Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Research

ESCO2 promotes lung adenocarcinoma progression by regulating hnRNPA1 acetylation

Authors: Hui-er Zhu, Tao Li, Shengnan Shi, De-xiong Chen, Weiping Chen, Hui Chen

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Emerging evidence indicates that metabolism reprogramming and abnormal acetylation modification play an important role in lung adenocarcinoma (LUAD) progression, although the mechanism is largely unknown.

Methods

Here, we used three public databases (Oncomine, Gene Expression Omnibus [GEO], The Cancer Genome Atlas [TCGA]) to analyze ESCO2 (establishment of cohesion 1 homolog 2) expression in LUAD. The biological function of ESCO2 was studiedusing cell proliferation, colony formation, cell migration, and invasion assays in vitro, and mouse xenograft models in vivo. ESCO2 interacting proteins were searched using gene set enrichment analysis (GSEA) and mass spectrometry. Pyruvate kinase M1/2 (PKM) mRNA splicing assay was performed using RT-PCR together with restriction digestion. LUAD cell metabolism was studied using glucose uptake assays and lactate production. ESCO2 expression was significantly upregulated in LUAD tissues, and higher ESCO2 expression indicated worse prognosis for patients with LUAD.

Results

We found that ESCO2 promoted LUAD cell proliferation and metastasis metabolic reprogramming in vitro and in vivo. Mechanistically, ESCO2 increased hnRNPA1 (heterogeneous nuclear ribonucleoprotein A1) binding to the intronic sequences flanking exon 9 (EI9) of PKM mRNA by inhibiting hnRNPA1 nuclear translocation, eventually inhibiting PKM1 isoform formation and inducing PKM2 isoform formation.

Conclusions

Our findings confirm that ESCO2 is a key factor in promoting LUAD malignant progression and suggest that it is a new target for treating LUAD.
Appendix
Available only for authorised users
Literature
2.
go back to reference Liang W, Zhang L, Jiang G, Wang Q, Liu L, Liu D, et al. Development and validation of a nomogram for predicting survival in patients with resected non-small-cell lung cancer. J Clin Oncol. 2015;33(8):861–9.PubMedCrossRef Liang W, Zhang L, Jiang G, Wang Q, Liu L, Liu D, et al. Development and validation of a nomogram for predicting survival in patients with resected non-small-cell lung cancer. J Clin Oncol. 2015;33(8):861–9.PubMedCrossRef
3.
go back to reference Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.PubMedCrossRef Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.PubMedCrossRef
4.
go back to reference Zhu HE, Yin JY, Chen DX, He S, Chen H. Agmatinase promotes the lung adenocarcinoma tumorigenesis by activating the NO-MAPKs-PI3K/Akt pathway. Cell Death Dis. 2019;10(11):854.PubMedPubMedCentralCrossRef Zhu HE, Yin JY, Chen DX, He S, Chen H. Agmatinase promotes the lung adenocarcinoma tumorigenesis by activating the NO-MAPKs-PI3K/Akt pathway. Cell Death Dis. 2019;10(11):854.PubMedPubMedCentralCrossRef
5.
go back to reference Shaw AT, Solomon BJ, Besse B, Bauer TM, Lin CC, Soo RA, et al. ALK resistance mutations and efficacy of Lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung Cancer. J Clin Oncol. 2019;37(16):1370–9.PubMedPubMedCentralCrossRef Shaw AT, Solomon BJ, Besse B, Bauer TM, Lin CC, Soo RA, et al. ALK resistance mutations and efficacy of Lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung Cancer. J Clin Oncol. 2019;37(16):1370–9.PubMedPubMedCentralCrossRef
6.
go back to reference Kobayashi K, Soejima K, Fukunaga K, Shintani Y, Sekine I, Shukuya T, et al. Key prognostic factors for EGFR-mutated non-adenocarcinoma lung cancer patients in the Japanese joint Committee of Lung Cancer Registry Database. Lung Cancer. 2020;146:236–43.PubMedCrossRef Kobayashi K, Soejima K, Fukunaga K, Shintani Y, Sekine I, Shukuya T, et al. Key prognostic factors for EGFR-mutated non-adenocarcinoma lung cancer patients in the Japanese joint Committee of Lung Cancer Registry Database. Lung Cancer. 2020;146:236–43.PubMedCrossRef
7.
go back to reference Chen JA, Riess JW. Optimal Management of Patients with advanced NSCLC harboring high PD-L1 expression and driver mutations. Curr Treat Options in Oncol. 2020;21(7):60.CrossRef Chen JA, Riess JW. Optimal Management of Patients with advanced NSCLC harboring high PD-L1 expression and driver mutations. Curr Treat Options in Oncol. 2020;21(7):60.CrossRef
8.
go back to reference Sankar K, Gadgeel SM, Qin A. Molecular therapeutic targets in non-small cell lung cancer. Expert Rev Anticancer Ther. 2020;20(8):647–61.PubMedCrossRef Sankar K, Gadgeel SM, Qin A. Molecular therapeutic targets in non-small cell lung cancer. Expert Rev Anticancer Ther. 2020;20(8):647–61.PubMedCrossRef
9.
go back to reference Noor ZS, Cummings AL, Johnson MM, Spiegel ML, Goldman JW. Targeted therapy for non-small cell lung Cancer. Semin Respir Crit Care Med. 2020;41(3):409–34.PubMedCrossRef Noor ZS, Cummings AL, Johnson MM, Spiegel ML, Goldman JW. Targeted therapy for non-small cell lung Cancer. Semin Respir Crit Care Med. 2020;41(3):409–34.PubMedCrossRef
10.
go back to reference Gu J, Yao W, Shi P, Zhang G, Owonikoko TK, Ramalingam SS, et al. MEK or ERK inhibition effectively abrogates emergence of acquired osimertinib resistance in the treatment of epidermal growth factor receptor-mutant lung cancers. Cancer. 2020;126(16):3788–99.PubMedCrossRef Gu J, Yao W, Shi P, Zhang G, Owonikoko TK, Ramalingam SS, et al. MEK or ERK inhibition effectively abrogates emergence of acquired osimertinib resistance in the treatment of epidermal growth factor receptor-mutant lung cancers. Cancer. 2020;126(16):3788–99.PubMedCrossRef
11.
go back to reference Calvayrac O, Pradines A, Pons E, Mazieres J, Guibert N. Molecular biomarkers for lung adenocarcinoma. Eur Respir J. 2017;49(4):1601734.PubMedCrossRef Calvayrac O, Pradines A, Pons E, Mazieres J, Guibert N. Molecular biomarkers for lung adenocarcinoma. Eur Respir J. 2017;49(4):1601734.PubMedCrossRef
12.
go back to reference Testa U, Castelli G, Pelosi E. Lung Cancers: Molecular Characterization, Clonal Heterogeneity and Evolution, and Cancer Stem Cells. Cancers (Basel). 2018;10(8):248.CrossRef Testa U, Castelli G, Pelosi E. Lung Cancers: Molecular Characterization, Clonal Heterogeneity and Evolution, and Cancer Stem Cells. Cancers (Basel). 2018;10(8):248.CrossRef
13.
go back to reference Alomer RM, da Silva EML, Chen J, Piekarz KM, McDonald K, Sansam CG, et al. Esco1 and Esco2 regulate distinct cohesin functions during cell cycle progression. Proc Natl Acad Sci U S A. 2017;114(37):9906–11.PubMedPubMedCentralCrossRef Alomer RM, da Silva EML, Chen J, Piekarz KM, McDonald K, Sansam CG, et al. Esco1 and Esco2 regulate distinct cohesin functions during cell cycle progression. Proc Natl Acad Sci U S A. 2017;114(37):9906–11.PubMedPubMedCentralCrossRef
14.
go back to reference Guo XB, Huang B, Pan YH, Su SG, Li Y. ESCO2 inhibits tumor metastasis via transcriptionally repressing MMP2 in colorectal cancer. Cancer Manag Res. 2018;10:6157–66.PubMedPubMedCentralCrossRef Guo XB, Huang B, Pan YH, Su SG, Li Y. ESCO2 inhibits tumor metastasis via transcriptionally repressing MMP2 in colorectal cancer. Cancer Manag Res. 2018;10:6157–66.PubMedPubMedCentralCrossRef
15.
go back to reference Wang QL, Liu L. Establishment of cohesion 1 homolog 2 facilitates cell aggressive behaviors and induces poor prognosis in renal cell carcinoma. J Clin Lab Anal. 2020;34(5):e23163.PubMedPubMedCentralCrossRef Wang QL, Liu L. Establishment of cohesion 1 homolog 2 facilitates cell aggressive behaviors and induces poor prognosis in renal cell carcinoma. J Clin Lab Anal. 2020;34(5):e23163.PubMedPubMedCentralCrossRef
16.
go back to reference Xiao B, Chen L, Ke Y, Hang J, Cao L, Zhang R, et al. Identification of methylation sites and signature genes with prognostic value for luminal breast cancer. BMC Cancer. 2018;18(1):405.PubMedPubMedCentralCrossRef Xiao B, Chen L, Ke Y, Hang J, Cao L, Zhang R, et al. Identification of methylation sites and signature genes with prognostic value for luminal breast cancer. BMC Cancer. 2018;18(1):405.PubMedPubMedCentralCrossRef
17.
go back to reference Zhang W, Cui Q, Qu W, Ding X, Jiang D, Liu H. TRIM58/cg26157385 methylation is associated with eight prognostic genes in lung squamous cell carcinoma. Oncol Rep. 2018;40(1):206–16.PubMedPubMedCentral Zhang W, Cui Q, Qu W, Ding X, Jiang D, Liu H. TRIM58/cg26157385 methylation is associated with eight prognostic genes in lung squamous cell carcinoma. Oncol Rep. 2018;40(1):206–16.PubMedPubMedCentral
18.
go back to reference Ryu B, Kim DS, Deluca AM, Alani RM. Comprehensive expression profiling of tumor cell lines identifies molecular signatures of melanoma progression. PLoS One. 2007;2(7):e594.PubMedPubMedCentralCrossRef Ryu B, Kim DS, Deluca AM, Alani RM. Comprehensive expression profiling of tumor cell lines identifies molecular signatures of melanoma progression. PLoS One. 2007;2(7):e594.PubMedPubMedCentralCrossRef
19.
go back to reference Chen H, Zhang L, He W, Liu T, Zhao Y, Chen H, et al. ESCO2 knockdown inhibits cell proliferation and induces apoptosis in human gastric cancer cells. Biochem Biophys Res Commun. 2018;496(2):475–81.PubMedCrossRef Chen H, Zhang L, He W, Liu T, Zhao Y, Chen H, et al. ESCO2 knockdown inhibits cell proliferation and induces apoptosis in human gastric cancer cells. Biochem Biophys Res Commun. 2018;496(2):475–81.PubMedCrossRef
20.
go back to reference Kedzierska H, Piekielko-Witkowska A. Splicing factors of SR and hnRNP families as regulators of apoptosis in cancer. Cancer Lett. 2017;396:53–65.PubMedCrossRef Kedzierska H, Piekielko-Witkowska A. Splicing factors of SR and hnRNP families as regulators of apoptosis in cancer. Cancer Lett. 2017;396:53–65.PubMedCrossRef
21.
go back to reference Kutluay SB, Emery A, Penumutchu SR, Townsend D, Tenneti K, Madison MK, et al. Genome-Wide Analysis of Heterogeneous Nuclear Ribonucleoprotein (hnRNP) Binding to HIV-1 RNA Reveals a Key Role for hnRNP H1 in Alternative Viral mRNA Splicing. J Virol. 2019;93(21):e01048–19.PubMedPubMedCentralCrossRef Kutluay SB, Emery A, Penumutchu SR, Townsend D, Tenneti K, Madison MK, et al. Genome-Wide Analysis of Heterogeneous Nuclear Ribonucleoprotein (hnRNP) Binding to HIV-1 RNA Reveals a Key Role for hnRNP H1 in Alternative Viral mRNA Splicing. J Virol. 2019;93(21):e01048–19.PubMedPubMedCentralCrossRef
23.
go back to reference David CJ, Chen M, Assanah M, Canoll P, Manley JL. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463(7279):364–8.PubMedCrossRef David CJ, Chen M, Assanah M, Canoll P, Manley JL. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463(7279):364–8.PubMedCrossRef
24.
go back to reference Kuranaga Y, Sugito N, Shinohara H, Tsujino T, Taniguchi K, Komura K, et al. SRSF3, a Splicer of the PKM Gene, Regulates Cell Growth and Maintenance of Cancer-Specific Energy Metabolism in Colon Cancer Cells. Int J Mol Sci. 2018;19(10):3012.PubMedCentralCrossRef Kuranaga Y, Sugito N, Shinohara H, Tsujino T, Taniguchi K, Komura K, et al. SRSF3, a Splicer of the PKM Gene, Regulates Cell Growth and Maintenance of Cancer-Specific Energy Metabolism in Colon Cancer Cells. Int J Mol Sci. 2018;19(10):3012.PubMedCentralCrossRef
25.
go back to reference Robbins Y, Greene S, Friedman J, Clavijo PE, Van Waes C, Fabian KP, et al. Tumor control via targeting PD-L1 with chimeric antigen receptor modified NK cells. Elife. 2020;9:e54854.PubMedPubMedCentralCrossRef Robbins Y, Greene S, Friedman J, Clavijo PE, Van Waes C, Fabian KP, et al. Tumor control via targeting PD-L1 with chimeric antigen receptor modified NK cells. Elife. 2020;9:e54854.PubMedPubMedCentralCrossRef
26.
go back to reference Huang JZ, Chen M, Chen D, Gao XC, Zhu S, Huang H, et al. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses Colon Cancer growth. Mol Cell. 2017;68(1):171–84 e6.PubMedCrossRef Huang JZ, Chen M, Chen D, Gao XC, Zhu S, Huang H, et al. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses Colon Cancer growth. Mol Cell. 2017;68(1):171–84 e6.PubMedCrossRef
27.
go back to reference Chabot B, LeBel C, Hutchison S, Nasim FH, Simard MJ. Heterogeneous nuclear ribonucleoprotein particle a/B proteins and the control of alternative splicing of the mammalian heterogeneous nuclear ribonucleoprotein particle A1 pre-mRNA. Prog Mol Subcell Biol. 2003;31:59–88.PubMedCrossRef Chabot B, LeBel C, Hutchison S, Nasim FH, Simard MJ. Heterogeneous nuclear ribonucleoprotein particle a/B proteins and the control of alternative splicing of the mammalian heterogeneous nuclear ribonucleoprotein particle A1 pre-mRNA. Prog Mol Subcell Biol. 2003;31:59–88.PubMedCrossRef
28.
go back to reference Izaurralde E, Jarmolowski A, Beisel C, Mattaj IW, Dreyfuss G, Fischer U. A role for the M9 transport signal of hnRNP A1 in mRNA nuclear export. J Cell Biol. 1997;137(1):27–35.PubMedPubMedCentralCrossRef Izaurralde E, Jarmolowski A, Beisel C, Mattaj IW, Dreyfuss G, Fischer U. A role for the M9 transport signal of hnRNP A1 in mRNA nuclear export. J Cell Biol. 1997;137(1):27–35.PubMedPubMedCentralCrossRef
29.
go back to reference Iijima M, Suzuki M, Tanabe A, Nishimura A, Yamada M. Two motifs essential for nuclear import of the hnRNP A1 nucleocytoplasmic shuttling sequence M9 core. FEBS Lett. 2006;580(5):1365–70.PubMedCrossRef Iijima M, Suzuki M, Tanabe A, Nishimura A, Yamada M. Two motifs essential for nuclear import of the hnRNP A1 nucleocytoplasmic shuttling sequence M9 core. FEBS Lett. 2006;580(5):1365–70.PubMedCrossRef
30.
go back to reference Chen M, David CJ, Manley JL. Concentration-dependent control of pyruvate kinase M mutually exclusive splicing by hnRNP proteins. Nat Struct Mol Biol. 2012;19(3):346–54.PubMedPubMedCentralCrossRef Chen M, David CJ, Manley JL. Concentration-dependent control of pyruvate kinase M mutually exclusive splicing by hnRNP proteins. Nat Struct Mol Biol. 2012;19(3):346–54.PubMedPubMedCentralCrossRef
31.
go back to reference Chen M, Sheng XJ, Qin YY, Zhu S, Wu QX, Jia L, et al. TBC1D8 amplification drives tumorigenesis through metabolism reprogramming in ovarian Cancer. Theranostics. 2019;9(3):676–90.PubMedPubMedCentralCrossRef Chen M, Sheng XJ, Qin YY, Zhu S, Wu QX, Jia L, et al. TBC1D8 amplification drives tumorigenesis through metabolism reprogramming in ovarian Cancer. Theranostics. 2019;9(3):676–90.PubMedPubMedCentralCrossRef
32.
go back to reference Guo R, Li Y, Ning J, Sun D, Lin L, Liu X. HnRNP A1/A2 and SF2/ASF regulate alternative splicing of interferon regulatory factor-3 and affect immunomodulatory functions in human non-small cell lung cancer cells. PLoS One. 2013;8(4):e62729.PubMedPubMedCentralCrossRef Guo R, Li Y, Ning J, Sun D, Lin L, Liu X. HnRNP A1/A2 and SF2/ASF regulate alternative splicing of interferon regulatory factor-3 and affect immunomodulatory functions in human non-small cell lung cancer cells. PLoS One. 2013;8(4):e62729.PubMedPubMedCentralCrossRef
33.
go back to reference Yang H, Zhu R, Zhao X, Liu L, Zhou Z, Zhao L, et al. Sirtuin-mediated deacetylation of hnRNP A1 suppresses glycolysis and growth in hepatocellular carcinoma. Oncogene. 2019;38(25):4915–31.PubMedCrossRef Yang H, Zhu R, Zhao X, Liu L, Zhou Z, Zhao L, et al. Sirtuin-mediated deacetylation of hnRNP A1 suppresses glycolysis and growth in hepatocellular carcinoma. Oncogene. 2019;38(25):4915–31.PubMedCrossRef
34.
go back to reference Carabet LA, Leblanc E, Lallous N, Morin H, Ghaidi F, Lee J, et al. Computer-Aided Discovery of Small Molecules Targeting the RNA Splicing Activity of hnRNP A1 in Castration-Resistant Prostate Cancer. Molecules. 2019;24(4):763.PubMedCentralCrossRef Carabet LA, Leblanc E, Lallous N, Morin H, Ghaidi F, Lee J, et al. Computer-Aided Discovery of Small Molecules Targeting the RNA Splicing Activity of hnRNP A1 in Castration-Resistant Prostate Cancer. Molecules. 2019;24(4):763.PubMedCentralCrossRef
35.
go back to reference Yu C, Guo J, Liu Y, Jia J, Jia R, Fan M. Oral squamous cancer cell exploits hnRNP A1 to regulate cell cycle and proliferation. J Cell Physiol. 2015;230(9):2252–61.PubMedCrossRef Yu C, Guo J, Liu Y, Jia J, Jia R, Fan M. Oral squamous cancer cell exploits hnRNP A1 to regulate cell cycle and proliferation. J Cell Physiol. 2015;230(9):2252–61.PubMedCrossRef
36.
go back to reference Taniguchi K, Sugito N, Shinohara H, Kuranaga Y, Inomata Y, Komura K, et al. Organ-Specific MicroRNAs (MIR122, 137, and 206) Contribute to Tissue Characteristics and Carcinogenesis by Regulating Pyruvate Kinase M1/2 (PKM) Expression. Int J Mol Sci. 2018;19(5):1276.PubMedCentralCrossRef Taniguchi K, Sugito N, Shinohara H, Kuranaga Y, Inomata Y, Komura K, et al. Organ-Specific MicroRNAs (MIR122, 137, and 206) Contribute to Tissue Characteristics and Carcinogenesis by Regulating Pyruvate Kinase M1/2 (PKM) Expression. Int J Mol Sci. 2018;19(5):1276.PubMedCentralCrossRef
37.
go back to reference Massari F, Ciccarese C, Santoni M, Iacovelli R, Mazzucchelli R, Piva F, et al. Metabolic phenotype of bladder cancer. Cancer Treat Rev. 2016;45:46–57.PubMedCrossRef Massari F, Ciccarese C, Santoni M, Iacovelli R, Mazzucchelli R, Piva F, et al. Metabolic phenotype of bladder cancer. Cancer Treat Rev. 2016;45:46–57.PubMedCrossRef
38.
go back to reference Calabretta S, Bielli P, Passacantilli I, Pilozzi E, Fendrich V, Capurso G, et al. Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene. 2016;35(16):2031–9.PubMedCrossRef Calabretta S, Bielli P, Passacantilli I, Pilozzi E, Fendrich V, Capurso G, et al. Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene. 2016;35(16):2031–9.PubMedCrossRef
39.
go back to reference Zhao X, Zhu Y, Hu J, Jiang L, Li L, Jia S, et al. Shikonin inhibits tumor growth in mice by suppressing pyruvate kinase M2-mediated aerobic glycolysis. Sci Rep. 2018;8(1):14517.PubMedPubMedCentralCrossRef Zhao X, Zhu Y, Hu J, Jiang L, Li L, Jia S, et al. Shikonin inhibits tumor growth in mice by suppressing pyruvate kinase M2-mediated aerobic glycolysis. Sci Rep. 2018;8(1):14517.PubMedPubMedCentralCrossRef
41.
go back to reference Rahman S, Jones MJ, Jallepalli PV. Cohesin recruits the Esco1 acetyltransferase genome wide to repress transcription and promote cohesion in somatic cells. Proc Natl Acad Sci U S A. 2015;112(36):11270–5.PubMedPubMedCentralCrossRef Rahman S, Jones MJ, Jallepalli PV. Cohesin recruits the Esco1 acetyltransferase genome wide to repress transcription and promote cohesion in somatic cells. Proc Natl Acad Sci U S A. 2015;112(36):11270–5.PubMedPubMedCentralCrossRef
42.
go back to reference Ooi L, Wood IC. Chromatin crosstalk in development and disease: lessons from REST. Nat Rev Genet. 2007;8(7):544–54.PubMedCrossRef Ooi L, Wood IC. Chromatin crosstalk in development and disease: lessons from REST. Nat Rev Genet. 2007;8(7):544–54.PubMedCrossRef
43.
go back to reference Qureshi IA, Gokhan S, Mehler MF. REST and CoREST are transcriptional and epigenetic regulators of seminal neural fate decisions. Cell Cycle. 2010;9(22):4477–86.PubMedPubMedCentralCrossRef Qureshi IA, Gokhan S, Mehler MF. REST and CoREST are transcriptional and epigenetic regulators of seminal neural fate decisions. Cell Cycle. 2010;9(22):4477–86.PubMedPubMedCentralCrossRef
Metadata
Title
ESCO2 promotes lung adenocarcinoma progression by regulating hnRNPA1 acetylation
Authors
Hui-er Zhu
Tao Li
Shengnan Shi
De-xiong Chen
Weiping Chen
Hui Chen
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01858-1

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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