MET-targeting antibody (emibetuzumab) and kinase inhibitor (merestinib) as single agent or in combination in a cancer model bearing MET exon 14 skipping

S Betty Yan, Suzane L Um, Victoria L Peek, Jennifer R Stephens, Wei Zeng, Bruce W Konicek, Ling Liu, Jason R Manro, Volker Wacheck, Richard A Walgren, S Betty Yan, Suzane L Um, Victoria L Peek, Jennifer R Stephens, Wei Zeng, Bruce W Konicek, Ling Liu, Jason R Manro, Volker Wacheck, Richard A Walgren

Abstract

Purpose Approximately 3% of lung cancer bears mutations leading to MET exon 14 skipping, an oncogenic driver which is further evidenced by case reports of patient response to MET kinase inhibitor treatment. Approximately 15% of tumors harboring MET exon14 skipping have concurrent MET amplification. Experimental Design Merestinib is a type II MET kinase inhibitor. Emibetuzumab, a bivalent anti-MET antibody, internalizes MET receptor. Each single agent and the combination were evaluated in the Hs746t gastric cancer line bearing MET exon14 skipping and MET amplification. Results Merestinib inhibited Hs746t cell proliferation (IC50=34 nM) and totally eliminated pMET at 100nM. Emibetuzumab showed little anti-proliferative activity against Hs746t cells (IC50>100nM), did not reduce pMET, and slightly reduced cell surface MET. In the Hs746t xenograft model, dose dependent differences in durability of response were seen with merestinib including durable tumor regression (91.8%) at 12 mg/kg qd. Emibetuzumab treatment (10mg/kg qw) provided transient tumor regression (37.7%), but tumors re-grew while on treatment. Concurrent combination of merestinib (6 mg/kg qd) and emibetuzumab resulted in 85% tumor regression, while a sequential combination (initiating merestinib first) resulted in longer duration of treatment response. Conclusions Data in this study support a clinical evaluation of merestinib in patients with MET exon 14 skipping (NCT02920996). As a type II MET kinase inhibitor, merestinib may provide a therapeutic option to treatment naïve patients or to patients who progress on type I MET inhibitor treatment. Data also support clinical evaluation of the sequential combination of merestinib with emibetuzumab when patients progress on single agent merestinib.

Keywords: Emibetuzumab; LY2801653; LY2875358; MET antibody; MET exon 14 skipping; MET kinase inhibitor; Merestinib.

Conflict of interest statement

Conflict of interest

All authors are full time employees of Eli Lilly and Company.

Ethical approval

All animal studies were performed in accordance with the International Association for Assessment and Accreditation of Laboratory Animal Care institutional guidelines. All in vivo experimental protocols were approved by the Eli Lilly and Company Animal Care and Use Committee.

This article does not contain any studies with human participants performed by any of the authors.

Figures

Fig. 1
Fig. 1
In vitro effect of MET inhibitors on expression of total MET, pMET (Y1234/1235), and pMET (Y1003). a Western blot analysis of Hs746t (exon 14 skipping and MET amplification) and MKN45 (MET amplification) cells treated for 24 h with merestinib, PF04217903, or emibetuzumab at indicated concentrations. Blots were probed with antibodies to total MET, phospho-MET (Y1234/1235), and phospho-MET (Y1003). b Western blot analysis of Hs746t cells treated for 24 h with single agent MET inhibitors (emibetuzumab, merestinib, or PF04217903) or hIgG4 at the indicated concentrations or combination of emibetuzumab with either merestinib or PF04217903. Blots were probed with antibodies to total MET, and phospho-MET (Y1003)
Fig. 2
Fig. 2
MET kinase inhibitors but not emibetuzumab inhibited MET signaling pathway in Hs746t cells in vitro by western blot analysis. Hs746t cells were treated for 24 h with 133 nM (20 μg/ml) or 667 nM (100 μg/ml) emibetuzumab or MET kinase inhibitors (merestinib, PF04217903, crizotinib, cabozantinib) at 15.6, 62.5, 250 or 1000 nM
Fig. 3
Fig. 3
In vivo single agent anti-tumor effect of merestinib, emibetuzumab, and crizotinib in the Hs746t xenograft model. Approximately 5 million cells were implanted in female athymic nude mice (n = 7 per group). Vehicle (), merestinib at 6 mg/kg (suboptimal dose – insufficient target coverage for 24 h) () or 12 mg/kg (optimal dose) () was dosed once daily orally, emibetuzumab at 10 mg/kg () was dosed once weekly intraperitoneally (IP), or crizotinib at 25 mg/kg () was dosed once daily orally. Dosing of inhibitors began on day 11, when tumors were of average size of 150 mm3, and continued for 21 days. The waterfall plot shows the response of individual animals to treatment on day 32. Missing bars are due to animals removed early from the study because of tumor burden. In the waterfall plot, the three horizontal dotted lines represent the three cut points for: Progressive Disease as ∆T/C > 10%; Stable Disease as ∆T/C ≤ 10% to regression <−50%; Partial Response as regression ≥ − 50% and tumor volume > 14 mm3; Complete Response as tumor volume ≤ 14 mm3. Statistical analysis comparing tumor volumes of treatment groups to vehicle control group () was performed on Day 32, where at least half of the animals (4/7) in the vehicle group still remained on study. Dosing for merestinib (12 mg/kg) and crizotinib continued to Day 64 and tumor growth monitored for another 5 weeks post-treatment (data not shown)
Fig. 4
Fig. 4
In vivo anti-tumor effect of the combination of merestinib and emibetuzumab in the Hs746t xenograft model. a Approximately 5 million cells were implanted in female athymic nude mice (n = 7 per group). Dosing of inhibitors began on day 14, when tumors reached average size 350 mm3, and continued for 28 days. As single agent, merestinib at 6 mg/kg (suboptimal dose – insufficient target coverage for 24 h) () was dosed once daily orally, and emibetuzumab at 10 mg/kg () was dosed once weekly intraperitoneally (IP). In combination, emibetuzumab at 10 mg/kg was dosed once weekly intraperitoneally (IP) concurrently with either merestinib at 6 mg/kg (suboptimal dose) () or merestinib at 12 mg/kg (optimal dose) () once daily orally. The waterfall plot shows the response of individual animals to treatment on day 27, when tumor volume analysis in comparison to the vehicle control group was performed. Missing bars are due to animals removed early from the study because of tumor burden. In the waterfall plot, the three horizontal dotted lines represent the three cut points for: Progressive Disease as ∆T/C > 10%; Stable Disease as ∆T/C ≤ 10% to regression <−50%; Partial Response as regression ≥ − 50% and tumor volume > 14 mm3; Complete Response as tumor volume ≤ 14 mm3. b Comparison of sequential and concurrent combination of merestinib (6 mg/kg once daily) with emibetuzumab (10 mg/kg once weekly). Approximately 5 million cells were implanted subcutaneously into the hind flank of female athymic nude mice (6 per group). Compound dosing began on day 21 when tumors reached an average size of 300–400 mm3. In the concurrent combination (), emibetuzumab was dosed for 7 cycles, with merestinib given for 50 days (dosing period is shown with the blue color double-arrowed horizontal bar). In the sequential combination () (dosing period and the sequence of the combination are shown with the red color double-arrowed horizontal bar), merestinib treatment was initially started as a single agent (Day 21), and upon tumor regression and regrowth to tumor size of 500 mm3, emibetuzumab was then added on for 6 cycles starting on Day 37. The last day of dosing was Day 70 and Day 78 for concurrent and sequential treatment group, respectively, and tumor growth was monitored post-treatment

References

    1. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–925. doi: 10.1038/nrm1261.
    1. Saito M, Shiraishi K, Kunitoh H, Takenoshita S, Yokota J, Kohno T. Gene aberrations for precision medicine against lung adenocarcinoma. Cancer Sci. 2016;107(6):713–720. doi: 10.1111/cas.12941.
    1. Li S, Choi YL, Gong Z, Liu X, Lira M, Kan Z, Oh E, Wang J, Ting JC, Ye X, Reinhart C, Liu X, Pei Y, Zhou W, Chen R, Fu S, et al. Comprehensive characterization of oncogenic drivers in Asian lung adenocarcinoma. J Thorac Oncol. 2016;11(12):2129–2140. doi: 10.1016/j.jtho.2016.08.142.
    1. Lee J, SHI O, Lee JM, Kim HC, Hong M, Kim SY, Jang J, Ahn S, Kang SY, Lee S, Kim ST, Kim B, Choi J, Kim KA, Lee J, Park C, et al. Gastrointestinal malignancies harbor actionable MET exon 14 deletions. Oncotarget. 2015;6(29):28211–28222.
    1. Onozato R, Kosaka T, Kuwano H, Sekido Y, Yatabe Y, Mitsudomi T. Activation of MET by gene amplification or by splice mutations deleting the juxtamembrane domain in primary resected lung cancers. J thoracic. Oncologia. 2009;4(1):5–11.
    1. Gow CH, Hsieh MS, SG W, Shih JY. A comprehensive analysis of clinical outcomes in lung cancer patients harboring a MET exon 14 skipping mutation compared to other driver mutations in an East Asian population. Lung Cancer. 2017;103:82–89. doi: 10.1016/j.lungcan.2016.12.001.
    1. Tong JHM, Yeung SF, Chan AWH, Chung LY, Chau SL, Lung RWM, Tong CYK, Chow C, Tin EKY, Yu YH, Li H, Pan Y, Chak RWP, Ng CSH, Mok TSK, To KF MET amplification and exon 14 splice site mutation define unique molecular subgroups of non-small lung carcinoma with poor prognosis. Clin Cancer Res. 2016;22(12):3048–3056. doi: 10.1158/1078-0432.CCR-15-2061.
    1. Schrock AB, Frampton GM, Suh J, Chalmers ZR, Rosenzwig M, Erlich RL, Halmos B, Goldman J, Forde P, Leuenberger K, Peled N, Kalemkerian GP, Ross JS, Stephens PJ, Miller VA, Ali SM, et al. Characterization of 298 lung cancer patients harboring MET exon 14 skipping (METex14) alterations. J Thorac Oncol. 2016;11(9):1493–1502. doi: 10.1016/j.jtho.2016.06.004.
    1. Awad MM, Oxnard GR, Jackman DM, Savukoski DO, Hall D, Shivdasani P, Heng JC, Dahlberg SE, Janne PA, Verma S, Christensen J, Hammerman PS, Sholl LM. MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression. J Clin Oncol. 2016;34(7):721–730. doi: 10.1200/JCO.2015.63.4600.
    1. Frampton GM, Ali SM, Rosenzweig M, Chmielecki J, Lu X, Bauer TM, Akimov M, Bufill J, Lee C, Jentz D, Hoover R, Ou ISH, Salgia R, Brennan T, Chalmers Z, Elvin JA, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015;5(8):850–859. doi: 10.1158/-15-0285.
    1. Van der Steen B, Giovannetti E, Pauwels P, Peters GJ, Hong DS, Cappuzzo F, Hirsch FR, Rolfo C. cMET exon 14 skipping: from the structure to the clinic. J Thorac Oncol. 2016;11(9):1423–1432. doi: 10.1016/j.jtho.2016.05.005.
    1. Heist RS, Shim HS, Gingipally S, Mino-Kenudson M, Le L, Gainor JF, Zheng Z, Aryee M, Xia J, Jia P, Jin H, Zhao Z, Pao W, Engelman JA, Iafrate AJ. MET exon 14 skipping in non-small cell lung cancer. Oncologist. 2016;21:481–486. doi: 10.1634/theoncologist.2015-0510.
    1. Shum E, Halmos B. Skipping the line: bringing MET exon 14 skipping mutations to the forefront of targeted therapy. Transl Cancer Res. 2016;5(2):110–116. doi: 10.21037/tcr.2016.03.12.
    1. Yan SB, Peek VL, Ajamie R, Buchanan SG, Graff JR, Heidler SA, Hui YH, Huss KL, Konicek BW, Manro JR, Shih C, Stewart JA, Stewart TR, Stout SL, Uhlik MT, Um SL, et al. LY2801653 is an orally bioavailable multi-kinase inhibitor with potent activity against MET, MST1R, and other oncoproteins, and displays anti-tumor activities in mouse xenograft models. Investig New Drugs. 2013;31(4):833–844. doi: 10.1007/s10637-012-9912-9.
    1. Wu W, Bi C, Credille KM, Manro JR, Peek VL, Donoho GP, Yan L, Wijsman JA, Yan SB, Walgren RA. Inhibition of tumor growth and metastasis in non-small cell lung cancer by LY2801653, an inhibitor of several oncokinases, including MET. Clin Cancer Res. 2013;19(20):5699–5710. doi: 10.1158/1078-0432.CCR-13-1758.
    1. Liu L, Zeng W, Wortinger MA, Yan SB, Cornwell P, Peek VL, Stephens JR, Tetreault JW, Xia J, Manro JR, Credille KM, Ballard DW, Brown-Augsburger P, Wacheck V, Chow CK, Huang L, et al. LY2875358, a neutralizing and internalizing anti-MET bivalent antibody, inhibits HGF-dependent and HGF-independent MET activation and tumor growth. Clin Cancer Res. 2014;20(23):6059–6070. doi: 10.1158/1078-0432.CCR-14-0543.
    1. Asaoka Y, Tada M, Ikenoue T, Seto M, Imai M, Miyabayashi K, Yamamoto K, Yamamoto S, Kudo Y, Mohri D, Isomura Y, Ijichi H, Tateishi K, Kanai F, Ogawa S, Omata M, et al. Gastric cancer cell line Hs746T harbors a splice site mutation of c-Met causing juxtamembrane domain deletion. Biochem Biophys Res Commun. 2010;394:1042–1046. doi: 10.1016/j.bbrc.2010.03.120.
    1. Egile C, Kenigsberg M, Delaisi C, Begassat F, Do-Vale V, Mestadier J, Bonche F, Benard T, Nicolas JP, Valence S, Lefranc C, Francesconi E, Castell C, Lefebvre AM, Nemecek C, Calvet L, et al. The selective intravenous inhibitor of the MET tyrosine kinase SAR125844 inhibits tumor growth in MET-amplified cancer. Mol Cancer Ther. 2015;14(2):384–394. doi: 10.1158/1535-7163.MCT-14-0428.
    1. Zhang Z, Wang J, Ji D, Wang C, Liu R, Wu Z, Liu L, Zhu D, Chang J, Geng R, Xiong L, Fang Q, Li J. Functional genetic approach identifies MET, HER3, IGF1R, INSR pathways as determinants of lapatinib unresponsiveness in HER2-positive gastric cancer. Clin Cancer Res. 2014;20(17):4559–4573. doi: 10.1158/1078-0432.CCR-13-3396.
    1. Siddiqui N, Sonenberg N. Signalling to eIF4E in cancer. Biochem Soc Trans. 2015;43:763–772. doi: 10.1042/BST20150126.
    1. Wang J, Goetsch L, Tucker L, Zhang Q, Gonzalez A, Vaidya KS, Oleksijew A, Boghaert ER, Song M, Sokolova I, Pestova E, Anderson M, Pappano WN, Ansell P, Bhathena A, Naumovski L, et al. Anti-c-Met monoclonal antibody ABT-700 breaks oncogene addiction in tumors with MET amplification. BMC Cancer. 2016;16:105. doi: 10.1186/s12885-016-2138-z.
    1. Wang J, Anderson MG, Oleksijew A, Vaidya KS, Boghaert ER, Tucker L, Zhang Q, Han EK, Palma JP, Naumovski L, Beilly EB. ABBV-399, a c-Met antibody drug conjugate that targets both MET amplified and c-Met overexpressing tumors, irrespective of MET pathway dependence. Clin Cancer Res. 2016;23(4):992–1000. doi: 10.1158/1078-0432.CCR-16-1568.
    1. Lee JM, Kim B, Lee SB, Jeong Y, YM O, Song YJ, Jung S, Choi J, Lee S, Cheong KH, Kim DU, Park HW, Han YK, Kim GW, Choi H, Song PH, et al. Cbl-independent degradation of MET: ways to avoid agonism of bivalent MEt-targeting antibody. Oncogene. 2014;33:34–43. doi: 10.1038/onc.2012.551.
    1. Pacchiana G, Chiriaco C, Stella MC, Petronzelli F, De Santis R, Galluzzo M, Carminati P, Comoglio PM, Michieli P, Vigna E. Monovalency unleashes the full therapeutic potential of the DN-30 anti-Met antibody. J Biol Chem. 2010;285(46):36149–36157. doi: 10.1074/jbc.M110.134031.
    1. Poulsen TT, Grandal MM, Skartved NJO, Hald R, Alifrangis L, Koefoed K, Lindsted T, Frohlich C, Pollmann SE, Eriksen KW, Dahlman A, Jacobsen HJ, Bouquin T, Pedersen MW, Horak ID, Lantto J, et al. Sym015: a highly efficacious antibody mixture against MET-amplified tumors. Clin Cancer Res. 2017;23(19):5923–5935. doi: 10.1158/1078-0432.CCR-17-0782.
    1. Heist RS, Sequist LV, Borger D, Gainor JF, Arellano RS, Le LP, Dias-Santagata D, Clark JW, Engelman JA, Shaw AT, Iafrate AJ. Acquired resistance to crizotinib in non-small cell lung cancer with MET exon 14 skipping. J Thorac Oncol. 2016;11(8):1242–1245. doi: 10.1016/j.jtho.2016.06.013.
    1. Ou SHI, Young L, Schrock AB, Johnson A, Klempner SJ, Zhu VW, Miller VA, Ali SM. Emergence of pre-existing MET Y1230C mutation as a resistance mechanism to crizotinib in NSCLC with MET exon 14 skipping. J Thorac Oncol. 2017;12(1):137–140. doi: 10.1016/j.jtho.2016.09.119.
    1. Bahcall M, Sim T, Paweletz CP, Patel JD, Alden RS, Kuang Y, Sacher AG, Kim ND, Lydon CA, Awad MM, Jaklitsch MT, Sholl LM, Janne PA, Oxnard GR. Acquired METD1228V mutation and resistance to MET inhibition in lung cancer. Cancer Discov. 2016;6(12):1334–1341. doi: 10.1158/-16-0686.
    1. Dong HJ, Li P, CL W, Zhou XY, HJ L, Zhou T. Response and acquired resistance to crizotinib in Chinese patientswith lung adenocarcinomas harboring MET Exon 14 splicing alternations. Lung Cancer. 2016;102:118–121. doi: 10.1016/j.lungcan.2016.11.006.
    1. Lu X, Peled N, Greer J, Wu W, Choi P, Berger AH, Wong S, Jen KY, Seo Y, Hann B, Brooks A, Meyerson M, Collisson EA. MET exon 14 mutation encodes an actionable therapeutic target in lung adenocarcinoma. Cancer Res. 2017;77(16):4498–4505. doi: 10.1158/0008-5472.CAN-16-1944.

Source: PubMed

3
구독하다