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

Open Access 01-12-2023 | Cytostatic Therapy | Research

Combination effects of amino acid transporter LAT1 inhibitor nanvuranlat and cytotoxic anticancer drug gemcitabine on pancreatic and biliary tract cancer cells

Authors: Kou Nishikubo, Ryuichi Ohgaki, Xingming Liu, Hiroki Okanishi, Minhui Xu, Hitoshi Endou, Yoshikatsu Kanai

Published in: Cancer Cell International | Issue 1/2023

Login to get access

Abstract

Background

Cytotoxic anticancer drugs widely used in cancer chemotherapy have some limitations, such as the development of side effects and drug resistance. Furthermore, monotherapy is often less effective against heterogeneous cancer tissues. Combination therapies of cytotoxic anticancer drugs with molecularly targeted drugs have been pursued to solve such fundamental problems. Nanvuranlat (JPH203 or KYT-0353), an inhibitor for L-type amino acid transporter 1 (LAT1; SLC7A5), has novel mechanisms of action to suppress the cancer cell proliferation and tumor growth by inhibiting the transport of large neutral amino acids into cancer cells. This study investigated the potential of the combined use of nanvuranlat and cytotoxic anticancer drugs.

Methods

The combination effects of cytotoxic anticancer drugs and nanvuranlat on cell growth were examined by a water-soluble tetrazolium salt assay in two-dimensional cultures of pancreatic and biliary tract cancer cell lines. To elucidate the pharmacological mechanisms underlying the combination of gemcitabine and nanvuranlat, we investigated apoptotic cell death and cell cycle by flow cytometry. The phosphorylation levels of amino acid-related signaling pathways were analyzed by Western blot. Furthermore, growth inhibition was examined in cancer cell spheroids.

Results

All the tested seven types of cytotoxic anticancer drugs combined with nanvuranlat significantly inhibited the cell growth of pancreatic cancer MIA PaCa-2 cells compared to their single treatment. Among them, the combined effects of gemcitabine and nanvuranlat were relatively high and confirmed in multiple pancreatic and biliary tract cell lines in two-dimensional cultures. The growth inhibitory effects were suggested to be additive but not synergistic under the tested conditions. Gemcitabine generally induced cell cycle arrest at the S phase and apoptotic cell death, while nanvuranlat induced cell cycle arrest at the G0/G1 phase and affected amino acid-related mTORC1 and GAAC signaling pathways. In combination, each anticancer drug basically exerted its own pharmacological activities, although gemcitabine more strongly influenced the cell cycle than nanvuranlat. The combination effects of growth inhibition were also verified in cancer cell spheroids.

Conclusions

Our study demonstrates the potential of first-in-class LAT1 inhibitor nanvuranlat as a concomitant drug with cytotoxic anticancer drugs, especially gemcitabine, on pancreatic and biliary tract cancers.
Appendix
Available only for authorised users
Literature
1.
go back to reference Undevia SD, Gomez-Abuin G, Ratain MJ. Pharmacokinetic variability of anticancer agents. Nat Rev Cancer. 2005;5:447–58.PubMedCrossRef Undevia SD, Gomez-Abuin G, Ratain MJ. Pharmacokinetic variability of anticancer agents. Nat Rev Cancer. 2005;5:447–58.PubMedCrossRef
3.
go back to reference Pan ST, Li ZL, He ZX, Qiu JX, Zhou SF. Molecular mechanisms for tumour resistance to chemotherapy. Clin Exp Pharmacol Physiol. 2016;43:723–37.PubMedCrossRef Pan ST, Li ZL, He ZX, Qiu JX, Zhou SF. Molecular mechanisms for tumour resistance to chemotherapy. Clin Exp Pharmacol Physiol. 2016;43:723–37.PubMedCrossRef
6.
7.
go back to reference Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.CrossRef Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.CrossRef
8.
go back to reference Hammel P, Huguet F, Van Laethem JL, Goldstein D, Glimelius B, Artru P, et al. Effect of chemoradiotherapy vs chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: the LAP07 randomized clinical trial. JAMA. 2016;315:1844–53.PubMedCrossRef Hammel P, Huguet F, Van Laethem JL, Goldstein D, Glimelius B, Artru P, et al. Effect of chemoradiotherapy vs chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: the LAP07 randomized clinical trial. JAMA. 2016;315:1844–53.PubMedCrossRef
9.
go back to reference Arima S, Shimizu K, Okamoto T, Toki M, Suzuki Y, Okano N, et al. A Multicenter phase II study of gemcitabine plus S-1 chemotherapy for advanced biliary tract cancer. Anticancer Res. 2017;37:909–14.PubMedCrossRef Arima S, Shimizu K, Okamoto T, Toki M, Suzuki Y, Okano N, et al. A Multicenter phase II study of gemcitabine plus S-1 chemotherapy for advanced biliary tract cancer. Anticancer Res. 2017;37:909–14.PubMedCrossRef
10.
go back to reference Morizane C, Okusaka T, Mizusawa J, Katayama H, Ueno M, Ikeda M, et al. Combination gemcitabine plus S-1 versus gemcitabine plus cisplatin for advanced/recurrent biliary tract cancer: the FUGA-BT (JCOG1113) randomized phase III clinical trial. Ann Oncol. 2019;30:1950–8.PubMedCrossRef Morizane C, Okusaka T, Mizusawa J, Katayama H, Ueno M, Ikeda M, et al. Combination gemcitabine plus S-1 versus gemcitabine plus cisplatin for advanced/recurrent biliary tract cancer: the FUGA-BT (JCOG1113) randomized phase III clinical trial. Ann Oncol. 2019;30:1950–8.PubMedCrossRef
13.
go back to reference Wolfson RL, Chantranupong L, Saxton RA, Shen K, Scaria SM, Cantor JR, et al. Sestrin2 is a leucine sensor for the mTORC1 pathway. Science. 2016;351:43–8.PubMedCrossRef Wolfson RL, Chantranupong L, Saxton RA, Shen K, Scaria SM, Cantor JR, et al. Sestrin2 is a leucine sensor for the mTORC1 pathway. Science. 2016;351:43–8.PubMedCrossRef
14.
go back to reference Kanai Y, Segawa H, Miyamoto KI, Uchino H, Takeda E, Endou H. Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen (CD98). J Biol Chem. 1998;273:23629–32.PubMedCrossRef Kanai Y, Segawa H, Miyamoto KI, Uchino H, Takeda E, Endou H. Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen (CD98). J Biol Chem. 1998;273:23629–32.PubMedCrossRef
15.
go back to reference Yanagida O, Kanai Y, Chairoungdua A, Kim DK, Segawa H, Nii T, et al. Human L-type amino acid transporter 1 (LAT1): characterization of function and expression in tumor cell lines. Biochim Biophys Acta. 2001;1514:291–302.PubMedCrossRef Yanagida O, Kanai Y, Chairoungdua A, Kim DK, Segawa H, Nii T, et al. Human L-type amino acid transporter 1 (LAT1): characterization of function and expression in tumor cell lines. Biochim Biophys Acta. 2001;1514:291–302.PubMedCrossRef
16.
go back to reference Kaira K, Sunose Y, Arakawa K, Ogawa T, Sunaga N, Shimizu K, et al. Prognostic significance of L-type amino-acid transporter 1 expression in surgically resected pancreatic cancer. Br J Cancer. 2012;107:632–8.PubMedPubMedCentralCrossRef Kaira K, Sunose Y, Arakawa K, Ogawa T, Sunaga N, Shimizu K, et al. Prognostic significance of L-type amino-acid transporter 1 expression in surgically resected pancreatic cancer. Br J Cancer. 2012;107:632–8.PubMedPubMedCentralCrossRef
17.
go back to reference Altan B, Kaira K, Watanabe A, Kubo N, Bao P, Dolgormaa G, et al. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. Cancer Chemother Pharmacol. 2018;81:141–53.PubMedCrossRef Altan B, Kaira K, Watanabe A, Kubo N, Bao P, Dolgormaa G, et al. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. Cancer Chemother Pharmacol. 2018;81:141–53.PubMedCrossRef
18.
go back to reference Kaira K, Sunose Y, Ohshima Y, Ishioka NS, Arakawa K, Ogawa T, et al. Clinical significance of L-type amino acid transporter 1 expression as a prognostic marker and potential of new targeting therapy in biliary tract cancer. BMC Cancer. 2013;13:482.PubMedPubMedCentralCrossRef Kaira K, Sunose Y, Ohshima Y, Ishioka NS, Arakawa K, Ogawa T, et al. Clinical significance of L-type amino acid transporter 1 expression as a prognostic marker and potential of new targeting therapy in biliary tract cancer. BMC Cancer. 2013;13:482.PubMedPubMedCentralCrossRef
19.
go back to reference Oda K, Hosoda N, Endo H, Saito K, Tsujihara K, Yamamura M, et al. L-Type amino acid transporter 1 inhibitors inhibit tumor cell growth. Cancer Sci. 2010;101:173–9.PubMedCrossRef Oda K, Hosoda N, Endo H, Saito K, Tsujihara K, Yamamura M, et al. L-Type amino acid transporter 1 inhibitors inhibit tumor cell growth. Cancer Sci. 2010;101:173–9.PubMedCrossRef
20.
go back to reference Kanai Y. Amino acid transporter LAT1 (SLC7A5) as a molecular target for cancer diagnosis and therapeutics. Pharmacol Ther. 2022;230:107964.PubMedCrossRef Kanai Y. Amino acid transporter LAT1 (SLC7A5) as a molecular target for cancer diagnosis and therapeutics. Pharmacol Ther. 2022;230:107964.PubMedCrossRef
21.
go back to reference Maimaiti M, Sakamoto S, Yamada Y, Sugiura M, Rii J, Takeuchi N, et al. Expression of L-type amino acid transporter 1 as a molecular target for prognostic and therapeutic indicators in bladder carcinoma. Sci Rep. 2020;10:1292.PubMedPubMedCentralCrossRef Maimaiti M, Sakamoto S, Yamada Y, Sugiura M, Rii J, Takeuchi N, et al. Expression of L-type amino acid transporter 1 as a molecular target for prognostic and therapeutic indicators in bladder carcinoma. Sci Rep. 2020;10:1292.PubMedPubMedCentralCrossRef
22.
go back to reference Cormerais Y, Pagnuzzi-Boncompagni M, Schrötter S, Giuliano S, Tambutté E, Endou H, et al. Inhibition of the amino-acid transporter LAT1 demonstrates anti-neoplastic activity in medulloblastoma. J Cell Mol Med. 2019;23:2711–8.PubMedPubMedCentralCrossRef Cormerais Y, Pagnuzzi-Boncompagni M, Schrötter S, Giuliano S, Tambutté E, Endou H, et al. Inhibition of the amino-acid transporter LAT1 demonstrates anti-neoplastic activity in medulloblastoma. J Cell Mol Med. 2019;23:2711–8.PubMedPubMedCentralCrossRef
23.
go back to reference Higuchi K, Sakamoto S, Ando K, Maimaiti M, Takeshita N, Okunushi K, et al. Characterization of the expression of LAT1 as a prognostic indicator and a therapeutic target in renal cell carcinoma. Sci Rep. 2019;9:16776.PubMedPubMedCentralCrossRef Higuchi K, Sakamoto S, Ando K, Maimaiti M, Takeshita N, Okunushi K, et al. Characterization of the expression of LAT1 as a prognostic indicator and a therapeutic target in renal cell carcinoma. Sci Rep. 2019;9:16776.PubMedPubMedCentralCrossRef
24.
go back to reference Häfliger P, Graff J, Rubin M, Stooss A, Dettmer MS, Altmann KH, et al. The LAT1 inhibitor JPH203 reduces growth of thyroid carcinoma in a fully immunocompetent mouse model. J Exp Clin Cancer Res. 2018;37:234.PubMedPubMedCentralCrossRef Häfliger P, Graff J, Rubin M, Stooss A, Dettmer MS, Altmann KH, et al. The LAT1 inhibitor JPH203 reduces growth of thyroid carcinoma in a fully immunocompetent mouse model. J Exp Clin Cancer Res. 2018;37:234.PubMedPubMedCentralCrossRef
25.
go back to reference Yothaisong S, Dokduang H, Anzai N, Hayashi K, Namwat N, Yongvanit P, et al. Inhibition of l-type amino acid transporter 1 activity as a new therapeutic target for cholangiocarcinoma treatment. Tumour Biol. 2017;39:1010428317694545.PubMedCrossRef Yothaisong S, Dokduang H, Anzai N, Hayashi K, Namwat N, Yongvanit P, et al. Inhibition of l-type amino acid transporter 1 activity as a new therapeutic target for cholangiocarcinoma treatment. Tumour Biol. 2017;39:1010428317694545.PubMedCrossRef
26.
go back to reference Hayashi K, Jutabha P, Maeda S, Supak Y, Ouchi M, Endou H, et al. LAT1 acts as a crucial transporter of amino acids in human thymic carcinoma cells. J Pharmacol Sci. 2016;132:201–4.PubMedCrossRef Hayashi K, Jutabha P, Maeda S, Supak Y, Ouchi M, Endou H, et al. LAT1 acts as a crucial transporter of amino acids in human thymic carcinoma cells. J Pharmacol Sci. 2016;132:201–4.PubMedCrossRef
27.
go back to reference Rosilio C, Nebout M, Imbert V, Griessinger E, Neffati Z, Benadiba J, et al. L-type amino-acid transporter 1 (LAT1): a therapeutic target supporting growth and survival of T-cell lymphoblastic lymphoma/T-cell acute lymphoblastic leukemia. Leukemia. 2015;29:1253–66.PubMedCrossRef Rosilio C, Nebout M, Imbert V, Griessinger E, Neffati Z, Benadiba J, et al. L-type amino-acid transporter 1 (LAT1): a therapeutic target supporting growth and survival of T-cell lymphoblastic lymphoma/T-cell acute lymphoblastic leukemia. Leukemia. 2015;29:1253–66.PubMedCrossRef
28.
go back to reference Muto Y, Furihata T, Kaneko M, Higuchi K, Okunushi K, Morio H, et al. Different response profiles of gastrointestinal cancer cells to an L-Type amino acid transporter inhibitor, JPH203. Anticancer Res. 2019;39:159–65.PubMedCrossRef Muto Y, Furihata T, Kaneko M, Higuchi K, Okunushi K, Morio H, et al. Different response profiles of gastrointestinal cancer cells to an L-Type amino acid transporter inhibitor, JPH203. Anticancer Res. 2019;39:159–65.PubMedCrossRef
29.
go back to reference Okanishi H, Ohgaki R, Okuda S, Endou H, Kanai Y. Proteomics and phosphoproteomics reveal key regulators associated with cytostatic effect of amino acid transporter LAT1 inhibitor. Cancer Sci. 2021;112:871–83.PubMedCrossRef Okanishi H, Ohgaki R, Okuda S, Endou H, Kanai Y. Proteomics and phosphoproteomics reveal key regulators associated with cytostatic effect of amino acid transporter LAT1 inhibitor. Cancer Sci. 2021;112:871–83.PubMedCrossRef
30.
go back to reference Nishikubo K, Ohgaki R, Okanishi H, Okuda S, Xu M, Endou H, et al. Pharmacologic inhibition of LAT1 predominantly suppresses transport of large neutral amino acids and downregulates global translation in cancer cells. J Cell Mol Med. 2022;26:5246–56.PubMedPubMedCentralCrossRef Nishikubo K, Ohgaki R, Okanishi H, Okuda S, Xu M, Endou H, et al. Pharmacologic inhibition of LAT1 predominantly suppresses transport of large neutral amino acids and downregulates global translation in cancer cells. J Cell Mol Med. 2022;26:5246–56.PubMedPubMedCentralCrossRef
31.
go back to reference Quan L, Ohgaki R, Hara S, Okuda S, Wei L, Okanishi H, et al. Amino acid transporter LAT1 in tumor-associated vascular endothelium promotes angiogenesis by regulating cell proliferation and VEGF-A-dependent mTORC1 activation. J Exp Clin Cancer Res. 2020;39:266.PubMedPubMedCentralCrossRef Quan L, Ohgaki R, Hara S, Okuda S, Wei L, Okanishi H, et al. Amino acid transporter LAT1 in tumor-associated vascular endothelium promotes angiogenesis by regulating cell proliferation and VEGF-A-dependent mTORC1 activation. J Exp Clin Cancer Res. 2020;39:266.PubMedPubMedCentralCrossRef
32.
go back to reference Saito Y, Li L, Coyaud E, Luna A, Sander C, Raught B, et al. LLGL2 rescues nutrient stress by promoting leucine uptake in ER+ breast cancer. Nature. 2019;569:275–9.PubMedCrossRef Saito Y, Li L, Coyaud E, Luna A, Sander C, Raught B, et al. LLGL2 rescues nutrient stress by promoting leucine uptake in ER+ breast cancer. Nature. 2019;569:275–9.PubMedCrossRef
33.
go back to reference Enomoto K, Sato F, Tamagawa S, Gunduz M, Onoda N, Uchino S, et al. A novel therapeutic approach for anaplastic thyroid cancer through inhibition of LAT1. Sci Rep. 2019;9:14616.PubMedPubMedCentralCrossRef Enomoto K, Sato F, Tamagawa S, Gunduz M, Onoda N, Uchino S, et al. A novel therapeutic approach for anaplastic thyroid cancer through inhibition of LAT1. Sci Rep. 2019;9:14616.PubMedPubMedCentralCrossRef
34.
go back to reference Furuse J, Ikeda M, Ueno M, Furukawa M, Morizane C, Takehara T, et al. Nanvuranlat, an L-type amino acid transporter (LAT1) inhibitor for patients with pretreated advanced refractory biliary tract cancer (BTC): primary endpoint results of a randomized, double-blind, placebo-controlled phase 2 study. J Clin Oncol. 2023;41(4suppl):494.CrossRef Furuse J, Ikeda M, Ueno M, Furukawa M, Morizane C, Takehara T, et al. Nanvuranlat, an L-type amino acid transporter (LAT1) inhibitor for patients with pretreated advanced refractory biliary tract cancer (BTC): primary endpoint results of a randomized, double-blind, placebo-controlled phase 2 study. J Clin Oncol. 2023;41(4suppl):494.CrossRef
35.
go back to reference Yamauchi K, Sakurai H, Kimura T, Wiriyasermkul P, Nagamori S, Kanai Y, et al. System L amino acid transporter inhibitor enhances anti-tumor activity of cisplatin in a head and neck squamous cell carcinoma cell line. Cancer Lett. 2009;276:95–101.PubMedCrossRef Yamauchi K, Sakurai H, Kimura T, Wiriyasermkul P, Nagamori S, Kanai Y, et al. System L amino acid transporter inhibitor enhances anti-tumor activity of cisplatin in a head and neck squamous cell carcinoma cell line. Cancer Lett. 2009;276:95–101.PubMedCrossRef
36.
go back to reference Bliss CI. The toxicity of poisons applied jointly. Ann Appl Biol. 1939;26:585–615.CrossRef Bliss CI. The toxicity of poisons applied jointly. Ann Appl Biol. 1939;26:585–615.CrossRef
37.
go back to reference Duarte D, Vale N. Evaluation of synergism in drug combinations and reference models for future orientations in oncology. Curr Res Pharmacol Drug Discov. 2022;3:100110.PubMedPubMedCentralCrossRef Duarte D, Vale N. Evaluation of synergism in drug combinations and reference models for future orientations in oncology. Curr Res Pharmacol Drug Discov. 2022;3:100110.PubMedPubMedCentralCrossRef
38.
go back to reference Mini E, Nobili S, Caciagli B, Landini I, Mazzei T. Cellular pharmacology of gemcitabine. Ann Oncol. 2006;17(Suppl 5):v7–12.PubMedCrossRef Mini E, Nobili S, Caciagli B, Landini I, Mazzei T. Cellular pharmacology of gemcitabine. Ann Oncol. 2006;17(Suppl 5):v7–12.PubMedCrossRef
39.
go back to reference Christensen HN. Role of amino acid transport and countertransport in nutrition and metabolism. Physiol Rev. 1990;70:43–77.PubMedCrossRef Christensen HN. Role of amino acid transport and countertransport in nutrition and metabolism. Physiol Rev. 1990;70:43–77.PubMedCrossRef
41.
go back to reference Ito D, Fujimoto K, Mori T, Kami K, Koizumi M, Toyoda E, et al. In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int J Cancer. 2006;118:2337–43.PubMedCrossRef Ito D, Fujimoto K, Mori T, Kami K, Koizumi M, Toyoda E, et al. In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int J Cancer. 2006;118:2337–43.PubMedCrossRef
42.
go back to reference Karavasilis V, Samantas E, Koliou GA, Kalogera-Fountzila A, Pentheroudakis G, Varthalitis I, et al. Gemcitabine combined with the mTOR inhibitor temsirolimus in patients with locally advanced or metastatic pancreatic cancer. A hellenic cooperative oncology group phase I/II study. Target Oncol. 2018;13:715–24.PubMedCrossRef Karavasilis V, Samantas E, Koliou GA, Kalogera-Fountzila A, Pentheroudakis G, Varthalitis I, et al. Gemcitabine combined with the mTOR inhibitor temsirolimus in patients with locally advanced or metastatic pancreatic cancer. A hellenic cooperative oncology group phase I/II study. Target Oncol. 2018;13:715–24.PubMedCrossRef
43.
go back to reference Cui J, Guo Y, Wu H, Xiong J, Peng T. Everolimus regulates the activity of gemcitabine-resistant pancreatic cancer cells by targeting the Warburg effect via PI3K/AKT/mTOR signaling. Mol Med. 2021;27:38.PubMedPubMedCentralCrossRef Cui J, Guo Y, Wu H, Xiong J, Peng T. Everolimus regulates the activity of gemcitabine-resistant pancreatic cancer cells by targeting the Warburg effect via PI3K/AKT/mTOR signaling. Mol Med. 2021;27:38.PubMedPubMedCentralCrossRef
44.
go back to reference Lin G, Lin KJ, Wang F, Chen TC, Yen TC, Yeh T, Sen. Synergistic antiproliferative effects of an mTOR inhibitor (rad001) plus gemcitabine on cholangiocarcinoma by decreasing choline kinase activity. Dis Model Mech. 2018;11:dmm033050.PubMedPubMedCentralCrossRef Lin G, Lin KJ, Wang F, Chen TC, Yen TC, Yeh T, Sen. Synergistic antiproliferative effects of an mTOR inhibitor (rad001) plus gemcitabine on cholangiocarcinoma by decreasing choline kinase activity. Dis Model Mech. 2018;11:dmm033050.PubMedPubMedCentralCrossRef
45.
go back to reference De Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.PubMedCrossRef De Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.PubMedCrossRef
Metadata
Title
Combination effects of amino acid transporter LAT1 inhibitor nanvuranlat and cytotoxic anticancer drug gemcitabine on pancreatic and biliary tract cancer cells
Authors
Kou Nishikubo
Ryuichi Ohgaki
Xingming Liu
Hiroki Okanishi
Minhui Xu
Hitoshi Endou
Yoshikatsu Kanai
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2023
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-023-02957-z

Other articles of this Issue 1/2023

Cancer Cell International 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