Skip to main content
Top
Published in: Breast Cancer Research and Treatment 2/2010

01-04-2010 | Preclinical study

The substrate domain of BCAR1 is essential for anti-estrogen-resistant proliferation of human breast cancer cells

Authors: Arend Brinkman, Danielle de Jong, Sietske Tuinman, Najat Azaouagh, Ton van Agthoven, Lambert C. J. Dorssers

Published in: Breast Cancer Research and Treatment | Issue 2/2010

Login to get access

Abstract

To unravel the mechanisms underlying failure of endocrine therapy of breast cancer, we have previously executed a functional genetic screen and identified the adaptor protein BCAR1 to be causative for tamoxifen resistance. As a consequence of the manifold of interactions with other proteins, we characterized the contribution of individual protein domains of BCAR1 to anti-estrogen-resistant proliferation of human breast cancer cells. We took advantage of the observation that the closely related family member HEF1 was unable to support long-term anti-estrogen-resistant cell proliferation. Chimerical proteins containing defined domains of BCAR1 and HEF1 were evaluated for anti-estrogen-resistant growth. Exchange of the SH3 and C-terminal domains did not modify the capacity to support cell proliferation. Full support of anti-estrogen resistant proliferation was observed for chimerical molecules containing the central part of BCAR1. The bi-partite SRC-binding site or the Serine-rich domain did not explain the differential capacity of BCAR1. These findings indicate that the differences between BCAR1 and HEF1 with respect to support of anti-estrogen resistance reside in the substrate domain which contains multiple sites for tyrosine phosphorylation. The crucial interactions required for anti-estrogen resistance occur within the substrate domain of BCAR1. Further deciphering of these interactions may resolve the growth regulatory mechanism and provide an explanation for the observation that primary tumors with high levels of BCAR1 are likely to fail on tamoxifen therapy. This information may also help to devise alternative personalized treatment strategies with improved outcome for breast cancer patients.
Literature
1.
go back to reference Group Early Breast Cancer Trialists’ Collaborative (2005) Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365:1687–1717CrossRef Group Early Breast Cancer Trialists’ Collaborative (2005) Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365:1687–1717CrossRef
2.
go back to reference Jaiyesimi IA, Buzdar AU, Decker DA et al (1995) Use of tamoxifen for breast cancer: twenty-eight years later. J Clin Oncol 13:513–529PubMed Jaiyesimi IA, Buzdar AU, Decker DA et al (1995) Use of tamoxifen for breast cancer: twenty-eight years later. J Clin Oncol 13:513–529PubMed
3.
go back to reference Ali S, Coombes RC (2002) Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev Cancer 2:101–112CrossRefPubMed Ali S, Coombes RC (2002) Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev Cancer 2:101–112CrossRefPubMed
4.
go back to reference Clarke R, Liu MC, Bouker KB et al (2003) Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene 22:7316–7339CrossRefPubMed Clarke R, Liu MC, Bouker KB et al (2003) Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene 22:7316–7339CrossRefPubMed
5.
go back to reference Osborne CK, Shou J, Massarweh S et al (2005) Crosstalk between estrogen receptor and growth factor receptor pathways as a cause for endocrine therapy resistance in breast cancer. Clin Cancer Res 11:865s–870sPubMed Osborne CK, Shou J, Massarweh S et al (2005) Crosstalk between estrogen receptor and growth factor receptor pathways as a cause for endocrine therapy resistance in breast cancer. Clin Cancer Res 11:865s–870sPubMed
6.
go back to reference Lewis JS, Jordan VC (2005) Selective estrogen receptor modulators (SERMs): mechanisms of anticarcinogenesis and drug resistance. Mutat Res 591:247–263PubMed Lewis JS, Jordan VC (2005) Selective estrogen receptor modulators (SERMs): mechanisms of anticarcinogenesis and drug resistance. Mutat Res 591:247–263PubMed
7.
go back to reference Riggins RB, Schrecengost RS, Guerrero MS et al (2007) Pathways to tamoxifen resistance. Cancer Lett 256:1–24CrossRefPubMed Riggins RB, Schrecengost RS, Guerrero MS et al (2007) Pathways to tamoxifen resistance. Cancer Lett 256:1–24CrossRefPubMed
8.
go back to reference Dorssers LCJ, Van Agthoven T, Dekker A et al (1993) Induction of antiestrogen resistance in human breast cancer cells by random insertional mutagenesis using defective retroviruses: identification of bcar-1, a common integration site. Mol Endocrinol 7:870–878CrossRefPubMed Dorssers LCJ, Van Agthoven T, Dekker A et al (1993) Induction of antiestrogen resistance in human breast cancer cells by random insertional mutagenesis using defective retroviruses: identification of bcar-1, a common integration site. Mol Endocrinol 7:870–878CrossRefPubMed
9.
go back to reference Van Agthoven T, Veldscholte J, Smid M et al (2009) Functional screen identifies genes associated with human breast cancer progression. Breast Cancer Res Treat 114:23–30CrossRefPubMed Van Agthoven T, Veldscholte J, Smid M et al (2009) Functional screen identifies genes associated with human breast cancer progression. Breast Cancer Res Treat 114:23–30CrossRefPubMed
10.
go back to reference Sakai R, Iwamatsu A, Hirano N et al (1994) A novel signaling molecule, p130, forms stable complexes in vivo with v-Crk and v-Src in a tyrosine phosphorylation- dependent manner. EMBO J 13:3748–3756PubMed Sakai R, Iwamatsu A, Hirano N et al (1994) A novel signaling molecule, p130, forms stable complexes in vivo with v-Crk and v-Src in a tyrosine phosphorylation- dependent manner. EMBO J 13:3748–3756PubMed
11.
go back to reference Sakai R, Iwamatsu A, Hirano N et al (1994) Characterization, partial purification, and peptide sequencing of p130, the main phosphoprotein associated with v-Crk oncoprotein. J Biol Chem 269:32740–32746PubMed Sakai R, Iwamatsu A, Hirano N et al (1994) Characterization, partial purification, and peptide sequencing of p130, the main phosphoprotein associated with v-Crk oncoprotein. J Biol Chem 269:32740–32746PubMed
12.
go back to reference Brinkman A, Van der Flier S, Kok EM et al (2000) BCAR1, a human homologue of the adapter protein p130Cas and antiestrogen resistance in breast cancer cells. J Natl Cancer Inst 92:112–120CrossRefPubMed Brinkman A, Van der Flier S, Kok EM et al (2000) BCAR1, a human homologue of the adapter protein p130Cas and antiestrogen resistance in breast cancer cells. J Natl Cancer Inst 92:112–120CrossRefPubMed
13.
go back to reference Ta HQ, Thomas KS, Schrecengost RS et al (2008) A novel association between p130Cas and resistance to the chemotherapeutic drug adriamycin in human breast cancer cells. Cancer Res 68:8796–8804CrossRefPubMed Ta HQ, Thomas KS, Schrecengost RS et al (2008) A novel association between p130Cas and resistance to the chemotherapeutic drug adriamycin in human breast cancer cells. Cancer Res 68:8796–8804CrossRefPubMed
14.
go back to reference Van der Flier S, Brinkman A, Look MP et al (2000) Bcar1/p130Cas protein and primary breast cancer: prognosis and response to tamoxifen treatment. J Natl Cancer Inst 92:120–127CrossRefPubMed Van der Flier S, Brinkman A, Look MP et al (2000) Bcar1/p130Cas protein and primary breast cancer: prognosis and response to tamoxifen treatment. J Natl Cancer Inst 92:120–127CrossRefPubMed
15.
go back to reference Van der Flier S, Van der Kwast TH, Claassen CJC et al (2001) Immunohistochemical study of the BCAR1/p130Cas protein in non-malignant and malignant human breast tissue. Int J Biol Markers 16:172–178PubMed Van der Flier S, Van der Kwast TH, Claassen CJC et al (2001) Immunohistochemical study of the BCAR1/p130Cas protein in non-malignant and malignant human breast tissue. Int J Biol Markers 16:172–178PubMed
16.
go back to reference Van der Flier S, Chan CMW, Brinkman A et al (2000) BCAR1/p130Cas expression in untreated and acquired tamoxifen-resistant human breast carcinomas. Int J Cancer 89:465–468CrossRefPubMed Van der Flier S, Chan CMW, Brinkman A et al (2000) BCAR1/p130Cas expression in untreated and acquired tamoxifen-resistant human breast carcinomas. Int J Cancer 89:465–468CrossRefPubMed
17.
go back to reference Grebenchtchikov N, Brinkman A, Van Broekhoven SPJ et al (2004) Development of an ELISA for measurement of BCAR1 protein in human breast cancer tissue. Clin Chem 50:1356–1363CrossRefPubMed Grebenchtchikov N, Brinkman A, Van Broekhoven SPJ et al (2004) Development of an ELISA for measurement of BCAR1 protein in human breast cancer tissue. Clin Chem 50:1356–1363CrossRefPubMed
18.
go back to reference Dorssers LCJ, Grebenchtchikov N, Brinkman A et al (2004) The prognostic value of BCAR1 in patients with primary breast cancer. Clin Cancer Res 10:6194–6202CrossRefPubMed Dorssers LCJ, Grebenchtchikov N, Brinkman A et al (2004) The prognostic value of BCAR1 in patients with primary breast cancer. Clin Cancer Res 10:6194–6202CrossRefPubMed
19.
go back to reference O’Neill GM, Fashena SJ, Golemis EA (2000) Integrin signalling: a new cas(t) of characters enters the stage. Trends Cell Biol 10:111–119CrossRefPubMed O’Neill GM, Fashena SJ, Golemis EA (2000) Integrin signalling: a new cas(t) of characters enters the stage. Trends Cell Biol 10:111–119CrossRefPubMed
20.
go back to reference Bouton AH, Riggins RB, Bruce-Staskal PJ (2001) Functions of the adapter protein Cas: signal convergence and the determination of cellular responses. Oncogene 20:6448–6458CrossRefPubMed Bouton AH, Riggins RB, Bruce-Staskal PJ (2001) Functions of the adapter protein Cas: signal convergence and the determination of cellular responses. Oncogene 20:6448–6458CrossRefPubMed
21.
go back to reference Singh MK, Dadke D, Nicolas E et al (2008) A novel Cas family member, HEPL, regulates FAK and cell spreading. Mol Biol Cell 19:1627–1636CrossRefPubMed Singh MK, Dadke D, Nicolas E et al (2008) A novel Cas family member, HEPL, regulates FAK and cell spreading. Mol Biol Cell 19:1627–1636CrossRefPubMed
22.
go back to reference Cabodi S, Moro L, Baj G et al (2004) p130Cas interacts with estrogen receptor alpha and modulates non-genomic estrogen signaling in breast cancer cells. J Cell Sci 117:1603–1611CrossRefPubMed Cabodi S, Moro L, Baj G et al (2004) p130Cas interacts with estrogen receptor alpha and modulates non-genomic estrogen signaling in breast cancer cells. J Cell Sci 117:1603–1611CrossRefPubMed
23.
go back to reference Defilippi P, Di Stefano P, Cabodi S (2006) p130Cas: a versatile scaffold in signaling networks. Trends Cell Biol 16:257–263CrossRefPubMed Defilippi P, Di Stefano P, Cabodi S (2006) p130Cas: a versatile scaffold in signaling networks. Trends Cell Biol 16:257–263CrossRefPubMed
24.
go back to reference Riggins RB, Thomas KS, Ta HQ et al (2006) Physical and functional interactions between Cas and c-Src induce tamoxifen resistance of breast cancer cells through pathways involving epidermal growth factor receptor and signal transducer and activator of transcription 5b. Cancer Res 66:7007–7015CrossRefPubMed Riggins RB, Thomas KS, Ta HQ et al (2006) Physical and functional interactions between Cas and c-Src induce tamoxifen resistance of breast cancer cells through pathways involving epidermal growth factor receptor and signal transducer and activator of transcription 5b. Cancer Res 66:7007–7015CrossRefPubMed
25.
go back to reference Cabodi S, Tinnirello A, Di Stefano P et al (2006) p130Cas as a new regulator of mammary epithelial cell proliferation, survival, and HER2-neu oncogene-dependent breast tumorigenesis. Cancer Res 66:4672–4680CrossRefPubMed Cabodi S, Tinnirello A, Di Stefano P et al (2006) p130Cas as a new regulator of mammary epithelial cell proliferation, survival, and HER2-neu oncogene-dependent breast tumorigenesis. Cancer Res 66:4672–4680CrossRefPubMed
26.
go back to reference Honda H, Oda H, Nakamoto T et al (1998) Cardiovascular anomaly, impaired actin bundling and resistance to Src- induced transformation in mice lacking p130Cas. Nat Genet 19:361–365CrossRefPubMed Honda H, Oda H, Nakamoto T et al (1998) Cardiovascular anomaly, impaired actin bundling and resistance to Src- induced transformation in mice lacking p130Cas. Nat Genet 19:361–365CrossRefPubMed
27.
go back to reference Kim M, Gans JD, Nogueira C et al (2006) Comparative oncogenomics identifies NEDD9 as a melanoma metastasis gene. Cell 125:1269–1281CrossRefPubMed Kim M, Gans JD, Nogueira C et al (2006) Comparative oncogenomics identifies NEDD9 as a melanoma metastasis gene. Cell 125:1269–1281CrossRefPubMed
28.
go back to reference O’Neill GM, Seo S, Serebriiskii IG et al (2007) A new central scaffold for metastasis: parsing HEF1/Cas-L/NEDD9. Cancer Res 67:8975–8979CrossRefPubMed O’Neill GM, Seo S, Serebriiskii IG et al (2007) A new central scaffold for metastasis: parsing HEF1/Cas-L/NEDD9. Cancer Res 67:8975–8979CrossRefPubMed
29.
go back to reference Singh M, Cowell L, Seo S et al (2007) Molecular basis for HEF1/NEDD9/Cas-L action as a multifunctional co-ordinator of invasion, apoptosis and cell cycle. Cell Biochem Biophys 48:54–72CrossRefPubMed Singh M, Cowell L, Seo S et al (2007) Molecular basis for HEF1/NEDD9/Cas-L action as a multifunctional co-ordinator of invasion, apoptosis and cell cycle. Cell Biochem Biophys 48:54–72CrossRefPubMed
30.
go back to reference Nakamoto T, Sakai R, Ozawa K et al (1996) Direct binding of C-terminal region of p130Cas to SH2 and SH3 domains of Src kinase. J Biol Chem 271:8959–8965CrossRefPubMed Nakamoto T, Sakai R, Ozawa K et al (1996) Direct binding of C-terminal region of p130Cas to SH2 and SH3 domains of Src kinase. J Biol Chem 271:8959–8965CrossRefPubMed
31.
go back to reference Burnham MR, Bruce-Staskal PJ, Harte MT et al (2000) Regulation of c-SRC activity and function by the adapter protein CAS. Mol Cell Biol 20:5865–5878CrossRefPubMed Burnham MR, Bruce-Staskal PJ, Harte MT et al (2000) Regulation of c-SRC activity and function by the adapter protein CAS. Mol Cell Biol 20:5865–5878CrossRefPubMed
32.
go back to reference Ruest PJ, Shin NY, Polte TR et al (2001) Mechanisms of CAS substrate domain tyrosine phosphorylation by FAK and Src. Mol Cell Biol 21:7641–7652CrossRefPubMed Ruest PJ, Shin NY, Polte TR et al (2001) Mechanisms of CAS substrate domain tyrosine phosphorylation by FAK and Src. Mol Cell Biol 21:7641–7652CrossRefPubMed
33.
go back to reference Sharma A, Mayer BJ (2008) Phosphorylation of p130Cas initiates Rac activation and membrane ruffling. BMC Cell Biol 9:50CrossRefPubMed Sharma A, Mayer BJ (2008) Phosphorylation of p130Cas initiates Rac activation and membrane ruffling. BMC Cell Biol 9:50CrossRefPubMed
34.
go back to reference Shen Y, Jia Z, Nagele RG et al (2006) SRC uses Cas to suppress Fhl1 in order to promote nonanchored growth and migration of tumor cells. Cancer Res 66:1543–1552CrossRefPubMed Shen Y, Jia Z, Nagele RG et al (2006) SRC uses Cas to suppress Fhl1 in order to promote nonanchored growth and migration of tumor cells. Cancer Res 66:1543–1552CrossRefPubMed
35.
go back to reference Shen Y, Khusial PR, Li X et al (2007) SRC utilizes Cas to block gap junctional communication mediated by connexin43. J Biol Chem 282:18914–18921CrossRefPubMed Shen Y, Khusial PR, Li X et al (2007) SRC utilizes Cas to block gap junctional communication mediated by connexin43. J Biol Chem 282:18914–18921CrossRefPubMed
36.
go back to reference Patwardhan P, Shen Y, Goldberg GS et al (2006) Individual Cas phosphorylation sites are dispensable for processive phosphorylation by Src and anchorage-independent cell growth. J Biol Chem 281:20689–20697CrossRefPubMed Patwardhan P, Shen Y, Goldberg GS et al (2006) Individual Cas phosphorylation sites are dispensable for processive phosphorylation by Src and anchorage-independent cell growth. J Biol Chem 281:20689–20697CrossRefPubMed
37.
go back to reference Cai D, Clayton LK, Smolyar A et al (1999) AND-34, a novel p130Cas-binding thymic stromal cell protein regulated by adhesion and inflammatory cytokines. J Immunol 163:2104–2112PubMed Cai D, Clayton LK, Smolyar A et al (1999) AND-34, a novel p130Cas-binding thymic stromal cell protein regulated by adhesion and inflammatory cytokines. J Immunol 163:2104–2112PubMed
38.
go back to reference Cai D, Iyer A, Felekkis KN et al (2003) AND-34/BCAR3, a GDP exchange factor whose overexpression confers antiestrogen resistance, activates Rac, PAK1, and the cyclin D1 promoter. Cancer Res 63:6802–6808PubMed Cai D, Iyer A, Felekkis KN et al (2003) AND-34/BCAR3, a GDP exchange factor whose overexpression confers antiestrogen resistance, activates Rac, PAK1, and the cyclin D1 promoter. Cancer Res 63:6802–6808PubMed
39.
go back to reference Gotoh T, Cai D, Tian X et al (2000) p130Cas regulates the activity of AND-34, a novel Ral, Rap1, and R-Ras guanine nucleotide exchange factor. J Biol Chem 275:30118–30123CrossRefPubMed Gotoh T, Cai D, Tian X et al (2000) p130Cas regulates the activity of AND-34, a novel Ral, Rap1, and R-Ras guanine nucleotide exchange factor. J Biol Chem 275:30118–30123CrossRefPubMed
40.
go back to reference Riggins RB, Quilliam LA, Bouton AH (2003) Synergistic promotion of c-Src activation and cell migration by Cas and AND-34/BCAR3. J Biol Chem 278:28264–28273CrossRefPubMed Riggins RB, Quilliam LA, Bouton AH (2003) Synergistic promotion of c-Src activation and cell migration by Cas and AND-34/BCAR3. J Biol Chem 278:28264–28273CrossRefPubMed
41.
go back to reference Schrecengost RS, Riggins RB, Thomas KS et al (2007) Breast cancer antiestrogen resistance-3 expression regulates breast cancer cell migration through promotion of p130Cas membrane localization and membrane ruffling. Cancer Res 67:6174–6182CrossRefPubMed Schrecengost RS, Riggins RB, Thomas KS et al (2007) Breast cancer antiestrogen resistance-3 expression regulates breast cancer cell migration through promotion of p130Cas membrane localization and membrane ruffling. Cancer Res 67:6174–6182CrossRefPubMed
42.
go back to reference Van Agthoven T, Van Agthoven TLA, Dekker A et al (1998) Identification of BCAR3 by a random search for genes involved in antiestrogen resistance of human breast cancer cells. EMBO J 17:2799–2808CrossRefPubMed Van Agthoven T, Van Agthoven TLA, Dekker A et al (1998) Identification of BCAR3 by a random search for genes involved in antiestrogen resistance of human breast cancer cells. EMBO J 17:2799–2808CrossRefPubMed
43.
go back to reference Van Agthoven T, Van Agthoven TLA, Portengen H et al (1992) Ectopic expression of epidermal growth factor receptors induces hormone independence in ZR-75–1 human breast cancer cells. Cancer Res 52:5082–5088PubMed Van Agthoven T, Van Agthoven TLA, Portengen H et al (1992) Ectopic expression of epidermal growth factor receptors induces hormone independence in ZR-75–1 human breast cancer cells. Cancer Res 52:5082–5088PubMed
44.
go back to reference Law SF, Zhang YZ, Klein-Szanto AJP et al (1998) Cell cycle-regulated processing of HEF1 to multiple protein forms differentially targeted to multiple subcellular compartments. Mol Cell Biol 18:3540–3551PubMed Law SF, Zhang YZ, Klein-Szanto AJP et al (1998) Cell cycle-regulated processing of HEF1 to multiple protein forms differentially targeted to multiple subcellular compartments. Mol Cell Biol 18:3540–3551PubMed
45.
go back to reference Riggins RB, DeBerry RM, Toosarvandani MD et al (2003) Src-Dependent Association of Cas and p85 Phosphatidylinositol 3′-Kinase in v-crk-Transformed Cells. Mol Cancer Res 1:428–437PubMed Riggins RB, DeBerry RM, Toosarvandani MD et al (2003) Src-Dependent Association of Cas and p85 Phosphatidylinositol 3′-Kinase in v-crk-Transformed Cells. Mol Cancer Res 1:428–437PubMed
47.
go back to reference Garron ML, Arsenieva D, Zhong J et al (2009) Structural insights into the association between BCAR3 and Cas family members, an atypical complex implicated in Anti-oestrogen resistance. J Mol Biol 386:190–203CrossRefPubMed Garron ML, Arsenieva D, Zhong J et al (2009) Structural insights into the association between BCAR3 and Cas family members, an atypical complex implicated in Anti-oestrogen resistance. J Mol Biol 386:190–203CrossRefPubMed
48.
go back to reference Law SF, Estojak J, Wang BL et al (1996) Human enhancer of filamentation 1, a novel p130 cas -like docking protein, associates with focal adhesion kinase and induces pseudohyphal growth in Saccharomyces cerevisiae. Mol Cell Biol 16:3327–3337PubMed Law SF, Estojak J, Wang BL et al (1996) Human enhancer of filamentation 1, a novel p130 cas -like docking protein, associates with focal adhesion kinase and induces pseudohyphal growth in Saccharomyces cerevisiae. Mol Cell Biol 16:3327–3337PubMed
49.
go back to reference Van Agthoven T, Sieuwerts AM, Meijer-Van Gelder ME et al (2009) Relevance of breast cancer antiestrogen resistance genes in human breast cancer progression and tamoxifen resistance. J Clin Oncol 27:542–549CrossRefPubMed Van Agthoven T, Sieuwerts AM, Meijer-Van Gelder ME et al (2009) Relevance of breast cancer antiestrogen resistance genes in human breast cancer progression and tamoxifen resistance. J Clin Oncol 27:542–549CrossRefPubMed
50.
go back to reference Nasertorabi F, Tars K, Becherer K et al (2006) Molecular basis for regulation of Src by the docking protein p130Cas. J Mol Recognit 19:30–38CrossRefPubMed Nasertorabi F, Tars K, Becherer K et al (2006) Molecular basis for regulation of Src by the docking protein p130Cas. J Mol Recognit 19:30–38CrossRefPubMed
Metadata
Title
The substrate domain of BCAR1 is essential for anti-estrogen-resistant proliferation of human breast cancer cells
Authors
Arend Brinkman
Danielle de Jong
Sietske Tuinman
Najat Azaouagh
Ton van Agthoven
Lambert C. J. Dorssers
Publication date
01-04-2010
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 2/2010
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-009-0403-4

Other articles of this Issue 2/2010

Breast Cancer Research and Treatment 2/2010 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