Pyrotinib combined with CDK4/6 inhibitor in HER2-positive metastatic gastric cancer: A promising strategy from AVATAR mouse to patients

Zuhua Chen, Yingying Xu, Jifang Gong, Furong Kou, Mengqi Zhang, Tiantian Tian, Xiaotian Zhang, Cheng Zhang, Jian Li, Zhongwu Li, Yumei Lai, Jianjun Zou, Xiaoyu Zhu, Jing Gao, Lin Shen, Zuhua Chen, Yingying Xu, Jifang Gong, Furong Kou, Mengqi Zhang, Tiantian Tian, Xiaotian Zhang, Cheng Zhang, Jian Li, Zhongwu Li, Yumei Lai, Jianjun Zou, Xiaoyu Zhu, Jing Gao, Lin Shen

Abstract

Background: Pyrotinib was well tolerated but its efficacy was unsatisfactory in patients with HER2-positive gastric cancer (GC) (NCT02378389). This study was to optimize the efficacy of pyrotinib.

Methods: Human GC cell lines and AVATAR mice were used to explore the refractory mechanisms of pyrotinib. A pyrotinib-combined strategy was proposed, which was validated in preclinical AVATAR mouse and in clinical patients enrolled in a phase I clinical trial (NCT03480256).

Results: Dysregulation of CCND1-CDK4/6-Rb axis might be the key to pyrotinib refractory. The strategy of pyrotinib combined with a CDK4/6 inhibitor SHR6390 was proposed and validated in preclinical AVATAR mouse, which was successfully verified in clinical patients. For five patients treated with pyrotinib plus SHR6390 who had available response evaluation, the best response was partial response in three patients, stable disease in one patient, and progressive disease in one patient. The progression-free survival times were 120, 200, 532, 109, and 57 days, respectively.

Conclusions: This translational study suggests that pyrotinib combined with SHR6390 may serve as a promising strategy for patients with HER2-positive GC.

Trial registration: The ClinicalTrials.gov identifiers are NCT02378389 (https://ichgcp.net/clinical-trials-registry/NCT02378389, registered in 11 February 2015) and NCT03480256 (https://ichgcp.net/clinical-trials-registry/NCT03480256, registered in 8 March 2018).

Keywords: CDK inhibitor; pyrotinib; refractory mechanisms.

Conflict of interest statement

Jianjun Zou and Xiaoyu Zhu are employees from Jiangsu Hengrui Medicine Co, Ltd. All other authors have no conflict of interest.

© 2020 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

Figures

FIGURE 1
FIGURE 1
Pyrotinib exerts selective antitumor activity in GC AVATAR models. A, Antitumor activity of pyrotinib in 10 GC AVATAR models with different expression of HER2 and EGFR (×200 magnification; scale bar represents 100 µm). Data are presented as mean ± SEM (n = five mice per group). B, The efficacy of pyrotinib in 10 AVATAR models with different molecular features of HER2 and EGFR IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; TGI, tumor growth inhibition.
FIGURE 2
FIGURE 2
The establishment and characteristics of the pyrotinib‐refractory AVATAR model. A, The process of generating a pyrotinib‐refractory AVATAR model (019R). B, H&E staining and immunohistochemical staining for Ki‐67 in 019P, 019S, and 019R (×100 magnification; scale bar represents 100 µm). C, The Pearson correlation between 019P, 019S, and 019R by transcriptome sequencing
FIGURE 3
FIGURE 3
CDK4/6 inhibitor SHR6390 sensitizes pyrotinib in pyrotinib refractory AVATAR model. A, Differentially expressed genes (DEGs) in the 019S and 019R detected by RNA sequencing. Green, Padj < .005 and log2 (fold change) < –1; Red, Padj < .005 and log2 (fold change) > 1. B, Quantitative real‐time PCR of CCND1, CDK4, and CDK6 in 019P and 019R. Data are presented as mean ± SD of three independent experiments. C and D, Expression and quantification of critical molecules in the ErbB family and cell‐cycle signaling pathway in 019P, 019S, and 019R. E, The efficacy of pyrotinib, SHR6390, and the combination therapy in 019R. Data are presented as mean ± SD (n = 5 mice per group). F and G, Expression and quantification of critical molecules in the ErbB family and cell cycle signaling pathway in 019R. *P < .05, **P < .01, and ***P < .001 according to repeated measures ANOVAs
FIGURE 4
FIGURE 4
The clinical responses of three HER2‐positive GC patients treated by pyrotinib combined with SHR6390. A, CT scans of tumor metastases in lung, liver, and hepatic hilar lymph nodes during four cycles of combination treatment in patient 1. C, CT scans of tumor metastases in lung, axillary, and supraclavicular lymph nodes during six cycles of combination treatment in patient 2. E, CT scans of tumor metastases in lung and retroperitoneal lymph nodes during four cycles of combination treatment in patient 3. B, D, and F, The dynamic change of CEA, CA199, CA72.4, and CA125 during combination treatment in patient 1, 2, and 3
FIGURE 5
FIGURE 5
The clinical responses of the other two HER2‐positive GC patients treated by pyrotinib combined with SHR6390. A, CT scans of tumor metastases in liver, left adrenal gland, and perigastric lymph nodes during three cycles of combination treatment in patient 4. C, CT scans of tumor metastases in liver during two cycles of combination treatment in patient 5. B and D, The dynamic change of CEA, CA199, CA72.4, and CA125 during combination treatment in patient 4 and 5. E, Treatment response of each patient after combination treatment and the duration of response. Patient number are shown to the left of the Y‐axis PR, partial response; SD, stable disease; PD, progressive disease. PFS, progression‐free survival.

References

    1. Citri A, Yarden Y. EGF‐ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol. 2006;7(7):505‐516.
    1. Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19(9):1523‐1529.
    1. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2‐positive advanced gastric or gastro‐oesophageal junction cancer (ToGA): a phase 3, open‐label, randomised controlled trial. Lancet. 2010;376(9742):687‐697.
    1. Tabernero J, Hoff PM, Shen L, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2‐positive metastatic gastric or gastro‐oesophageal junction cancer (JACOB): final analysis of a double‐blind, randomised, placebo‐controlled phase 3 study. Lancet Oncol. 2018;19(10):1372‐1384.
    1. Thuss‐Patience PC, Shah MA, Ohtsu A, et al. Trastuzumab emtansine versus taxane use for previously treated HER2‐positive locally advanced or metastatic gastric or gastro‐oesophageal junction adenocarcinoma (GATSBY): an international randomised, open‐label, adaptive, phase 2/3 study. Lancet Oncol. 2017;18(5):640‐653.
    1. Satoh T, Xu RH, Chung HC, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second‐line treatment of HER2‐amplified advanced gastric cancer in Asian populations: tyTAN–a randomized, phase III study. J Clin Oncol. 2014;32(19):2039‐2049.
    1. Hecht JR, Bang YJ, Qin SK, et al. Lapatinib in combination with capecitabine plus oxaliplatin in human epidermal growth factor receptor 2‐positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma: tRIO‐013/LOGiC–A randomized phase III trial. J Clin Oncol. 2016;34(5):443‐451.
    1. Ma F, Li Q, Chen S, et al. Phase I study and biomarker analysis of pyrotinib, a novel irreversible pan‐ErbB receptor tyrosine kinase inhibitor, in patients with human epidermal growth factor receptor 2‐positive metastatic breast cancer. J Clin Oncol. 2017;35(27):3105‐3112.
    1. Zhu Y, Tian T, Li Z, et al. Establishment and characterization of patient‐derived tumor xenograft using gastroscopic biopsies in gastric cancer. Sci Rep. 2015;5:8542.
    1. Chen Z, Huang W, Tian T, et al. Characterization and validation of potential therapeutic targets based on the molecular signature of patient‐derived xenografts in gastric cancer. J Hematol Oncol. 2018;11(1):20.
    1. Liu Z, Chen Z, Wang J, et al. Mouse avatar models of esophageal squamous cell carcinoma proved the potential for EGFR‐TKI afatinib and uncovered Src family kinases involved in acquired resistance. J Hematol Oncol. 2018;11(1):109.
    1. Cobleigh MA, Vogel CL, Tripathy D, et al. Multinational study of the efficacy and safety of humanized anti‐HER2 monoclonal antibody in women who have HER2‐overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol. 1999;17(9):2639‐2648.
    1. Baselga J, Cortes J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366(2):109‐119.
    1. Urruticoechea A, Rizwanullah M, Im SA, et al. Randomized phase III trial of trastuzumab plus capecitabine with or without pertuzumab in patients with human epidermal growth factor receptor 2‐positive metastatic breast cancer who experienced disease progression during or after trastuzumab‐based therapy. J Clin Oncol. 2017;35(26):3030‐3038.
    1. Perez EA, Barrios C, Eiermann W, et al. Trastuzumab emtansine with or without pertuzumab versus trastuzumab plus taxane for human epidermal growth factor receptor 2‐positive, advanced breast cancer: primary results from the phase III MARIANNE Study. J Clin Oncol. 2017;35(2):141‐148.
    1. Chen K, Yang D, Li X, et al. Mutational landscape of gastric adenocarcinoma in Chinese: implications for prognosis and therapy. Proc Natl Acad Sci USA. 2015;112(4):1107‐1112.
    1. Lordick F, Janjigian YY. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nat Rev Clin Oncol. 2016;13(6):348‐360.
    1. Leto SM, Sassi F, Catalano I, et al. Sustained inhibition of HER3 and EGFR is necessary to induce regression of HER2‐amplified gastrointestinal carcinomas. Clin Cancer Res. 2015;21(24):5519‐5531.
    1. Iida M, Bahrar H, Brand TM, et al. Targeting the HER family with Pan‐HER effectively overcomes resistance to cetuximab. Mol Cancer Ther. 2016;15(9):2175‐2186.
    1. Goel S, Wang Q, Watt AC, et al. Overcoming therapeutic resistance in HER2‐positive breast cancers with CDK4/6 inhibitors. Cancer Cell. 2016;29(3):255‐269.
    1. Cristofanilli M, Turner NC, Bondarenko I, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone‐receptor‐positive, HER2‐negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA‐3): final analysis of the multicentre, double‐blind, phase 3 randomised controlled trial. Lancet Oncol. 2016;17(4):425‐439.
    1. O'Sullivan CC, Suman VJ, Goetz MP. The emerging role of CDK4/6i in HER2‐positive breast cancer. Ther Adv Med Oncol. 2019;11:1758835919887665.
    1. Gianni L, Bisagni G, Colleoni M, et al. Neoadjuvant treatment with trastuzumab and pertuzumab plus palbociclib and fulvestrant in HER2‐positive, ER‐positive breast cancer (NA‐PHER2): an exploratory, open‐label, phase 2 study. Lancet Oncol. 2018;19(2):249‐256.
    1. Zhang K, Hong R, Kaping L, et al. CDK4/6 inhibitor palbociclib enhances the effect of pyrotinib in HER2‐positive breast cancer. Cancer Lett. 2019;447:130‐140.
    1. Liu B, Chen D, Chen S, et al. Transcriptional activation of cyclin D1 via HER2/HER3 contributes to EGFR‐TKI resistance in lung cancer. Biochem Pharmacol. 2020;178:114095.
    1. Inoue K, Fry EA. Aberrant expression of cyclin D1 in cancer. Sign Transduct Insights. 2015;4:1‐13.
    1. Thery JC, Spano JP, Azria D, et al. Resistance to human epidermal growth factor receptor type 2‐targeted therapies. Eur J Cancer. 2014;50(5):892‐901.
    1. Nagata Y, Lan KH, Zhou X, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell. 2004;6(2):117‐127.
    1. Berns K, Horlings HM, Hennessy BT, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. 2007;12(4):395‐402.
    1. Mercogliano MF, De Martino M, Venturutti L, et al. TNFalpha‐induced mucin 4 expression elicits trastuzumab resistance in HER2‐positive breast cancer. Clin Cancer Res. 2017;23(3):636‐648.

Source: PubMed

3
Subscribe