Skip to main content
Top
Published in: Investigational New Drugs 6/2013

Open Access 01-12-2013 | PRECLINICAL STUDIES

Chemo-sensitisation of HeLa cells to Etoposide by a Benzoxazine in the absence of DNA-PK inhibition

Authors: Cheree Fitzgibbon, Saleh Ihmaid, Jasim Al-Rawi, Terri Meehan-Andrews, Christopher Bradley

Published in: Investigational New Drugs | Issue 6/2013

Login to get access

Summary

The benzoaxines have been developed from structurally similar chromones as specific inhibitors of the PI3K family to sensitize cancer cells to the effects of chemotherapeutic agents; most have been shown to do this through specific inhibition of DNA-PK and DNA repair mechanisms. In this study we examined the benzoxazine, 2-((3-methoxybut-3en-2-yl)amino)-8methyl-4H-benzo[1,3]oxazin-4one (LTUSI54). This compound had no DNA-PK or PI3K inhibitory activity but still sensitized HeLa cells to the effects of Etoposide. LTUSI54 works synergistically with Etoposide to inhibit growth of HeLa cells and sub G1 analysis indicates that this is not due to an increase in apoptosis. LTUSI54 neither enhances DSB formation due to Etoposide nor does it delay the repair of such damage. Cell cycle analysis shows a clear G2 block with Etoposide alone while, in combination with LTUSI54 there is an additional S phase arrest. Phospho-kinase analysis indicated that LTUSI54 engages key regulators of cell cycle progression, specifically p38α, p53 and ERK 1/2. From our results we hypothesize that LTUSI54 is promoting the cell cycle arrest through activation of p38α pathways, independent of p53 mechanisms. This results in a decrease in p53 phosphorylation and hence, restricted apoptosis. Changes in cell number appear to be the result of p38α pathways disrupting cell cycle progression, at the S and G2 checkpoints. Further investigation into the finer mechanisms by which LTUSI54 effects cell cycle progression would be of great interest in assessing this compound as a chemosensitising agent.
Literature
1.
go back to reference Willmore E, Frank AJ, Padget K, Tilby MJ, Austin CA (1998) Etoposide targets topoisomerase IIa and IIb in leukemic cells: isoform-specific cleavable complexes visualized and quantified in situ by a novel immunofluorescence technique. Mol Pharmacol 53:78–85 Willmore E, Frank AJ, Padget K, Tilby MJ, Austin CA (1998) Etoposide targets topoisomerase IIa and IIb in leukemic cells: isoform-specific cleavable complexes visualized and quantified in situ by a novel immunofluorescence technique. Mol Pharmacol 53:78–85
2.
go back to reference Wilmore E, Sd C, Sunter NJ, Tilby MJ, Jackson GH, Austin CA, Durkacz BW (2004) A novel DNA-dependent protein kinase inhibitoe, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood 103(12):4659–4665CrossRef Wilmore E, Sd C, Sunter NJ, Tilby MJ, Jackson GH, Austin CA, Durkacz BW (2004) A novel DNA-dependent protein kinase inhibitoe, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood 103(12):4659–4665CrossRef
3.
go back to reference Tanaka THH, Traganos F, Seiter K, Darzynkiewicz Z (2007) Induction of ATM activation, histone H2AX phosphorylation and apoptosis by etoposide. Relation to cell cycle phase. Cell Cycle 6(3):6CrossRef Tanaka THH, Traganos F, Seiter K, Darzynkiewicz Z (2007) Induction of ATM activation, histone H2AX phosphorylation and apoptosis by etoposide. Relation to cell cycle phase. Cell Cycle 6(3):6CrossRef
4.
go back to reference Karlsson-Rosenthal C, Millar JBA (2006) Cdc25: mechanisms of checkpoint inhibition and recovery. Trends Cell Biol 16(6):285–292CrossRefPubMed Karlsson-Rosenthal C, Millar JBA (2006) Cdc25: mechanisms of checkpoint inhibition and recovery. Trends Cell Biol 16(6):285–292CrossRefPubMed
5.
go back to reference Gabrielli B, Brooks K, Pavey S (2012) Defective cell cycle checkpoints as targets for anti-cancer therapies. Front Pharmacol 3 Gabrielli B, Brooks K, Pavey S (2012) Defective cell cycle checkpoints as targets for anti-cancer therapies. Front Pharmacol 3
6.
go back to reference Alexander BM, Pinnell N, Wen PY, D’Anrea A (2012) Targeting DNA repair and the cell cycle in gliolastoma. J Neuron Oncolocy 107:463–477CrossRef Alexander BM, Pinnell N, Wen PY, D’Anrea A (2012) Targeting DNA repair and the cell cycle in gliolastoma. J Neuron Oncolocy 107:463–477CrossRef
7.
go back to reference Kawabe T (2004) G2 checkpoint abrogators as anticancer drugs. Mol Cancer Ther 3(4):513–519PubMed Kawabe T (2004) G2 checkpoint abrogators as anticancer drugs. Mol Cancer Ther 3(4):513–519PubMed
8.
go back to reference Kung G, Konstantinidis K, Kitsis RN (2011) Programed necrosis, not apoptosis, in the heart. Circ Res 108:1017–1036CrossRefPubMed Kung G, Konstantinidis K, Kitsis RN (2011) Programed necrosis, not apoptosis, in the heart. Circ Res 108:1017–1036CrossRefPubMed
9.
go back to reference Munck JM, Batey MA, Zhao Y, Jenkins H, Richardson CJ, Cano C, Tavecchio M, Barbeau J, Bardos J, Cornell L, Griffin RJ, Menear K, Slade A, Thommes P, Martin NM, Newell DR, Smith GC, Curtin NJ (2012) Chemosensitization of cancer cells by KU-0060648, a dual inhibitor of DNA-PK and PI-3K. Mol Cancer Ther 11(8):1789–1798PubMedCentralCrossRefPubMed Munck JM, Batey MA, Zhao Y, Jenkins H, Richardson CJ, Cano C, Tavecchio M, Barbeau J, Bardos J, Cornell L, Griffin RJ, Menear K, Slade A, Thommes P, Martin NM, Newell DR, Smith GC, Curtin NJ (2012) Chemosensitization of cancer cells by KU-0060648, a dual inhibitor of DNA-PK and PI-3K. Mol Cancer Ther 11(8):1789–1798PubMedCentralCrossRefPubMed
10.
go back to reference Griffin RJ, Fontana G, Golding BT, Guiard S, Hardcastle IR, Leahy JJJ, Martin N, Richardson C, Rigoreau L, Stockley M, Smith GCM (2005) Selective benzopyranone and pyrimido[2,1-a]isoquinolin-4-one inhibitors of DNA-dependent protein kinase: synthesis, structure-activity studies, and radiosensitization of a human tumor cell line in vitro. J Med Chem 48:569–585CrossRefPubMed Griffin RJ, Fontana G, Golding BT, Guiard S, Hardcastle IR, Leahy JJJ, Martin N, Richardson C, Rigoreau L, Stockley M, Smith GCM (2005) Selective benzopyranone and pyrimido[2,1-a]isoquinolin-4-one inhibitors of DNA-dependent protein kinase: synthesis, structure-activity studies, and radiosensitization of a human tumor cell line in vitro. J Med Chem 48:569–585CrossRefPubMed
11.
go back to reference Ding J, Miao Z-H, Meng L-H, Geng M-Y (2006) Emerging cancer therapeutic opportunities target DNA-repair systems. Trends Pharmacol Sci 27(6):338–344CrossRefPubMed Ding J, Miao Z-H, Meng L-H, Geng M-Y (2006) Emerging cancer therapeutic opportunities target DNA-repair systems. Trends Pharmacol Sci 27(6):338–344CrossRefPubMed
12.
go back to reference Martin SA, Lord CJ, Ashworth A (2008) DNA repair deficiency as a therapeutic target in cancer. Curr Opin Genet Dev 18(1):80–86CrossRefPubMed Martin SA, Lord CJ, Ashworth A (2008) DNA repair deficiency as a therapeutic target in cancer. Curr Opin Genet Dev 18(1):80–86CrossRefPubMed
13.
go back to reference Sánchez-Pérez I (2006) DNA repair inhibitors in cancer treatment. Clin Transl Oncol 8(9):642–646CrossRefPubMed Sánchez-Pérez I (2006) DNA repair inhibitors in cancer treatment. Clin Transl Oncol 8(9):642–646CrossRefPubMed
14.
go back to reference Veuger SJ, Curtin NJ, Richardson CJ, Smith GC, Durkacz BW (2003) Radiosensitization and DNA repair inhibition by the combined use of novel inhibitors of DNA-dependent protein kinase and poly (ADP-ribose) polymerase-1. Can Res 63(18):6008–6015 Veuger SJ, Curtin NJ, Richardson CJ, Smith GC, Durkacz BW (2003) Radiosensitization and DNA repair inhibition by the combined use of novel inhibitors of DNA-dependent protein kinase and poly (ADP-ribose) polymerase-1. Can Res 63(18):6008–6015
15.
go back to reference Hosoya N, Miyagawa K (2009) Clinical importance of DNA repair inhibitors in cancer therapy. Memo 2(1):9–14CrossRef Hosoya N, Miyagawa K (2009) Clinical importance of DNA repair inhibitors in cancer therapy. Memo 2(1):9–14CrossRef
16.
go back to reference Workman P, Clarke PA, Raynaud FI, van Montfort RL (2010) Drugging the PI3 kinome: from chemical tools to drugs in the clinic. Can Res 70(6):2146–2157CrossRef Workman P, Clarke PA, Raynaud FI, van Montfort RL (2010) Drugging the PI3 kinome: from chemical tools to drugs in the clinic. Can Res 70(6):2146–2157CrossRef
17.
go back to reference Sarbassov DD, Guertin DA, Ali SM, Sabatini DM (2005) Phosphorylation and regulation of Akt/PKB by the Rictor-mTOR complex. Science 307(5712):1098–1101CrossRefPubMed Sarbassov DD, Guertin DA, Ali SM, Sabatini DM (2005) Phosphorylation and regulation of Akt/PKB by the Rictor-mTOR complex. Science 307(5712):1098–1101CrossRefPubMed
18.
go back to reference Chiosis G, Rosen N, Sepp-Lorenzino L (2001) LY294002-geldanamycin heterodimers as selective inhibitors of the PI3K and PI3K-related family. Bioorg Med Chem Lett 11(7):909–913CrossRefPubMed Chiosis G, Rosen N, Sepp-Lorenzino L (2001) LY294002-geldanamycin heterodimers as selective inhibitors of the PI3K and PI3K-related family. Bioorg Med Chem Lett 11(7):909–913CrossRefPubMed
19.
go back to reference Lee CM, Fuhrman CB, Planelles V, Peltier MR, Gaffney DK, Soisson AP, Dodson MK, Tolley HD, Green CL, Zempolich KA (2006) Phosphatidylinositol 3-kinase inhibition by LY294002 radiosensitizes human cancer cell lines. Clin Cancer Res 12(1):250–256CrossRefPubMed Lee CM, Fuhrman CB, Planelles V, Peltier MR, Gaffney DK, Soisson AP, Dodson MK, Tolley HD, Green CL, Zempolich KA (2006) Phosphatidylinositol 3-kinase inhibition by LY294002 radiosensitizes human cancer cell lines. Clin Cancer Res 12(1):250–256CrossRefPubMed
20.
go back to reference Freshney RI (1992) Animal cell culture: a practical approach. IRL Press at Oxford University Press Freshney RI (1992) Animal cell culture: a practical approach. IRL Press at Oxford University Press
21.
go back to reference MacPhail SH, Banath JP, Yu TY, Chu EHM, Lambur H, Olive PL (2003) Expression of phosphorylated histone H2AX in culture cell lines following exposure to X-rays. Int J Radiat Biol 79(5):351–358CrossRefPubMed MacPhail SH, Banath JP, Yu TY, Chu EHM, Lambur H, Olive PL (2003) Expression of phosphorylated histone H2AX in culture cell lines following exposure to X-rays. Int J Radiat Biol 79(5):351–358CrossRefPubMed
22.
go back to reference Riccardi C, Nicoletti I (2006) Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat Protoc 1(3):1458–1461CrossRefPubMed Riccardi C, Nicoletti I (2006) Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat Protoc 1(3):1458–1461CrossRefPubMed
23.
go back to reference Ihmaid S, Al-Rawi J, Bradley C, Angove MJ, Robertson MN, Clark RL (2011) Synthesis, structural elucidation, DNA-PK inhibition, homology modelling and anti-platelet activity of morpholino-substituted-1,3-naphth-oxazines. Bioorg Med Chem 19(13):3983–3994CrossRefPubMed Ihmaid S, Al-Rawi J, Bradley C, Angove MJ, Robertson MN, Clark RL (2011) Synthesis, structural elucidation, DNA-PK inhibition, homology modelling and anti-platelet activity of morpholino-substituted-1,3-naphth-oxazines. Bioorg Med Chem 19(13):3983–3994CrossRefPubMed
24.
go back to reference Ihmaid SK, Al-Rawi JMA, Bradley CJ, Angove MJ, Robertson MN (2012) Synthesis, DNA-PK inhibition, anti-platelet activity studies of 2-(N-substituted-3-aminopyridine)-substituted-1,3-benzoxazines and DNA-PK and PI3K inhibition, homology modelling studies of 2-morpholino-(7,8-di and 8-substituted)-1,3-benzoxazines. Eur J Med Chem 57:85–101CrossRefPubMed Ihmaid SK, Al-Rawi JMA, Bradley CJ, Angove MJ, Robertson MN (2012) Synthesis, DNA-PK inhibition, anti-platelet activity studies of 2-(N-substituted-3-aminopyridine)-substituted-1,3-benzoxazines and DNA-PK and PI3K inhibition, homology modelling studies of 2-morpholino-(7,8-di and 8-substituted)-1,3-benzoxazines. Eur J Med Chem 57:85–101CrossRefPubMed
25.
go back to reference Clapham KM, Bardos J, Finlay MRV, Golding BT, Griffen EJ, Griffin RJ, Hardcastle IR, Menear KA, Ting A, Turner P (2011) DNA-dependent protein kinase (DNA-PK) inhibitors: structure–activity relationships for O-alkoxyphenylchromen-4-one probes of the ATP-binding domain. Bioorg Med Chem Lett 21(3):966–970CrossRefPubMed Clapham KM, Bardos J, Finlay MRV, Golding BT, Griffen EJ, Griffin RJ, Hardcastle IR, Menear KA, Ting A, Turner P (2011) DNA-dependent protein kinase (DNA-PK) inhibitors: structure–activity relationships for O-alkoxyphenylchromen-4-one probes of the ATP-binding domain. Bioorg Med Chem Lett 21(3):966–970CrossRefPubMed
26.
go back to reference Lock RB, Stribinskiene L (1996) Dual modes of death induced by etoposide in human epithelial tumor cells allow Bcl-2 to inhibit apoptosis without affecting clonogenic survival. Cancer Res 56:4006–4012PubMed Lock RB, Stribinskiene L (1996) Dual modes of death induced by etoposide in human epithelial tumor cells allow Bcl-2 to inhibit apoptosis without affecting clonogenic survival. Cancer Res 56:4006–4012PubMed
27.
go back to reference Kao J, Lavaf A, Lan C-H, Fu S (2010) Inhibition of γ-H2AX after ionizing radiation as a biological surrogate of impaired upstream DNA damage signaling and radiosensitivity. J Cancer Mol 5(2):49–54 Kao J, Lavaf A, Lan C-H, Fu S (2010) Inhibition of γ-H2AX after ionizing radiation as a biological surrogate of impaired upstream DNA damage signaling and radiosensitivity. J Cancer Mol 5(2):49–54
28.
go back to reference Banáth JP, Olive PL (2003) Expression of phosphorylated histone H2AX as a surrogate of call killing by drugs that create DNA double-strand breaks. Cancer Res 63:4347–4350PubMed Banáth JP, Olive PL (2003) Expression of phosphorylated histone H2AX as a surrogate of call killing by drugs that create DNA double-strand breaks. Cancer Res 63:4347–4350PubMed
29.
go back to reference Celeste A, Fernandez-Capetillo O, Kruhlak MJ, Pilch DR, Staudt DW, Lee A, Bonner RF, Bonner WM, Nussenzweig A (2003) Histone H2AX phosphyrlation is dispensable for the inital recognition of DNA breaks. Nat Cell Biol 5(7):675–679CrossRefPubMed Celeste A, Fernandez-Capetillo O, Kruhlak MJ, Pilch DR, Staudt DW, Lee A, Bonner RF, Bonner WM, Nussenzweig A (2003) Histone H2AX phosphyrlation is dispensable for the inital recognition of DNA breaks. Nat Cell Biol 5(7):675–679CrossRefPubMed
30.
go back to reference Collins I, Garrett MD (2005) Targeting the cell division cycle in cancer: CDK and cell cycle checkpoint kinase inhibitors. Curr Opin Pharmacol 5(4):366–373CrossRefPubMed Collins I, Garrett MD (2005) Targeting the cell division cycle in cancer: CDK and cell cycle checkpoint kinase inhibitors. Curr Opin Pharmacol 5(4):366–373CrossRefPubMed
31.
go back to reference de Klein A, Muijtjens M, van Os R, Verhoeven Y, Smit B, Carr AM, Lehmann AR, Hoeijmakers JHJ (2000) Targeted disruption of the cell-cycle checkpoint gene ATR leads to early embryonic lethality in mice. Curr Biol 10(8):479–482CrossRefPubMed de Klein A, Muijtjens M, van Os R, Verhoeven Y, Smit B, Carr AM, Lehmann AR, Hoeijmakers JHJ (2000) Targeted disruption of the cell-cycle checkpoint gene ATR leads to early embryonic lethality in mice. Curr Biol 10(8):479–482CrossRefPubMed
32.
go back to reference Yves P (2004) Camptothecins and topoisomerase I: a foot in the door. Targeting the genome beyond topoisomerase I with camptothecins and novel anticancer drugs: importance of DNA replication, repair and cell cycle checkpoints. Curr Med Chem Anti Cancer Agents 4(5):429–434CrossRef Yves P (2004) Camptothecins and topoisomerase I: a foot in the door. Targeting the genome beyond topoisomerase I with camptothecins and novel anticancer drugs: importance of DNA replication, repair and cell cycle checkpoints. Curr Med Chem Anti Cancer Agents 4(5):429–434CrossRef
33.
go back to reference Schwartz GK, Shah MA (2005) Targeting the cell cycle: a new approach to cancer therapy. J Clin Oncol 23(36):9408–9421CrossRefPubMed Schwartz GK, Shah MA (2005) Targeting the cell cycle: a new approach to cancer therapy. J Clin Oncol 23(36):9408–9421CrossRefPubMed
35.
go back to reference Johnstone RW, Ruefli AA, Lowe SW (2002) Apoptosis: a link between cancer genetics and chemotherapy. Cell 108(2):153–164CrossRefPubMed Johnstone RW, Ruefli AA, Lowe SW (2002) Apoptosis: a link between cancer genetics and chemotherapy. Cell 108(2):153–164CrossRefPubMed
36.
go back to reference Shieh S-Y, Ikeda M, Taya Y, Prives C (1997) DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell 91(3):325–334CrossRefPubMed Shieh S-Y, Ikeda M, Taya Y, Prives C (1997) DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell 91(3):325–334CrossRefPubMed
37.
go back to reference Banin S, Moyal L, Shieh S-Y, Taya Y, Anderson CW, Chessa L, Smorodinsky NI, Prives C, Reiss Y, Shiloh Y, Ziv Y (1998) Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 281(5383):1674–1677CrossRefPubMed Banin S, Moyal L, Shieh S-Y, Taya Y, Anderson CW, Chessa L, Smorodinsky NI, Prives C, Reiss Y, Shiloh Y, Ziv Y (1998) Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 281(5383):1674–1677CrossRefPubMed
38.
39.
40.
go back to reference Reinhardt HC, Aslanian AS, Lees JA, Yaffe MB (2007) p53-deficient cells rely on ATM- and ATR-mediated checkpoint signaling through the p38MAPK/MK2 pathway for survival after DNA damage. Cancer Cell 11(2):175–189PubMedCentralCrossRefPubMed Reinhardt HC, Aslanian AS, Lees JA, Yaffe MB (2007) p53-deficient cells rely on ATM- and ATR-mediated checkpoint signaling through the p38MAPK/MK2 pathway for survival after DNA damage. Cancer Cell 11(2):175–189PubMedCentralCrossRefPubMed
41.
go back to reference Skorski T (2007) DNA damage-dependent apoptosis. In: Srivastava R (ed) Apoptosis, cell signaling, and human diseases. Humana Press, pp 263–272 Skorski T (2007) DNA damage-dependent apoptosis. In: Srivastava R (ed) Apoptosis, cell signaling, and human diseases. Humana Press, pp 263–272
43.
go back to reference Manke IA, Nguyen A, Lim D, Stewart MQ, Elia AEH, Yaffe MB (2005) MAPKAP kinase-2 is a cell cycle checkpoint kinase that regulates the G2/M transition and S phase progression in response to UV irradiation. Mol Cell 17(1):37–48CrossRefPubMed Manke IA, Nguyen A, Lim D, Stewart MQ, Elia AEH, Yaffe MB (2005) MAPKAP kinase-2 is a cell cycle checkpoint kinase that regulates the G2/M transition and S phase progression in response to UV irradiation. Mol Cell 17(1):37–48CrossRefPubMed
44.
46.
go back to reference Dolado I, Nebreda AR (2008) Regulation of tumorigenesis by p38α MAP kinase. Stress Activated Protein Kinases Top Curr Genet 20:99–128CrossRef Dolado I, Nebreda AR (2008) Regulation of tumorigenesis by p38α MAP kinase. Stress Activated Protein Kinases Top Curr Genet 20:99–128CrossRef
47.
go back to reference Mikhailov A, Patel D, McCance DJ, Rieder CL (2007) The G2 p38-mediated stress-activated checkpoint pathway becomes attenuated in transformed cells. Curr Biol 17(24):2162–2168PubMedCentralCrossRefPubMed Mikhailov A, Patel D, McCance DJ, Rieder CL (2007) The G2 p38-mediated stress-activated checkpoint pathway becomes attenuated in transformed cells. Curr Biol 17(24):2162–2168PubMedCentralCrossRefPubMed
48.
go back to reference Weber HO, Ludwig RL, Morrison D, Kotlyarov A, Gaestel M, Vousden KH (2005) HDM2 phosphorylation by MAPKAP kinase 2. Oncogene 24(12):1965–1972CrossRefPubMed Weber HO, Ludwig RL, Morrison D, Kotlyarov A, Gaestel M, Vousden KH (2005) HDM2 phosphorylation by MAPKAP kinase 2. Oncogene 24(12):1965–1972CrossRefPubMed
49.
go back to reference Mebratu Y, Tesfaigzi Y (2009) How ERK1/2 activation controls cell proliferation and cell death is subcellular localization the answer? Cell Cycle 8(8):1168–1175PubMedCentralCrossRefPubMed Mebratu Y, Tesfaigzi Y (2009) How ERK1/2 activation controls cell proliferation and cell death is subcellular localization the answer? Cell Cycle 8(8):1168–1175PubMedCentralCrossRefPubMed
50.
go back to reference Meloche S, Pouyssegur J (2007) The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1-to S-phase transition. Oncogene 26(22):3227–3239CrossRefPubMed Meloche S, Pouyssegur J (2007) The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1-to S-phase transition. Oncogene 26(22):3227–3239CrossRefPubMed
51.
go back to reference Balmanno K, Cook S (2009) Tumour cell survival signalling by the ERK1/2 pathway. Cell Death Differ 16(3):368–377CrossRefPubMed Balmanno K, Cook S (2009) Tumour cell survival signalling by the ERK1/2 pathway. Cell Death Differ 16(3):368–377CrossRefPubMed
Metadata
Title
Chemo-sensitisation of HeLa cells to Etoposide by a Benzoxazine in the absence of DNA-PK inhibition
Authors
Cheree Fitzgibbon
Saleh Ihmaid
Jasim Al-Rawi
Terri Meehan-Andrews
Christopher Bradley
Publication date
01-12-2013
Publisher
Springer US
Published in
Investigational New Drugs / Issue 6/2013
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-013-0031-z

Other articles of this Issue 6/2013

Investigational New Drugs 6/2013 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