Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Metastasis | Research

HOXD9 promotes the growth, invasion and metastasis of gastric cancer cells by transcriptional activation of RUFY3

Authors: Huiqiong Zhu, Weiyu Dai, Jiaying Li, Li Xiang, Xiaosheng Wu, Weimei Tang, Yaying Chen, Qiong Yang, Mengwei Liu, Yizhi Xiao, Wenjing Zhang, Jianjiao Lin, Jing Wang, Guangnan Liu, Yong Sun, Ping Jiang, Guoxin Li, Aimin Li, Side Liu, Ye Chen, Jide Wang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

The transcription factor HOXD9 is one of the members of the HOX family, which plays an important role in neoplastic processes. However, the role of HOXD9 in the growth and metastasis of gastric cancer (GC) remains to be elucidated.

Methods

In vitro functional role of HOXD9 and RURY3 in GC cells was determined using the TMA-based immunohistochemistry, western blot, EdU incorporation, gelatin zymography, luciferase, chromatin Immunoprecipitation (ChIP) and cell invasion assays. In vivo tumor growth and metastasis were conducted in nude mice.

Results

HOXD9 is overexpressed in GC cells and tissues. The high expression of HOXD9 was correlated with poor survival in GC patients. Functionally, HOXD9 expression significantly promoted the proliferation, invasion and migration of GC cells. Mechanically, HOXD9 directly associated with the RUFY3 promoter to increase the transcriptional activity of RUFY3. Inhibition of RUFY3 attenuated the proliferation, migration and invasiveness of HOXD9-overexpressing GC cells in vitro and in vivo. Moreover, both HOXD9 and RUFY3 were highly expressed in cancer cells but not in normal gastric tissues, with their expressions being positively correlated.

Conclusions

The evidence presented here suggests that the HOXD9-RUFY3 axis promotes the development and progression of human GC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Moens CB, Selleri L. Hox cofactors in vertebrate development. Dev Biol. 2006;291(2):193–206.CrossRef Moens CB, Selleri L. Hox cofactors in vertebrate development. Dev Biol. 2006;291(2):193–206.CrossRef
2.
go back to reference Mansour MA, Senga T. HOXD8 exerts a tumor-suppressing role in colorectal cancer as an apoptotic inducer. Int J Biochem Cell Biol. 2017;88:1–13.CrossRef Mansour MA, Senga T. HOXD8 exerts a tumor-suppressing role in colorectal cancer as an apoptotic inducer. Int J Biochem Cell Biol. 2017;88:1–13.CrossRef
3.
go back to reference Hombria JC, Lovegrove B. Beyond homeosis--HOX function in morphogenesis and organogenesis. Differentiation. 2003;71(8):461–76.CrossRef Hombria JC, Lovegrove B. Beyond homeosis--HOX function in morphogenesis and organogenesis. Differentiation. 2003;71(8):461–76.CrossRef
4.
go back to reference Liu H, Zhang M, Xu S, Zhang J, Zou J, Yang C, et al. HOXC8 promotes proliferation and migration through transcriptional up-regulation of TGFbeta1 in non-small cell lung cancer. Oncogenesis. 2018;7(2):1.CrossRef Liu H, Zhang M, Xu S, Zhang J, Zou J, Yang C, et al. HOXC8 promotes proliferation and migration through transcriptional up-regulation of TGFbeta1 in non-small cell lung cancer. Oncogenesis. 2018;7(2):1.CrossRef
5.
go back to reference Rieger E, Bijl JJ, van Oostveen JW, Soyer HP, Oudejans CB, Jiwa NM, et al. Expression of the homeobox gene HOXC4 in keratinocytes of normal skin and epithelial skin tumors is correlated with differentiation. J Invest Dermatol. 1994;103(3):341–6.CrossRef Rieger E, Bijl JJ, van Oostveen JW, Soyer HP, Oudejans CB, Jiwa NM, et al. Expression of the homeobox gene HOXC4 in keratinocytes of normal skin and epithelial skin tumors is correlated with differentiation. J Invest Dermatol. 1994;103(3):341–6.CrossRef
6.
go back to reference Castronovo V, Kusaka M, Chariot A, Gielen J, Sobel M. Homeobox genes: potential candidates for the transcriptional control of the transformed and invasive phenotype. Biochem Pharmacol. 1994;47(1):137–43.CrossRef Castronovo V, Kusaka M, Chariot A, Gielen J, Sobel M. Homeobox genes: potential candidates for the transcriptional control of the transformed and invasive phenotype. Biochem Pharmacol. 1994;47(1):137–43.CrossRef
7.
go back to reference Faiella A, Zappavigna V, Mavilio F, Boncinelli E. Inhibition of retinoic acid-induced activation of 3′ human HOXB genes by antisense oligonucleotides affects sequential activation of genes located upstream in the four HOX clusters. Proc Natl Acad Sci U S A. 1994;91(12):5335–9.CrossRef Faiella A, Zappavigna V, Mavilio F, Boncinelli E. Inhibition of retinoic acid-induced activation of 3′ human HOXB genes by antisense oligonucleotides affects sequential activation of genes located upstream in the four HOX clusters. Proc Natl Acad Sci U S A. 1994;91(12):5335–9.CrossRef
8.
go back to reference Liu DB, Gu ZD, Cao XZ, Liu H, Li JY. Immunocytochemical detection of HoxD9 and Pbx1 homeodomain protein expression in Chinese esophageal squamous cell carcinomas. World J Gastroenterol. 2005;11(10):1562–6.CrossRef Liu DB, Gu ZD, Cao XZ, Liu H, Li JY. Immunocytochemical detection of HoxD9 and Pbx1 homeodomain protein expression in Chinese esophageal squamous cell carcinomas. World J Gastroenterol. 2005;11(10):1562–6.CrossRef
9.
go back to reference Tabuse M, Ohta S, Ohashi Y, Fukaya R, Misawa A, Yoshida K, et al. Functional analysis of HOXD9 in human gliomas and glioma cancer stem cells. Mol Cancer. 2011;10:60.CrossRef Tabuse M, Ohta S, Ohashi Y, Fukaya R, Misawa A, Yoshida K, et al. Functional analysis of HOXD9 in human gliomas and glioma cancer stem cells. Mol Cancer. 2011;10:60.CrossRef
10.
go back to reference Lv X, Li L, Lv L, Qu X, Jin S, Li K, et al. HOXD9 promotes epithelial-mesenchymal transition and cancer metastasis by ZEB1 regulation in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:133.CrossRef Lv X, Li L, Lv L, Qu X, Jin S, Li K, et al. HOXD9 promotes epithelial-mesenchymal transition and cancer metastasis by ZEB1 regulation in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:133.CrossRef
11.
go back to reference Wei Z, Sun M, Liu X, Zhang J, Jin Y. Rufy3, a protein specifically expressed in neurons, interacts with actin-bundling protein Fascin to control the growth of axons. J Neurochem. 2014;130(5):678–92.CrossRef Wei Z, Sun M, Liu X, Zhang J, Jin Y. Rufy3, a protein specifically expressed in neurons, interacts with actin-bundling protein Fascin to control the growth of axons. J Neurochem. 2014;130(5):678–92.CrossRef
12.
go back to reference Honda A, Usui H, Sakimura K, Igarashi M. Rufy3 is an adapter protein for small GTPases that activates a Rac guanine nucleotide exchange factor to control neuronal polarity. J Biol Chem. 2017;292(51):20936–46.CrossRef Honda A, Usui H, Sakimura K, Igarashi M. Rufy3 is an adapter protein for small GTPases that activates a Rac guanine nucleotide exchange factor to control neuronal polarity. J Biol Chem. 2017;292(51):20936–46.CrossRef
13.
go back to reference Yoshida H, Okumura N, Kitagishi Y, Shirafuji N, Matsuda S. Rab5(Q79L) interacts with the carboxyl terminus of RUFY3. Int J Biol Sci. 2010;6(2):187–9.CrossRef Yoshida H, Okumura N, Kitagishi Y, Shirafuji N, Matsuda S. Rab5(Q79L) interacts with the carboxyl terminus of RUFY3. Int J Biol Sci. 2010;6(2):187–9.CrossRef
14.
go back to reference Xie R, Wang J, Tang W, Li Y, Peng Y, Zhang H, et al. Rufy3 promotes metastasis through epithelial-mesenchymal transition in colorectal cancer. Cancer Lett. 2017;390:30–8.CrossRef Xie R, Wang J, Tang W, Li Y, Peng Y, Zhang H, et al. Rufy3 promotes metastasis through epithelial-mesenchymal transition in colorectal cancer. Cancer Lett. 2017;390:30–8.CrossRef
15.
go back to reference Wang G, Zhang Q, Song Y, Wang X, Guo Q, Zhang J, et al. PAK1 regulates RUFY3-mediated gastric cancer cell migration and invasion. Cell Death Dis. 2015;6:e1682.CrossRef Wang G, Zhang Q, Song Y, Wang X, Guo Q, Zhang J, et al. PAK1 regulates RUFY3-mediated gastric cancer cell migration and invasion. Cell Death Dis. 2015;6:e1682.CrossRef
16.
go back to reference Xie R, Wang J, Liu X, Wu L, Zhang H, Tang W, et al. RUFY3 interaction with FOXK1 promotes invasion and metastasis in colorectal cancer. Sci Rep. 2017;7(1):3709.CrossRef Xie R, Wang J, Liu X, Wu L, Zhang H, Tang W, et al. RUFY3 interaction with FOXK1 promotes invasion and metastasis in colorectal cancer. Sci Rep. 2017;7(1):3709.CrossRef
17.
go back to reference Zhang H, Wu X, Xiao Y, Wu L, Peng Y, Tang W, et al. Coexpression of FOXK1 and vimentin promotes EMT, migration, and invasion in gastric cancer cells. J Mol Med. 2018. Zhang H, Wu X, Xiao Y, Wu L, Peng Y, Tang W, et al. Coexpression of FOXK1 and vimentin promotes EMT, migration, and invasion in gastric cancer cells. J Mol Med. 2018.
18.
go back to reference Peng Y, Zhang P, Huang X, Yan Q, Wu M, Xie R, et al. Direct regulation of FOXK1 by C-Jun promotes proliferation, invasion and metastasis in gastric cancer cells. Cell Death Dis. 2016;7(11):e2480.CrossRef Peng Y, Zhang P, Huang X, Yan Q, Wu M, Xie R, et al. Direct regulation of FOXK1 by C-Jun promotes proliferation, invasion and metastasis in gastric cancer cells. Cell Death Dis. 2016;7(11):e2480.CrossRef
19.
go back to reference Liang L, Li X, Zhang X, Lv Z, He G, Zhao W, et al. MicroRNA-137, an HMGA1 target, suppresses colorectal cancer cell invasion and metastasis in mice by directly targeting FMNL2. Gastroenterology. 2013;144(3):624–35 e4.CrossRef Liang L, Li X, Zhang X, Lv Z, He G, Zhao W, et al. MicroRNA-137, an HMGA1 target, suppresses colorectal cancer cell invasion and metastasis in mice by directly targeting FMNL2. Gastroenterology. 2013;144(3):624–35 e4.CrossRef
20.
go back to reference Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science. 2002;298(5600):1911–2.CrossRef Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science. 2002;298(5600):1911–2.CrossRef
21.
go back to reference Huang X, Xiang L, Li Y, Zhao Y, Zhu H, Xiao Y, et al. Snail/FOXK1/Cyr61 signaling Axis regulates the epithelial-mesenchymal transition and metastasis in colorectal Cancer. Cell Physiol Biochem. 2018;47(2):590–603.CrossRef Huang X, Xiang L, Li Y, Zhao Y, Zhu H, Xiao Y, et al. Snail/FOXK1/Cyr61 signaling Axis regulates the epithelial-mesenchymal transition and metastasis in colorectal Cancer. Cell Physiol Biochem. 2018;47(2):590–603.CrossRef
22.
go back to reference Wu M, Wang J, Tang W, Zhan X, Li Y, Peng Y, et al. FOXK1 interaction with FHL2 promotes proliferation, invasion and metastasis in colorectal cancer. Oncogenesis. 2016;5(11):e271.CrossRef Wu M, Wang J, Tang W, Zhan X, Li Y, Peng Y, et al. FOXK1 interaction with FHL2 promotes proliferation, invasion and metastasis in colorectal cancer. Oncogenesis. 2016;5(11):e271.CrossRef
23.
go back to reference Bhatlekar S, Viswanathan V, Fields JZ, Boman BM. Overexpression of HOXA4 and HOXA9 genes promotes self-renewal and contributes to colon cancer stem cell overpopulation. J Cell Physiol. 2018;233(2):727–35.CrossRef Bhatlekar S, Viswanathan V, Fields JZ, Boman BM. Overexpression of HOXA4 and HOXA9 genes promotes self-renewal and contributes to colon cancer stem cell overpopulation. J Cell Physiol. 2018;233(2):727–35.CrossRef
24.
go back to reference Quagliata L, Quintavalle C, Lanzafame M, Matter MS, Novello C, di Tommaso L, et al. High expression of HOXA13 correlates with poorly differentiated hepatocellular carcinomas and modulates sorafenib response in in vitro models. Lab Investig. 2018;98(1):95–105.CrossRef Quagliata L, Quintavalle C, Lanzafame M, Matter MS, Novello C, di Tommaso L, et al. High expression of HOXA13 correlates with poorly differentiated hepatocellular carcinomas and modulates sorafenib response in in vitro models. Lab Investig. 2018;98(1):95–105.CrossRef
25.
go back to reference Shah M, Cardenas R, Wang B, Persson J, Mongan NP, Grabowska A, et al. HOXC8 regulates self-renewal, differentiation and transformation of breast cancer stem cells. Mol Cancer. 2017;16(1):38.CrossRef Shah M, Cardenas R, Wang B, Persson J, Mongan NP, Grabowska A, et al. HOXC8 regulates self-renewal, differentiation and transformation of breast cancer stem cells. Mol Cancer. 2017;16(1):38.CrossRef
26.
go back to reference Kelemen LE, Lawrenson K, Tyrer J, Li Q, Lee JM, Seo JH, et al. Genome-wide significant risk associations for mucinous ovarian carcinoma. Nat Genet. 2015;47(8):888–97.CrossRef Kelemen LE, Lawrenson K, Tyrer J, Li Q, Lee JM, Seo JH, et al. Genome-wide significant risk associations for mucinous ovarian carcinoma. Nat Genet. 2015;47(8):888–97.CrossRef
27.
go back to reference Li H, Huang CJ, Choo KB. Expression of homeobox genes in cervical cancer. Gynecol Oncol. 2002;84(2):216–21.CrossRef Li H, Huang CJ, Choo KB. Expression of homeobox genes in cervical cancer. Gynecol Oncol. 2002;84(2):216–21.CrossRef
28.
go back to reference Charlaftis N, Suddason T, Wu X, Anwar S, Karin M, Gallagher E. The MEKK1 PHD ubiquitinates TAB1 to activate MAPKs in response to cytokines. EMBO J. 2014;33(21):2581–96.CrossRef Charlaftis N, Suddason T, Wu X, Anwar S, Karin M, Gallagher E. The MEKK1 PHD ubiquitinates TAB1 to activate MAPKs in response to cytokines. EMBO J. 2014;33(21):2581–96.CrossRef
29.
go back to reference Hu XM, Liu YN, Zhang HL, Cao SB, Zhang T, Chen LP, et al. CXCL12/CXCR4 chemokine signaling in spinal glia induces pain hypersensitivity through MAPKs-mediated neuroinflammation in bone cancer rats. J Neurochem. 2015;132(4):452–63.CrossRef Hu XM, Liu YN, Zhang HL, Cao SB, Zhang T, Chen LP, et al. CXCL12/CXCR4 chemokine signaling in spinal glia induces pain hypersensitivity through MAPKs-mediated neuroinflammation in bone cancer rats. J Neurochem. 2015;132(4):452–63.CrossRef
30.
go back to reference Zhen X, Uryu K, Wang HY, Friedman E. D1 dopamine receptor agonists mediate activation of p38 mitogen-activated protein kinase and c-Jun amino-terminal kinase by a protein kinase A-dependent mechanism in SK-N-MC human neuroblastoma cells. Mol Pharmacol. 1998;54(3):453–8.CrossRef Zhen X, Uryu K, Wang HY, Friedman E. D1 dopamine receptor agonists mediate activation of p38 mitogen-activated protein kinase and c-Jun amino-terminal kinase by a protein kinase A-dependent mechanism in SK-N-MC human neuroblastoma cells. Mol Pharmacol. 1998;54(3):453–8.CrossRef
31.
go back to reference Jerjees DA, Alabdullah M, Alkaabi M, Abduljabbar R, Muftah A, Nolan C, et al. ERK1/2 is related to oestrogen receptor and predicts outcome in hormone-treated breast cancer. Breast Cancer Res Treat. 2014;147(1):25–37.CrossRef Jerjees DA, Alabdullah M, Alkaabi M, Abduljabbar R, Muftah A, Nolan C, et al. ERK1/2 is related to oestrogen receptor and predicts outcome in hormone-treated breast cancer. Breast Cancer Res Treat. 2014;147(1):25–37.CrossRef
32.
go back to reference Choi JH, Jeong YJ, Yu AR, Yoon KS, Choe W, Ha J, et al. Fluoxetine induces apoptosis through endoplasmic reticulum stress via mitogen-activated protein kinase activation and histone hyperacetylation in SK-N-BE(2)-M17 human neuroblastoma cells. Apoptosis. 2017;22(9):1079–97.CrossRef Choi JH, Jeong YJ, Yu AR, Yoon KS, Choe W, Ha J, et al. Fluoxetine induces apoptosis through endoplasmic reticulum stress via mitogen-activated protein kinase activation and histone hyperacetylation in SK-N-BE(2)-M17 human neuroblastoma cells. Apoptosis. 2017;22(9):1079–97.CrossRef
33.
go back to reference Yi YS, Baek KS, Cho JY. L1 cell adhesion molecule induces melanoma cell motility by activation of mitogen-activated protein kinase pathways. Die Pharmazie. 2014;69(6):461–7.PubMed Yi YS, Baek KS, Cho JY. L1 cell adhesion molecule induces melanoma cell motility by activation of mitogen-activated protein kinase pathways. Die Pharmazie. 2014;69(6):461–7.PubMed
34.
go back to reference Luo J, Miller MW. Transforming growth factor beta1-regulated cell proliferation and expression of neural cell adhesion molecule in B104 neuroblastoma cells: differential effects of ethanol. J Neurochem. 1999;72(6):2286–93.CrossRef Luo J, Miller MW. Transforming growth factor beta1-regulated cell proliferation and expression of neural cell adhesion molecule in B104 neuroblastoma cells: differential effects of ethanol. J Neurochem. 1999;72(6):2286–93.CrossRef
35.
go back to reference Cong Q, Jia H, Li P, Qiu S, Yeh J, Wang Y, et al. p38alpha MAPK regulates proliferation and differentiation of osteoclast progenitors and bone remodeling in an aging-dependent manner. Sci Rep. 2017;7:45964.CrossRef Cong Q, Jia H, Li P, Qiu S, Yeh J, Wang Y, et al. p38alpha MAPK regulates proliferation and differentiation of osteoclast progenitors and bone remodeling in an aging-dependent manner. Sci Rep. 2017;7:45964.CrossRef
36.
go back to reference McEwen DG, Peifer M. Puckered, a Drosophila MAPK phosphatase, ensures cell viability by antagonizing JNK-induced apoptosis. Development. 2005;132(17):3935–46.CrossRef McEwen DG, Peifer M. Puckered, a Drosophila MAPK phosphatase, ensures cell viability by antagonizing JNK-induced apoptosis. Development. 2005;132(17):3935–46.CrossRef
37.
go back to reference Hao W, Yuan X, Yu L, Gao C, Sun X, Wang D, et al. Licochalcone A-induced human gastric cancer BGC-823 cells apoptosis by regulating ROS-mediated MAPKs and PI3K/AKT signaling pathways. Sci Rep. 2015;5:10336.CrossRef Hao W, Yuan X, Yu L, Gao C, Sun X, Wang D, et al. Licochalcone A-induced human gastric cancer BGC-823 cells apoptosis by regulating ROS-mediated MAPKs and PI3K/AKT signaling pathways. Sci Rep. 2015;5:10336.CrossRef
38.
go back to reference Long W, Foulds CE, Qin J, Liu J, Ding C, Lonard DM, et al. ERK3 signals through SRC-3 coactivator to promote human lung cancer cell invasion. J Clin Invest. 2012;122(5):1869–80.CrossRef Long W, Foulds CE, Qin J, Liu J, Ding C, Lonard DM, et al. ERK3 signals through SRC-3 coactivator to promote human lung cancer cell invasion. J Clin Invest. 2012;122(5):1869–80.CrossRef
39.
go back to reference Fister S, Gunthert AR, Aicher B, Paulini KW, Emons G, Grundker C. GnRH-II antagonists induce apoptosis in human endometrial, ovarian, and breast cancer cells via activation of stress-induced MAPKs p38 and JNK and proapoptotic protein Bax. Cancer Res. 2009;69(16):6473–81.CrossRef Fister S, Gunthert AR, Aicher B, Paulini KW, Emons G, Grundker C. GnRH-II antagonists induce apoptosis in human endometrial, ovarian, and breast cancer cells via activation of stress-induced MAPKs p38 and JNK and proapoptotic protein Bax. Cancer Res. 2009;69(16):6473–81.CrossRef
40.
go back to reference Yang L, Ling Y, Zhang Z, Zhao Q, Tang J, Ji H, et al. ZL11n is a novel nitric oxide-releasing derivative of farnesylthiosalicylic acid that induces apoptosis in human hepatoma HepG2 cells via MAPK/mitochondrial pathways. Biochem Biophys Res Commun. 2011;409(4):752–7.CrossRef Yang L, Ling Y, Zhang Z, Zhao Q, Tang J, Ji H, et al. ZL11n is a novel nitric oxide-releasing derivative of farnesylthiosalicylic acid that induces apoptosis in human hepatoma HepG2 cells via MAPK/mitochondrial pathways. Biochem Biophys Res Commun. 2011;409(4):752–7.CrossRef
41.
go back to reference Zhi Q, Chen H, Liu F, Han Y, Wan D, Xu Z, et al. Podocalyxin-like protein promotes gastric cancer progression through interacting with RUN and FYVE domain containing 1 protein. Cancer Sci. 2019;110(1):118–34.PubMed Zhi Q, Chen H, Liu F, Han Y, Wan D, Xu Z, et al. Podocalyxin-like protein promotes gastric cancer progression through interacting with RUN and FYVE domain containing 1 protein. Cancer Sci. 2019;110(1):118–34.PubMed
42.
go back to reference Yang J, Kim O, Wu J, Qiu Y. Interaction between tyrosine kinase Etk and a RUN domain- and FYVE domain-containing protein RUFY1. A possible role of ETK in regulation of vesicle trafficking. J Biol Chem. 2002;277(33):30219–26.CrossRef Yang J, Kim O, Wu J, Qiu Y. Interaction between tyrosine kinase Etk and a RUN domain- and FYVE domain-containing protein RUFY1. A possible role of ETK in regulation of vesicle trafficking. J Biol Chem. 2002;277(33):30219–26.CrossRef
Metadata
Title
HOXD9 promotes the growth, invasion and metastasis of gastric cancer cells by transcriptional activation of RUFY3
Authors
Huiqiong Zhu
Weiyu Dai
Jiaying Li
Li Xiang
Xiaosheng Wu
Weimei Tang
Yaying Chen
Qiong Yang
Mengwei Liu
Yizhi Xiao
Wenjing Zhang
Jianjiao Lin
Jing Wang
Guangnan Liu
Yong Sun
Ping Jiang
Guoxin Li
Aimin Li
Side Liu
Ye Chen
Jide Wang
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1399-1

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 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