Skip to main content
Top
Published in: Breast Cancer Research 6/2005

Open Access 01-12-2005 | Research article

Effects of combined treatment with rapamycin and cotylenin A, a novel differentiation-inducing agent, on human breast carcinoma MCF-7 cells and xenografts

Authors: Takashi Kasukabe, Junko Okabe-Kado, Nobuo Kato, Takeshi Sassa, Yoshio Honma

Published in: Breast Cancer Research | Issue 6/2005

Login to get access

Abstract

Introduction

Rapamycin, an inhibitor of the serine/threonine kinase target of rapamycin, induces G1 arrest and/or apoptosis. Although rapamycin and its analogues are attractive candidates for cancer therapy, their sensitivities with respect to growth inhibition differ markedly among various cancer cells. Using human breast carcinoma cell line MCF-7 as an experimental model system, we examined the growth-inhibitory effects of combinations of various agents and rapamycin to find the agent that most potently enhances the growth-inhibitory effect of rapamycin.

Method

We evaluated the growth-inhibitory effect of rapamycin plus various agents, including cotylenin A (a novel inducer of differentiation of myeloid leukaemia cells) to MCF-7 cells, using either MTT assay or trypan blue dye exclusion test. The cell cycle was analyzed using propidium iodide-stained nuclei. Expressions of several genes in MCF-7 cells with rapamycin plus cotylenin A were studied using cDNA microarray analysis and RT-PCR. The in vitro results of MCF-7 cells treated with rapamycin plus cotylenin A were further confirmed in vivo in a mouse xenograft model.

Results

We found that the sensitivity of rapamycin to MCF-7 cells was markedly affected by cotylenin A. This treatment induced growth arrest of the cells at the G1 phase, rather than apoptosis, and induced senescence-associated β-galactosidase activity. We examined the gene expression profiles associated with exposure to rapamycin and cotylenin A using cDNA microarrays. We found that expressions of cyclin G2, transforming growth factor-β-induced 68 kDa protein, BCL2-interacting killer, and growth factor receptor-bound 7 were markedly induced in MCF-7 cells treated with rapamycin plus cotylenin A. Furthermore, combined treatment with rapamycin and cotylenin A significantly inhibited the growth of MCF-7 cells as xenografts, without apparent adverse effects.

Conclusion

Rapamycin and cotylenin A cooperatively induced growth arrest in breast carcinoma MCF-7 cells in vitro, and treatment with rapamycin and cotylenin A combined more strongly inhibited the growth of MCF-7 cells as xenografts in vivo than treatment with rapamycin or cotylenin A alone, suggesting that this combination may have therapeutic value in treating breast cancer. We also identified several genes that were markedly modulated in MCF-7 cells treated with rapamycin plus cotylenin A.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jemal A, Clegg LX, Ward E, Ries LAG, Wu X, Jamison PM, Wingo P A, Howe HE, Anderson RN, Edwards BK: Annual report to nation on the states of cancer, 1975-with a special feature regarding survival. Cancer. 2004, 101: 3-27. 10.1002/cncr.20288.CrossRefPubMed Jemal A, Clegg LX, Ward E, Ries LAG, Wu X, Jamison PM, Wingo P A, Howe HE, Anderson RN, Edwards BK: Annual report to nation on the states of cancer, 1975-with a special feature regarding survival. Cancer. 2004, 101: 3-27. 10.1002/cncr.20288.CrossRefPubMed
2.
go back to reference Christensen GL, Jepsen JS, Fog CK, Christensen IJ, Lykkesfeldt AE: Sequential versus combined treatment of human breast cancer cells with antiestrogens and the vitamin D analogue EB1089 and evaluation of predictive markers for vitamin D treatment. Breast Cancer Res Treat. 2004, 85: 53-63. 10.1023/B:BREA.0000021047.37869.95.CrossRefPubMed Christensen GL, Jepsen JS, Fog CK, Christensen IJ, Lykkesfeldt AE: Sequential versus combined treatment of human breast cancer cells with antiestrogens and the vitamin D analogue EB1089 and evaluation of predictive markers for vitamin D treatment. Breast Cancer Res Treat. 2004, 85: 53-63. 10.1023/B:BREA.0000021047.37869.95.CrossRefPubMed
3.
go back to reference Dabrosin C, Johansson A, Olinger K: Decreased secretion of cathepsin D in breast cancer in vivo by tamoxifen: mediated by the mannose-6-phosphate/IGF-II receptor?. Breast Cancer Res Treat. 2004, 85: 229-238. 10.1023/B:BREA.0000025417.82291.36.CrossRefPubMed Dabrosin C, Johansson A, Olinger K: Decreased secretion of cathepsin D in breast cancer in vivo by tamoxifen: mediated by the mannose-6-phosphate/IGF-II receptor?. Breast Cancer Res Treat. 2004, 85: 229-238. 10.1023/B:BREA.0000025417.82291.36.CrossRefPubMed
4.
go back to reference Yu K, Toral-Barza L, Discafani C, Zhang W-G, Skotnicki J, Frost P, Gibbons JJ: mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer. 2001, 8: 249-258. 10.1677/erc.0.0080249.CrossRefPubMed Yu K, Toral-Barza L, Discafani C, Zhang W-G, Skotnicki J, Frost P, Gibbons JJ: mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer. 2001, 8: 249-258. 10.1677/erc.0.0080249.CrossRefPubMed
5.
go back to reference Noh W-C, Mondesire WH, Peng J, Jian W, Zhang H, Dong J, Mills GB, Hung M-C, Meric-Bernstam F: Determinants of rapamycin sensitivity in breast cancer cells. Clin Cancer Res. 2004, 10: 1013-1023. 10.1158/1078-0432.CCR-03-0043.CrossRefPubMed Noh W-C, Mondesire WH, Peng J, Jian W, Zhang H, Dong J, Mills GB, Hung M-C, Meric-Bernstam F: Determinants of rapamycin sensitivity in breast cancer cells. Clin Cancer Res. 2004, 10: 1013-1023. 10.1158/1078-0432.CCR-03-0043.CrossRefPubMed
6.
go back to reference Huang S, Houghton PJ: Inhibitors of mammalian target of rapamycin as novel antitumor agents. Curr Opin Investig Drugs. 2002, 3: 295-304.PubMed Huang S, Houghton PJ: Inhibitors of mammalian target of rapamycin as novel antitumor agents. Curr Opin Investig Drugs. 2002, 3: 295-304.PubMed
7.
go back to reference Huang S, Houghton P: Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol. 2003, 3: 371-377. 10.1016/S1471-4892(03)00071-7.CrossRefPubMed Huang S, Houghton P: Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol. 2003, 3: 371-377. 10.1016/S1471-4892(03)00071-7.CrossRefPubMed
8.
go back to reference Okabe-Kado J, Hayashi M, Honma Y, Hozumi M: Enhancement by hemin on the sensitivity of K562 human leukemic cells to 1-β-D-arabinofuranocylcytosine. Cancer Res. 1986, 46: 1239-1243.PubMed Okabe-Kado J, Hayashi M, Honma Y, Hozumi M: Enhancement by hemin on the sensitivity of K562 human leukemic cells to 1-β-D-arabinofuranocylcytosine. Cancer Res. 1986, 46: 1239-1243.PubMed
9.
go back to reference Okabe-Kado J, Hayashi M, Honma Y, Hozumi M, Tsuruo T: Inhibition by erythroid differentiation factor (activin A) of p-glycoprotein expression in multidrug-resistant human K562 erythroleukemia cells. Cancer Res. 1991, 51: 2582-2586.PubMed Okabe-Kado J, Hayashi M, Honma Y, Hozumi M, Tsuruo T: Inhibition by erythroid differentiation factor (activin A) of p-glycoprotein expression in multidrug-resistant human K562 erythroleukemia cells. Cancer Res. 1991, 51: 2582-2586.PubMed
10.
go back to reference Sassa T, Tojyo T, Munakata K: Isolation of a new plant growth substance with cytokinin-like activity. Nature. 1970, 227: 379-10.1038/227379a0.CrossRefPubMed Sassa T, Tojyo T, Munakata K: Isolation of a new plant growth substance with cytokinin-like activity. Nature. 1970, 227: 379-10.1038/227379a0.CrossRefPubMed
11.
go back to reference Sassa T, Ooi T, Nukina M, Ikeda M, Kato N: Structural confirmation of cotylenin A, a novel fusicoccane-diterpene glycoside with potent plant growth-regulating activity from Cladosporium fungus sp. 501-7W. Biosci Biotechnol Biochem. 1998, 62: 1815-1818. 10.1271/bbb.62.1815.CrossRefPubMed Sassa T, Ooi T, Nukina M, Ikeda M, Kato N: Structural confirmation of cotylenin A, a novel fusicoccane-diterpene glycoside with potent plant growth-regulating activity from Cladosporium fungus sp. 501-7W. Biosci Biotechnol Biochem. 1998, 62: 1815-1818. 10.1271/bbb.62.1815.CrossRefPubMed
12.
go back to reference Asahi K, Honma Y, Hazeki K, Sassa T, Kubohara Y, Sakurai A, Takahashi N: Cotylenin A, a plant-growth regulator, induces differentiation in murine and human myeloid leukemia cells. Biochem Biophys Res Commun. 1997, 238: 758-763. 10.1006/bbrc.1997.7385.CrossRefPubMed Asahi K, Honma Y, Hazeki K, Sassa T, Kubohara Y, Sakurai A, Takahashi N: Cotylenin A, a plant-growth regulator, induces differentiation in murine and human myeloid leukemia cells. Biochem Biophys Res Commun. 1997, 238: 758-763. 10.1006/bbrc.1997.7385.CrossRefPubMed
13.
go back to reference Yamamoto-Yamaguchi Y, Yamada K, Ishii Y, Asahi K, Tomoyasu S, Honma Y: Induction of monocytic differentiation of myeloid leukaemia cells by cotylenin A, a plant growth regulator. Br J Haematol. 2001, 112: 697-705. 10.1046/j.1365-2141.2001.02601.x.CrossRefPubMed Yamamoto-Yamaguchi Y, Yamada K, Ishii Y, Asahi K, Tomoyasu S, Honma Y: Induction of monocytic differentiation of myeloid leukaemia cells by cotylenin A, a plant growth regulator. Br J Haematol. 2001, 112: 697-705. 10.1046/j.1365-2141.2001.02601.x.CrossRefPubMed
14.
go back to reference Honma Y: Cotylenin A: a plant growth regulator as a differentiation-inducing agent against myeloid leukemia. Leuk Lymphoma. 2002, 43: 1169-1178. 10.1080/10428190290026222.CrossRefPubMed Honma Y: Cotylenin A: a plant growth regulator as a differentiation-inducing agent against myeloid leukemia. Leuk Lymphoma. 2002, 43: 1169-1178. 10.1080/10428190290026222.CrossRefPubMed
15.
go back to reference Yamada K, Honma Y, Asahi K, Sassa T, Hino K, Tomoyasu S: Differentiation of human acute myeloid leukaemia cells in primary culture in response to cotylenin A, a plant growth regulator. Br J Haematol. 2001, 114: 814-821. 10.1046/j.1365-2141.2001.03029.x.CrossRefPubMed Yamada K, Honma Y, Asahi K, Sassa T, Hino K, Tomoyasu S: Differentiation of human acute myeloid leukaemia cells in primary culture in response to cotylenin A, a plant growth regulator. Br J Haematol. 2001, 114: 814-821. 10.1046/j.1365-2141.2001.03029.x.CrossRefPubMed
16.
go back to reference Honma Y, Ishii Y, Sassa T, Asahi K: Treatment of human promyelocytic leukemia in the SCID mouse model with cotylenin A, an inducer of myelomonocytic differentiation of leukemia cells. Leuk Res. 2003, 27: 1019-1025. 10.1016/S0145-2126(03)00071-7.CrossRefPubMed Honma Y, Ishii Y, Sassa T, Asahi K: Treatment of human promyelocytic leukemia in the SCID mouse model with cotylenin A, an inducer of myelomonocytic differentiation of leukemia cells. Leuk Res. 2003, 27: 1019-1025. 10.1016/S0145-2126(03)00071-7.CrossRefPubMed
17.
go back to reference Honma Y, Ishii Y, Yamamoto-Yamaguchi Y, Sassa T, Asahi K: Cotylenin A, a differentiation-inducing agent, and IFN-α cooperatively induce apoptosis and have an antitumor effect on human non-small cell lung carcinoma cells in nude mice. Cancer Res. 2003, 63: 3659-3666.PubMed Honma Y, Ishii Y, Yamamoto-Yamaguchi Y, Sassa T, Asahi K: Cotylenin A, a differentiation-inducing agent, and IFN-α cooperatively induce apoptosis and have an antitumor effect on human non-small cell lung carcinoma cells in nude mice. Cancer Res. 2003, 63: 3659-3666.PubMed
18.
go back to reference Chou TC, Talalay P: Quantitative analysis of dose-effect relationship: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984, 22: 27-55. 10.1016/0065-2571(84)90007-4.CrossRefPubMed Chou TC, Talalay P: Quantitative analysis of dose-effect relationship: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984, 22: 27-55. 10.1016/0065-2571(84)90007-4.CrossRefPubMed
19.
go back to reference Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubeji I, Perrira-Smith O, et al: A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995, 92: 9363-9367.CrossRefPubMedPubMedCentral Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubeji I, Perrira-Smith O, et al: A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995, 92: 9363-9367.CrossRefPubMedPubMedCentral
20.
go back to reference Nakamaki T, Okabe-Kado J, Yamamoto-Yamaguchi Y, Hino K, Tomoyasu S, Honma Y, Kasukabe T: Role of MmTRA1b/phospholipid scramblase 1 gene expression in the induction of differentiation of human myeloid leukemia cells into granulocytes. Exp Hematol. 2002, 30: 421-429. 10.1016/S0301-472X(02)00779-8.CrossRefPubMed Nakamaki T, Okabe-Kado J, Yamamoto-Yamaguchi Y, Hino K, Tomoyasu S, Honma Y, Kasukabe T: Role of MmTRA1b/phospholipid scramblase 1 gene expression in the induction of differentiation of human myeloid leukemia cells into granulocytes. Exp Hematol. 2002, 30: 421-429. 10.1016/S0301-472X(02)00779-8.CrossRefPubMed
21.
go back to reference Li C, Shridhar K, Liu J: Molecular characterization of oncostatin M-induced growth arrest of MCF cells expressing a temperature-sensitive mutant of p53. Breast Cancer Res Treat. 2003, 80: 23-37. 10.1023/A:1024483017549.CrossRefPubMed Li C, Shridhar K, Liu J: Molecular characterization of oncostatin M-induced growth arrest of MCF cells expressing a temperature-sensitive mutant of p53. Breast Cancer Res Treat. 2003, 80: 23-37. 10.1023/A:1024483017549.CrossRefPubMed
22.
go back to reference Gera JF, Mellinghoff IK, Shi Y, Rettig MB, Trans C, Hsu J, Sawyers CL, Lichtenstein AK: AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem. 2004, 279: 2737-2746. 10.1074/jbc.M309999200.CrossRefPubMed Gera JF, Mellinghoff IK, Shi Y, Rettig MB, Trans C, Hsu J, Sawyers CL, Lichtenstein AK: AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem. 2004, 279: 2737-2746. 10.1074/jbc.M309999200.CrossRefPubMed
23.
go back to reference Skonier J, Bennett K, Rothwell V, Kosowski S, Plowman G, Wallace P, Edelhoff S, Disteche C, Neubauer M, Marquaedt H: beta ig-h3: a transforming growth factor-beta-responsive gene encoding a secreted protein that inhibits cell attachment in vitro and suppresses the growth of CHO cells in nude mice. DNA Cell Biol. 1994, 13: 571-584.CrossRefPubMed Skonier J, Bennett K, Rothwell V, Kosowski S, Plowman G, Wallace P, Edelhoff S, Disteche C, Neubauer M, Marquaedt H: beta ig-h3: a transforming growth factor-beta-responsive gene encoding a secreted protein that inhibits cell attachment in vitro and suppresses the growth of CHO cells in nude mice. DNA Cell Biol. 1994, 13: 571-584.CrossRefPubMed
24.
go back to reference Boyd JM, Gallo GJ, Elangovan B, Houghton AB, Malstrom S, Avery BJ, Ebb RG, Subramanian T, Chittenden T, Lutz RJ: Bik, a novel death-inducing protein shares a distinct sequence motif with Bcl-2 family proteins and interacts with viral and cellular survival-promoting proteins. Oncogene. 1995, 11: 1921-1928.PubMed Boyd JM, Gallo GJ, Elangovan B, Houghton AB, Malstrom S, Avery BJ, Ebb RG, Subramanian T, Chittenden T, Lutz RJ: Bik, a novel death-inducing protein shares a distinct sequence motif with Bcl-2 family proteins and interacts with viral and cellular survival-promoting proteins. Oncogene. 1995, 11: 1921-1928.PubMed
25.
go back to reference Horne MC, Donaldson KL, Goolsby GM, Tran D, Mulheisen M, Hell JW, Wahl AF: Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated cell cycle arrest. J Biol Chem. 1997, 272: 12650-12661. 10.1074/jbc.272.19.12650.CrossRefPubMed Horne MC, Donaldson KL, Goolsby GM, Tran D, Mulheisen M, Hell JW, Wahl AF: Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated cell cycle arrest. J Biol Chem. 1997, 272: 12650-12661. 10.1074/jbc.272.19.12650.CrossRefPubMed
26.
go back to reference Martinez-Gac L, Marques M, Garcia Z, Campanero MR, Carrera AC: Control of cyclin G2 mRNA expression by forkhead transcription factors: novel mechanism for cell cycle control by phosphoinositide 3-kinase and forkhead. Mol Cell Biol. 2004, 24: 2181-2189. 10.1128/MCB.24.5.2181-2189.2004.CrossRefPubMedPubMedCentral Martinez-Gac L, Marques M, Garcia Z, Campanero MR, Carrera AC: Control of cyclin G2 mRNA expression by forkhead transcription factors: novel mechanism for cell cycle control by phosphoinositide 3-kinase and forkhead. Mol Cell Biol. 2004, 24: 2181-2189. 10.1128/MCB.24.5.2181-2189.2004.CrossRefPubMedPubMedCentral
27.
go back to reference Shen T-L, Han DC, Guan J-L: Association of Grb7 with phosphoinositides and its role in the regulation of cell migration. J Biol Chem. 2002, 277: 29069-29077. 10.1074/jbc.M203085200.CrossRefPubMed Shen T-L, Han DC, Guan J-L: Association of Grb7 with phosphoinositides and its role in the regulation of cell migration. J Biol Chem. 2002, 277: 29069-29077. 10.1074/jbc.M203085200.CrossRefPubMed
28.
go back to reference Patwardhan S, Gashler A, Siegel MG, Chang LC, Joseph LJ, Shows TB, Le Beau MM, Sukhatme VP: EGR3, a novel member of the Egr family of genes encoding immediate-early transcription factors. Oncogene. 1991, 6: 917-928.PubMed Patwardhan S, Gashler A, Siegel MG, Chang LC, Joseph LJ, Shows TB, Le Beau MM, Sukhatme VP: EGR3, a novel member of the Egr family of genes encoding immediate-early transcription factors. Oncogene. 1991, 6: 917-928.PubMed
29.
go back to reference Luo J, Soh J, Xing W, Mao Y, Matsuno T, Weinstein IB: PM-3, a benzo-γ-pyran derivative isolated propolis, inhibits growth of MCF-7 human breast cancer cells. Anticancer Res. 2001, 21: 1665-1672.PubMed Luo J, Soh J, Xing W, Mao Y, Matsuno T, Weinstein IB: PM-3, a benzo-γ-pyran derivative isolated propolis, inhibits growth of MCF-7 human breast cancer cells. Anticancer Res. 2001, 21: 1665-1672.PubMed
30.
go back to reference Carlson B, Lahusen T, Singh S, Loaiza-Perez A, Worland PJ, Pestell R, Albanese C, Sausville WA, Senderowicz AM: Down-regulation of cyclin D1 by transcriptional repression in MCF-7 human breast carcinoma cells induced by flavopiridol. Cancer Res. 1999, 59: 4634-4641.PubMed Carlson B, Lahusen T, Singh S, Loaiza-Perez A, Worland PJ, Pestell R, Albanese C, Sausville WA, Senderowicz AM: Down-regulation of cyclin D1 by transcriptional repression in MCF-7 human breast carcinoma cells induced by flavopiridol. Cancer Res. 1999, 59: 4634-4641.PubMed
31.
go back to reference Monks A, Harris E, Hose C, Connelly J, Sausville ED: Genotoxic profiling of MCF-7 breast cancer cell line elucidates gene expression modifications underlying toxicity of the anticancer drug 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole. Mol Pharmacol. 2003, 63: 766-772. 10.1124/mol.63.3.766.CrossRefPubMed Monks A, Harris E, Hose C, Connelly J, Sausville ED: Genotoxic profiling of MCF-7 breast cancer cell line elucidates gene expression modifications underlying toxicity of the anticancer drug 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole. Mol Pharmacol. 2003, 63: 766-772. 10.1124/mol.63.3.766.CrossRefPubMed
32.
go back to reference Bennin DA, Arachchige Don AS, Brake T, McKenzie JL, Rosenbaum H, Ortiz L, DePaoli-Roach AA, Horne MC: Cyclin G2 associates with protein phosphatase 2A catalytic and regulatory B' subunits in active complexes and induces nuclear aberrations and a G1/S phase cell cycle arrest. J Biol Chem. 2002, 277: 27449-27467. 10.1074/jbc.M111693200.CrossRefPubMed Bennin DA, Arachchige Don AS, Brake T, McKenzie JL, Rosenbaum H, Ortiz L, DePaoli-Roach AA, Horne MC: Cyclin G2 associates with protein phosphatase 2A catalytic and regulatory B' subunits in active complexes and induces nuclear aberrations and a G1/S phase cell cycle arrest. J Biol Chem. 2002, 277: 27449-27467. 10.1074/jbc.M111693200.CrossRefPubMed
33.
go back to reference Frasor J, Danes JM, Komm B, Chang KCN, Lyttle CR, Katzenellenbogen BS: Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic into control of proliferation and cell phenotype. Endocrinology. 2003, 144: 4562-4574. 10.1210/en.2003-0567.CrossRefPubMed Frasor J, Danes JM, Komm B, Chang KCN, Lyttle CR, Katzenellenbogen BS: Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic into control of proliferation and cell phenotype. Endocrinology. 2003, 144: 4562-4574. 10.1210/en.2003-0567.CrossRefPubMed
34.
go back to reference Maxwell PJ, Longley DB, Latif T, Boyer J, Allen W, Lynch M, McDermott U, Harkin DP, Allegra CJ, Johnston PG: Identification of 5-fluorouracil-inducible target genes using cDNA microarray profiling. Cancer Res. 2003, 63: 4602-4606.PubMed Maxwell PJ, Longley DB, Latif T, Boyer J, Allen W, Lynch M, McDermott U, Harkin DP, Allegra CJ, Johnston PG: Identification of 5-fluorouracil-inducible target genes using cDNA microarray profiling. Cancer Res. 2003, 63: 4602-4606.PubMed
35.
go back to reference Dieudonne SC, Kerr JM, Xu T, Sommer B, DeRubeis AR, Kuznetsov SA, Kim IS, Gehron Robey P, Young MF: Differential display of human marrow stroma cells reveals unique mRNA expression patterns in response to dexamethasone. J Cell Biochem. 1999, 76: 231-243. 10.1002/(SICI)1097-4644(20000201)76:2<231::AID-JCB7>3.0.CO;2-X.CrossRefPubMed Dieudonne SC, Kerr JM, Xu T, Sommer B, DeRubeis AR, Kuznetsov SA, Kim IS, Gehron Robey P, Young MF: Differential display of human marrow stroma cells reveals unique mRNA expression patterns in response to dexamethasone. J Cell Biochem. 1999, 76: 231-243. 10.1002/(SICI)1097-4644(20000201)76:2<231::AID-JCB7>3.0.CO;2-X.CrossRefPubMed
36.
go back to reference Kim J, Kim S, Jeong H, Lee B, Choi J, Park R, Park J, Kim I: RGD peptides released from βig-h3, a TGF-β-induced cell adhesive molecule, mediate apoptosis. Oncogene. 2003, 22: 2045-2053. 10.1038/sj.onc.1206269.CrossRefPubMed Kim J, Kim S, Jeong H, Lee B, Choi J, Park R, Park J, Kim I: RGD peptides released from βig-h3, a TGF-β-induced cell adhesive molecule, mediate apoptosis. Oncogene. 2003, 22: 2045-2053. 10.1038/sj.onc.1206269.CrossRefPubMed
37.
go back to reference Zou Y, Peng H, Zhou B, Wen Y, Wang S, Tsai E, Hung M: Systemic tumor suppression by the proapoptotic gene bik. Cancer Res. 2002, 62: 8-12.PubMed Zou Y, Peng H, Zhou B, Wen Y, Wang S, Tsai E, Hung M: Systemic tumor suppression by the proapoptotic gene bik. Cancer Res. 2002, 62: 8-12.PubMed
38.
go back to reference Shen TL, Guan JL: Grb7 in intracellular signaling and its role in cell regulation. Front Biosci. 2004, 9: 192-200.CrossRefPubMed Shen TL, Guan JL: Grb7 in intracellular signaling and its role in cell regulation. Front Biosci. 2004, 9: 192-200.CrossRefPubMed
39.
go back to reference Kairouz R, Parmar J, Lyons RJ, Swarbrick A, Musgrove EA, Daly RJ: Hormonal regulation of the Grb14 signal modulator and its role in cell cycle progression of MCF-7 human breast cancer cells. J Cell Physiol. 2005, 203: 85-93. 10.1002/jcp.20199.CrossRefPubMed Kairouz R, Parmar J, Lyons RJ, Swarbrick A, Musgrove EA, Daly RJ: Hormonal regulation of the Grb14 signal modulator and its role in cell cycle progression of MCF-7 human breast cancer cells. J Cell Physiol. 2005, 203: 85-93. 10.1002/jcp.20199.CrossRefPubMed
40.
go back to reference Inoue A, Omoto Y, Yamaguchi Y, Kiyama R, Hayashi S: Transcription factor EGR3 is involved in the estrogen-signaling pathway in breast cancer cells. J Mol Endocrinol. 2004, 32: 649-661. 10.1677/jme.0.0320649.CrossRefPubMed Inoue A, Omoto Y, Yamaguchi Y, Kiyama R, Hayashi S: Transcription factor EGR3 is involved in the estrogen-signaling pathway in breast cancer cells. J Mol Endocrinol. 2004, 32: 649-661. 10.1677/jme.0.0320649.CrossRefPubMed
41.
Metadata
Title
Effects of combined treatment with rapamycin and cotylenin A, a novel differentiation-inducing agent, on human breast carcinoma MCF-7 cells and xenografts
Authors
Takashi Kasukabe
Junko Okabe-Kado
Nobuo Kato
Takeshi Sassa
Yoshio Honma
Publication date
01-12-2005
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 6/2005
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr1344

Other articles of this Issue 6/2005

Breast Cancer Research 6/2005 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