Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

Dacomitinib potentiates the efficacy of conventional chemotherapeutic agents via inhibiting the drug efflux function of ABCG2 in vitro and in vivo

Authors: Xiaoran Guo, Kenneth K. W. To, Zhen Chen, Xiaokun Wang, Jianye Zhang, Min Luo, Fang Wang, Shirong Yan, Liwu Fu

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

ATP-binding cassette subfamily G member 2 (ABCG2), a member of the ABC transporter superfamily proteins, mediates multidrug resistance (MDR) by transporting substrate anticancer drugs out of cancer cells and decreasing their intracellular accumulation. MDR is a major hurdle to successful chemotherapy. A logical approach to overcome MDR is to inhibit the transporter. However, no safe and effective MDR inhibitor has been approved in the clinic.

Methods

The MTT assay was used to evaluate cell cytotoxicity and MDR reversal effect. Drug efflux and intracellular drug accumulation were measured by flow cytometry. The H460/MX20 cell xenograft model was established to evaluate the enhancement of anticancer efficacy of topotecan by dacomitinib in vivo. To ascertain the interaction of dacomitinib with the substrate binding sites of ABCG2, the competition of dacomitinib for photolabeling of ABCG2 with [125I]- iodoarylazidoprazosin (IAAP) was performed. Vanadate-sensitive ATPase activity of ABCG2 was measured in the presence of a range of different concentrations of dacomitinib to evaluate the effect of dacomitinib on ATP hydrolysis as the energy source of the transporter. A flow cytometry-based assay and western blotting were employed to study whether dacomitininb could inhibit the expression level of ABCG2. The mRNA expression levels of ABCG2 were analyzed by real-time quantitative RT-PCR. The protein expression level of AKT, ERK and their phosphorylations were detected by Western blotting.

Results

Here, we found that dacomitinib, an irreversible pan-ErbB tyrosine kinase inhibitor (TKI) in phase III clinical trial, could enhance the efficacy of conventional chemotherapeutic agents specifically in ABCG2-overexpressing MDR cancer cells but not in the parental sensitive cells. Dacomitinib was found to significantly increase the accumulation of ABCG2 probe substrates [doxorubicin (DOX),Rhodamine 123 (Rho 123) and Hoechst 33342] by inhibiting the transporter efflux function. Moreover, dacomitinib stimulated ABCG2 ATPase activity and competed with [125I]-IAAP photolabeling of ABCG2 in a concentration-dependent manner. However, dacomitinib did not alter ABCG2 expression at protein and mRNA levels or inhibit ErbB downstream signaling of AKT and ERK. Importantly, dacomitinib significantly enhanced the efficacy of topotecan in ABCG2-overexpressing H460/MX20 cell xenografts in nude mice without incurring additional toxicity.

Conclusions

These results suggest that dacomitinib reverses ABCG2-mediated MDR by inhibiting ABCG2 efflux function and increasing intracellular accumulation of anticancer agents. Our findings advocate further clinical investigation of combinations of dacomitinib and conventional chemotherapy in cancer patients with ABCG2-overexpressing MDR tumors.
Literature
1.
go back to reference Hasanabady MH, Kalalinia F. ABCG2 inhibition as a therapeutic approach for overcoming multidrug resistance in cancer. J Biosci. 2016;41:313–24.CrossRefPubMed Hasanabady MH, Kalalinia F. ABCG2 inhibition as a therapeutic approach for overcoming multidrug resistance in cancer. J Biosci. 2016;41:313–24.CrossRefPubMed
3.
go back to reference Choudhuri S, Klaassen CD. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int J Toxicol. 2006;25:231–59.CrossRefPubMed Choudhuri S, Klaassen CD. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int J Toxicol. 2006;25:231–59.CrossRefPubMed
4.
go back to reference Gatti L, Beretta GL, Cossa G, Zunino F, Perego P. ABC transporters as potential targets for modulation of drug resistance. Mini-Rev Med Chem. 2009;9:1102.CrossRefPubMed Gatti L, Beretta GL, Cossa G, Zunino F, Perego P. ABC transporters as potential targets for modulation of drug resistance. Mini-Rev Med Chem. 2009;9:1102.CrossRefPubMed
5.
go back to reference Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A. 1998;95:15665–70.CrossRefPubMedPubMedCentral Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A. 1998;95:15665–70.CrossRefPubMedPubMedCentral
6.
go back to reference Jain HD, Zhang C, Zhou S, Zhou H, Ma J, Liu X, et al. Synthesis and structure-activity relationship studies on tryprostatin A, an inhibitor of breast cancer resistance protein. Bioorg Med Chem. 2008;16:4626–51.CrossRefPubMedPubMedCentral Jain HD, Zhang C, Zhou S, Zhou H, Ma J, Liu X, et al. Synthesis and structure-activity relationship studies on tryprostatin A, an inhibitor of breast cancer resistance protein. Bioorg Med Chem. 2008;16:4626–51.CrossRefPubMedPubMedCentral
7.
go back to reference Borst P, Elferink RO. Mammalian ABC transporters in health and disease. Annu Rev Biochem. 2002;71:537–92.CrossRefPubMed Borst P, Elferink RO. Mammalian ABC transporters in health and disease. Annu Rev Biochem. 2002;71:537–92.CrossRefPubMed
8.
go back to reference Han JY, Lim HS, Yoo YK, Shin ES, Park YH, Lee SY, et al. Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Cancer. 2007;110:138–47.CrossRefPubMed Han JY, Lim HS, Yoo YK, Shin ES, Park YH, Lee SY, et al. Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Cancer. 2007;110:138–47.CrossRefPubMed
9.
go back to reference Zhou Y, Sridhar R, Shan L, Sha W, Gu X, Sukumar S. Loperamide, an FDA-approved antidiarrhea drug, effectively reverses the resistance of multidrug resistant MCF-7/MDR1 human breast cancer cells to doxorubicin-induced cytotoxicity. Cancer Investig. 2012;30:119–25.CrossRef Zhou Y, Sridhar R, Shan L, Sha W, Gu X, Sukumar S. Loperamide, an FDA-approved antidiarrhea drug, effectively reverses the resistance of multidrug resistant MCF-7/MDR1 human breast cancer cells to doxorubicin-induced cytotoxicity. Cancer Investig. 2012;30:119–25.CrossRef
10.
go back to reference Li J, Xu LZ, He KL, Guo WJ, Zheng YH, Xia P, et al. Reversal effects of nomegestrol acetate on multidrug resistance in adriamycin-resistant MCF7 breast cancer cell line. Breast Cancer Res. 2001;3:1–11.CrossRef Li J, Xu LZ, He KL, Guo WJ, Zheng YH, Xia P, et al. Reversal effects of nomegestrol acetate on multidrug resistance in adriamycin-resistant MCF7 breast cancer cell line. Breast Cancer Res. 2001;3:1–11.CrossRef
11.
go back to reference Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nature reviews. Drug Des Discov. 2006;5:219–34.CrossRef Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nature reviews. Drug Des Discov. 2006;5:219–34.CrossRef
12.
go back to reference Dai CL, Tiwari AK, Wu CP, Su XD, Wang SR, Liu DG, et al. Lapatinib (Tykerb, GW572016) reverses multidrug resistance in cancer cells by inhibiting the activity of ATP-binding cassette subfamily B member 1 and G member 2. Cancer Res. 2008;68:7905.CrossRefPubMedPubMedCentral Dai CL, Tiwari AK, Wu CP, Su XD, Wang SR, Liu DG, et al. Lapatinib (Tykerb, GW572016) reverses multidrug resistance in cancer cells by inhibiting the activity of ATP-binding cassette subfamily B member 1 and G member 2. Cancer Res. 2008;68:7905.CrossRefPubMedPubMedCentral
13.
go back to reference Yang K, Chen Y, To KK, Wang F, Li D, Chen L, et al. Alectinib (CH5424802) antagonizes ABCB1- and ABCG2-mediated multidrug resistance in vitro, in vivo and ex vivo. Exp Mol Med. 2017;49:e303.CrossRefPubMedPubMedCentral Yang K, Chen Y, To KK, Wang F, Li D, Chen L, et al. Alectinib (CH5424802) antagonizes ABCB1- and ABCG2-mediated multidrug resistance in vitro, in vivo and ex vivo. Exp Mol Med. 2017;49:e303.CrossRefPubMedPubMedCentral
14.
go back to reference Tong X, Wang F, Liang S, Zhang X, He J, Chen X, et al. Apatinib (YN968D1) enhances the efficacy of conventional chemotherapeutical drugs in side population cells and ABCB1-overexpressing leukemia cells. Biochem Pharmacol. 2012;83:586–97.CrossRefPubMed Tong X, Wang F, Liang S, Zhang X, He J, Chen X, et al. Apatinib (YN968D1) enhances the efficacy of conventional chemotherapeutical drugs in side population cells and ABCB1-overexpressing leukemia cells. Biochem Pharmacol. 2012;83:586–97.CrossRefPubMed
15.
go back to reference Wu Y, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:1454–66.CrossRefPubMed Wu Y, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:1454–66.CrossRefPubMed
16.
go back to reference Momeny M, Zarrinrad G, Moghaddaskho F, Poursheikhani A, Sankanian G, Zaghal A, et al. Dacomitinib, a pan-inhibitor of ErbB receptors, suppresses growth and invasive capacity of chemoresistant ovarian carcinoma cells. Sci Rep. 2017;7:4204.CrossRefPubMedPubMedCentral Momeny M, Zarrinrad G, Moghaddaskho F, Poursheikhani A, Sankanian G, Zaghal A, et al. Dacomitinib, a pan-inhibitor of ErbB receptors, suppresses growth and invasive capacity of chemoresistant ovarian carcinoma cells. Sci Rep. 2017;7:4204.CrossRefPubMedPubMedCentral
17.
go back to reference Abdul Razak AR, Soulières D, Laurie SA, Hotte SJ, Singh S, Winquist E, et al. A phase II trial of dacomitinib, an oral pan-human EGF receptor (HER) inhibitor, as first-line treatment in recurrent and/or metastatic squamous-cell carcinoma of the head and neck. Annals of Oncology Official Journal of the European Society for Medical Oncology. 2013;24:761. Abdul Razak AR, Soulières D, Laurie SA, Hotte SJ, Singh S, Winquist E, et al. A phase II trial of dacomitinib, an oral pan-human EGF receptor (HER) inhibitor, as first-line treatment in recurrent and/or metastatic squamous-cell carcinoma of the head and neck. Annals of Oncology Official Journal of the European Society for Medical Oncology. 2013;24:761.
18.
go back to reference Yu HA, Ahn M, Cho BC, Gerber DE, Natale RB, Socinski MA, et al. Phase 2 study of intermittent pulse dacomitinib in patients with advanced non-small cell lung cancers. Lung Cancer. 2017;112:195–9.CrossRefPubMed Yu HA, Ahn M, Cho BC, Gerber DE, Natale RB, Socinski MA, et al. Phase 2 study of intermittent pulse dacomitinib in patients with advanced non-small cell lung cancers. Lung Cancer. 2017;112:195–9.CrossRefPubMed
19.
go back to reference Calvo E, Soria JC, Ma WW, Wang T, Bahleda R, Tolcher AW, et al. A Phase I Clinical Trial and Independent Patient-Derived Xenograft Study of Combined Targeted Treatment with Dacomitinib and Figitumumab in Advanced Solid Tumors. Clin Cancer Res. 2017;23:1177.CrossRefPubMed Calvo E, Soria JC, Ma WW, Wang T, Bahleda R, Tolcher AW, et al. A Phase I Clinical Trial and Independent Patient-Derived Xenograft Study of Combined Targeted Treatment with Dacomitinib and Figitumumab in Advanced Solid Tumors. Clin Cancer Res. 2017;23:1177.CrossRefPubMed
20.
go back to reference Henrich C, Bokesch H, Dean MS, Robey R, Goncharova E, Wilson J, et al. A high-throughput cell-based assay for inhibitors of ABCG2 activity. J Biomol Screen. 2006;11:176–83.CrossRefPubMed Henrich C, Bokesch H, Dean MS, Robey R, Goncharova E, Wilson J, et al. A high-throughput cell-based assay for inhibitors of ABCG2 activity. J Biomol Screen. 2006;11:176–83.CrossRefPubMed
21.
go back to reference Zhang JY, Wu HY, Xia XK, Liang YJ, Yan YY, She ZG, et al. Anthracenedione derivative 1403P-3 induces apoptosis in KB and KBv200 cells via reactive oxygen species-independent mitochondrial pathway and death receptor pathway. Cancer Biol Ther. 2007;6:1413.PubMed Zhang JY, Wu HY, Xia XK, Liang YJ, Yan YY, She ZG, et al. Anthracenedione derivative 1403P-3 induces apoptosis in KB and KBv200 cells via reactive oxygen species-independent mitochondrial pathway and death receptor pathway. Cancer Biol Ther. 2007;6:1413.PubMed
22.
go back to reference Robey RW, Honjo Y, Morisaki K, Nadjem TA, Runge S, Risbood M, et al. Mutations at amino-acid 482 in the ABCG2 gene affect substrate and antagonist specificity. Br J Cancer. 2003;89:1971.CrossRefPubMedPubMedCentral Robey RW, Honjo Y, Morisaki K, Nadjem TA, Runge S, Risbood M, et al. Mutations at amino-acid 482 in the ABCG2 gene affect substrate and antagonist specificity. Br J Cancer. 2003;89:1971.CrossRefPubMedPubMedCentral
23.
go back to reference Robey RW, Shukla S, Finley EM, Oldham RK, Barnett D, Ambudkar SV, et al. Inhibition of P-glycoprotein (ABCB1)- and multidrug resistance-associated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1((R)). Biochem Pharmacol. 2008;75:1302.CrossRefPubMed Robey RW, Shukla S, Finley EM, Oldham RK, Barnett D, Ambudkar SV, et al. Inhibition of P-glycoprotein (ABCB1)- and multidrug resistance-associated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1((R)). Biochem Pharmacol. 2008;75:1302.CrossRefPubMed
24.
go back to reference Robey RW, Honjo Y, Morisaki K, Nadjem TA, Runge S, Risbood M, et al. Mutations at amino-acid 482 in the ABCG2 gene affect substrate and antagonist specificity. Br J Cancer. 2003;89:1971–8.CrossRefPubMedPubMedCentral Robey RW, Honjo Y, Morisaki K, Nadjem TA, Runge S, Risbood M, et al. Mutations at amino-acid 482 in the ABCG2 gene affect substrate and antagonist specificity. Br J Cancer. 2003;89:1971–8.CrossRefPubMedPubMedCentral
25.
go back to reference Gerlier D, Thomasset N. Use of MTT colorimetric assay to measure cell activation. J Immunol Methods. 1986;94:57–63.CrossRefPubMed Gerlier D, Thomasset N. Use of MTT colorimetric assay to measure cell activation. J Immunol Methods. 1986;94:57–63.CrossRefPubMed
26.
go back to reference Rabindran SK, Ross DD, Doyle LA, Yang W, Greenberger LM. Fumitremorgin C reverses multidrug resistance in cells transfected with the breast cancer resistance protein. Cancer Res. 2000;60:47–50.PubMed Rabindran SK, Ross DD, Doyle LA, Yang W, Greenberger LM. Fumitremorgin C reverses multidrug resistance in cells transfected with the breast cancer resistance protein. Cancer Res. 2000;60:47–50.PubMed
27.
go back to reference Zhang W, Chen Z, Chen L, Wang F, Li F, Wang X, et al. ABCG2-overexpressing H460/MX20 cell xenografts in athymic nude mice maintained original biochemical and cytological characteristics. Sci Rep. 2017;7:40064.CrossRefPubMedPubMedCentral Zhang W, Chen Z, Chen L, Wang F, Li F, Wang X, et al. ABCG2-overexpressing H460/MX20 cell xenografts in athymic nude mice maintained original biochemical and cytological characteristics. Sci Rep. 2017;7:40064.CrossRefPubMedPubMedCentral
28.
go back to reference Dai CL, Liang YJ, Wang YS, Tiwari AK, Yan YY, Wang F, et al. Sensitization of ABCG2-overexpressing cells to conventional chemotherapeutic agent by sunitinib was associated with inhibiting the function of ABCG2. Cancer Lett. 2009;279:74.CrossRefPubMed Dai CL, Liang YJ, Wang YS, Tiwari AK, Yan YY, Wang F, et al. Sensitization of ABCG2-overexpressing cells to conventional chemotherapeutic agent by sunitinib was associated with inhibiting the function of ABCG2. Cancer Lett. 2009;279:74.CrossRefPubMed
30.
go back to reference Hrycyna CA, Ramachandra M, Ambudkar SV, Ko YH, Pedersen PL, Pastan I, et al. Mechanism of action of human P-glycoprotein ATPase activity. Photochemical cleavage during a catalytic transition state using orthovanadate reveals cross-talk between the two ATP sites. J Biol Chem. 1998;273:16631–4.CrossRefPubMed Hrycyna CA, Ramachandra M, Ambudkar SV, Ko YH, Pedersen PL, Pastan I, et al. Mechanism of action of human P-glycoprotein ATPase activity. Photochemical cleavage during a catalytic transition state using orthovanadate reveals cross-talk between the two ATP sites. J Biol Chem. 1998;273:16631–4.CrossRefPubMed
31.
go back to reference Zhou W, Zhang X, Cheng C, Wang F, Wang X, Liang Y, et al. Crizotinib (PF-02341066) reverses multidrug resistance in cancer cells by inhibiting the function of P-glycoprotein. Br J Pharmacol. 2012;166:1669–83.CrossRefPubMedPubMedCentral Zhou W, Zhang X, Cheng C, Wang F, Wang X, Liang Y, et al. Crizotinib (PF-02341066) reverses multidrug resistance in cancer cells by inhibiting the function of P-glycoprotein. Br J Pharmacol. 2012;166:1669–83.CrossRefPubMedPubMedCentral
32.
go back to reference Shukla S, Robey RW, Bates SE, Ambudkar SV. The calcium channel blockers, 1,4-dihydropyridines, are substrates of the multidrug resistance-linked ABC drug transporter, ABCG2. Biochemistry. 2006;45:8940–51.CrossRefPubMed Shukla S, Robey RW, Bates SE, Ambudkar SV. The calcium channel blockers, 1,4-dihydropyridines, are substrates of the multidrug resistance-linked ABC drug transporter, ABCG2. Biochemistry. 2006;45:8940–51.CrossRefPubMed
33.
go back to reference Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2012;25:402–8.CrossRef Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2012;25:402–8.CrossRef
34.
go back to reference Sarkadi B, Homolya L, Szakacs G, Varadi A. Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system. Physiol Rev. 2006;86:1179–236.CrossRefPubMed Sarkadi B, Homolya L, Szakacs G, Varadi A. Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system. Physiol Rev. 2006;86:1179–236.CrossRefPubMed
35.
go back to reference Boyer MJ, Janne PA, Mok T, O'Byrne KJ, Pazares LG, Ramalingam SS, et al. ARCHER: Dacomitinib (D; PF-00299804) versus erlotinib (E) for advanced (adv) non-small cell lung cancer (NSCLC)—A randomized double-blind phase III study. J Clin Oncol. 2012;30(suppl; abstr TPS7615). Boyer MJ, Janne PA, Mok T, O'Byrne KJ, Pazares LG, Ramalingam SS, et al. ARCHER: Dacomitinib (D; PF-00299804) versus erlotinib (E) for advanced (adv) non-small cell lung cancer (NSCLC)—A randomized double-blind phase III study. J Clin Oncol. 2012;30(suppl; abstr TPS7615).
36.
go back to reference Boyer M, Jänne PA, Mok T, O'Byrne K, Paz-Ares L, Ramalingam SS, et al. Rationale and study design of ARCHER: a randomized, double-blind, Phase III study of dacomitinib versus erlotinib for advanced non-small-cell lung cancer. Clin Inv. 2013;3:29–35. Boyer M, Jänne PA, Mok T, O'Byrne K, Paz-Ares L, Ramalingam SS, et al. Rationale and study design of ARCHER: a randomized, double-blind, Phase III study of dacomitinib versus erlotinib for advanced non-small-cell lung cancer. Clin Inv. 2013;3:29–35.
37.
go back to reference Ramalingam SS, Blackhall F, Krzakowski M, Barrios CH, Park K, Bover I, et al. Randomized Phase II Study of Dacomitinib (PF-00299804), an Irreversible Pan–Human Epidermal Growth Factor Receptor Inhibitor, Versus Erlotinib in Patients With Advanced Non–Small-Cell Lung Cancer. J Clin Oncol. 2012;30:3337.CrossRefPubMedPubMedCentral Ramalingam SS, Blackhall F, Krzakowski M, Barrios CH, Park K, Bover I, et al. Randomized Phase II Study of Dacomitinib (PF-00299804), an Irreversible Pan–Human Epidermal Growth Factor Receptor Inhibitor, Versus Erlotinib in Patients With Advanced Non–Small-Cell Lung Cancer. J Clin Oncol. 2012;30:3337.CrossRefPubMedPubMedCentral
Metadata
Title
Dacomitinib potentiates the efficacy of conventional chemotherapeutic agents via inhibiting the drug efflux function of ABCG2 in vitro and in vivo
Authors
Xiaoran Guo
Kenneth K. W. To
Zhen Chen
Xiaokun Wang
Jianye Zhang
Min Luo
Fang Wang
Shirong Yan
Liwu Fu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0690-x

Other articles of this Issue 1/2018

Journal of Experimental & Clinical 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