Skip to main content
Top
Published in: Investigational New Drugs 3/2014

01-06-2014 | PRECLINICAL STUDIES

NPS-1034, a novel MET inhibitor, inhibits the activated MET receptor and its constitutively active mutants

Authors: Jae-Sik Shin, Seung-Woo Hong, Jai-Hee Moon, Jin-Sun Kim, Kyung-Ah Jung, Seung-Mi Kim, Dae-Hee Lee, InKi Kim, Seon-Joo Yoon, Chang-Gyu Lee, Eun-Kyoung Choi, Joo-Young Lee, Kyu-pyo Kim, Yong Sang Hong, Jae-Lyun Lee, Bongcheol Kim, Eun Kyung Choi, Jung Shin Lee, Dong-Hoon Jin, Tae Won Kim

Published in: Investigational New Drugs | Issue 3/2014

Login to get access

Summary

The MET proto-oncogene product, which is the receptor for hepatocyte growth factor (HGF), has been implicated in tumorigenesis and metastatic progression. Point mutations in MET lead to the aberrant activation of the receptor in many types of human malignancies, and the deregulated activity of MET has been correlated with tumor growth, invasion, and metastasis. MET has therefore attracted considerable attention as a potential target in anticancer therapy. Here, we report that a novel MET kinase inhibitor, NPS-1034, inhibits various constitutively active mutant forms of MET as well as HGF-activated wild-type MET. NPS-1034 inhibited the proliferation of cells expressing activated MET and promoted the regression of tumors formed from such cells in a mouse xenograft model through anti-angiogenic and pro-apoptotic actions. NPS-1034 also inhibited HGF-stimulated activation of MET signaling in the presence or absence of serum. Furthermore, when tested on 27 different MET variants, NPS-1034 inhibited 15 of the 17 MET variants that exhibited autophosphorylation with nanomolar potency; only the F1218I and M1149T variants were not inhibited by NPS-1034. Notably, NPS-1034 inhibited three MET variants that are resistant to the MET inhibitors SU11274, NVP-BVU972, and PHA665752. Together, these results suggest that NPS-1034 can be used as a potent therapeutic agent for human malignancies bearing MET point mutations or expressing activated MET.
Appendix
Available only for authorised users
Literature
2.
go back to reference Otsuka T, Takayama H, Sharp R, Celli G, LaRochelle WJ, Bottaro DP, Ellmore N, Vieira W, Owens JW, Anver M, Merlino G (1998) c-Met autocrine activation induces development of malignant melanoma and acquisition of the metastatic phenotype. Cancer Res 58(22):5157–5167PubMed Otsuka T, Takayama H, Sharp R, Celli G, LaRochelle WJ, Bottaro DP, Ellmore N, Vieira W, Owens JW, Anver M, Merlino G (1998) c-Met autocrine activation induces development of malignant melanoma and acquisition of the metastatic phenotype. Cancer Res 58(22):5157–5167PubMed
3.
go back to reference Sattler M, Salgia R (2007) c-Met and hepatocyte growth factor: potential as novel targets in cancer therapy. Curr Oncol Rep 9(2):102–108CrossRefPubMed Sattler M, Salgia R (2007) c-Met and hepatocyte growth factor: potential as novel targets in cancer therapy. Curr Oncol Rep 9(2):102–108CrossRefPubMed
5.
go back to reference Toiyama Y, Yasuda H, Saigusa S, Matushita K, Fujikawa H, Tanaka K, Mohri Y, Inoue Y, Goel A, Kusunoki M (2012) Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int J Cancer 130(12):2912–2921. doi:10.1002/ijc.26330 CrossRefPubMed Toiyama Y, Yasuda H, Saigusa S, Matushita K, Fujikawa H, Tanaka K, Mohri Y, Inoue Y, Goel A, Kusunoki M (2012) Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int J Cancer 130(12):2912–2921. doi:10.​1002/​ijc.​26330 CrossRefPubMed
7.
go back to reference Lorenzato A, Olivero M, Patane S, Rosso E, Oliaro A, Comoglio PM, Di Renzo MF (2002) Novel somatic mutations of the MET oncogene in human carcinoma metastases activating cell motility and invasion. Cancer Res 62(23):7025–7030PubMed Lorenzato A, Olivero M, Patane S, Rosso E, Oliaro A, Comoglio PM, Di Renzo MF (2002) Novel somatic mutations of the MET oncogene in human carcinoma metastases activating cell motility and invasion. Cancer Res 62(23):7025–7030PubMed
8.
go back to reference Bellon SF, Kaplan-Lefko P, Yang Y, Zhang Y, Moriguchi J, Rex K, Johnson CW, Rose PE, Long AM, O’Connor AB, Gu Y, Coxon A, Kim TS, Tasker A, Burgess TL, Dussault I (2008) c-Met inhibitors with novel binding mode show activity against several hereditary papillary renal cell carcinoma-related mutations. J Biol Chem 283(5):2675–2683. doi:10.1074/jbc.M705774200 CrossRefPubMed Bellon SF, Kaplan-Lefko P, Yang Y, Zhang Y, Moriguchi J, Rex K, Johnson CW, Rose PE, Long AM, O’Connor AB, Gu Y, Coxon A, Kim TS, Tasker A, Burgess TL, Dussault I (2008) c-Met inhibitors with novel binding mode show activity against several hereditary papillary renal cell carcinoma-related mutations. J Biol Chem 283(5):2675–2683. doi:10.​1074/​jbc.​M705774200 CrossRefPubMed
9.
go back to reference Park WS, Dong SM, Kim SY, Na EY, Shin MS, Pi JH, Kim BJ, Bae JH, Hong YK, Lee KS, Lee SH, Yoo NJ, Jang JJ, Pack S, Zhuang Z, Schmidt L, Zbar B, Lee JY (1999) Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res 59(2):307–310PubMed Park WS, Dong SM, Kim SY, Na EY, Shin MS, Pi JH, Kim BJ, Bae JH, Hong YK, Lee KS, Lee SH, Yoo NJ, Jang JJ, Pack S, Zhuang Z, Schmidt L, Zbar B, Lee JY (1999) Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res 59(2):307–310PubMed
10.
go back to reference Di Renzo MF, Olivero M, Martone T, Maffe A, Maggiora P, Stefani AD, Valente G, Giordano S, Cortesina G, Comoglio PM (2000) Somatic mutations of the MET oncogene are selected during metastatic spread of human HNSC carcinomas. Oncogene 19(12):1547–1555. doi:10.1038/sj.onc.1203455 CrossRefPubMed Di Renzo MF, Olivero M, Martone T, Maffe A, Maggiora P, Stefani AD, Valente G, Giordano S, Cortesina G, Comoglio PM (2000) Somatic mutations of the MET oncogene are selected during metastatic spread of human HNSC carcinomas. Oncogene 19(12):1547–1555. doi:10.​1038/​sj.​onc.​1203455 CrossRefPubMed
11.
go back to reference Ma PC, Kijima T, Maulik G, Fox EA, Sattler M, Griffin JD, Johnson BE, Salgia R (2003) c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Res 63(19):6272–6281PubMed Ma PC, Kijima T, Maulik G, Fox EA, Sattler M, Griffin JD, Johnson BE, Salgia R (2003) c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Res 63(19):6272–6281PubMed
12.
go back to reference Yuen HF, Abramczyk O, Montgomery G, Chan KK, Huang YH, Sasazuki T, Shirasawa S, Gopesh S, Chan KW, Fennell D, Janne P, El-Tanani M, Murray JT (2012) Impact of oncogenic driver mutations on feedback between the PI3K and MEK pathways in cancer cells. Biosci Rep 32(4):413–422. doi:10.1042/BSR20120050 PubMedCentralCrossRefPubMed Yuen HF, Abramczyk O, Montgomery G, Chan KK, Huang YH, Sasazuki T, Shirasawa S, Gopesh S, Chan KW, Fennell D, Janne P, El-Tanani M, Murray JT (2012) Impact of oncogenic driver mutations on feedback between the PI3K and MEK pathways in cancer cells. Biosci Rep 32(4):413–422. doi:10.​1042/​BSR20120050 PubMedCentralCrossRefPubMed
14.
go back to reference Graveel C, Su Y, Koeman J, Wang LM, Tessarollo L, Fiscella M, Birchmeier C, Swiatek P, Bronson R, Vande Woude G (2004) Activating Met mutations produce unique tumor profiles in mice with selective duplication of the mutant allele. Proc Natl Acad Sci U S A 101(49):17198–17203. doi:10.1073/pnas.0407651101 PubMedCentralCrossRefPubMed Graveel C, Su Y, Koeman J, Wang LM, Tessarollo L, Fiscella M, Birchmeier C, Swiatek P, Bronson R, Vande Woude G (2004) Activating Met mutations produce unique tumor profiles in mice with selective duplication of the mutant allele. Proc Natl Acad Sci U S A 101(49):17198–17203. doi:10.​1073/​pnas.​0407651101 PubMedCentralCrossRefPubMed
15.
go back to reference Berthou S, Aebersold DM, Schmidt LS, Stroka D, Heigl C, Streit B, Stalder D, Gruber G, Liang C, Howlett AR, Candinas D, Greiner RH, Lipson KE, Zimmer Y (2004) The Met kinase inhibitor SU11274 exhibits a selective inhibition pattern toward different receptor mutated variants. Oncogene 23(31):5387–5393. doi:10.1038/sj.onc.1207691 CrossRefPubMed Berthou S, Aebersold DM, Schmidt LS, Stroka D, Heigl C, Streit B, Stalder D, Gruber G, Liang C, Howlett AR, Candinas D, Greiner RH, Lipson KE, Zimmer Y (2004) The Met kinase inhibitor SU11274 exhibits a selective inhibition pattern toward different receptor mutated variants. Oncogene 23(31):5387–5393. doi:10.​1038/​sj.​onc.​1207691 CrossRefPubMed
18.
go back to reference Tiedt R, Degenkolbe E, Furet P, Appleton BA, Wagner S, Schoepfer J, Buck E, Ruddy DA, Monahan JE, Jones MD, Blank J, Haasen D, Drueckes P, Wartmann M, McCarthy C, Sellers WR, Hofmann F (2011) A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients. Cancer Res 71(15):5255–5264. doi:10.1158/0008-5472.CAN-10-4433 CrossRefPubMed Tiedt R, Degenkolbe E, Furet P, Appleton BA, Wagner S, Schoepfer J, Buck E, Ruddy DA, Monahan JE, Jones MD, Blank J, Haasen D, Drueckes P, Wartmann M, McCarthy C, Sellers WR, Hofmann F (2011) A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients. Cancer Res 71(15):5255–5264. doi:10.​1158/​0008-5472.​CAN-10-4433 CrossRefPubMed
19.
go back to reference Pan BS, Chan GK, Chenard M, Chi A, Davis LJ, Deshmukh SV, Gibbs JB, Gil S, Hang G, Hatch H, Jewell JP, Kariv I, Katz JD, Kunii K, Lu W, Lutterbach BA, Paweletz CP, Qu X, Reilly JF, Szewczak AA, Zeng Q, Kohl NE, Dinsmore CJ (2010) MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Res 70(4):1524–1533. doi:10.1158/0008-5472.CAN-09-2541 CrossRefPubMed Pan BS, Chan GK, Chenard M, Chi A, Davis LJ, Deshmukh SV, Gibbs JB, Gil S, Hang G, Hatch H, Jewell JP, Kariv I, Katz JD, Kunii K, Lu W, Lutterbach BA, Paweletz CP, Qu X, Reilly JF, Szewczak AA, Zeng Q, Kohl NE, Dinsmore CJ (2010) MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Res 70(4):1524–1533. doi:10.​1158/​0008-5472.​CAN-09-2541 CrossRefPubMed
20.
go back to reference Hong SW, Kim CJ, Park WS, Shin JS, Lee SD, Ko SG, Jung SI, Park IC, An SK, Lee WK, Lee WJ, Jin DH, Lee MS (2009) p34SEI-1 inhibits apoptosis through the stabilization of the X-linked inhibitor of apoptosis protein: p34SEI-1 as a novel target for anti-breast cancer strategies. Cancer Res 69(3):741–746. doi:10.1158/0008-5472.CAN-08-1189 CrossRefPubMed Hong SW, Kim CJ, Park WS, Shin JS, Lee SD, Ko SG, Jung SI, Park IC, An SK, Lee WK, Lee WJ, Jin DH, Lee MS (2009) p34SEI-1 inhibits apoptosis through the stabilization of the X-linked inhibitor of apoptosis protein: p34SEI-1 as a novel target for anti-breast cancer strategies. Cancer Res 69(3):741–746. doi:10.​1158/​0008-5472.​CAN-08-1189 CrossRefPubMed
21.
go back to reference Moonen JR, Krenning G, Brinker MG, Koerts JA, van Luyn MJ, Harmsen MC (2010) Endothelial progenitor cells give rise to pro-angiogenic smooth muscle-like progeny. Cardiovasc Res 86(3):506–515. doi:10.1093/cvr/cvq012 CrossRefPubMed Moonen JR, Krenning G, Brinker MG, Koerts JA, van Luyn MJ, Harmsen MC (2010) Endothelial progenitor cells give rise to pro-angiogenic smooth muscle-like progeny. Cardiovasc Res 86(3):506–515. doi:10.​1093/​cvr/​cvq012 CrossRefPubMed
22.
go back to reference Kong-Beltran M, Seshagiri S, Zha J, Zhu W, Bhawe K, Mendoza N, Holcomb T, Pujara K, Stinson J, Fu L, Severin C, Rangell L, Schwall R, Amler L, Wickramasinghe D, Yauch R (2006) Somatic mutations lead to an oncogenic deletion of met in lung cancer. Cancer Res 66(1):283–289. doi:10.1158/0008-5472.CAN-05-2749 CrossRefPubMed Kong-Beltran M, Seshagiri S, Zha J, Zhu W, Bhawe K, Mendoza N, Holcomb T, Pujara K, Stinson J, Fu L, Severin C, Rangell L, Schwall R, Amler L, Wickramasinghe D, Yauch R (2006) Somatic mutations lead to an oncogenic deletion of met in lung cancer. Cancer Res 66(1):283–289. doi:10.​1158/​0008-5472.​CAN-05-2749 CrossRefPubMed
23.
go back to reference Zwick E, Bange J, Ullrich A (2001) Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer 8(3):161–173CrossRefPubMed Zwick E, Bange J, Ullrich A (2001) Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer 8(3):161–173CrossRefPubMed
24.
go back to reference Christensen JG, Schreck R, Burrows J, Kuruganti P, Chan E, Le P, Chen J, Wang X, Ruslim L, Blake R, Lipson KE, Ramphal J, Do S, Cui JJ, Cherrington JM, Mendel DB (2003) A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res 63(21):7345–7355PubMed Christensen JG, Schreck R, Burrows J, Kuruganti P, Chan E, Le P, Chen J, Wang X, Ruslim L, Blake R, Lipson KE, Ramphal J, Do S, Cui JJ, Cherrington JM, Mendel DB (2003) A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res 63(21):7345–7355PubMed
25.
go back to reference Crosswell HE, Dasgupta A, Alvarado CS, Watt T, Christensen JG, De P, Durden DL, Findley HW (2009) PHA665752, a small-molecule inhibitor of c-Met, inhibits hepatocyte growth factor-stimulated migration and proliferation of c-Met-positive neuroblastoma cells. BMC Cancer 9:411. doi:10.1186/1471-2407-9-411 PubMedCentralCrossRefPubMed Crosswell HE, Dasgupta A, Alvarado CS, Watt T, Christensen JG, De P, Durden DL, Findley HW (2009) PHA665752, a small-molecule inhibitor of c-Met, inhibits hepatocyte growth factor-stimulated migration and proliferation of c-Met-positive neuroblastoma cells. BMC Cancer 9:411. doi:10.​1186/​1471-2407-9-411 PubMedCentralCrossRefPubMed
26.
go back to reference Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, Kim WJ, Okimoto RA, Bell DW, Sgroi DC, Christensen JG, Settleman J, Haber DA (2006) Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A 103(7):2316–2321. doi:10.1073/pnas.0508776103 PubMedCentralCrossRefPubMed Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, Kim WJ, Okimoto RA, Bell DW, Sgroi DC, Christensen JG, Settleman J, Haber DA (2006) Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A 103(7):2316–2321. doi:10.​1073/​pnas.​0508776103 PubMedCentralCrossRefPubMed
27.
go back to reference Jun HT, Sun J, Rex K, Radinsky R, Kendall R, Coxon A, Burgess TL (2007) AMG 102, a fully human anti-hepatocyte growth factor/scatter factor neutralizing antibody, enhances the efficacy of temozolomide or docetaxel in U-87 MG cells and xenografts. Clin Cancer Res 13(22 Pt 1):6735–6742. doi:10.1158/1078-0432.CCR-06-2969 CrossRefPubMed Jun HT, Sun J, Rex K, Radinsky R, Kendall R, Coxon A, Burgess TL (2007) AMG 102, a fully human anti-hepatocyte growth factor/scatter factor neutralizing antibody, enhances the efficacy of temozolomide or docetaxel in U-87 MG cells and xenografts. Clin Cancer Res 13(22 Pt 1):6735–6742. doi:10.​1158/​1078-0432.​CCR-06-2969 CrossRefPubMed
28.
go back to reference Guo A, Villen J, Kornhauser J, Lee KA, Stokes MP, Rikova K, Possemato A, Nardone J, Innocenti G, Wetzel R, Wang Y, MacNeill J, Mitchell J, Gygi SP, Rush J, Polakiewicz RD, Comb MJ (2008) Signaling networks assembled by oncogenic EGFR and c-Met. Proc Natl Acad Sci U S A 105(2):692–697. doi:10.1073/pnas.0707270105 PubMedCentralCrossRefPubMed Guo A, Villen J, Kornhauser J, Lee KA, Stokes MP, Rikova K, Possemato A, Nardone J, Innocenti G, Wetzel R, Wang Y, MacNeill J, Mitchell J, Gygi SP, Rush J, Polakiewicz RD, Comb MJ (2008) Signaling networks assembled by oncogenic EGFR and c-Met. Proc Natl Acad Sci U S A 105(2):692–697. doi:10.​1073/​pnas.​0707270105 PubMedCentralCrossRefPubMed
29.
go back to reference Jo M, Stolz DB, Esplen JE, Dorko K, Michalopoulos GK, Strom SC (2000) Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells. J Biol Chem 275(12):8806–8811CrossRefPubMed Jo M, Stolz DB, Esplen JE, Dorko K, Michalopoulos GK, Strom SC (2000) Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells. J Biol Chem 275(12):8806–8811CrossRefPubMed
30.
go back to reference Stommel JM, Kimmelman AC, Ying H, Nabioullin R, Ponugoti AH, Wiedemeyer R, Stegh AH, Bradner JE, Ligon KL, Brennan C, Chin L, DePinho RA (2007) Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 318(5848):287–290. doi:10.1126/science.1142946 CrossRefPubMed Stommel JM, Kimmelman AC, Ying H, Nabioullin R, Ponugoti AH, Wiedemeyer R, Stegh AH, Bradner JE, Ligon KL, Brennan C, Chin L, DePinho RA (2007) Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 318(5848):287–290. doi:10.​1126/​science.​1142946 CrossRefPubMed
31.
go back to reference Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, Chitale D, Motoi N, Szoke J, Broderick S, Balak M, Chang WC, Yu CJ, Gazdar A, Pass H, Rusch V, Gerald W, Huang SF, Yang PC, Miller V, Ladanyi M, Yang CH, Pao W (2007) MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A 104(52):20932–20937. doi:10.1073/pnas.0710370104 PubMedCentralCrossRefPubMed Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, Chitale D, Motoi N, Szoke J, Broderick S, Balak M, Chang WC, Yu CJ, Gazdar A, Pass H, Rusch V, Gerald W, Huang SF, Yang PC, Miller V, Ladanyi M, Yang CH, Pao W (2007) MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A 104(52):20932–20937. doi:10.​1073/​pnas.​0710370104 PubMedCentralCrossRefPubMed
32.
go back to reference Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, Kosaka T, Holmes AJ, Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson BE, Cantley LC, Janne PA (2007) MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316(5827):1039–1043. doi:10.1126/science.1141478 CrossRefPubMed Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, Kosaka T, Holmes AJ, Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson BE, Cantley LC, Janne PA (2007) MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316(5827):1039–1043. doi:10.​1126/​science.​1141478 CrossRefPubMed
Metadata
Title
NPS-1034, a novel MET inhibitor, inhibits the activated MET receptor and its constitutively active mutants
Authors
Jae-Sik Shin
Seung-Woo Hong
Jai-Hee Moon
Jin-Sun Kim
Kyung-Ah Jung
Seung-Mi Kim
Dae-Hee Lee
InKi Kim
Seon-Joo Yoon
Chang-Gyu Lee
Eun-Kyoung Choi
Joo-Young Lee
Kyu-pyo Kim
Yong Sang Hong
Jae-Lyun Lee
Bongcheol Kim
Eun Kyung Choi
Jung Shin Lee
Dong-Hoon Jin
Tae Won Kim
Publication date
01-06-2014
Publisher
Springer US
Published in
Investigational New Drugs / Issue 3/2014
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-013-0039-4

Other articles of this Issue 3/2014

Investigational New Drugs 3/2014 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