Skip to main content
Top
Published in: Molecular Cancer 1/2015

Open Access 01-12-2015 | Research

Extracellular nucleotides as novel, underappreciated pro-metastatic factors that stimulate purinergic signaling in human lung cancer cells

Authors: Gabriela Schneider, Talita Glaser, Claudiana Lameu, Ahmed Abdelbaset-Ismail, Zachariah Payne Sellers, Marcin Moniuszko, Henning Ulrich, Mariusz Z. Ratajczak

Published in: Molecular Cancer | Issue 1/2015

Login to get access

Abstract

Background

One of the challenging problems of current radio-chemotherapy is recurrence and metastasis of cancer cells that survive initial treatment. We propose that one of the unwanted effects of radiochemotherapy is the release from damaged (“leaky”) cells of nucleotides such as ATP and UTP that exert pro-metastatic functions and can directly stimulate chemotaxis of cancer cells.

Methods

To address this problem in a model of human lung cancer (LC), we employed several complementary in vitro and in vivo approaches to demonstrate the role of extracellular nucleotides (EXNs) in LC cell line metastasis and tumor progression. We measured concentrations of EXNs in several organs before and after radiochemotherapy. The purinergic receptor agonists and antagonists, inhibiting all or selected subtypes of receptors, were employed in in vitro and in vivo pro-metastatic assays.

Results

We found that EXNs accumulate in several organs in response to radiochemotherapy, and RT-PCR analysis revealed that most of the P1 and P2 receptor subtypes are expressed in human LC cells. EXNs were found to induce chemotaxis and adhesion of LC cells, and an autocrine loop was identified that promotes the proliferation of LC cells. Most importantly, metastasis of these cells could be inhibited in immunodeficient mice in the presence of specific small molecule inhibitors of purinergic receptors.

Conclusions

Based on this result, EXNs are novel pro-metastatic factors released particularly during radiochemotherapy, and inhibition of their pro-metastatic effects via purinergic signaling could become an important part of anti-metastatic treatment.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ratajczak MZ, Jadczyk T, Schneider G, Kakar SS, Kucia M. Induction of a tumor-metastasis-receptive microenvironment as an unwanted and underestimated side effect of treatment by chemotherapy or radiotherapy. J Ovarian Res. 2013;6:95.PubMedCentralCrossRefPubMed Ratajczak MZ, Jadczyk T, Schneider G, Kakar SS, Kucia M. Induction of a tumor-metastasis-receptive microenvironment as an unwanted and underestimated side effect of treatment by chemotherapy or radiotherapy. J Ovarian Res. 2013;6:95.PubMedCentralCrossRefPubMed
2.
go back to reference Hodgson DC. Long-term toxicity of chemotherapy and radiotherapy in lymphoma survivors: optimizing treatment for individual patients. Clin Adv Hematol Oncol. 2015;13:103–12.PubMed Hodgson DC. Long-term toxicity of chemotherapy and radiotherapy in lymphoma survivors: optimizing treatment for individual patients. Clin Adv Hematol Oncol. 2015;13:103–12.PubMed
3.
go back to reference Schneider G, Bryndza E, Abdel-Latif A, Ratajczak J, Maj M, Tarnowski M, et al. Bioactive lipids S1P and C1P are prometastatic factors in human rhabdomyosarcoma, and their tissue levels increase in response to radio/chemotherapy. Mol Cancer Res. 2013;11:793–807.PubMedCentralCrossRefPubMed Schneider G, Bryndza E, Abdel-Latif A, Ratajczak J, Maj M, Tarnowski M, et al. Bioactive lipids S1P and C1P are prometastatic factors in human rhabdomyosarcoma, and their tissue levels increase in response to radio/chemotherapy. Mol Cancer Res. 2013;11:793–807.PubMedCentralCrossRefPubMed
4.
go back to reference Schneider G, Sellers ZP, Abdel-Latif A, Morris AJ, Ratajczak MZ. Bioactive lipids, LPC and LPA, are novel prometastatic factors and their tissue levels increase in response to radio/chemotherapy. Mol Cancer Res. 2014;12:1560–73.PubMedCentralCrossRefPubMed Schneider G, Sellers ZP, Abdel-Latif A, Morris AJ, Ratajczak MZ. Bioactive lipids, LPC and LPA, are novel prometastatic factors and their tissue levels increase in response to radio/chemotherapy. Mol Cancer Res. 2014;12:1560–73.PubMedCentralCrossRefPubMed
5.
go back to reference Gunjal PM, Schneider G, Ismail AA, Kakar SS, Kucia M, Ratajczak MZ. Evidence for induction of a tumor metastasis-receptive microenvironment for ovarian cancer cells in bone marrow and other organs as an unwanted and underestimated side effect of chemotherapy/radiotherapy. J Ovarian Res. 2015;8:20.PubMedCentralCrossRefPubMed Gunjal PM, Schneider G, Ismail AA, Kakar SS, Kucia M, Ratajczak MZ. Evidence for induction of a tumor metastasis-receptive microenvironment for ovarian cancer cells in bone marrow and other organs as an unwanted and underestimated side effect of chemotherapy/radiotherapy. J Ovarian Res. 2015;8:20.PubMedCentralCrossRefPubMed
6.
go back to reference Glaser T, Cappellari AR, Pillat MM, Iser IC, Wink MR, Battastini AM, et al. Perspectives of purinergic signaling in stem cell differentiation and tissue regeneration. Purinergic Signal. 2012;8:523–37.PubMedCentralCrossRefPubMed Glaser T, Cappellari AR, Pillat MM, Iser IC, Wink MR, Battastini AM, et al. Perspectives of purinergic signaling in stem cell differentiation and tissue regeneration. Purinergic Signal. 2012;8:523–37.PubMedCentralCrossRefPubMed
7.
go back to reference Guzman-Aranguez A, Santano C, Martin-Gil A, Fonseca B, Pintor J. Nucleotides in the eye: focus on functional aspects and therapeutic perspectives. J Pharmacol Exp Ther. 2011;345:331–41.CrossRef Guzman-Aranguez A, Santano C, Martin-Gil A, Fonseca B, Pintor J. Nucleotides in the eye: focus on functional aspects and therapeutic perspectives. J Pharmacol Exp Ther. 2011;345:331–41.CrossRef
8.
go back to reference Burnstock G, Knight GE. Cellular distribution and functions of P2 receptor subtypes in different systems. Int Rev Cytol. 2004;240:31–304.CrossRefPubMed Burnstock G, Knight GE. Cellular distribution and functions of P2 receptor subtypes in different systems. Int Rev Cytol. 2004;240:31–304.CrossRefPubMed
9.
go back to reference Kronlage M, Song J, Sorokin L, Isfort K, Schwerdtle T, Leipziger J, et al. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci Signal. 2010;3:ra55.PubMed Kronlage M, Song J, Sorokin L, Isfort K, Schwerdtle T, Leipziger J, et al. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci Signal. 2010;3:ra55.PubMed
10.
go back to reference Ulrich H. Purinergic receptors in stem cell biology. M.A. Hayat. (Org.). Stem Cells and Cancer Stem Cells. Dordrecht, Heidelberg: Springer; 2012. Ulrich H. Purinergic receptors in stem cell biology. M.A. Hayat. (Org.). Stem Cells and Cancer Stem Cells. Dordrecht, Heidelberg: Springer; 2012.
11.
go back to reference Qian Y, Wang X, Liu Y, Li Y, Colvin RA, Tong L, et al. Extracellular ATP is internalized by macropinocytosis and induces intracellular ATP increase and drug resistance in cancer cells. Cancer Lett. 2014;351:242–51.CrossRefPubMed Qian Y, Wang X, Liu Y, Li Y, Colvin RA, Tong L, et al. Extracellular ATP is internalized by macropinocytosis and induces intracellular ATP increase and drug resistance in cancer cells. Cancer Lett. 2014;351:242–51.CrossRefPubMed
12.
go back to reference Nascimento IC, Glaser T, Nery AA, Pillat MM, Pesquero JB, Ulrich H. Kinin-B1 and B2 receptor activity in proliferation and neural phenotype determination of mouse embryonic stem cells. Cytometry A. 2015;87:989–1000. Nascimento IC, Glaser T, Nery AA, Pillat MM, Pesquero JB, Ulrich H. Kinin-B1 and B2 receptor activity in proliferation and neural phenotype determination of mouse embryonic stem cells. Cytometry A. 2015;87:989–1000.
13.
go back to reference Ratajczak M, Tarnowski M, Staniszewska M, Sroczynski T, Banach B. Mechanisms of cancer metastasis: involvement of cancer stem cells? Minerva Med. 2010;101:179–91.PubMed Ratajczak M, Tarnowski M, Staniszewska M, Sroczynski T, Banach B. Mechanisms of cancer metastasis: involvement of cancer stem cells? Minerva Med. 2010;101:179–91.PubMed
14.
go back to reference Kim D, Kim S, Koh H, Yoon SO, Chung AS, Cho KS, et al. Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. Faseb J. 2001;15:1953–62.CrossRefPubMed Kim D, Kim S, Koh H, Yoon SO, Chung AS, Cho KS, et al. Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. Faseb J. 2001;15:1953–62.CrossRefPubMed
15.
go back to reference Kukreja P, Abdel-Mageed AB, Mondal D, Liu K, Agrawal KC. Up-regulation of CXCR4 expression in PC-3 cells by stromal-derived factor-1alpha (CXCL12) increases endothelial adhesion and transendothelial migration: role of MEK/ERK signaling pathway-dependent NF-kappaB activation. Cancer Res. 2005;65:9891–8.CrossRefPubMed Kukreja P, Abdel-Mageed AB, Mondal D, Liu K, Agrawal KC. Up-regulation of CXCR4 expression in PC-3 cells by stromal-derived factor-1alpha (CXCL12) increases endothelial adhesion and transendothelial migration: role of MEK/ERK signaling pathway-dependent NF-kappaB activation. Cancer Res. 2005;65:9891–8.CrossRefPubMed
16.
go back to reference Zhou JZ, Riquelme MA, Gao X, Ellies LG, Sun LZ, Jiang JX. Differential impact of adenosine nucleotides released by osteocytes on breast cancer growth and bone metastasis. Oncogene. 2015;34:1831–42.PubMedCentralCrossRefPubMed Zhou JZ, Riquelme MA, Gao X, Ellies LG, Sun LZ, Jiang JX. Differential impact of adenosine nucleotides released by osteocytes on breast cancer growth and bone metastasis. Oncogene. 2015;34:1831–42.PubMedCentralCrossRefPubMed
17.
go back to reference Takai E, Tsukimoto M, Harada H, Kojima S. Autocrine signaling via release of ATP and activation of P2X7 receptor influences motile activity of human lung cancer cells. Purinergic Signal. 2014;10:487–97.PubMedCentralCrossRefPubMed Takai E, Tsukimoto M, Harada H, Kojima S. Autocrine signaling via release of ATP and activation of P2X7 receptor influences motile activity of human lung cancer cells. Purinergic Signal. 2014;10:487–97.PubMedCentralCrossRefPubMed
19.
go back to reference Bortolato M, Yardley MM, Khoja S, Godar SC, Asatryan L, Finn DA, et al. Pharmacological insights into the role of P2X4 receptors in behavioural regulation: lessons from ivermectin. Int J Neuropsychopharmacol. 2013;16:1059–70.PubMedCentralCrossRefPubMed Bortolato M, Yardley MM, Khoja S, Godar SC, Asatryan L, Finn DA, et al. Pharmacological insights into the role of P2X4 receptors in behavioural regulation: lessons from ivermectin. Int J Neuropsychopharmacol. 2013;16:1059–70.PubMedCentralCrossRefPubMed
21.
go back to reference Schafer R, Sedehizade F, Welte T, Reiser G. ATP- and UTP-activated P2Y receptors differently regulate proliferation of human lung epithelial tumor cells. Am J Physiol Lung Cell Mol Physiol. 2003;285:L376–85.CrossRefPubMed Schafer R, Sedehizade F, Welte T, Reiser G. ATP- and UTP-activated P2Y receptors differently regulate proliferation of human lung epithelial tumor cells. Am J Physiol Lung Cell Mol Physiol. 2003;285:L376–85.CrossRefPubMed
22.
go back to reference Buzzi N, Bilbao PS, Boland R, de Boland AR. Extracellular ATP activates MAP kinase cascades through a P2Y purinergic receptor in the human intestinal Caco-2 cell line. Biochim Biophys Acta. 1790;2009:1651–9. Buzzi N, Bilbao PS, Boland R, de Boland AR. Extracellular ATP activates MAP kinase cascades through a P2Y purinergic receptor in the human intestinal Caco-2 cell line. Biochim Biophys Acta. 1790;2009:1651–9.
24.
go back to reference Huang SH, Perez-Ordonez B, Weinreb I, Hope A, Massey C, Waldron JN, et al. Natural course of distant metastases following radiotherapy or chemoradiotherapy in HPV-related oropharyngeal cancer. Oral Oncol. 2013;49:79–85.CrossRefPubMed Huang SH, Perez-Ordonez B, Weinreb I, Hope A, Massey C, Waldron JN, et al. Natural course of distant metastases following radiotherapy or chemoradiotherapy in HPV-related oropharyngeal cancer. Oral Oncol. 2013;49:79–85.CrossRefPubMed
25.
go back to reference Dou Y, Wu HJ, Li HQ, Qin S, Wang YE, Li J, et al. Microglial migration mediated by ATP-induced ATP release from lysosomes. Cell Res. 2012;22:1022–33.PubMedCentralCrossRefPubMed Dou Y, Wu HJ, Li HQ, Qin S, Wang YE, Li J, et al. Microglial migration mediated by ATP-induced ATP release from lysosomes. Cell Res. 2012;22:1022–33.PubMedCentralCrossRefPubMed
28.
go back to reference Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8:3723–8.CrossRefPubMed Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8:3723–8.CrossRefPubMed
30.
go back to reference Al-Rayahi IA, Sanyi RH. The overlapping roles of antimicrobial peptides and complement in recruitment and activation of tumor-associated inflammatory cells. Front Immunol. 2015;6:2.PubMedCentralCrossRefPubMed Al-Rayahi IA, Sanyi RH. The overlapping roles of antimicrobial peptides and complement in recruitment and activation of tumor-associated inflammatory cells. Front Immunol. 2015;6:2.PubMedCentralCrossRefPubMed
31.
go back to reference Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT. Cancer and the complement cascade. Mol Cancer Res. 2010;8:1453–65.CrossRefPubMed Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT. Cancer and the complement cascade. Mol Cancer Res. 2010;8:1453–65.CrossRefPubMed
32.
go back to reference Surace L, Lysenko V, Fontana AO, Cecconi V, Janssen H, Bicvic A, et al. Complement is a central mediator of radiotherapy-induced tumor-specific immunity and clinical response. Immunity. 2015;42:767–77.CrossRefPubMed Surace L, Lysenko V, Fontana AO, Cecconi V, Janssen H, Bicvic A, et al. Complement is a central mediator of radiotherapy-induced tumor-specific immunity and clinical response. Immunity. 2015;42:767–77.CrossRefPubMed
33.
34.
go back to reference Burnstock G, Williams M. P2 purinergic receptors: modulation of cell function and therapeutic potential. J Pharmacol Exp Ther. 2000;295:862–9.PubMed Burnstock G, Williams M. P2 purinergic receptors: modulation of cell function and therapeutic potential. J Pharmacol Exp Ther. 2000;295:862–9.PubMed
35.
go back to reference Virtanen SS, Kukkonen-Macchi A, Vainio M, Elima K, Harkonen PL, Jalkanen S, et al. Adenosine inhibits tumor cell invasion via receptor-independent mechanisms. Mol Cancer Res. 2014;12:1863–74.CrossRefPubMed Virtanen SS, Kukkonen-Macchi A, Vainio M, Elima K, Harkonen PL, Jalkanen S, et al. Adenosine inhibits tumor cell invasion via receptor-independent mechanisms. Mol Cancer Res. 2014;12:1863–74.CrossRefPubMed
36.
go back to reference Buzzi N, Boland R, Russo de Boland A. Signal transduction pathways associated with ATP-induced proliferation of colon adenocarcinoma cells. Biochim Biophys Acta. 2010;1800:946–55.CrossRefPubMed Buzzi N, Boland R, Russo de Boland A. Signal transduction pathways associated with ATP-induced proliferation of colon adenocarcinoma cells. Biochim Biophys Acta. 2010;1800:946–55.CrossRefPubMed
37.
go back to reference Hattori F, Ohshima Y, Seki S, Tsukimoto M, Sato M, Takenouchi T, et al. Feasibility study of B16 melanoma therapy using oxidized ATP to target purinergic receptor P2X7. Eur J Pharmacol. 2012;695:20–6.CrossRefPubMed Hattori F, Ohshima Y, Seki S, Tsukimoto M, Sato M, Takenouchi T, et al. Feasibility study of B16 melanoma therapy using oxidized ATP to target purinergic receptor P2X7. Eur J Pharmacol. 2012;695:20–6.CrossRefPubMed
38.
go back to reference Yang D, Song J, Wu L, Ma Y, Song C, Dovat S, et al. Induction of senescence by adenosine suppressing the growth of lung cancer cells. Biochem Biophys Res Commun. 2013;440:62–7.CrossRefPubMed Yang D, Song J, Wu L, Ma Y, Song C, Dovat S, et al. Induction of senescence by adenosine suppressing the growth of lung cancer cells. Biochem Biophys Res Commun. 2013;440:62–7.CrossRefPubMed
39.
go back to reference Hatta Y, Takahashi M, Enomoto Y, Takahashi N, Sawada U, Horie T. Adenosine triphosphate (ATP) enhances the antitumor effect of etoposide (VP16) in lung cancer cells. Oncol Rep. 2004;12:1139–42.PubMed Hatta Y, Takahashi M, Enomoto Y, Takahashi N, Sawada U, Horie T. Adenosine triphosphate (ATP) enhances the antitumor effect of etoposide (VP16) in lung cancer cells. Oncol Rep. 2004;12:1139–42.PubMed
40.
go back to reference Swennen EL, Ummels V, Buss I, Jaehde U, Bast A, Dagnelie PC. ATP sensitizes H460 lung carcinoma cells to cisplatin-induced apoptosis. Chem Biol Interact. 2010;184:338–45.CrossRefPubMed Swennen EL, Ummels V, Buss I, Jaehde U, Bast A, Dagnelie PC. ATP sensitizes H460 lung carcinoma cells to cisplatin-induced apoptosis. Chem Biol Interact. 2010;184:338–45.CrossRefPubMed
41.
go back to reference Jin H, Eun SY, Lee JS, Park SW, Lee JH, Chang KC, et al. P2Y2 receptor activation by nucleotides released from highly metastatic breast cancer cells increases tumor growth and invasion via crosstalk with endothelial cells. Breast Cancer Res. 2014;16:R77.PubMedCentralCrossRefPubMed Jin H, Eun SY, Lee JS, Park SW, Lee JH, Chang KC, et al. P2Y2 receptor activation by nucleotides released from highly metastatic breast cancer cells increases tumor growth and invasion via crosstalk with endothelial cells. Breast Cancer Res. 2014;16:R77.PubMedCentralCrossRefPubMed
42.
go back to reference Patel L, Thaker A. The effects of adenosine A2B receptor inhibition on VEGF and nitric oxide axis-mediated renal function in diabetic nephropathy. Ren Fail. 2014;36:916–24.CrossRefPubMed Patel L, Thaker A. The effects of adenosine A2B receptor inhibition on VEGF and nitric oxide axis-mediated renal function in diabetic nephropathy. Ren Fail. 2014;36:916–24.CrossRefPubMed
43.
go back to reference Ferrari D, Gulinelli S, Salvestrini V, Lucchetti G, Zini R, Manfredini R, et al. Purinergic stimulation of human mesenchymal stem cells potentiates their chemotactic response to CXCL12 and increases the homing capacity and production of proinflammatory cytokines. Exp Hematol. 2011;39:360–74. 74 e1-5.CrossRefPubMed Ferrari D, Gulinelli S, Salvestrini V, Lucchetti G, Zini R, Manfredini R, et al. Purinergic stimulation of human mesenchymal stem cells potentiates their chemotactic response to CXCL12 and increases the homing capacity and production of proinflammatory cytokines. Exp Hematol. 2011;39:360–74. 74 e1-5.CrossRefPubMed
44.
go back to reference Gast RE, Konig S, Rose K, Ferenz KB, Krieglstein J. Binding of ATP to vascular endothelial growth factor isoform VEGF-A165 is essential for inducing proliferation of human umbilical vein endothelial cells. BMC Biochem. 2011;12:28.PubMedCentralCrossRefPubMed Gast RE, Konig S, Rose K, Ferenz KB, Krieglstein J. Binding of ATP to vascular endothelial growth factor isoform VEGF-A165 is essential for inducing proliferation of human umbilical vein endothelial cells. BMC Biochem. 2011;12:28.PubMedCentralCrossRefPubMed
45.
go back to reference Bergamin LS, Braganhol E, Zanin RF, Edelweiss MI, Battastini AM. Ectonucleotidases in tumor cells and tumor-associated immune cells: an overview. J Biomed Biotechnol. 2012;2012:959848.PubMedCentralCrossRefPubMed Bergamin LS, Braganhol E, Zanin RF, Edelweiss MI, Battastini AM. Ectonucleotidases in tumor cells and tumor-associated immune cells: an overview. J Biomed Biotechnol. 2012;2012:959848.PubMedCentralCrossRefPubMed
47.
go back to reference Yegutkin GG. Enzymes involved in metabolism of extracellular nucleotides and nucleosides: functional implications and measurement of activities. Crit Rev Biochem Mol Biol. 2014;49:473–97.CrossRefPubMed Yegutkin GG. Enzymes involved in metabolism of extracellular nucleotides and nucleosides: functional implications and measurement of activities. Crit Rev Biochem Mol Biol. 2014;49:473–97.CrossRefPubMed
48.
go back to reference Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di Virgilio F. Increased level of extracellular ATP at tumor sites: in vivo imaging with plasma membrane luciferase. PLoS One. 2008;3:e2599.PubMedCentralCrossRefPubMed Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di Virgilio F. Increased level of extracellular ATP at tumor sites: in vivo imaging with plasma membrane luciferase. PLoS One. 2008;3:e2599.PubMedCentralCrossRefPubMed
49.
go back to reference Tafani M, Schito L, Pellegrini L, Villanova L, Marfe G, Anwar T, et al. Hypoxia-increased RAGE and P2X7R expression regulates tumor cell invasion through phosphorylation of Erk1/2 and Akt and nuclear translocation of NF-{kappa}B. Carcinogenesis. 2011;32:1167–75.CrossRefPubMed Tafani M, Schito L, Pellegrini L, Villanova L, Marfe G, Anwar T, et al. Hypoxia-increased RAGE and P2X7R expression regulates tumor cell invasion through phosphorylation of Erk1/2 and Akt and nuclear translocation of NF-{kappa}B. Carcinogenesis. 2011;32:1167–75.CrossRefPubMed
Metadata
Title
Extracellular nucleotides as novel, underappreciated pro-metastatic factors that stimulate purinergic signaling in human lung cancer cells
Authors
Gabriela Schneider
Talita Glaser
Claudiana Lameu
Ahmed Abdelbaset-Ismail
Zachariah Payne Sellers
Marcin Moniuszko
Henning Ulrich
Mariusz Z. Ratajczak
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2015
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0469-z

Other articles of this Issue 1/2015

Molecular Cancer 1/2015 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