Targeting HER2 Exon 20 Insertion-Mutant Lung Adenocarcinoma with a Novel Tyrosine Kinase Inhibitor Mobocertinib

Han Han, Shuai Li, Ting Chen, Michael Fitzgerald, Shengwu Liu, Chengwei Peng, Kwan Ho Tang, Shougen Cao, Johara Chouitar, Jiansheng Wu, David Peng, Jiehui Deng, Zhendong Gao, Theresa E Baker, Fei Li, Hua Zhang, Yuanwang Pan, Hailin Ding, Hai Hu, Val Pyon, Cassandra Thakurdin, Eleni Papadopoulos, Sittinon Tang, Francois Gonzalvez, Haiquan Chen, Victor M Rivera, Rachael Brake, Sylvie Vincent, Kwok-Kin Wong, Han Han, Shuai Li, Ting Chen, Michael Fitzgerald, Shengwu Liu, Chengwei Peng, Kwan Ho Tang, Shougen Cao, Johara Chouitar, Jiansheng Wu, David Peng, Jiehui Deng, Zhendong Gao, Theresa E Baker, Fei Li, Hua Zhang, Yuanwang Pan, Hailin Ding, Hai Hu, Val Pyon, Cassandra Thakurdin, Eleni Papadopoulos, Sittinon Tang, Francois Gonzalvez, Haiquan Chen, Victor M Rivera, Rachael Brake, Sylvie Vincent, Kwok-Kin Wong

Abstract

No targeted treatments are currently approved for HER2 exon 20 insertion-mutant lung adenocarcinoma patients. Mobocertinib (TAK-788) is a potent irreversible tyrosine kinase inhibitor (TKI) designed to target human epidermal growth factor receptor 2 (HER2/ERBB2) exon 20 insertion mutations. However, the function of mobocertinib on HER2 exon 20 insertion-mutant lung cancer is still unclear. Here we conducted systematic characterization of preclinical models to understand the activity profile of mobocertinib against HER2 exon 20 insertions. In HER2 exon 20 insertion-mutant cell lines, the IC50 of mobocertinib was higher than poziotinib and comparable with or slightly lower than afatinib, neratinib, and pyrotinib. Mobocertinib had the lowest HER2 exon 20 insertion IC50/wild-type (WT) EGFR IC50 ratio, indicating that mobocertinib displayed the best selectivity profile in these models. Also, mobocertinib showed strong inhibitory activity in HER2 exon 20YVMA allograft and patient-derived xenograft models. In genetically engineered mouse models, HER2 exon 20G776>VC lung tumors exhibited a sustained complete response to mobocertinib, whereas HER2 exon 20YVMA tumors showed only partial and transient response. Combined treatment with a second antibody-drug conjugate (ADC) against HER2, ado-trastuzumab emtansine (T-DM1), synergized with mobocertinib in HER2 exon 20YVMA tumors. In addition to the tumor cell autonomous effect, sustained tumor growth control derived from M1 macrophage infiltration and CD4+ T-cell activation. These findings support the ongoing clinical development of mobocertinib (NCT02716116) and provide a rationale for future clinical evaluation of T-DM1 combinational therapy in HER2 exon 20YVMA insertion-mutant lung adenocarcinoma patients. SIGNIFICANCE: This study elucidates the potent inhibitory activity of mobocertinib against HER2 exon 20 insertion-mutant lung cancer and the synergic effect of combined mobocertinib and T-DM1, providing a strong rationale for clinical investigation.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
HER2 exon 20 insertion mutations are sensitive to mobocertinib. A, IC50 of mobocertinib, osimertinib, neratinib, poziotinib, afatinib, and pyrotinib on Ba/F3-HER2 exon 20 insertion–mutant cell lines. B, IC50 of mobocertinib, poziotinib, osimertinib, and afatinib on H1781 (HER2G776>VC) cell line. C, The ratio of Ba/F3-HER2 exon 20 insertion–mutant IC50 to EGFR WT IC50. D, Western blot of HER2 signaling (total HER2, pHER2, total AKT, pAKT, total ERK, pERK, and actin) of H1781 (HER2 Exon 20G776>VC) cell line treated with 0.01 μmol/L, 0.1 μmol/L, and 1 μmol/L mobocertinib for 6 hours. E, Western blot of HER2 signaling (total HER2, pHER2, total AKT, pAKT, total ERK, pERK, and actin) of Ba/F3-HER2 exon 20YVMA cell line treated with 0.1 μmol/L of mobocertinib for 6 hours. F, Quantification of colony formation assay on H1781 (HER2 exon 20G776>VC) cell line treated with DMSO control, 0.01 μmol/L, and 0.1 μmol/L of mobocertinib. G, Tumor volumes of Ba/F3-HER2 exon 20YVMA allograft treated with vehicle, mobocertinib 50 and 100 mpk. H, Tumor volume change of NSCLC HER2 exon 20YVMA PDX treated with vehicle, 15 and 30 mpk mobocertinib. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 2.
Figure 2.
HER2 exon 20G776>VC GEMM showed sustained response to mobocertinib. A, Working model of genetically engineered mice with HER2 exon 20G776>VC insertion mutation using the LoxP-STOP-LoxP system. B, Different time points of tumor volume quantification on MRI of HER2 exon 20G776>VC–mutant mice after induction. C, Representative image of hematoxylin and eosin (H&E) staining and immunochemistry of TTF1, p63, and SPC on HER2 exon 20G776>VC–mutant tumors. D, Dot plot of tumor volume changes on MRI from HER2 exon 20G776>VC insertion–mutant GEMMs treated with mobocertinib for 2 and 4 weeks. E, Tumor volume change for up to 16 weeks on MRI from HER2 exon 20G776>VC insertion–mutant GEMMs treated with mobocertinib. ****, P < 0.0001.
Figure 3.
Figure 3.
HER2 exon 20YVMA GEMM initially responded to mobocertinib but became acquired resistant upon continuous treatment. A, Tumor volume change on MRI from HER2 exon 20YVMA insertion–mutant GEMMs treated with mobocertinib. B, Representative immunochemistry image and quantification of HER2 protein on HER2 exon 20YVMA insertion–mutant GEMMs treated with vehicle or mobocertinib together with the acquired resistant tumors. C, Representative immunochemistry image and quantification of pHER2 protein on HER2 exon 20YVMA insertion–mutant GEMMs treated with vehicle or mobocertinib together with the acquired resistant tumors. D, Representative immunochemistry image and quantification of pERK protein on HER2 exon 20YVMA insertion–mutant GEMMs treated with vehicle or mobocertinib together with the acquired resistant tumors. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 4.
Figure 4.
Pathways enriched in HER2 exon 20YVMA acquired resistant tumors and mobocertinib increased HER2 expression at the cell membrane. A, Dot plot of enriched pathways (NOM P < 0.05 and FDR q < 0.25) in GSEA result from mobocertinib acquired resistant tumors versus response (treated for 3 days) tumor nodules. B, Enrichment plot of “G2M Checkpoint” gene set. C, Enrichment plot of “Mitotic Spindle” gene set. D, Enrichment plot of “mTOR1 Signaling” gene set. E, Mean fluorescence intensity of surface HER2 receptor on H1781 (HER2G776>VC) cell line treated with 0.01, 0.1, and 1 μmol/L mobocertinib using flow cytometry. F, Mean fluorescence intensity of surface HER2 receptor on the Ba/F3 HER2 exon 20YVMA cell line treated with 0.01, 0.1, and 1 μmol/L mobocertinib using flow cytometry. G, Relative mRNA change of HER2 on Ba/F3-HER2 exon 20YVMA cell line treated with 0.01, 0.1, and 1 μmol/L mobocertinib using real-time PCR. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 5.
Figure 5.
Mobocertinib and T-DM1 combination is the most effective among all three combinations. A, Dot plot of tumor volume change on MRI at the time point of weeks 2, 4, and 6 from HER2 exon 20YVMA insertion–mutant GEMMs treated with monotherapy (mobocertinib, T-DM1, alisertib, and sapanisertib) and combinations (mobocertinib combined with T-DM1, alisertib, and sapanisertib). B, Representative MRI image of HER2 exon 20YVMA insertion–mutant GEMMs treated with mobocertinib and T-DM1 combination. C, Survival curve of HER2 exon 20YVMA insertion–mutant GEMMs treated with vehicle, mobocertinib, T-DM1, and combination. D, Tumor volume change of acquired resistant HER2 exon 20YVMA insertion–mutant tumors treated with monotherapies (mobocertinib and T-DM1) and combinations (mobocertinib plus alisertib or T-DM1). E, Representative MRI image of acquired resistant HER2 exon 20YVMA insertion–mutant tumors treated with mobocertinib and T-DM1 combination therapy. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; NS, nonsignificant.
Figure 6.
Figure 6.
M1 macrophage infiltration and CD4+ T-cell activation contributed to the synergic effect of mobocertinib and T-DM1 combination. A, Dot plot of enriched pathways (NOM P < 0.05 and FDR q < 0.25) in GSEA resulting from mobocertinib and T-DM1–treated tumors versus response (treated for 3 days) tumor nodules. B, Heat map of 22 different immune cell populations from RNA-seq data of tumor nodules treated with vehicle, mobocertinib, and mobocertinib plus T-DM1 combination using CIBERSORT analysis. C–H, Comprehensive immuno-profiling of HER2 exon 20YVMA insertion-mutant GEMMs treated with vehicle, mobocertinib, T-DM1, and combination therapy using multiparameter flow cytometry. I, Multi-immunofluorescence of DAPI, TTF1, and F4/80 on mobocertinib and T-DM1–treated tumor. J, Dot plot of tumor volume change on MRI from HER2 exon 20YVMA insertion–mutant GEMMs treated with vehicle, Clodrosome, mobocertinib, mobocertinib plus T-DM1, and mobocertinib combined with T-DM1 plus Clodrosome. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; NS, nonsignificant.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30.
    1. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014;511:543–50.
    1. Recondo G, Facchinetti F, Olaussen KA, Besse B, Friboulet L. Making the first move in EGFR-driven or ALK-driven NSCLC: first-generation or next-generation TKI? Nat Rev Clin Oncol 2018;15:694–708.
    1. Arcila ME, Chaft JE, Nafa K, Roy-Chowdhuri S, Lau C, Zaidinski M, et al. . Prevalence, clinicopathologic associations, and molecular spectrum of ERBB2 (HER2) tyrosine kinase mutations in lung adenocarcinomas. Clin Cancer Res 2012;18:4910–8.
    1. Tomizawa K, Suda K, Onozato R, Kosaka T, Endoh H, Sekido Y, et al. . Prognostic and predictive implications of HER2/ERBB2/neu gene mutations in lung cancers. Lung Cancer 2011;74:139–44.
    1. Mazieres J, Peters S, Lepage B, Cortot AB, Barlesi F, Beau-Faller M, et al. . Lung cancer that harbors an HER2 mutation: epidemiologic characteristics and therapeutic perspectives. J Clin Oncol 2013;31:1997–2003.
    1. Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ, Wistuba II, et al. . Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014;311:1998–2006.
    1. Shigematsu H, Takahashi T, Nomura M, Majmudar K, Suzuki M, Lee H, et al. . Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res 2005;65:1642–6.
    1. Hyman DM, Piha-Paul SA, Won H, Rodon J, Saura C, Shapiro GI, et al. . HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 2018;554:189–94.
    1. Mazieres J, Barlesi F, Filleron T, Besse B, Monnet I, Beau-Faller M, et al. . Lung cancer patients with HER2 mutations treated with chemotherapy and HER2-targeted drugs: results from the European EUHER2 cohort. Ann Oncol 2016;27:281–6.
    1. Wang Y, Jiang T, Qin Z, Jiang J, Wang Q, Yang S, et al. . HER2 exon 20 insertions in non-small-cell lung cancer are sensitive to the irreversible pan-HER receptor tyrosine kinase inhibitor pyrotinib. Ann Oncol 2019;30:447–55.
    1. Prelaj A, Bottiglieri A, Proto C, Lo Russo G, Signorelli D, Ferrara R, et al. . Poziotinib for EGFR and HER2 exon 20 insertion mutation in advanced NSCLC: results from the expanded access program. Eur J Cancer 2021;149:235–48.
    1. Neal J, Doebele R, Riely G, Spira A, Horn L, Piotrowska Z, et al. . P1.13-44 safety, PK, and preliminary antitumor activity of the oral EGFR/HER2 exon 20 inhibitor TAK-788 in NSCLC. J Thorac Oncol 2018;13:S599.
    1. Riely GJ, Neal JW, Camidge DR, Spira AI, Piotrowska Z, Costa DB, et al. . Activity and safety of mobocertinib (TAK-788) in previously treated non-small cell lung cancer with EGFR exon 20 insertion mutations from a phase 1/2 trial. Cancer Discov 2021;11:1688–99.
    1. Akbay EA, Moslehi J, Christensen CL, Saha S, Tchaicha JH, Ramkissoon SH, et al. . D-2-hydroxyglutarate produced by mutant IDH2 causes cardiomyopathy and neurodegeneration in mice. Genes Dev 2014;28:479–90.
    1. Chen Z, Cheng K, Walton Z, Wang Y, Ebi H, Shimamura T, et al. . A murine lung cancer co-clinical trial identifies genetic modifiers of therapeutic response. Nature 2012;483:613–7.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 2011;12:323.
    1. Misharin AV, Morales-Nebreda L, Mutlu GM, Budinger GR, Perlman H. Flow cytometric analysis of macrophages and dendritic cell subsets in the mouse lung. Am J Respir Cell Mol Biol 2013;49:503–10.
    1. Ji H, Zhao X, Yuza Y, Shimamura T, Li D, Protopopov A, et al. . Epidermal growth factor receptor variant III mutations in lung tumorigenesis and sensitivity to tyrosine kinase inhibitors. Proc Natl Acad Sci U S A 2006;103:7817–22.
    1. Shimamura T, Ji H, Minami Y, Thomas RK, Lowell AM, Shah K, et al. . Non-small-cell lung cancer and Ba/F3 transformed cells harboring the ERBB2 G776insV_G/C mutation are sensitive to the dual-specific epidermal growth factor receptor and ERBB2 inhibitor HKI-272. Cancer Res 2006;66:6487–91.
    1. Prelaj A, Bottiglieri A, Proto C, Lo Russo G, Signorelli D, Ferrara R, et al. . Poziotinib in advanced NSCLC with EGFR or HER2 exon 20 insertion mutation: initial results from a single site expanded access program [abstract 1388P]. Ann Oncol 2020;31:S882.
    1. Perera SA, Li D, Shimamura T, Raso MG, Ji H, Chen L, et al. . HER2YVMA drives rapid development of adenosquamous lung tumors in mice that are sensitive to BIBW2992 and rapamycin combination therapy. Proc Natl Acad Sci U S A 2009;106:474–9.
    1. Shah KN, Bhatt R, Rotow J, Rohrberg J, Olivas V, Wang VE, et al. . Aurora kinase A drives the evolution of resistance to third-generation EGFR inhibitors in lung cancer. Nat Med 2019;25:111–8.
    1. Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, et al. . The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 2012;485:55–61.
    1. Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, et al. . Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008;68:9280–90.
    1. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. . Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 2015;12:453–7.
    1. Shi Y, Fan X, Deng H, Brezski RJ, Rycyzyn M, Jordan RE, et al. . Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcgamma receptors on macrophages. J Immunol 2015;194:4379–86.
    1. van Rooijen N, Sanders A, van den Berg TK. Apoptosis of macrophages induced by liposome-mediated intracellular delivery of clodronate and propamidine. J Immunol Methods 1996;193:93–9.
    1. Gonzalvez F, Vincent S, Baker TE, Gould AE, Li S, Wardwell SD, et al. . Mobocertinib (TAK-788): a targeted inhibitor of EGFR exon 20 insertion mutants in non-small cell lung cancer. Cancer Discov 2021;11:1672–87.
    1. Dziadziuszko R, Smit EF, Dafni U, Wolf J, Wasag B, Biernat W, et al. . Afatinib in NSCLC with HER2 mutations: results of the prospective, open-label phase II NICHE trial of European Thoracic Oncology Platform (ETOP). J Thorac Oncol 2019;14:1086–94.
    1. Liu SV, Villaruz LC, Lee VHF, Zhu VW, Baik CS, Sacher A, et al. . First analysis of RAIN-701: study of tarloxotinib in patients with non-small cell lung cancer (NSCLC) EGFR Exon 20 insertion, HER2-activating mutations & other solid tumours with NRG1/ERBB gene fusions [abstract LBA61]. Ann Oncol 2020;31:S1189.
    1. Nagamoto Y, Miyamoto M, Togashi N, Taira T, Jimbo T, Isoyama T, et al. . Preclinical evaluation of DS-2087b, a novel and selective inhibitor of EGFR/HER2 exon 20 insertions [abstract 11P]. Ann Oncol 2020;31:S248.
    1. Robichaux JP, Elamin YY, Vijayan RSK, Nilsson MB, Hu L, He J, et al. . Pan-cancer landscape and analysis of ERBB2 mutations identifies poziotinib as a clinically active inhibitor and enhancer of T-DM1 activity. Cancer Cell 2019;36:444–57.
    1. Zhao S, Fang W, Pan H, Yang Y, Liang Y, Yang L, et al. . Conformational landscapes of HER2 exon 20 insertions explain their sensitivity to kinase inhibitors in lung adenocarcinoma. J Thorac Oncol 2020;15:962–72.
    1. Yuan B, Zhao J, Zhou C, Wang X, Zhu B, Zhuo M, et al. . Co-Occurring alterations of ERBB2 exon 20 insertion in non-small cell lung cancer (NSCLC) and the potential indicator of response to afatinib. Front Oncol 2020;10:729.
    1. Fang W, Zhao S, Liang Y, Yang Y, Yang L, Dong X, et al. . Mutation variants and co-mutations as genomic modifiers of response to afatinib in HER2-mutant lung adenocarcinoma. Oncologist 2020;25:e545–54.
    1. Scaltriti M, Verma C, Guzman M, Jimenez J, Parra JL, Pedersen K, et al. . Lapatinib, a HER2 tyrosine kinase inhibitor, induces stabilization and accumulation of HER2 and potentiates trastuzumab-dependent cell cytotoxicity. Oncogene 2009;28:803–14.
    1. Li BT, Michelini F, Misale S, Cocco E, Baldino L, Cai Y, et al. . HER2-mediated internalization of cytotoxic agents in ERBB2 amplified or mutant lung cancers. Cancer Discov 2020;10:674–87.
    1. Li F, Huang Q, Luster TA, Hu H, Zhang H, Ng WL, et al. . In Vivo epigenetic CRISPR screen identifies Asf1a as an immunotherapeutic target in Kras-mutant lung adenocarcinoma. Cancer Discov 2020;10:270–87.
    1. Mills CD. Anatomy of a discovery: m1 and m2 macrophages. Front Immunol 2015;6:212.
    1. Wang W, Marinis JM, Beal AM, Savadkar S, Wu Y, Khan M, et al. . RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer. Cancer Cell 2018;34:757–74.

Source: PubMed

3
Prenumerera