Skip to main content
Top
Published in: Molecular Cancer 1/2023

Open Access 01-12-2023 | Acidosis | Research

Acidosis-mediated increase in IFN-γ-induced PD-L1 expression on cancer cells as an immune escape mechanism in solid tumors

Authors: Philipp Knopf, Dimitri Stowbur, Sabrina H. L. Hoffmann, Natalie Hermann, Andreas Maurer, Valentina Bucher, Marilena Poxleitner, Bredi Tako, Dominik Sonanini, Balaji Krishnamachary, Sanhita Sinharay, Birgit Fehrenbacher, Irene Gonzalez-Menendez, Felix Reckmann, David Bomze, Lukas Flatz, Daniela Kramer, Martin Schaller, Stephan Forchhammer, Zaver M. Bhujwalla, Leticia Quintanilla-Martinez, Klaus Schulze-Osthoff, Mark D. Pagel, Marieke F. Fransen, Martin Röcken, André F. Martins, Bernd J. Pichler, Kamran Ghoreschi, Manfred Kneilling

Published in: Molecular Cancer | Issue 1/2023

Login to get access

Abstract

Immune checkpoint inhibitors have revolutionized cancer therapy, yet the efficacy of these treatments is often limited by the heterogeneous and hypoxic tumor microenvironment (TME) of solid tumors. In the TME, programmed death-ligand 1 (PD-L1) expression on cancer cells is mainly regulated by Interferon-gamma (IFN-γ), which induces T cell exhaustion and enables tumor immune evasion. In this study, we demonstrate that acidosis, a common characteristic of solid tumors, significantly increases IFN-γ-induced PD-L1 expression on aggressive cancer cells, thus promoting immune escape. Using preclinical models, we found that acidosis enhances the genomic expression and phosphorylation of signal transducer and activator of transcription 1 (STAT1), and the translation of STAT1 mRNA by eukaryotic initiation factor 4F (elF4F), resulting in an increased PD-L1 expression. We observed this effect in murine and human anti-PD-L1-responsive tumor cell lines, but not in anti-PD-L1-nonresponsive tumor cell lines. In vivo studies fully validated our in vitro findings and revealed that neutralizing the acidic extracellular tumor pH by sodium bicarbonate treatment suppresses IFN-γ-induced PD-L1 expression and promotes immune cell infiltration in responsive tumors and thus reduces tumor growth. However, this effect was not observed in anti-PD-L1-nonresponsive tumors. In vivo experiments in tumor-bearing IFN-γ−/− mice validated the dependency on immune cell-derived IFN-γ for acidosis-mediated cancer cell PD-L1 induction and tumor immune escape. Thus, acidosis and IFN-γ-induced elevation of PD-L1 expression on cancer cells represent a previously unknown immune escape mechanism that may serve as a novel biomarker for anti-PD-L1/PD-1 treatment response. These findings have important implications for the development of new strategies to enhance the efficacy of immunotherapy in cancer patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Hamid O, et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol. 2019;30(4):582–8.PubMedPubMedCentralCrossRef Hamid O, et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol. 2019;30(4):582–8.PubMedPubMedCentralCrossRef
2.
go back to reference Grossman JE, et al. Is PD-L1 a consistent biomarker for anti-PD-1 therapy? The model of balstilimab in a virally-driven tumor. Oncogene. 2021;40(8):1393–5.PubMedPubMedCentralCrossRef Grossman JE, et al. Is PD-L1 a consistent biomarker for anti-PD-1 therapy? The model of balstilimab in a virally-driven tumor. Oncogene. 2021;40(8):1393–5.PubMedPubMedCentralCrossRef
3.
go back to reference Tewari KS, et al. Survival with cemiplimab in recurrent cervical cancer. N Engl J Med. 2022;386(6):544–55.PubMedCrossRef Tewari KS, et al. Survival with cemiplimab in recurrent cervical cancer. N Engl J Med. 2022;386(6):544–55.PubMedCrossRef
5.
go back to reference Mirza MR, et al. Dostarlimab for primary advanced or recurrent endometrial cancer. N Engl J Med. 2023;388(23):2145–58.PubMedCrossRef Mirza MR, et al. Dostarlimab for primary advanced or recurrent endometrial cancer. N Engl J Med. 2023;388(23):2145–58.PubMedCrossRef
7.
go back to reference Ott PA, et al. T-Cell-Inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37(4):318–27.PubMedCrossRef Ott PA, et al. T-Cell-Inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37(4):318–27.PubMedCrossRef
8.
go back to reference Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–56.PubMedCrossRef Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–56.PubMedCrossRef
9.
go back to reference Green MR, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010;116(17):3268–77.PubMedPubMedCentralCrossRef Green MR, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010;116(17):3268–77.PubMedPubMedCentralCrossRef
13.
15.
go back to reference Noman MZ, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90.PubMedPubMedCentralCrossRef Noman MZ, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90.PubMedPubMedCentralCrossRef
17.
go back to reference Williams JB, et al. Tumor heterogeneity and clonal cooperation influence the immune selection of IFN-γ-signaling mutant cancer cells. Nat Commun. 2020;11(1):602.PubMedPubMedCentralCrossRef Williams JB, et al. Tumor heterogeneity and clonal cooperation influence the immune selection of IFN-γ-signaling mutant cancer cells. Nat Commun. 2020;11(1):602.PubMedPubMedCentralCrossRef
19.
go back to reference Corbet C, Feron O. Tumour acidosis: from the passenger to the driver’s seat. Nat Rev Cancer. 2017;17(10):577–93.PubMedCrossRef Corbet C, Feron O. Tumour acidosis: from the passenger to the driver’s seat. Nat Rev Cancer. 2017;17(10):577–93.PubMedCrossRef
20.
go back to reference Huber V, et al. Cancer acidity: an ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol. 2017;43:74–89.PubMedCrossRef Huber V, et al. Cancer acidity: an ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol. 2017;43:74–89.PubMedCrossRef
21.
go back to reference Feng J, et al. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene. 2017;36(42):5829–39.PubMedCrossRef Feng J, et al. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene. 2017;36(42):5829–39.PubMedCrossRef
22.
go back to reference Daneshmandi S, Wegiel B, Seth P. Blockade of Lactate Dehydrogenase-A (LDH-A) improves efficacy of anti-programmed cell death-1 (PD-1) therapy in melanoma. Cancers (Basel). 2019;11(4):450.PubMedCrossRef Daneshmandi S, Wegiel B, Seth P. Blockade of Lactate Dehydrogenase-A (LDH-A) improves efficacy of anti-programmed cell death-1 (PD-1) therapy in melanoma. Cancers (Basel). 2019;11(4):450.PubMedCrossRef
23.
go back to reference Kwon Y-J, et al. The acidic tumor microenvironment enhances PD-L1 expression via activation of STAT3 in MDA-MB-231 breast cancer cells. BMC Cancer. 2022;22(1):852.PubMedPubMedCentralCrossRef Kwon Y-J, et al. The acidic tumor microenvironment enhances PD-L1 expression via activation of STAT3 in MDA-MB-231 breast cancer cells. BMC Cancer. 2022;22(1):852.PubMedPubMedCentralCrossRef
24.
go back to reference Pilon-Thomas S, et al. Neutralization of tumor acidity improves antitumor responses to immunotherapy. Cancer Res. 2016;76(6):1381–90.PubMedCrossRef Pilon-Thomas S, et al. Neutralization of tumor acidity improves antitumor responses to immunotherapy. Cancer Res. 2016;76(6):1381–90.PubMedCrossRef
26.
go back to reference Giatromanolaki A, et al. Programmed death-1 receptor (PD-1) and PD-ligand-1 (PD-L1) expression in non-small cell lung cancer and the immune-suppressive effect of anaerobic glycolysis. Med Oncol. 2019;36(9):76.PubMedCrossRef Giatromanolaki A, et al. Programmed death-1 receptor (PD-1) and PD-ligand-1 (PD-L1) expression in non-small cell lung cancer and the immune-suppressive effect of anaerobic glycolysis. Med Oncol. 2019;36(9):76.PubMedCrossRef
27.
go back to reference Estrella V, et al. Acidity generated by the tumor microenvironment drives local invasion. Can Res. 2013;73(5):1524–35.CrossRef Estrella V, et al. Acidity generated by the tumor microenvironment drives local invasion. Can Res. 2013;73(5):1524–35.CrossRef
28.
go back to reference Wu M, et al. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 2022;15(1):24.PubMedPubMedCentralCrossRef Wu M, et al. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 2022;15(1):24.PubMedPubMedCentralCrossRef
30.
go back to reference Peng Q, et al. PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade. Nat Commun. 2020;11(1):4835.PubMedPubMedCentralCrossRef Peng Q, et al. PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade. Nat Commun. 2020;11(1):4835.PubMedPubMedCentralCrossRef
31.
go back to reference Ding L, et al. PD-1/PD-L1 inhibitors-based treatment for advanced renal cell carcinoma: mechanisms affecting efficacy and combination therapies. Cancer Med. 2021;10(18):6384–401.PubMedPubMedCentralCrossRef Ding L, et al. PD-1/PD-L1 inhibitors-based treatment for advanced renal cell carcinoma: mechanisms affecting efficacy and combination therapies. Cancer Med. 2021;10(18):6384–401.PubMedPubMedCentralCrossRef
32.
go back to reference Mortezaee K, Majidpoor J, Kharazinejad E. The impact of hypoxia on tumor-mediated bypassing anti-PD-(L)1 therapy. Biomed Pharmacother. 2023;162:114646.PubMedCrossRef Mortezaee K, Majidpoor J, Kharazinejad E. The impact of hypoxia on tumor-mediated bypassing anti-PD-(L)1 therapy. Biomed Pharmacother. 2023;162:114646.PubMedCrossRef
33.
go back to reference Dalton DK, et al. Multiple defects of immune cell function in mice with disrupted interferon-gamma genes. Science. 1993;259(5102):1739–42.PubMedCrossRef Dalton DK, et al. Multiple defects of immune cell function in mice with disrupted interferon-gamma genes. Science. 1993;259(5102):1739–42.PubMedCrossRef
34.
go back to reference Shah T, et al. CEST-FISP: a novel technique for rapid chemical exchange saturation transfer MRI at 7 T. Magn Reson Med. 2011;65(2):432–7.PubMedCrossRef Shah T, et al. CEST-FISP: a novel technique for rapid chemical exchange saturation transfer MRI at 7 T. Magn Reson Med. 2011;65(2):432–7.PubMedCrossRef
35.
go back to reference Jones KM, et al. Respiration gating and Bloch fitting improve pH measurements with acidoCEST MRI in an ovarian orthotopic tumor model. Proc SPIE Int Soc Opt Eng. 2016;9788:97855154. Jones KM, et al. Respiration gating and Bloch fitting improve pH measurements with acidoCEST MRI in an ovarian orthotopic tumor model. Proc SPIE Int Soc Opt Eng. 2016;9788:97855154.
36.
go back to reference Nishisho T, et al. The a3 isoform vacuolar type H+-ATPase promotes distant metastasis in the mouse B16 melanoma cells. Mol Cancer Res. 2011;9(7):845–55.PubMedCrossRef Nishisho T, et al. The a3 isoform vacuolar type H+-ATPase promotes distant metastasis in the mouse B16 melanoma cells. Mol Cancer Res. 2011;9(7):845–55.PubMedCrossRef
37.
38.
go back to reference Mosely SI, et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res. 2017;5(1):29–41.PubMedCrossRef Mosely SI, et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res. 2017;5(1):29–41.PubMedCrossRef
40.
go back to reference Cerezo M, et al. Translational control of tumor immune escape via the eIF4F-STAT1-PD-L1 axis in melanoma. Nat Med. 2018;24(12):1877–86.PubMedCrossRef Cerezo M, et al. Translational control of tumor immune escape via the eIF4F-STAT1-PD-L1 axis in melanoma. Nat Med. 2018;24(12):1877–86.PubMedCrossRef
41.
go back to reference Lopez PJ, et al. Translation inhibitors stabilize Escherichia coli mRNAs independently of ribosome protection. Proc Natl Acad Sci U S A. 1998;95(11):6067–72.PubMedPubMedCentralCrossRef Lopez PJ, et al. Translation inhibitors stabilize Escherichia coli mRNAs independently of ribosome protection. Proc Natl Acad Sci U S A. 1998;95(11):6067–72.PubMedPubMedCentralCrossRef
42.
go back to reference Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and Anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. 2018;8:86.PubMedPubMedCentralCrossRef Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and Anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. 2018;8:86.PubMedPubMedCentralCrossRef
43.
go back to reference Fischer K, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007;109(9):3812–9.PubMedCrossRef Fischer K, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007;109(9):3812–9.PubMedCrossRef
44.
go back to reference Dietl K, et al. Lactic acid and acidification inhibit TNF secretion and glycolysis of human monocytes. J Immunol. 2010;184(3):1200–9.PubMedCrossRef Dietl K, et al. Lactic acid and acidification inhibit TNF secretion and glycolysis of human monocytes. J Immunol. 2010;184(3):1200–9.PubMedCrossRef
45.
go back to reference Hirahara K, et al. Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity. 2012;36(6):1017–30.PubMedPubMedCentralCrossRef Hirahara K, et al. Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity. 2012;36(6):1017–30.PubMedPubMedCentralCrossRef
47.
go back to reference Chen LQ, et al. Evaluations of extracellular pH within in vivo tumors using acidoCEST MRI. Magn Reson Med. 2014;72(5):1408–17.PubMedCrossRef Chen LQ, et al. Evaluations of extracellular pH within in vivo tumors using acidoCEST MRI. Magn Reson Med. 2014;72(5):1408–17.PubMedCrossRef
48.
go back to reference Cappellesso F, et al. Targeting the bicarbonate transporter SLC4A4 overcomes immunosuppression and immunotherapy resistance in pancreatic cancer. Nat Cancer. 2022;3(12):1464–83.PubMedPubMedCentralCrossRef Cappellesso F, et al. Targeting the bicarbonate transporter SLC4A4 overcomes immunosuppression and immunotherapy resistance in pancreatic cancer. Nat Cancer. 2022;3(12):1464–83.PubMedPubMedCentralCrossRef
50.
go back to reference Reiniger L, et al. Tumor necrosis correlates with PD-L1 and PD-1 expression in lung adenocarcinoma. Acta Oncol. 2019;58(8):1087–94.PubMedCrossRef Reiniger L, et al. Tumor necrosis correlates with PD-L1 and PD-1 expression in lung adenocarcinoma. Acta Oncol. 2019;58(8):1087–94.PubMedCrossRef
51.
go back to reference Pötzl J, et al. Reversal of tumor acidosis by systemic buffering reactivates NK cells to express IFN-γ and induces NK cell-dependent lymphoma control without other immunotherapies. Int J Cancer. 2017;140(9):2125–33.PubMedCrossRef Pötzl J, et al. Reversal of tumor acidosis by systemic buffering reactivates NK cells to express IFN-γ and induces NK cell-dependent lymphoma control without other immunotherapies. Int J Cancer. 2017;140(9):2125–33.PubMedCrossRef
52.
go back to reference Bidani A, et al. Evidence for pH sensitivity of tumor necrosis factor-alpha release by alveolar macrophages. Lung. 1998;176(2):111–21.PubMedCrossRef Bidani A, et al. Evidence for pH sensitivity of tumor necrosis factor-alpha release by alveolar macrophages. Lung. 1998;176(2):111–21.PubMedCrossRef
53.
go back to reference Hartley G, et al. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother. 2017;66(4):523–35.PubMedPubMedCentralCrossRef Hartley G, et al. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother. 2017;66(4):523–35.PubMedPubMedCentralCrossRef
55.
go back to reference Hosogi S, et al. An inhibitor of Na(+)/H(+) exchanger (NHE), ethyl-isopropyl amiloride (EIPA), diminishes proliferation of MKN28 human gastric cancer cells by decreasing the cytosolic Cl(-) concentration via DIDS-sensitive pathways. Cell Physiol Biochem. 2012;30(5):1241–53.PubMedCrossRef Hosogi S, et al. An inhibitor of Na(+)/H(+) exchanger (NHE), ethyl-isopropyl amiloride (EIPA), diminishes proliferation of MKN28 human gastric cancer cells by decreasing the cytosolic Cl(-) concentration via DIDS-sensitive pathways. Cell Physiol Biochem. 2012;30(5):1241–53.PubMedCrossRef
58.
go back to reference Juneja VR, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med. 2017;214(4):895–904.PubMedPubMedCentralCrossRef Juneja VR, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med. 2017;214(4):895–904.PubMedPubMedCentralCrossRef
59.
go back to reference Jin Y, et al. Different syngeneic tumors show distinctive intrinsic tumor-immunity and mechanisms of actions (MOA) of anti-PD-1 treatment. Sci Rep. 2022;12(1):3278.PubMedPubMedCentralCrossRef Jin Y, et al. Different syngeneic tumors show distinctive intrinsic tumor-immunity and mechanisms of actions (MOA) of anti-PD-1 treatment. Sci Rep. 2022;12(1):3278.PubMedPubMedCentralCrossRef
61.
go back to reference Zheng X, et al. Disulfiram improves the Anti-PD-1 therapy efficacy by regulating PD-L1 expression via epigenetically reactivation of IRF7 in triple negative breast cancer. Front Oncol. 2021;11(734853):11. Zheng X, et al. Disulfiram improves the Anti-PD-1 therapy efficacy by regulating PD-L1 expression via epigenetically reactivation of IRF7 in triple negative breast cancer. Front Oncol. 2021;11(734853):11.
62.
go back to reference Chen DS, Mellman I. Elements of cancer immunity and the cancer–immune set point. Nature. 2017;541(7637):321–30.PubMedCrossRef Chen DS, Mellman I. Elements of cancer immunity and the cancer–immune set point. Nature. 2017;541(7637):321–30.PubMedCrossRef
63.
go back to reference Higgs BW, et al. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin Cancer Res. 2018;24(16):3857–66.PubMedCrossRef Higgs BW, et al. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin Cancer Res. 2018;24(16):3857–66.PubMedCrossRef
64.
go back to reference Johnson DB, et al. Quantitative spatial profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 predicts improved outcomes of anti-PD-1 therapies in metastatic melanoma. Clin Cancer Res. 2018;24(21):5250–60.PubMedPubMedCentralCrossRef Johnson DB, et al. Quantitative spatial profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 predicts improved outcomes of anti-PD-1 therapies in metastatic melanoma. Clin Cancer Res. 2018;24(21):5250–60.PubMedPubMedCentralCrossRef
65.
go back to reference Bosticardo M, et al. Biased activation of human T lymphocytes due to low extracellular pH is antagonized by B7/CD28 costimulation. Eur J Immunol. 2001;31(9):2829–38.PubMedCrossRef Bosticardo M, et al. Biased activation of human T lymphocytes due to low extracellular pH is antagonized by B7/CD28 costimulation. Eur J Immunol. 2001;31(9):2829–38.PubMedCrossRef
66.
go back to reference Calcinotto A, et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012;72(11):2746–56.PubMedCrossRef Calcinotto A, et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012;72(11):2746–56.PubMedCrossRef
67.
Metadata
Title
Acidosis-mediated increase in IFN-γ-induced PD-L1 expression on cancer cells as an immune escape mechanism in solid tumors
Authors
Philipp Knopf
Dimitri Stowbur
Sabrina H. L. Hoffmann
Natalie Hermann
Andreas Maurer
Valentina Bucher
Marilena Poxleitner
Bredi Tako
Dominik Sonanini
Balaji Krishnamachary
Sanhita Sinharay
Birgit Fehrenbacher
Irene Gonzalez-Menendez
Felix Reckmann
David Bomze
Lukas Flatz
Daniela Kramer
Martin Schaller
Stephan Forchhammer
Zaver M. Bhujwalla
Leticia Quintanilla-Martinez
Klaus Schulze-Osthoff
Mark D. Pagel
Marieke F. Fransen
Martin Röcken
André F. Martins
Bernd J. Pichler
Kamran Ghoreschi
Manfred Kneilling
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2023
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-023-01900-0

Other articles of this Issue 1/2023

Molecular Cancer 1/2023 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