Antitumor and antiangiogenic effect of the dual EGFR and HER-2 tyrosine kinase inhibitor lapatinib in a lung cancer model

Roque Diaz, Paul A Nguewa, Ricardo Parrondo, Carlos Perez-Stable, Irene Manrique, Miriam Redrado, Raul Catena, Maria Collantes, Ivan Peñuelas, Juan Antonio Díaz-González, Alfonso Calvo, Roque Diaz, Paul A Nguewa, Ricardo Parrondo, Carlos Perez-Stable, Irene Manrique, Miriam Redrado, Raul Catena, Maria Collantes, Ivan Peñuelas, Juan Antonio Díaz-González, Alfonso Calvo

Abstract

Background: There is strong evidence demonstrating that activation of epidermal growth factor receptors (EGFRs) leads to tumor growth, progression, invasion and metastasis. Erlotinib and gefitinib, two EGFR-targeted agents, have been shown to be relevant drugs for lung cancer treatment. Recent studies demonstrate that lapatinib, a dual tyrosine kinase inhibitor of EGFR and HER-2 receptors, is clinically effective against HER-2-overexpressing metastatic breast cancer. In this report, we investigated the activity of lapatinib against non-small cell lung cancer (NSCLC).

Methods: We selected the lung cancer cell line A549, which harbors genomic amplification of EGFR and HER-2. Proliferation, cell cycle analysis, clonogenic assays, and signaling cascade analyses (by western blot) were performed in vitro. In vivo experiments with A549 cells xenotransplanted into nude mice treated with lapatinib (with or without radiotherapy) were also carried out.

Results: Lapatinib dramatically reduced cell proliferation (P < 0.0001), DNA synthesis (P < 0.006), and colony formation capacity (P < 0.0001) in A549 cells in vitro. Furthermore, lapatinib induced G1 cell cycle arrest (P < 0.0001) and apoptotic cell death (P < 0.0006) and reduced cyclin A and B1 levels, which are regulators of S and G2/M cell cycle stages, respectively. Stimulation of apoptosis in lapatinib-treated A549 cells was correlated with increased cleaved PARP, active caspase-3, and proapoptotic Bak-1 levels, and reduction in the antiapoptotic IAP-2 and Bcl-xL protein levels. We also demonstrate that lapatinib altered EGFR/HER-2 signaling pathways reducing p-EGFR, p-HER-2, p-ERK1/2, p-AKT, c-Myc and PCNA levels. In vivo experiments revealed that A549 tumor-bearing mice treated with lapatinib had significantly less active tumors (as assessed by PET analysis) (P < 0.04) and smaller in size than controls. In addition, tumors from lapatinib-treated mice showed a dramatic reduction in angiogenesis (P < 0.0001).

Conclusion: Overall, these data suggest that lapatinib may be a clinically useful agent for the treatment of lung cancer.

Figures

Figure 1
Figure 1
FISH analysis showing 4 signals for HER-2 (red) and 4 signals for the chromosome 17 centromere (green) in A549 cells.
Figure 2
Figure 2
Cytotoxicity of lapatinib in A549 cells exposed to different concentrations (0.05, 0.5 and 5 μM) of the drug, for 24 h and 72 h. At the indicated time point, cell viability was measured by MTT, indicating that lapatinib significantly reduces A549 cell proliferation (**: P < 0.01; ***:P < 0.001).
Figure 3
Figure 3
Analysis of cell survival after 2 μM lapatinib treatment. A. A549 cells exposed to 2 μM lapatinib for 24 h exhibited a reduced cell growth proliferation (P < 0.001); B. After 10 days of exposure to lapatinib, the number of colonies was dramatically abrogated (***: P < 0.0001).
Figure 4
Figure 4
Cell cycle study of A549 cells treated with lapatinib. A. Lapatinib significantly alters cell cycle phases (G1 arrest and DNA synthesis reduction) analyzed by flow cytometry after propidium iodide staining (**: P < 0.01; ***: P < 0.001); B. Western blot showing the effect of lapatinib on the expression of the cell cycle regulators cyclins A, B1 and D1. In treated cells, the lower levels of cyclins A and B1, regulators of S and G2/M stages, respectively, corroborated the results described by cytometry. Levels of cyclin D1, a regulator of the G1 phase, were very low and remained unchanged upon treatment.
Figure 5
Figure 5
Intracellular signaling changes induced by lapatinib in A549 cells, analyzed by western blot. A. Immunoblots showing decreased levels of p-EGFR and p-HER-2 after stimulation with 100 ng/ml EGF and treatment with lapatinib. Downstream targets p-AKT, p-ERK1/2, c-Myc and PCNA were also reduced upon exposure to the drug. B. After lapatinib treatment, the proapoptotic protein Bak-1 was increased, the levels of the antiapoptotic proteins IAP-2 and Bcl-xL were reduced, and PARP was cleaved, thus demonstrating that the apoptotic pathway is switched on by this drug in A549 lung cancer cells.
Figure 6
Figure 6
In vivo tumor growth assays. A. After tumor implantation into immunocompromised nude mice, animals were treated with lapatinib for four weeks at the indicated concentration. Tumor volumes in treated mice were smaller than those found in controls; B. Lapatinib significantly reduced tumor metabolism (P = 0.037), which was shown by the standardized glucose uptake values (SUV) measured with micro-PET.
Figure 7
Figure 7
In vivo effect of radiotherapy alone or in combination with lapatinib, in A549 tumor-bearing mice. Lapatinib reduced modestly tumor growth in irradiated animals compared to only irradiated mice.
Figure 8
Figure 8
Lapatinib alters angiogenesis and the number of circulating endothelial progenitors (CEPs) in mice xenotransplanted with A549 tumor cells. A. Representative images of CD31-stained tumors from controls, lapatinib-treated, radiotherapy-treated and radiotherapy plus lapatinib-treated mice. B. Lapatinib dramatically reduced the CD31-positive area in the tumors (*: P < 0.05; **: P < 0.01; ***: P < 0.001); C. Quantification of CEPs in A549 tumor-bearing mice by flow cytometry from the peripheral blood. Lapatinib tended to reduce the number of CEPs compared to controls. Interestingly, it significantly diminished the number of CEPs (P = 0.0167) that were increased after radiotherapy treatment.

References

    1. Pajares MJ, Zudaire I, Lozano MD, Agorreta J, Bastarrika G, Torre W, Remirez A, Pio R, Zulueta JJ, Montuenga LM. Molecular profiling of computed tomography screen-detected lung nodules shows multiple malignant features. Cancer Epidemiol Biomarkers Prev. 2006;15:373–380. doi: 10.1158/1055-9965.EPI-05-0320.
    1. Hirsch FR, Scagliotti GV, Langer CJ, Varella-Garcia M, Franklin WA. Epidermal growth factor family of receptors in preneoplasia and lung cancer: perspectives for targeted therapies. Lung Cancer. 2003;41(Suppl 1):S29–S42. doi: 10.1016/S0169-5002(03)00137-5.
    1. Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M, Insa A, Massuti B, Gonzalez-Larriba JL, Paz-Ares L, Bover I, Garcia-Campelo R, Moreno MA, Catot S, Rolfo C, Reguart N, Palmero R, Sánchez JM, Bastus R, Mayo C, Bertran-Alamillo J, Molina MA, Sanchez JJ, Taron M. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361:958–967. doi: 10.1056/NEJMoa0904554.
    1. Rocha-Lima CM, Soares HP, Raez LE, Singal R. EGFR targeting of solid tumors. Cancer Control. 2007;14:295–304.
    1. Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis. Eur J Cancer. 2001;37(Suppl 4):S9–15. doi: 10.1016/S0959-8049(01)00231-3.
    1. Hirsch FR, Varella-Garcia M, Bunn PA Jr, Di Maria MV, Veve R, Bremmes RM, Baron AE, Zeng C, Franklin WA. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol. 2003;21:3798–3807. doi: 10.1200/JCO.2003.11.069.
    1. Greshock J, Cheng J, Rusnak D, Martin AM, Wooster R, Gilmer T, Lee K, Weber BL, Zaks T. Genome-wide DNA copy number predictors of lapatinib sensitivity in tumor-derived cell lines. Mol Cancer Ther. 2008;7:935–943. doi: 10.1158/1535-7163.MCT-07-2072.
    1. She Y, Lee F, Chen J, Haimovitz-Friedman A, Miller VA, Rusch VR, Kris MG, Sirotnak FM. The epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 selectively potentiates radiation response of human tumors in nude mice, with a marked improvement in therapeutic index. Clin Cancer Res. 2003;9:3773–3778.
    1. Rusnak DW, Alligood KJ, Mullin RJ, Spehar GM, Arenas-Elliott C, Martin AM, Degenhardt Y, Rudolph SK, Haws TF Jr, Hudson-Curtis BL, Gilmer TM. Assessment of epidermal growth factor receptor (EGFR, ErbB1) and HER2 (ErbB2) protein expression levels and response to lapatinib (Tykerb, GW572016) in an expanded panel of human normal and tumour cell lines. Cell Prolif. 2007;40:580–594. doi: 10.1111/j.1365-2184.2007.00455.x.
    1. Harrington KJ, El Hariry IA, Holford CS, Lusinchi A, Nutting CM, Rosine D, Tanay M, Deutsch E, Matthews J, D'Ambrosio C, Turner SJ, Pandeshwara JS, Bourhis J. Phase I study of lapatinib in combination with chemoradiation in patients with locally advanced squamous cell carcinoma of the head and neck. J Clin Oncol. 2009;27:1100–1107. doi: 10.1200/JCO.2008.17.5349.
    1. Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, Abbruzzese J, Traxler P, Buchdunger E, Radinsky R, Fidler IJ. Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res. 2000;60:2926–2935.
    1. Tortora G, Ciardiello F, Gasparini G. Combined targeting of EGFR-dependent and VEGF-dependent pathways: rationale, preclinical studies and clinical applications. Nat Clin Pract Oncol. 2008;5:521–530. doi: 10.1038/ncponc1161.
    1. Epperly MW, Guo H, Shen H, Niu Y, Zhang X, Jefferson M, Sikora CA, Greenberger JS. Bone marrow origin of cells with capacity for homing and differentiation to esophageal squamous epithelium. Radiat Res. 2004;162:233–240. doi: 10.1667/RR3224.
    1. Epperly MW, Shields D, Niu Y, Carlos T, Greenberger JS. Bone marrow from CD18-/- (MAC-1-/-) homozygous deletion recombinant negative mice demonstrates increased longevity in long-term bone marrow culture and decreased contribution to irradiation pulmonary damage. In Vivo. 2006;20:431–438.
    1. Kelly K, Crowley J, Bunn PA Jr, Presant CA, Grevstad PK, Moinpour CM, Ramsey SD, Wozniak AJ, Weiss GR, Moore DF, Israel VK, Livingston RB, Gandara DR. Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non--small-cell lung cancer: a Southwest Oncology Group trial. J Clin Oncol. 2001;19:3210–3218.
    1. Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, Zhu J, Johnson DH. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med. 2002;346:92–98. doi: 10.1056/NEJMoa011954.
    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer Statistics, 2009. CA Cancer J Clin. 2009;59(4):225–49. doi: 10.3322/caac.20006.
    1. Rusch V, Klimstra D, Venkatraman E, Pisters PW, Langenfeld J, Dmitrovsky E. Overexpression of the epidermal growth factor receptor and its ligand transforming growth factor alpha is frequent in resectable non-small cell lung cancer but does not predict tumor progression. Clin Cancer Res. 1997;3:515–522.
    1. Thatcher N, Chang A, Parikh P, Rodrigues PJ, Ciuleanu T, von Pawel J, Thongprasert S, Tan EH, Pemberton K, Archer V, Carroll K. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer) Lancet. 2005;366:1527–1537. doi: 10.1016/S0140-6736(05)67625-8.
    1. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, Skarlos D, Campone M, Davidson N, Berger M, Oliva C, Rubin SD, Stein S, Cameron D. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–2743. doi: 10.1056/NEJMoa064320.
    1. Gomez HL, Doval DC, Chavez MA, Ang PC, Aziz Z, Nag S, Ng C, Franco SX, Chow LW, Arbushites MC, Casey MA, Berger MS, Stein SH, Sledge GW. Efficacy and safety of lapatinib as first-line therapy for ErbB2-amplified locally advanced or metastatic breast cancer. J Clin Oncol. 2008;26:2999–3005. doi: 10.1200/JCO.2007.14.0590.
    1. Lin NU, Carey LA, Liu MC, Younger J, Come SE, Ewend M, Harris GJ, Bullitt E, Abbeele AD Van den, Henson JW, Li X, Gelman R, Burstein HJ, Kasparian E, Kirsch DG, Crawford A, Hochberg F, Winer EP. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2008;26:1993–1999. doi: 10.1200/JCO.2007.12.3588.
    1. Ling YH, Li T, Yuan Z, Haigentz M Jr, Weber TK, Perez-Soler R. Erlotinib, an effective epidermal growth factor receptor tyrosine kinase inhibitor, induces p27KIP1 up-regulation and nuclear translocation in association with cell growth inhibition and G1/S phase arrest in human non-small-cell lung cancer cell lines. Mol Pharmacol. 2007;72:248–258. doi: 10.1124/mol.107.034827.
    1. Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, Untch M, Rusnak DW, Spehar G, Mullin RJ, Keith BR, Gilmer TM, Berger M, Podratz KC, Slamon DJ. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–1639. doi: 10.1158/0008-5472.CAN-05-1182.
    1. Kim JW, Kim HP, Im SA, Kang S, Hur HS, Yoon YK, Oh DY, Kim JH, Lee DS, Kim TY, Bang YJ. The growth inhibitory effect of lapatinib, a dual inhibitor of EGFR and HER2 tyrosine kinase, in gastric cancer cell lines. Cancer Lett. 2008;272:296–306. doi: 10.1016/j.canlet.2008.07.018.
    1. Qi Y, Tu Y, Yang D, Chen Q, Xiao J, Chen Y, Fu J, Xiao X, Zhou Z. Cyclin A but not cyclin D1 is essential for c-myc-modulated cell-cycle progression. J Cell Physiol. 2007;210:63–71. doi: 10.1002/jcp.20816.
    1. Adams JM, Cory S. Bcl-2-regulated apoptosis: mechanism and therapeutic potential. Curr Opin Immunol. 2007;19:488–496. doi: 10.1016/j.coi.2007.05.004.
    1. Vucic D, Fairbrother WJ. The inhibitor of apoptosis proteins as therapeutic targets in cancer. Clin Cancer Res. 2007;13:5995–6000. doi: 10.1158/1078-0432.CCR-07-0729.
    1. Martin AP, Miller A, Emad L, Rahmani M, Walker T, Mitchell C, Hagan MP, Park MA, Yacoub A, Fisher PB, Grant S, Dent P. Lapatinib resistance in HCT116 cells is mediated by elevated MCL-1 expression and decreased BAK activation and not by ERBB receptor kinase mutation. Mol Pharmacol. 2008;74:807–822. doi: 10.1124/mol.108.047365.
    1. Sunaga N, Oriuchi N, Kaira K, Yanagitani N, Tomizawa Y, Hisada T, Ishizuka T, Endo K, Mori M. Usefulness of FDG-PET for early prediction of the response to gefitinib in non-small cell lung cancer. Lung Cancer. 2008;59:203–210. doi: 10.1016/j.lungcan.2007.08.012.
    1. Yokoi K, Thaker PH, Yazici S, Rebhun RR, Nam DH, He J, Kim SJ, Abbruzzese JL, Hamilton SR, Fidler IJ. Dual inhibition of epidermal growth factor receptor and vascular endothelial growth factor receptor phosphorylation by AEE788 reduces growth and metastasis of human colon carcinoma in an orthotopic nude mouse model. Cancer Res. 2005;65:3716–3725. doi: 10.1158/0008-5472.CAN-04-3700.
    1. Olaussen KA, Commo F, Tailler M, Lacroix L, Vitale I, Raza SQ, Richon C, Dessen P, Lazar V, Soria JC, Kroemer G. Synergistic proapoptotic effects of the two tyrosine kinase inhibitors pazopanib and lapatinib on multiple carcinoma cell lines. Oncogene. 2009;28:4249–4260. doi: 10.1038/onc.2009.277.
    1. Bertolini F, Shaked Y, Mancuso P, Kerbel RS. The multifaceted circulating endothelial cell in cancer: towards marker and target identification. Nat Rev Cancer. 2006;6:835–845. doi: 10.1038/nrc1971.
    1. Dome B, Timar J, Dobos J, Meszaros L, Raso E, Paku S, Kenessey I, Ostoros G, Magyar M, Ladanyi A, Bogos K, Tovari J. Identification and clinical significance of circulating endothelial progenitor cells in human non-small cell lung cancer. Cancer Res. 2006;66:7341–7347. doi: 10.1158/0008-5472.CAN-05-4654.
    1. Woywodt A, Scheer J, Hambach L, Buchholz S, Ganser A, Haller H, Hertenstein B, Haubitz M. Circulating endothelial cells as a marker of endothelial damage in allogeneic hematopoietic stem cell transplantation. Blood. 2004;103:3603–3605. doi: 10.1182/blood-2003-10-3479.
    1. Takatsuka H, Wakae T, Mori A, Okada M, Okamoto T, Kakishita E. Effects of total body irradiation on the vascular endothelium. Clin Transplant. 2002;16:374–377. doi: 10.1034/j.1399-0012.2002.02035.x.
    1. Bertolini F, Paul S, Mancuso P, Monestiroli S, Gobbi A, Shaked Y, Kerbel RS. Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells. Cancer Res. 2003;63:4342–4346.

Source: PubMed

3
Iratkozz fel