Skip to main content
Top
Published in: Breast Cancer Research 1/2018

Open Access 01-12-2018 | Research article

NOTCH3 expression is linked to breast cancer seeding and distant metastasis

Authors: Alexey A. Leontovich, Mohammad Jalalirad, Jeffrey L. Salisbury, Lisa Mills, Candace Haddox, Mark Schroeder, Ann Tuma, Maria E. Guicciardi, Luca Zammataro, Mario W. Gambino, Angela Amato, Aldo Di Leonardo, James McCubrey, Carol A. Lange, Minetta Liu, Tufia Haddad, Matthew Goetz, Judy Boughey, Jann Sarkaria, Liewei Wang, James N. Ingle, Evanthia Galanis, Antonino B. D’Assoro

Published in: Breast Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Development of distant metastases involves a complex multistep biological process termed the invasion-metastasis cascade, which includes dissemination of cancer cells from the primary tumor to secondary organs. NOTCH developmental signaling plays a critical role in promoting epithelial-to-mesenchymal transition, tumor stemness, and metastasis. Although all four NOTCH receptors show oncogenic properties, the unique role of each of these receptors in the sequential stepwise events that typify the invasion-metastasis cascade remains elusive.

Methods

We have established metastatic xenografts expressing high endogenous levels of NOTCH3 using estrogen receptor alpha-positive (ERα+) MCF-7 breast cancer cells with constitutive active Raf-1/mitogen-associated protein kinase (MAPK) signaling (vMCF-7Raf-1) and MDA-MB-231 triple-negative breast cancer (TNBC) cells. The critical role of NOTCH3 in inducing an invasive phenotype and poor outcome was corroborated in unique TNBC cells resulting from a patient-derived brain metastasis (TNBC-M25) and in publicly available claudin-low breast tumor specimens collected from participants in the Molecular Taxonomy of Breast Cancer International Consortium database.

Results

In this study, we identified an association between NOTCH3 expression and development of metastases in ERα+ and TNBC models. ERα+ breast tumor xenografts with a constitutive active Raf-1/MAPK signaling developed spontaneous lung metastases through the clonal expansion of cancer cells expressing a NOTCH3 reprogramming network. Abrogation of NOTCH3 expression significantly reduced the self-renewal and invasive capacity of ex vivo breast cancer cells, restoring a luminal CD44low/CD24high/ERαhigh phenotype. Forced expression of the mitotic Aurora kinase A (AURKA), which promotes breast cancer metastases, failed to restore the invasive capacity of NOTCH3-null cells, demonstrating that NOTCH3 expression is required for an invasive phenotype. Likewise, pharmacologic inhibition of NOTCH signaling also impaired TNBC cell seeding and metastatic growth. Significantly, the role of aberrant NOTCH3 expression in promoting tumor self-renewal, invasiveness, and poor outcome was corroborated in unique TNBC cells from a patient-derived brain metastasis and in publicly available claudin-low breast tumor specimens.

Conclusions

These findings demonstrate the key role of NOTCH3 oncogenic signaling in the genesis of breast cancer metastasis and provide a compelling preclinical rationale for the design of novel therapeutic strategies that will selectively target NOTCH3 to halt metastatic seeding and to improve the clinical outcomes of patients with breast cancer.
Appendix
Available only for authorised users
Literature
3.
go back to reference McKee MJ, Keith K, Deal AM, Garrett AL, Wheless AA, Green RL, et al. A multidisciplinary breast cancer brain metastases clinic: the University of North Carolina experience. Oncologist. 2016;21(1):16–20.CrossRefPubMed McKee MJ, Keith K, Deal AM, Garrett AL, Wheless AA, Green RL, et al. A multidisciplinary breast cancer brain metastases clinic: the University of North Carolina experience. Oncologist. 2016;21(1):16–20.CrossRefPubMed
4.
go back to reference Small GW, Shi YY, Higgins LS, Orlowski RZ. Mitogen-activated protein kinase phosphatase-1 is a mediator of breast cancer chemoresistance. Cancer Res. 2007;67(9):4459–66.CrossRefPubMed Small GW, Shi YY, Higgins LS, Orlowski RZ. Mitogen-activated protein kinase phosphatase-1 is a mediator of breast cancer chemoresistance. Cancer Res. 2007;67(9):4459–66.CrossRefPubMed
5.
go back to reference Leontovich AA, Zhang S, Quatraro C, Iankov I, Veroux PF, Gambino MW, et al. Raf-1 oncogenic signaling is linked to activation of mesenchymal to epithelial transition pathway in metastatic breast cancer cells. Int J Oncol. 2012;40:1858–64.PubMedPubMedCentral Leontovich AA, Zhang S, Quatraro C, Iankov I, Veroux PF, Gambino MW, et al. Raf-1 oncogenic signaling is linked to activation of mesenchymal to epithelial transition pathway in metastatic breast cancer cells. Int J Oncol. 2012;40:1858–64.PubMedPubMedCentral
6.
go back to reference Omarini C, Bettelli S, Caprera C, Manfredini S, Caggia F, Guaitoli G, et al. Clinical and molecular predictors of long-term response in HER2 positive metastatic breast cancerpatients. Cancer Biol Ther. 2018;1-8. Omarini C, Bettelli S, Caprera C, Manfredini S, Caggia F, Guaitoli G, et al. Clinical and molecular predictors of long-term response in HER2 positive metastatic breast cancerpatients. Cancer Biol Ther. 2018;1-8.
7.
go back to reference Mittal S, Sharma A, Balaji SA, Gowda MC, Dighe RR, Kumar RV, et al. Coordinate hyperactivation of Notch1 and Ras/MAPK pathways correlates with poor patient survival: novel therapeutic strategy for aggressive breast cancers. Mol Cancer Ther. 2014;13(12):3198–209.CrossRefPubMedPubMedCentral Mittal S, Sharma A, Balaji SA, Gowda MC, Dighe RR, Kumar RV, et al. Coordinate hyperactivation of Notch1 and Ras/MAPK pathways correlates with poor patient survival: novel therapeutic strategy for aggressive breast cancers. Mol Cancer Ther. 2014;13(12):3198–209.CrossRefPubMedPubMedCentral
9.
go back to reference Li H, Solomon E, Duhachek Muggy S, Sun D, Zolkiewska A. Metalloprotease-disintegrin ADAM12 expression is regulated by Notch signaling via microRNA-29. J Biol Chem. 2011;286(24):21500–10.CrossRefPubMedPubMedCentral Li H, Solomon E, Duhachek Muggy S, Sun D, Zolkiewska A. Metalloprotease-disintegrin ADAM12 expression is regulated by Notch signaling via microRNA-29. J Biol Chem. 2011;286(24):21500–10.CrossRefPubMedPubMedCentral
10.
11.
go back to reference Haruki N, Kawaguchi KS, Eichenberger S, Massion PP, Olson S, Gonzalez A, et al. Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res. 2005;65(9):3555–61.CrossRefPubMed Haruki N, Kawaguchi KS, Eichenberger S, Massion PP, Olson S, Gonzalez A, et al. Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res. 2005;65(9):3555–61.CrossRefPubMed
12.
go back to reference Bigas A, Guiu J, Gama-Norton L. Notch and Wnt signaling in the emergence of hematopoietic stem cells. Blood Cells Mol Dis. 2013;51(4):264–70.CrossRefPubMed Bigas A, Guiu J, Gama-Norton L. Notch and Wnt signaling in the emergence of hematopoietic stem cells. Blood Cells Mol Dis. 2013;51(4):264–70.CrossRefPubMed
13.
go back to reference Bui QT, Im JH, Jeong SB, Kim YM, Lim SC, Kim B, et al. Essential role of Notch4/STAT3 signaling in epithelial-mesenchymal transition of tamoxifen-resistant human breast cancer. Cancer Lett. 2017;390:115–25.CrossRefPubMed Bui QT, Im JH, Jeong SB, Kim YM, Lim SC, Kim B, et al. Essential role of Notch4/STAT3 signaling in epithelial-mesenchymal transition of tamoxifen-resistant human breast cancer. Cancer Lett. 2017;390:115–25.CrossRefPubMed
14.
go back to reference Shao S, Zhao X, Zhang X, Luo M, Zuo X, Huang S, et al. Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol Cancer. 2015;14:28.CrossRefPubMedPubMedCentral Shao S, Zhao X, Zhang X, Luo M, Zuo X, Huang S, et al. Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol Cancer. 2015;14:28.CrossRefPubMedPubMedCentral
15.
go back to reference Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–15.CrossRefPubMedPubMedCentral Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–15.CrossRefPubMedPubMedCentral
16.
go back to reference Hwang-Verslues WW, Kuo WH, Chang PH, Pan CC, Wang HH, Tsai ST, et al. Multiple lineages of human breast cancer stem/progenitor cells identified by profiling with stem cell markers. PLoS One. 2009;4(12):e8377.CrossRefPubMedPubMedCentral Hwang-Verslues WW, Kuo WH, Chang PH, Pan CC, Wang HH, Tsai ST, et al. Multiple lineages of human breast cancer stem/progenitor cells identified by profiling with stem cell markers. PLoS One. 2009;4(12):e8377.CrossRefPubMedPubMedCentral
17.
go back to reference Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, et al. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007;11(3):259–73.CrossRefPubMed Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, et al. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007;11(3):259–73.CrossRefPubMed
18.
go back to reference Bolós V, Mira E, Martínez-Poveda B, Luxán G, Cañamero M, Martínez-A C, et al. Notch activation stimulates migration of breast cancer cells and promotes tumor growth. Breast Cancer Res. 2013;15(4):R54.CrossRefPubMedPubMedCentral Bolós V, Mira E, Martínez-Poveda B, Luxán G, Cañamero M, Martínez-A C, et al. Notch activation stimulates migration of breast cancer cells and promotes tumor growth. Breast Cancer Res. 2013;15(4):R54.CrossRefPubMedPubMedCentral
19.
go back to reference Zanotti S, Canalis E. Notch regulation of bone development and remodeling and related skeletal disorders. Calcif Tissue Int. 2012;90(2):69–75.CrossRefPubMed Zanotti S, Canalis E. Notch regulation of bone development and remodeling and related skeletal disorders. Calcif Tissue Int. 2012;90(2):69–75.CrossRefPubMed
21.
go back to reference Chiaramonte R, Colombo M, Bulfamante G, Falleni M, Tosi D, Garavelli S, et al. Notch pathway promotes ovarian cancer growth and migration via CXCR4/SDF1α chemokine system. Int J Biochem Cell Biol. 2015;66:134–40.CrossRefPubMed Chiaramonte R, Colombo M, Bulfamante G, Falleni M, Tosi D, Garavelli S, et al. Notch pathway promotes ovarian cancer growth and migration via CXCR4/SDF1α chemokine system. Int J Biochem Cell Biol. 2015;66:134–40.CrossRefPubMed
22.
go back to reference Mukherjee D, Zhao J. The role of chemokine receptor CXCR4 in breast cancer metastasis. Am J Cancer Res. 2013;3(1):46–57.PubMedPubMedCentral Mukherjee D, Zhao J. The role of chemokine receptor CXCR4 in breast cancer metastasis. Am J Cancer Res. 2013;3(1):46–57.PubMedPubMedCentral
23.
go back to reference Shima H, Yamada A, Ishikawa T, Endo I. Are breast cancer stem cells the key to resolving clinical issues in breast cancer therapy? Gland Surg. 2017;6(1):82–8.CrossRefPubMedPubMedCentral Shima H, Yamada A, Ishikawa T, Endo I. Are breast cancer stem cells the key to resolving clinical issues in breast cancer therapy? Gland Surg. 2017;6(1):82–8.CrossRefPubMedPubMedCentral
25.
go back to reference Yen WC, Fischer MM, Axelrod F, Bond C, Cain J, Cancilla B, et al. Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21(9):2084–95.CrossRefPubMed Yen WC, Fischer MM, Axelrod F, Bond C, Cain J, Cancilla B, et al. Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21(9):2084–95.CrossRefPubMed
26.
go back to reference D’Angelo RC, Ouzounova M, Davis A, Choi D, Tchuenkam SM, Kim G, et al. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Mol Cancer Ther. 2015;14(3):779–87.CrossRefPubMedPubMedCentral D’Angelo RC, Ouzounova M, Davis A, Choi D, Tchuenkam SM, Kim G, et al. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Mol Cancer Ther. 2015;14(3):779–87.CrossRefPubMedPubMedCentral
27.
go back to reference Takebe N, Nguyen D, Yang SX. Targeting notch signaling pathway in cancer: clinical development advances and challenges. Pharmacol Ther. 2014;141(2):140–9.CrossRefPubMed Takebe N, Nguyen D, Yang SX. Targeting notch signaling pathway in cancer: clinical development advances and challenges. Pharmacol Ther. 2014;141(2):140–9.CrossRefPubMed
28.
go back to reference Previs RA, Coleman RL, Harris AL, Sood AK. Molecular pathways: translational and therapeutic implications of the Notch signaling pathway in cancer. Clin Cancer Res. 2015;21(5):955–61.CrossRefPubMed Previs RA, Coleman RL, Harris AL, Sood AK. Molecular pathways: translational and therapeutic implications of the Notch signaling pathway in cancer. Clin Cancer Res. 2015;21(5):955–61.CrossRefPubMed
29.
go back to reference D’Assoro AB, Liu T, Quatraro C, Amato A, Opyrchal M, Leontovich A, et al. The mitotic kinase Aurora-A promotes distant metastases by inducing epithelial-to-mesenchymal transition in ERα+ breast cancer cells. Oncogene. 2014;33:599–610.CrossRefPubMed D’Assoro AB, Liu T, Quatraro C, Amato A, Opyrchal M, Leontovich A, et al. The mitotic kinase Aurora-A promotes distant metastases by inducing epithelial-to-mesenchymal transition in ERα+ breast cancer cells. Oncogene. 2014;33:599–610.CrossRefPubMed
30.
go back to reference Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486(7403):346–52.CrossRefPubMedPubMedCentral Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486(7403):346–52.CrossRefPubMedPubMedCentral
31.
go back to reference Leontovich AA, Zhang S, Quatraro C, Iankov I, Veroux PF, Gambino MW, et al. Raf-1 oncogenic signaling is linked to activation of mesenchymal to epithelial transition pathway in metastatic breast cancer cells. Int J Oncol. 2012;40(6):1858–64.PubMedPubMedCentral Leontovich AA, Zhang S, Quatraro C, Iankov I, Veroux PF, Gambino MW, et al. Raf-1 oncogenic signaling is linked to activation of mesenchymal to epithelial transition pathway in metastatic breast cancer cells. Int J Oncol. 2012;40(6):1858–64.PubMedPubMedCentral
32.
go back to reference Liu T, Yu J, Deng M, Yin Y, Zhang H, Luo K, et al. CDK4/6-dependent activation of DUB3 regulates cancer metastasis through SNAIL1. Nat Commun. 2017;8:13923.CrossRefPubMedPubMedCentral Liu T, Yu J, Deng M, Yin Y, Zhang H, Luo K, et al. CDK4/6-dependent activation of DUB3 regulates cancer metastasis through SNAIL1. Nat Commun. 2017;8:13923.CrossRefPubMedPubMedCentral
33.
go back to reference Opyrchal M, Salisbury JL, Iankov I, Goetz MP, McCubrey J, Gambino MW, et al. Inhibition of Cdk2 kinase activity selectively targets the CD44+/CD24−/Low stem-like subpopulation and restores chemosensitivity of SUM149PT triple-negative breast cancer cells. Int J Oncol. 2014;45(3):1193–9.CrossRefPubMedPubMedCentral Opyrchal M, Salisbury JL, Iankov I, Goetz MP, McCubrey J, Gambino MW, et al. Inhibition of Cdk2 kinase activity selectively targets the CD44+/CD24−/Low stem-like subpopulation and restores chemosensitivity of SUM149PT triple-negative breast cancer cells. Int J Oncol. 2014;45(3):1193–9.CrossRefPubMedPubMedCentral
34.
go back to reference Opyrchal M, Gil M, Salisbury JL, Goetz MP, Suman V, Degnim A, et al. Molecular targeting of the Aurora-A/SMAD5 oncogenic axis restores chemosensitivity in human breast cancer cells. Oncotarget. 2017;8(53):91803–16.CrossRefPubMedPubMedCentral Opyrchal M, Gil M, Salisbury JL, Goetz MP, Suman V, Degnim A, et al. Molecular targeting of the Aurora-A/SMAD5 oncogenic axis restores chemosensitivity in human breast cancer cells. Oncotarget. 2017;8(53):91803–16.CrossRefPubMedPubMedCentral
35.
go back to reference D’Assoro AB, Barrett SL, Folk C, Negron VC, Boeneman K, Busby R, et al. Amplified centrosomes in breast cancer: a potential indicator of tumor aggressiveness. Breast Cancer Res Treat. 2002;75(1):25–34.CrossRefPubMed D’Assoro AB, Barrett SL, Folk C, Negron VC, Boeneman K, Busby R, et al. Amplified centrosomes in breast cancer: a potential indicator of tumor aggressiveness. Breast Cancer Res Treat. 2002;75(1):25–34.CrossRefPubMed
36.
go back to reference Lingle WL, Barrett SL, Negron VC, D’Assoro AB, Boeneman K, Liu W, et al. Centrosome amplification drives chromosomal instability in breast tumor development. Proc Natl Acad Sci U S A. 2002;99(4):1978–83.CrossRefPubMedPubMedCentral Lingle WL, Barrett SL, Negron VC, D’Assoro AB, Boeneman K, Liu W, et al. Centrosome amplification drives chromosomal instability in breast tumor development. Proc Natl Acad Sci U S A. 2002;99(4):1978–83.CrossRefPubMedPubMedCentral
37.
go back to reference Opyrchal M, Salisbury JL, Zhang S, McCubrey J, Hawse J, Goetz MP, et al. Aurora-A mitotic kinase induces endocrine resistance through down-regulation of ERα expression in initially ERα+ breast cancer cells. PLoS One. 2014;9(5):e96995.CrossRefPubMedPubMedCentral Opyrchal M, Salisbury JL, Zhang S, McCubrey J, Hawse J, Goetz MP, et al. Aurora-A mitotic kinase induces endocrine resistance through down-regulation of ERα expression in initially ERα+ breast cancer cells. PLoS One. 2014;9(5):e96995.CrossRefPubMedPubMedCentral
38.
go back to reference Sansone P, Ceccarelli C, Berishaj M, Chang Q, Rajasekhar VK, Perna F, et al. Self-renewal of CD133hi cells by IL6/Notch3 signalling regulates endocrine resistance in metastatic breast cancer. Nat Commun. 2016;7:10442.CrossRefPubMedPubMedCentral Sansone P, Ceccarelli C, Berishaj M, Chang Q, Rajasekhar VK, Perna F, et al. Self-renewal of CD133hi cells by IL6/Notch3 signalling regulates endocrine resistance in metastatic breast cancer. Nat Commun. 2016;7:10442.CrossRefPubMedPubMedCentral
40.
go back to reference Iankov ID, Kurokawa CB, D’Assoro AB, Ingle JN, Domingo-Musibay E, Allen C, et al. Inhibition of the Aurora A kinase augments the anti-tumor efficacy of oncolytic measles virotherapy. Cancer Gene Ther. 2015;22(9):438–44.CrossRefPubMedPubMedCentral Iankov ID, Kurokawa CB, D’Assoro AB, Ingle JN, Domingo-Musibay E, Allen C, et al. Inhibition of the Aurora A kinase augments the anti-tumor efficacy of oncolytic measles virotherapy. Cancer Gene Ther. 2015;22(9):438–44.CrossRefPubMedPubMedCentral
47.
go back to reference Bartholomeusz C, Xie X, Pitner MK, Kondo K, Dadbin A, Lee J, Saso H, Smith PD, Dalby KN, Ueno NT. MEK inhibitor selumetinib (AZD6244; ARRY-142886) prevents lung metastasis in a triple-negative breast cancer xenograft model. Mol Cancer Ther. 2015;14(12):2773–81.CrossRefPubMedPubMedCentral Bartholomeusz C, Xie X, Pitner MK, Kondo K, Dadbin A, Lee J, Saso H, Smith PD, Dalby KN, Ueno NT. MEK inhibitor selumetinib (AZD6244; ARRY-142886) prevents lung metastasis in a triple-negative breast cancer xenograft model. Mol Cancer Ther. 2015;14(12):2773–81.CrossRefPubMedPubMedCentral
48.
go back to reference Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial-to-mesenchymal transition. Breast Cancer Res. 2011;13(3):211.CrossRefPubMedPubMedCentral Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial-to-mesenchymal transition. Breast Cancer Res. 2011;13(3):211.CrossRefPubMedPubMedCentral
49.
go back to reference Chiba S, Okuda M, Mussman JG, Fukasawa K. Genomic convergence and suppression of centrosome hyperamplification in primary p53−/− cells in prolonged culture. Exp Cell Res. 2000;258(2):310–21.CrossRefPubMed Chiba S, Okuda M, Mussman JG, Fukasawa K. Genomic convergence and suppression of centrosome hyperamplification in primary p53−/− cells in prolonged culture. Exp Cell Res. 2000;258(2):310–21.CrossRefPubMed
51.
go back to reference Mu X, Isaac C, Greco N, Huard J, Weiss K. Notch signaling is associated with ALDH activity and an aggressive metastatic phenotype in murine osteosarcoma cells. Front Oncol. 2013;3:143.CrossRefPubMedPubMedCentral Mu X, Isaac C, Greco N, Huard J, Weiss K. Notch signaling is associated with ALDH activity and an aggressive metastatic phenotype in murine osteosarcoma cells. Front Oncol. 2013;3:143.CrossRefPubMedPubMedCentral
52.
go back to reference Hamilton SR, Fard SF, Paiwand FF, Tolg C, Veiseh M, Wang C, et al. The hyaluronan receptors CD44 and Rhamm (CD168) form complexes with ERK1,2 that sustain high basal motility in breast cancer cells. J Biol Chem. 2007;282(22):16667–80.CrossRefPubMedPubMedCentral Hamilton SR, Fard SF, Paiwand FF, Tolg C, Veiseh M, Wang C, et al. The hyaluronan receptors CD44 and Rhamm (CD168) form complexes with ERK1,2 that sustain high basal motility in breast cancer cells. J Biol Chem. 2007;282(22):16667–80.CrossRefPubMedPubMedCentral
53.
go back to reference Choy L, Hagenbeek TJ, Solon M, French D, Finkle D, Shelton A, et al. Constitutive NOTCH3 signaling promotes the growth of basal breast cancers. Cancer Res. 2017;77(6):1439–52.CrossRefPubMed Choy L, Hagenbeek TJ, Solon M, French D, Finkle D, Shelton A, et al. Constitutive NOTCH3 signaling promotes the growth of basal breast cancers. Cancer Res. 2017;77(6):1439–52.CrossRefPubMed
Metadata
Title
NOTCH3 expression is linked to breast cancer seeding and distant metastasis
Authors
Alexey A. Leontovich
Mohammad Jalalirad
Jeffrey L. Salisbury
Lisa Mills
Candace Haddox
Mark Schroeder
Ann Tuma
Maria E. Guicciardi
Luca Zammataro
Mario W. Gambino
Angela Amato
Aldo Di Leonardo
James McCubrey
Carol A. Lange
Minetta Liu
Tufia Haddad
Matthew Goetz
Judy Boughey
Jann Sarkaria
Liewei Wang
James N. Ingle
Evanthia Galanis
Antonino B. D’Assoro
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2018
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-018-1020-0

Other articles of this Issue 1/2018

Breast Cancer Research 1/2018 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