vivo. British Journal of Cancer, 97(6), 761–768.PubMedCrossRef Mellor, P., et al. (2007). Modulatory effects of heparin and short-length oligosaccharides of heparin on the metastasis and growth of LMD MDA-MB 231 breast cancer cells in vivo. British Journal of Cancer, 97(6), 761–768.PubMedCrossRef
85.
go back to reference Heinze, E., et al. (2009). Antibody-mediated FOXP3 protein therapy induces apoptosis in cancer cells in vitro and inhibits metastasis in vivo. International Journal of Oncology, 35(1), 167–173.PubMed Heinze, E., et al. (2009). Antibody-mediated FOXP3 protein therapy induces apoptosis in cancer cells in vitro and inhibits metastasis in vivo. International Journal of Oncology, 35(1), 167–173.PubMed
86.
go back to reference Jung, D. J., et al. (2010). Foxp3 expression in p53-dependent DNA damage responses. Journal of Biological Chemistry, 285(11), 7995–8002.PubMedCrossRef Jung, D. J., et al. (2010). Foxp3 expression in p53-dependent DNA damage responses. Journal of Biological Chemistry, 285(11), 7995–8002.PubMedCrossRef
87.
go back to reference Liu, Y., et al. (2009). Activating transcription factor 2 and c-Jun-mediated induction of FoxP3 for experimental therapy of mammary tumor in the mouse. Cancer Research, 69(14), 5954–5960.PubMedCrossRef Liu, Y., et al. (2009). Activating transcription factor 2 and c-Jun-mediated induction of FoxP3 for experimental therapy of mammary tumor in the mouse. Cancer Research, 69(14), 5954–5960.PubMedCrossRef
Metadata
Title
The role of FOXP3 in the development and metastatic spread of breast cancer
Authors
Stephen Douglass
Simi Ali
Annette P. Meeson
David Browell
John A. Kirby
Publication date
01-12-2012
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 3-4/2012
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-012-9395-3

Other articles of this Issue 3-4/2012

Cancer and Metastasis Reviews 3-4/2012 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