Discovery of a novel ALK/ROS1/FAK inhibitor, APG-2449, in preclinical non-small cell lung cancer and ovarian cancer models

Douglas D Fang, Ran Tao, Guangfeng Wang, Yuanbao Li, Kaixiang Zhang, Chunhua Xu, Guoqin Zhai, Qixin Wang, Jingwen Wang, Chunyang Tang, Ping Min, Dengkun Xiong, Jianyong Chen, Shaomeng Wang, Dajun Yang, Yifan Zhai, Douglas D Fang, Ran Tao, Guangfeng Wang, Yuanbao Li, Kaixiang Zhang, Chunhua Xu, Guoqin Zhai, Qixin Wang, Jingwen Wang, Chunyang Tang, Ping Min, Dengkun Xiong, Jianyong Chen, Shaomeng Wang, Dajun Yang, Yifan Zhai

Abstract

Background: Tyrosine kinase inhibitors (TKIs) are mainstays of cancer treatment. However, their clinical benefits are often constrained by acquired resistance. To overcome such outcomes, we have rationally engineered APG-2449 as a novel multikinase inhibitor that is highly potent against oncogenic alterations of anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 receptor tyrosine kinase (ROS1), and focal adhesion kinase (FAK). Here we present the preclinical evaluation of APG-2449, which exhibits antiproliferative activity in cells carrying ALK fusion or secondary mutations.

Methods: KINOMEscan® and LANCE TR-FRET were used to characterize targets and selectivity of APG-2449. Water-soluble tetrazolium salt (WST-8) viability assay and xenograft tumorigenicity were employed to evaluate therapeutic efficacy of monotherapy or drug combination in preclinical models of solid tumors. Western blot, pharmacokinetic, and flow cytometry analyses, as well as RNA sequencing were used to explore pharmacokinetic-pharmacodynamic correlations and the mechanism of actions driving drug combination synergy.

Results: In mice bearing wild-type or ALK/ROS1-mutant non-small-cell lung cancer (NSCLC), APG-2449 demonstrates potent antitumor activity, with correlations between pharmacokinetics and pharmacodynamics in vivo. Through FAK inhibition, APG-2449 sensitizes ovarian xenograft tumors to paclitaxel by reducing CD44+ and aldehyde dehydrogenase 1-positive (ALDH1+) cancer stem cell populations, including ovarian tumors insensitive to carboplatin. In epidermal growth factor receptor (EGFR)-mutated NSCLC xenograft models, APG-2449 enhances EGFR TKI-induced tumor growth inhibition, while the ternary combination of APG-2449 with EGFR (osimertinib) and mitogen-activated extracellular signal-regulated kinase (MEK; trametinib) inhibitors overcomes osimertinib resistance. Mechanistically, phosphorylation of ALK, ROS1, and FAK, as well as their downstream components, is effectively inhibited by APG-2449.

Conclusions: Taken together, our studies demonstrate that APG-2449 exerts potent and durable antitumor activity in human NSCLC and ovarian tumor models when administered alone or in combination with other therapies. A phase 1 clinical trial has been initiated to evaluate the safety and preliminary efficacy of APG-2449 in patients with advanced solid tumors, including ALK+ NSCLC refractory to earlier-generation ALK inhibitors.

Trial registration: Clinicaltrial.gov registration: NCT03917043 (date of first registration, 16/04/2019) and Chinese clinical trial registration: CTR20190468 (date of first registration, 09/04/2019).

Keywords: Anaplastic lymphoma kinase (ALK); Focal adhesion kinase (FAK); ROS proto-oncogene 1 receptor tyrosine kinase (ROS1); Solid tumors; Targeted therapies.

Conflict of interest statement

D. D. Fang, R. Tao, G. Wang, Y. Li, K. Zhang, C. Xu, G. Zhai, Q. Wang, J. Wang, C. Tang, P. Min, D. Xiong, J. Chen, D. Yang, and Y Zhai are full-time employees of Ascentage Pharma and equity shareholders of Ascentage Pharma Group International, the ultimate parent of Ascentage Pharma. S. Wang is a cofounder of Ascentage Pharma Group International, owns stock in the company, and receives grants and personal fees. He is a member of its board of directors, is its Chief Scientific Advisor, and is also a paid consultant. S. Wang and J. Chen hold an issued and licensed patent (US10709705B2) filed by the University of Michigan on APG-2449 and its analogs and receive royalties from the University of Michigan on this patent. The University of Michigan owns equity in, and has received research contracts from, affiliates of Ascentage Pharma for which S. Wang is the principal investigator. All other authors declare that they have no other competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
APG-2449 exhibits potent inhibitory activity against ALK/ROS1/FAK kinases and proliferation of relevant cell lines. A, Chemical structure of APG-2449. B, Dissociation constants (Kd values, nM) of APG-2449 calculated from KINOMEscan®. C, TREEspot interaction map of KINOMEscan selectivity panel for APG-2449 at 100 nM. Red spheres: kinases inhibited to < 35% of controls. Circle size reflects binding affinity. D, Inhibition of wild-type (wt) ALK and ALK mutants by APG-2449 and second-generation ALK inhibitors alectinib and ceritinib, using LANCE TR-FRET assay (IC50). E, Antiproliferative activity of APG-2449 (log IC50) in a panel of cancer cell lines. Data represent the mean of at least 2 independent experiments performed with triplicates. LANCE TR-FRET, lanthanide chelate excite time-resolved fluorescence resonance energy transfer
Fig. 2
Fig. 2
APG-2449 demonstrates antitumor activity and pharmacokinetic/pharmacodynamic correlation in ALK- or ROS1-positive xenografts in vivo. Antitumor activity of APG-2449 was evaluated in mice bearing subcutaneous tumors derived from H3122 NSCLC cells carrying EML4-ALK fusion gene (A; treated for 3 weeks, n = 7 per treatment group; T/C values were assessed on Day 21), KARPAS-299 cells carrying NPM-ALK fusion gene (B; treated for 3 weeks, n = 7 per treatment group; T/C values were assessed on Day 13), or Ba/F3 cells carrying CD74-ROS1 fusion gene (C; the control group was treated for 2 weeks and the other groups for 3 weeks, n = 5 per treatment group; T/C values were assessed on Day 15). D, Pharmacokinetics of APG-2449 in plasma (left panel) or tumors (right panel) of mice carrying KARPAS-299-derived CDX tissues (treated once, n = 3 per treatment group). E, Western blot analysis of ALK signaling pathway in tumors collected 24 hours after administration from the same experiment as shown in (D). Each lane represents a tumor collected from an individual animal. F, Western blot analysis of ROS1 signaling pathway in HCC78 cells carrying SLC34A2-ROS1 fusion following treatments in vitro. Dimethyl sulfoxide (DMSO) was included as a control. EML4, echinoderm microtubule-associated protein-like 4; NPM, nucleophosmin; SLC34A2, solute carrier family 34 member 2
Fig. 3
Fig. 3
Secondary ALK or ROS1 mutations conferring drug resistance are responsive to APG-2449 in vivo. Antitumor activity of APG-2449 against acquired, drug-resistant mutations was evaluated in subcutaneous xenograft models derived from Ba/F3 cells carrying EML4-ALKL1196M mutation (A; treated for 2 weeks, n = 6 per treatment group), Ba/F3 cells carrying EML4-ALKG1202R mutation (B; treated for 2 weeks, n = 5 per treatment group), Ba/F3 cells carrying CD74-ROS1L2026M mutation (C; treated for 3 weeks while 2 groups were terminated earlier as indicated, n = 4 or 5 per treatment group), NSCLC PDX LD1–0006-390,637 harboring ALKL1196M_G1202R double mutations (D; treated for 3 weeks, n = 5 per treatment group), or NSCLC PDX LU-01-0582R with acquired resistance to crizotinib (E; treated for 4 weeks, n = 3 or 4 per treatment group)
Fig. 4
Fig. 4
Combination of APG-2449 and paclitaxel inhibits tumor growth of FAK-expressing ovarian cancer xenografts in mice. Western blotting of FAK signaling pathway in PA-1 ovarian cells treated with APG-2449 or defactinib for 4 hours in vitro (A). Efficacy studies in subcutaneous CDX models derived from PA-1 (B; treated for 3 weeks, n = 5 per treatment group) or OVCAR-3 (C; treated for 4 weeks, n = 5 per treatment group; T/C values assessed on Day 28) ovarian cancer cells. D, A mouse trial experiment was conducted in a panel of 6 ovarian cancer PDX models (treated for 3–6 weeks, n = 2 per treatment group). E, Heatmap of genes significantly changed in ovarian PDX tumors that responded to APG-2449 plus paclitaxel combination (synergy ratio > 2) versus nonresponders (synergy ratio < 2). p < 0.05 (false discovery rate < 0.05). F, Western blot analysis of FAK signaling pathways in tumor samples responsive (OV2018) and nonresponsive (OV1396) to APG-2449 plus paclitaxel treatment; tumor samples collected 4 hours after the last administration from the experimental animals shown in panel D. G, IHC staining of CD44- or ALDH1A1-positive cells in OVCAR-3 tumors collected 4 hours after the last treatment from tumor-bearing mice treated with APG-2449, paclitaxel or the combination for 10 days. H, SKOV-3 cells were treated with increasing doses of APG-2449 for 72 hours. I, SKOV-3 cells were treated with APG-2449 (1 μM), paclitaxel (5 nM), or the combination for 72 hours. CD44+ and ALDH1A+ cells were determined by flow cytometry. ***p < 0.001, **p < 0.01 vs. DMSO controls. Date presented are representative of 2 independent experiments as the mean ± SEM of triplicate biological replicates
Fig. 5
Fig. 5
APG-2449 synergizes with EGFR inhibitors and overcomes osimertinib-resistance when combined with osimertinib/trametinib in xenograft models. Combination treatments with APG-2449 and EGFR inhibitors were evaluated in an HCC827 CDX model (A; treated for 3 weeks, n = 5 per treatment group), NCI-H1975 CDX model (B; treated for 3 weeks, n = 5 per treatment group), and NSCLC PDX model LD1–0006-215,676 harboring EGFRL858R_T790M and ROS1 fusion (C; treated for 61 days, n = 6 per treatment group) in mice. D, Tumor growth curve of NCI-H1975 CDX model rechallenged with APG-2449 plus osimertinib (Day 47- Day 67) upon disease progression from 21-day osimertinib treatment (Day 1-Day 21). E, Western blot analysis of EGFR and FAK downstream signaling in NCI-H1975 xenografts collected 4 hours after the last administration from tumor-bearing mice treated with indicated compounds for 2 weeks. Each lane represents an individual animal. F, Cell viability (IC50) of PC-9 (parental) or PC-9/OR (osimertinib-resistant) cells treated with osimertinib at indicated concentrations for 3 days. Data are representative of 3 independent experiments and shown as the mean ± SEM of triplicates. ****p < 0.0001. G, Cell viability of PC-9/OR cells treated with indicated compounds for 72 hours. H, PC-9/OR xenograft models were treated with the indicated compounds for 3 weeks (n = 5 per treatment group)
Fig. 6
Fig. 6
Scheme for the mechanisms of action of APG-2449. Created with BioRender (Toronto, Ontario, Canada). CSC, cancer stem cell; EML, echinoderm microtubule-associated protein-like; MEK-ERK, mitogen-activated protein kinase kinase-extracellular signal-regulated kinase; PI3K-AKT, phosphatidylinositol 3-kinase and protein kinase B; SHP, SH2 containing protein tyrosine phosphatase; STAT, signal transducer and activator of transcription

References

    1. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 2018;15:81–94. doi: 10.1038/nrclinonc.2017.166.
    1. Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, et al. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13:1011–1019. doi: 10.1016/S1470-2045(12)70344-3.
    1. Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693–1703. doi: 10.1056/NEJMoa1006448.
    1. Luo QY, Zhou SN, Pan WT, Sun J, Yang LQ, Zhang L, et al. A multi-kinase inhibitor APG-2449 enhances the antitumor effect of ibrutinib in esophageal squamous cell carcinoma via EGFR/FAK pathway inhibition. Biochem Pharmacol. 2021;183:114318. doi: 10.1016/j.bcp.2020.114318.
    1. Bergethon K, Shaw AT, Ou SH, Katayama R, Lovly CM, McDonald NT, et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol. 2012;30:863–870. doi: 10.1200/JCO.2011.35.6345.
    1. Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci. 2003;116:1409–1416. doi: 10.1242/jcs.00373.
    1. Schaller MD. Cellular functions of FAK kinases: insight into molecular mechanisms and novel functions. J Cell Sci. 2010;123:1007–1013. doi: 10.1242/jcs.045112.
    1. Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: mechanistic findings and clinical applications. Nat Rev Cancer. 2014;14:598–610. doi: 10.1038/nrc3792.
    1. Zhou J, Yi Q, Tang L. The roles of nuclear focal adhesion kinase (FAK) on Cancer: a focused review. J Exp Clin Cancer Res. 2019;38:250. doi: 10.1186/s13046-019-1265-1.
    1. Levy A, Alhazzani K, Dondapati P, Alaseem A, Cheema K, Thallapureddy K, et al. Focal adhesion kinase in ovarian cancer: a potential therapeutic target for platinum and taxane-resistant tumors. Curr Cancer Drug Targets. 2019;19:179–188. doi: 10.2174/1568009618666180706165222.
    1. Mohanty A, Pharaon RR, Nam A, Salgia S, Kulkarni P, Massarelli E. FAK-targeted and combination therapies for the treatment of cancer: an overview of phase I and II clinical trials. Expert Opin Investig Drugs. 2020;29:399–409. doi: 10.1080/13543784.2020.1740680.
    1. Kolev VN, Tam WF, Wright QG, McDermott SP, Vidal CM, Shapiro IM, et al. Inhibition of FAK kinase activity preferentially targets cancer stem cells. Oncotarget. 2017;8:51733–51747. doi: 10.18632/oncotarget.18517.
    1. Roberts WG, Ung E, Whalen P, Cooper B, Hulford C, Autry C, et al. Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Cancer Res. 2008;68:1935–1944. doi: 10.1158/0008-5472.CAN-07-5155.
    1. Patel MR, Infante JR, Moore KN, Keegan M, Poli A, Padval M, et al. Phase 1/1b study of the FAK inhibitor defactinib (VS-6063) in combination with weekly paclitaxel for advanced ovarian cancer. J Clin Oncol. 2014;32:5521. doi: 10.1200/jco.2014.32.15_suppl.5521.
    1. Laszlo V, Valko Z, Ozsvar J, Kovacs I, Garay T, Hoda MA, et al. The FAK inhibitor BI 853520 inhibits spheroid formation and orthotopic tumor growth in malignant pleural mesothelioma. J Mol Med (Berl) 2019;97:231–242. doi: 10.1007/s00109-018-1725-7.
    1. Zhang J, He DH, Zajac-Kaye M, Hochwald SN. A small molecule FAK kinase inhibitor, GSK2256098, inhibits growth and survival of pancreatic ductal adenocarcinoma cells. Cell Cycle. 2014;13:3143–3149. doi: 10.4161/15384101.2014.949550.
    1. Jones SF, Siu LL, Bendell JC, Cleary JM, Razak AR, Infante JR, et al. A phase I study of VS-6063, a second-generation focal adhesion kinase inhibitor, in patients with advanced solid tumors. Investig New Drugs. 2015;33:1100–1107. doi: 10.1007/s10637-015-0282-y.
    1. Infante JR, Camidge DR, Mileshkin LR, Chen EX, Hicks RJ, Rischin D, et al. Safety, pharmacokinetic, and pharmacodynamic phase I dose-escalation trial of PF-00562271, an inhibitor of focal adhesion kinase, in advanced solid tumors. J Clin Oncol. 2012;30:1527–1533. doi: 10.1200/JCO.2011.38.9346.
    1. Gerber DE, Camidge DR, Morgensztern D, Cetnar J, Kelly RJ, Ramalingam SS, et al. Phase 2 study of the focal adhesion kinase inhibitor defactinib (VS-6063) in previously treated advanced KRAS mutant non-small cell lung cancer. Lung Cancer. 2020;139:60–67. doi: 10.1016/j.lungcan.2019.10.033.
    1. Ichihara E, Westover D, Meador CB, Yan Y, Bauer JA, Lu P, et al. SFK/FAK signaling attenuates osimertinib efficacy in both drug-sensitive and drug-resistant models of EGFR-mutant lung cancer. Cancer Res. 2017;77:2990–3000. doi: 10.1158/0008-5472.CAN-16-2300.
    1. Murakami Y, Sonoda K, Abe H, Watari K, Kusakabe D, Azuma K, et al. The activation of SRC family kinases and focal adhesion kinase with the loss of the amplified, mutated EGFR gene contributes to the resistance to afatinib, erlotinib and osimertinib in human lung cancer cells. Oncotarget. 2017;8:70736–70751. doi: 10.18632/oncotarget.19982.
    1. Fang DD, Tang Q, Kong Y, Rong T, Wang Q, Li N, et al. MDM2 inhibitor APG-115 exerts potent antitumor activity and synergizes with standard-of-care agents in preclinical acute myeloid leukemia models. Cell Death Discov. 2021;7:90. doi: 10.1038/s41420-021-00465-5.
    1. Fang DD, Tang Q, Kong Y, Wang Q, Gu J, Fang X, et al. MDM2 inhibitor APG-115 synergizes with PD-1 blockade through enhancing antitumor immunity in the tumor microenvironment. J Immunother Cancer. 2019;7:327. doi: 10.1186/s40425-019-0750-6.
    1. Chen J, Zhou Y, Dong X, Liu L, Bai L, McEachern D, et al. Discovery of CJ-2360 as a potent and orally active inhibitor of anaplastic lymphoma kinase capable of achieving complete tumor regression. J Med Chem. 2020;63:13994–14016. doi: 10.1021/acs.jmedchem.0c01550.
    1. Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21:1318–1325. doi: 10.1038/nm.3954.
    1. Mayer B, Muche R. Formal sample size calculation and its limited validity in animal studies of medical basic research. Tierarztl Prax Ausg K Kleintiere Heimtiere. 2013;41:367–374. doi: 10.1055/s-0038-1623732.
    1. Gainor JF, Dardaei L, Yoda S, Friboulet L, Leshchiner I, Katayama R, et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov. 2016;6:1118–1133. doi: 10.1158/-16-0596.
    1. Lin JJ, Riely GJ, Shaw AT. Targeting ALK: precision medicine takes on drug resistance. Cancer Discov. 2017;7:137–155. doi: 10.1158/-16-1123.
    1. Stone RL, Baggerly KA, Armaiz-Pena GN, Kang Y, Sanguino AM, Thanapprapasr D, et al. Focal adhesion kinase: an alternative focus for anti-angiogenesis therapy in ovarian cancer. Cancer Biol Ther. 2014;15:919–929. doi: 10.4161/cbt.28882.
    1. Miyazaki T, Kato H, Nakajima M, Sohda M, Fukai Y, Masuda N, et al. FAK overexpression is correlated with tumour invasiveness and lymph node metastasis in oesophageal squamous cell carcinoma. Br J Cancer. 2003;89:140–145. doi: 10.1038/sj.bjc.6601050.
    1. Nallanthighal S, Heiserman JP, Cheon DJ. The role of the extracellular matrix in cancer stemness. Front Cell Dev Biol. 2019;7:86. doi: 10.3389/fcell.2019.00086.
    1. Amelio I, Cutruzzola F, Antonov A, Agostini M, Melino G. Serine and glycine metabolism in cancer. Trends Biochem Sci. 2014;39:191–198. doi: 10.1016/j.tibs.2014.02.004.
    1. Bartakova A, Michalova K, Presl J, Vlasak P, Kostun J, Bouda J. CD44 as a cancer stem cell marker and its prognostic value in patients with ovarian carcinoma. J Obstet Gynaecol. 2018;38:110–114. doi: 10.1080/01443615.2017.1336753.
    1. Kang Y, Hu W, Ivan C, Dalton HJ, Miyake T, Pecot CV, et al. Role of focal adhesion kinase in regulating YB-1-mediated paclitaxel resistance in ovarian cancer. J Natl Cancer Inst. 2013;105:1485–1495. doi: 10.1093/jnci/djt210.
    1. Diaz Osterman CJ, Ozmadenci D, Kleinschmidt EG, Taylor KN, Barrie AM, Jiang S, et al. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy. Elife. 2019;8:e47327. doi: 10.7554/eLife.47327.
    1. Fan PD, Narzisi G, Jayaprakash AD, Venturini E, Robine N, Smibert P, et al. YES1 amplification is a mechanism of acquired resistance to EGFR inhibitors identified by transposon mutagenesis and clinical genomics. Proc Natl Acad Sci U S A. 2018;115:E6030–E60E8.
    1. Rosenzweig SA. Acquired resistance to drugs targeting tyrosine kinases. Adv Cancer Res. 2018;138:71–98. doi: 10.1016/bs.acr.2018.02.003.
    1. Murphy JM, Rodriguez YAR, Jeong K, Ahn EE, Lim SS. Targeting focal adhesion kinase in cancer cells and the tumor microenvironment. Exp Mol Med. 2020;52:877–886. doi: 10.1038/s12276-020-0447-4.
    1. Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011;17:313–319. doi: 10.1038/nm.2304.
    1. Zong X, Nephew KP. Ovarian cancer stem cells: role in metastasis and opportunity for therapeutic targeting. Cancers (Basel) 2019;11:934. doi: 10.3390/cancers11070934.
    1. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 2005;65:9328–9337. doi: 10.1158/0008-5472.CAN-05-1343.
    1. Steg AD, Bevis KS, Katre AA, Ziebarth A, Dobbin ZC, Alvarez RD, et al. Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin Cancer Res. 2012;18:869–881. doi: 10.1158/1078-0432.CCR-11-2188.
    1. Van Zyl B, Tang D, Bowden NA. Biomarkers of platinum resistance in ovarian cancer: what can we use to improve treatment. Endocr Relat Cancer. 2018;25:R303–RR18. doi: 10.1530/ERC-17-0336.
    1. Taylor KN, Schlaepfer DD. Adaptive resistance to chemotherapy, a multi-FAK-torial linkage. Mol Cancer Ther. 2018;17:719–723. doi: 10.1158/1535-7163.MCT-17-1177.
    1. Parte SC, Batra SK, Kakar SS. Characterization of stem cell and cancer stem cell populations in ovary and ovarian tumors. J Ovarian Res. 2018;11:69. doi: 10.1186/s13048-018-0439-3.
    1. Tong X, Tanino R, Sun R, Tsubata Y, Okimoto T, Takechi M, et al. Protein tyrosine kinase 2: a novel therapeutic target to overcome acquired EGFR-TKI resistance in non-small cell lung cancer. Respir Res. 2019;20:270. doi: 10.1186/s12931-019-1244-2.
    1. Ercan D, Xu C, Yanagita M, Monast CS, Pratilas CA, Montero J, et al. Reactivation of ERK signaling causes resistance to EGFR kinase inhibitors. Cancer Discov. 2012;2:934–947. doi: 10.1158/-12-0103.
    1. Tricker EM, Xu C, Uddin S, Capelletti M, Ercan D, Ogino A, et al. Combined EGFR/MEK inhibition prevents the emergence of resistance in EGFR-mutant lung cancer. Cancer Discov. 2015;5:960–971. doi: 10.1158/-15-0063.

Source: PubMed

3
Abonnere