Skip to main content

Advertisement

Log in

Bay846, a new irreversible small molecule inhibitor of EGFR and Her2, is highly effective against malignant brain tumor models

  • PRECLINICAL STUDIES
  • Published:
Investigational New Drugs Aims and scope Submit manuscript

Summary

The epidermal growth factor receptor (EGFR) pathway is aberrantly activated in tumors and plays a key role in promoting tumor growth. Small molecule inhibitors which bind reversibly to EGFR have demonstrated limited clinical activity. Thus, there is a continued need to develop novel EGFR inhibitors with improved anti-tumor activity. Bay846 is a newly developed small molecule inhibitor that binds irreversibly to the tyrosine kinase domains of EGFR and Her2. The in vitro and in vivo efficacy of Bay846 was tested using a panel of nine human malignant brain tumor (glioma) models. Lapatinib, a reversible inhibitor of EGFR and Her2, was included for comparison. Six glioma cell lines were sensitive to Bay846 treatment. Bay846 strongly suppressed tumor cell growth in vitro by inducing cell lysis/death rather than cell cycle arrest. Consistent with this, Bay846 had potent anti-tumor activity which led to regressions in tumor size. The active, phosphorylated form of EGFR was reduced by Bay846 treatment in vitro and in tumors. Importantly, the efficacy of Bay846 was significantly greater than lapatinib in all assays. Bay846-sensitivity was associated with expression of a wild-type PTEN in conjunction with high levels of an oncogenic EGFR variant (A289V or EGFRvIII). These studies demonstrate that targeting the EGFR pathway with the irreversible inhibitor Bay846 has great potential to increase the efficacy of this cancer therapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Mitsudomi T, Yatabe Y (2010) Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J 277:301–308

    Article  CAS  PubMed  Google Scholar 

  2. Gazdar AF (2010) Epidermal growth factor receptor inhibition in lung cancer: the evolving role of individualized therapy. Cancer Metastasis Rev 29:37–48

    Article  CAS  PubMed  Google Scholar 

  3. Schneider-Merck T, Trepel M (2010) Lapatinib. Recent Results Cancer Res 184:45–59

    Article  CAS  PubMed  Google Scholar 

  4. Moon C, Chae YK, Lee J (2010) Targeting epidermal growth factor receptor in head and neck cancer: lessons learned from cetuximab. Exp Biol Med (Maywood) 235:907–920

    Article  CAS  Google Scholar 

  5. Karpel-Massler G, Schmidt U, Unterberg A, Halatsch ME (2009) Therapeutic inhibition of the epidermal growth factor receptor in high-grade gliomas: where do we stand? Mol Cancer Res 7:1000–1012

    Article  CAS  PubMed  Google Scholar 

  6. Hynes NE, Macdonald G (2009) ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol 21:177–184

    Article  CAS  PubMed  Google Scholar 

  7. Iyer R, Bharthuar A (2010) A review of erlotinib–an oral, selective epidermal growth factor receptor tyrosine kinase inhibitor. Expert Opin Pharmacother 11:311–320

    Article  CAS  PubMed  Google Scholar 

  8. Belani CP (2010) The role of irreversible EGFR inhibitors in the treatment of non-small cell lung cancer: overcoming resistance to reversible EGFR inhibitors. Cancer Invest 28:413–423

    CAS  PubMed  Google Scholar 

  9. Thiessen B, Stewart C, Tsao M, Kamel-Reid S, Schaiquevich P, Mason W et al (2009) A phase I/II trial of GW572016 (lapatinib) in recurrent glioblastoma multiforme: clinical outcomes, pharmacokinetics and molecular correlation. Cancer Chemother Pharmacol 65:353–361

    Article  Google Scholar 

  10. Ross HJ, Blumenschein GR Jr, Aisner J, Damjanov N, Dowlati A, Garst J et al (2010) Randomized phase II multicenter trial of two schedules of lapatinib as first- or second-line monotherapy in patients with advanced or metastatic non-small cell lung cancer. Clin Cancer Res 16:1938–1949

    Article  CAS  PubMed  Google Scholar 

  11. Eller JL, Longo SL, Kyle MM, Bassano D, Hicklin DJ, Canute GW (2005) Anti-epidermal growth factor receptor monoclonal antibody cetuximab augments radiation effects in glioblastoma multiforme in vitro and in vivo. Neurosurgery 56:155–162

    PubMed  Google Scholar 

  12. Eller JL, Longo SL, Hicklin DJ, Canute GW (2002) Activity of anti-EGFR monoclonal antibody C225 against glioblastoma multiforme. Neurosurgery 51:1005–1014

    PubMed  Google Scholar 

  13. Sarkaria JN, Yang L, Grogan PT, Kitange GJ, Carlson BL, Schroeder MA et al (2007) Identification of molecular characteristics correlated with glioblastoma sensitivity to EGFR kinase inhibition through use of an intracranial xenograft test panel. Mol Cancer Ther 6:1167–1174

    Article  CAS  PubMed  Google Scholar 

  14. Ishii N, Maier D, Merlo A, Tada M, Sawamura Y, Diserens AC et al (1999) Frequent co-alterations of TP53, p16/CDKN2A, p14 ARF, PTEN tumor suppressor genes in human glioma cell lines. Brain Pathol 9:469–479

    Article  CAS  PubMed  Google Scholar 

  15. Nishikawa R, Ji XD, Harmon RC, Lazar CS, Gill GN, Cavenee WK et al (1994) A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci USA 91:7727–7731

    Article  CAS  PubMed  Google Scholar 

  16. Han Y, Caday CG, Nanda A, Cavenee WK, Huang HJ (1996) Tyrphostin AG 1478 preferentially inhibits human glioma cells expressing truncated rather than wild-type epidermal growth factor receptors. Cancer Res 56:3859–3861

    CAS  PubMed  Google Scholar 

  17. Cherry T, Longo SL, Tovar-Spinoza Z, Post DE (2010) Second-generation HIF-activated oncolytic adenoviruses with improved replication, oncolytic, and antitumor efficacy. Gene Ther 17:1430–1441

    Article  CAS  PubMed  Google Scholar 

  18. Lee JC, Vivanco I, Beroukhim R, Huang JH, Feng WL, DeBiasi RM et al (2006) Epidermal growth factor receptor activation in glioblastoma through novel missense mutations in the extracellular domain. PLoS Med 3:e485

    Article  PubMed  Google Scholar 

  19. Goncalves A, Esteyries S, Taylor-Smedra B, Lagarde A, Ayadi M, Monges G et al (2008) A polymorphism of EGFR extracellular domain is associated with progression free-survival in metastatic colorectal cancer patients receiving cetuximab-based treatment. BMC Cancer 8:169

    Article  PubMed  Google Scholar 

  20. Wiechen K, Karaaslan S, Turzynski A, Dietel M (1999) Suppression of the c-erbB-2 gene product decreases transformation abilities but not the proliferation and secretion of proteases of SK-OV-3 ovarian cancer cells. Br J Cancer 81:790–795

    Article  CAS  PubMed  Google Scholar 

  21. Takai N, Jain A, Kawamata N, Popoviciu LM, Said JW, Whittaker S et al (2005) 2 C4, a monoclonal antibody against HER2, disrupts the HER kinase signaling pathway and inhibits ovarian carcinoma cell growth. Cancer 104:2701–2708

    Article  CAS  PubMed  Google Scholar 

  22. Szollosi J, Balazs M, Feuerstein BG, Benz CC, Waldman FM (1995) ERBB-2 (HER2/neu) gene copy number, p185HER-2 overexpression, and intratumor heterogeneity in human breast cancer. Cancer Res 55:5400–5407

    CAS  PubMed  Google Scholar 

  23. Liu G, Ying H, Zeng G, Wheeler CJ, Black KL, Yu JS (2004) HER-2, gp100, and MAGE-1 are expressed in human glioblastoma and recognized by cytotoxic T cells. Cancer Res 64:4980–4986

    Article  CAS  PubMed  Google Scholar 

  24. Olayioye MA (2001) Update on HER-2 as a target for cancer therapy: intracellular signaling pathways of ErbB2/HER-2 and family members. Breast Cancer Res 3:385–389

    Article  CAS  PubMed  Google Scholar 

  25. Fojo T, Parkinson DR (2010) Biologically targeted cancer therapy and marginal benefits: are we making too much of too little or are we achieving too little by giving too much? Clin Cancer Res 16:5972–5980

    Article  CAS  PubMed  Google Scholar 

  26. Idbaih A, Aimard J, Boisselier B, Marie Y, Paris S, Criniere E et al (2009) Epidermal growth factor receptor extracellular domain mutations in primary glioblastoma. Neuropathol Appl Neurobiol 35:208–213

    Article  CAS  PubMed  Google Scholar 

  27. Johns TG, Perera RM, Vernes SC, Vitali AA, Cao DX, Cavenee WK et al (2007) The efficacy of epidermal growth factor receptor-specific antibodies against glioma xenografts is influenced by receptor levels, activation status, and heterodimerization. Clin Cancer Res 13:1911–1925

    Article  CAS  PubMed  Google Scholar 

  28. Andersson U, Guo D, Malmer B, Bergenheim AT, Brännström T, Hedman H et al (2004) Epidermal growth factor receptor family (EGFR, ErbB2-4) in gliomas and meningiomas. Acta Neuropathol 108:135–142

    Article  CAS  PubMed  Google Scholar 

  29. Torp SH, Gulati S, Johannessen E, Dalen A (2007) Coexpression of c-erbB 1-4 receptor proteins in human glioblastomas. An immunohistochemical study. J Exp Clin Cancer Res 26:353–359

    CAS  PubMed  Google Scholar 

  30. Mineo JF, Bordron A, Baroncini M, Maurage CA, Ramirez C, Siminski RM et al (2007) Low HER2-expressing glioblastomas are more often secondary to anaplastic transformation of low-grade glioma. J Neurooncol 85:281–287

    Article  PubMed  Google Scholar 

  31. (2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455: 1061-1068

  32. Mineo JF, Bordron A, Quintin-Roué I, Loisel S, Ster KL, Buhé V et al (2004) Recombinant humanised anti-HER2/neu antibody (Herceptin(R)) induces cellular death of glioblastomas. Br J Cancer 91:1195–1199

    CAS  PubMed  Google Scholar 

  33. Wheeler DL, Huang S, Kruser TJ, Nechrebecki MM, Armstrong EA, Benavente S et al (2008) Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members. Oncogene 27:3944–3956

    Article  CAS  PubMed  Google Scholar 

  34. Quesnelle KM, Grandis JR (2011) Dual kinase inhibition of EGFR and HER2 overcomes resistance to cetuximab in a novel in vivo model of acquired cetuximab resistance. Clin Cancer Res 17:5935–5944

    Article  CAS  PubMed  Google Scholar 

  35. Kong A, Calleja V, Leboucher P, Harris A, Parker PJ, Larijani B (2008) HER2 oncogenic function escapes EGFR tyrosine kinase inhibitors via activation of alternative HER receptors in breast cancer cells. PLoS One 3:e2881

    Article  PubMed  Google Scholar 

  36. Westphal M, Meima L, Szonyi E, Lofgren J, Meissner H, Hamel W et al (1997) Heregulins and the ErbB-2/3/4 receptors in gliomas. J Neurooncol 35:335–346

    Article  CAS  PubMed  Google Scholar 

  37. Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M et al (2006) Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res 66:1630–1639

    Article  CAS  PubMed  Google Scholar 

  38. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P et al (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321:1807–1812

    Article  CAS  PubMed  Google Scholar 

  39. Boström J, Cobbers JMJL, Wolter M, Tabatabai G, Weber RG, Lichter P et al (1998) Mutation of the PTEN (MMAC1) tumor suppressor gene in a subset of glioblastomas but not in meningiomas with loss of chromosome arm 10q. Cancer Res 58:29–33

    PubMed  Google Scholar 

  40. Duerr EM, Rollbrocker B, Hayashi Y, Peters N, Meyer-Puttlitz B, Louis DN et al (1998) PTEN mutations in gliomas and glioneuronal tumors. Oncogene 16:2259–2264

    Article  CAS  PubMed  Google Scholar 

  41. Wang SI, Puc J, Li J, Bruce JN, Cairns P, Sidransky D et al (1997) Somatic mutations of PTEN in glioblastoma multiforme. Cancer Res 57:4183–4186

    CAS  PubMed  Google Scholar 

  42. Haas-Kogan DA, Prados MD, Tihan T, Eberhard DA, Jelluma N, Arvold ND et al (2005) Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib. J Natl Cancer Inst 97:880–887

    Article  CAS  PubMed  Google Scholar 

  43. Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu SJ, Dia EQ et al (2005) Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353:2012–2024

    Article  CAS  PubMed  Google Scholar 

  44. Fan QW, Weiss WA (2010) Targeting the RTK-PI3K-mTOR axis in malignant glioma: overcoming resistance. Curr Top Microbiol Immunol 347:279–296

    Article  CAS  PubMed  Google Scholar 

  45. Xia W, Husain I, Liu L, Bacus S, Saini S, Spohn J et al (2007) Lapatinib antitumor activity is not dependent upon phosphatase and tensin homologue deleted on chromosome 10 in ErbB2-overexpressing breast cancers. Cancer Res 67:1170–1175

    Article  CAS  PubMed  Google Scholar 

  46. Dave B, Migliaccio I, Gutierrez MC, Wu MF, Chamness GC, Wong H et al (2010) Loss of Phosphatase and Tensin Homolog or Phosphoinositol-3 Kinase Activation and Response to Trastuzumab or Lapatinib in Human Epidermal Growth Factor Receptor 2-Overexpressing Locally Advanced Breast Cancers. J Clin Oncol 29:166–173

    Article  PubMed  Google Scholar 

  47. Bouchalova K, Cizkova M, Cwiertka K, Trojanec R, Friedecky D, Hajduch M (2010) Lapatinib in breast cancer - the predictive significance of HER1 (EGFR), HER2, PTEN and PIK3CA genes and lapatinib plasma level assessment. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 154:281–288

    Article  CAS  PubMed  Google Scholar 

  48. Carrasco-Garcia E, Saceda M, Grasso S, Rocamora-Reverte L, Conde M, Gomez-Martinez A et al (2011) Small tyrosine kinase inhibitors interrupt EGFR signaling by interacting with erbB3 and erbB4 in glioblastoma cell lines. Exp Cell Res 317:1476–1489

    Article  CAS  PubMed  Google Scholar 

  49. Brown JM (2000) Exploiting the hypoxic cancer cell: mechanisms and therapeutic strategies. Mol Med Today 6:157–162

    Article  CAS  PubMed  Google Scholar 

  50. Gril B, Palmieri D, Bronder JL, Herring JM, Vega-Valle E, Feigenbaum L et al (2008) Effect of lapatinib on the outgrowth of metastatic breast cancer cells to the brain. J Natl Cancer Inst 100:1092–1103

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Financial support was provided by Bayer Healthcare (DEP and GWC), an American Cancer Society Institutional Research grant from the Upstate Cancer Institute at SUNY Upstate Medical University (DEP), and the Dept. of Neurosurgery at SUNY Upstate Medical University. We thank Bayer Healthcare for providing Bay846, Rebecca Sager for assistance with the LDH studies, and Ed Shillitoe for critical reading of the manuscript.

Financial support

Bayer Healthcare (DEP and GWC), an American Cancer Society Institutional Research grant from the Upstate Cancer Institute at SUNY Upstate Medical University (DEP), and Dept. of Neurosurgery, SUNY Upstate Medical University.

Conflict of Interest

The authors declare that financial support and the Bay846 compound was provided by Bayer Healthcare. KP, HS, and OP are employees of Bayer Healthcare. The commercial funder had no role in the analysis or interpretation of data.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dawn E. Post.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplemental Fig. 1

Cell surface EGFR expression levels. Cell surface EGFR levels were quantified by flow cytometry using an anti-EGFR specific antibody (open curves). An isotype matched IgG antibody was used as a negative control (red shaded curves). The relative EGFR level (SFI) was calculated as the ratio of the mean fluorescence value obtained with the specific EGFR antibody versus the control antibody. (PPT 87 kb)

Supplemental Fig. 2

Cell surface Her2 expression levels. Cell surface Her2 levels were quantified by flow cytometry using an anti-Her2 specific antibody (open curves). An isotype matched IgG antibody was used as a negative control (red shaded curves). The relative Her2 level (SFI) was calculated as the ratio of the mean fluorescence value obtained with the specific Her2 antibody versus the control antibody. The study was performed twice for each cell line, except for GBM39 and U87 which were measured one time, and the resulting SFI was averaged. Representative data for each cell line is shown. (PPT 86 kb)

Supplemental Fig. 3

Cell cycle profile of cell lines which were resistant to Bay846 and lapatinib treatment. Cells were treated with Bay846, lapatinib, or untreated at the indicated drug concentrations for 48 h. The relative DNA content per cell was obtained by staining cells with propidium iodide and then measuring fluorescence using a LSRII flow cytometer. The cell cycle profile was analyzed using FACSDiva software (Becton-Dickinson). See Fig. 2 for cells lines which were sensitive to Bay846 and lapatinib. (PPT 120 kb)

Supplemental Fig. 4

Total and phospho-EGFR levels in cells lines which were resistant to Bay846 and lapatinib treatment. Cells were treated with Bay846, lapatinib, or untreated. Bay846 and lapatinib were used at a concentration of 1 μM. Total cell lysates were examined by western blotting for total and phospho-(p1086 and p1173) EGFR, PTEN, and actin (loading control) at Day 2 (Bai, U87, Arn) or Day 3 [Mor(lo)]. Asterisks (*) indicate non-specific bands that are cross-reactive with the anti-PTEN antibody. See Fig. 4 for cells lines which were sensitive to Bay846 and lapatinib. (PPT 166 kb)

Supplemental Table 1

Primers used for sequencing PTEN and EGFR cDNAs. The PCR product size represents the expected size in base pairs of a wild-type cDNA sequence. (DOC 30 kb)

Supplemental Table 2

Inhibition of GBM cell growth by Bay846 and lapatinib measured by MTT assay at 3 days post-treatment. Data is the average percent cell growth inhibition ± SD and is shown graphically in Fig. 1. Asterisks indicate a significant increase in cell growth inhibition by Bay846 compared to lapatinib (p < 0.002, t-test). (DOC 32 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Longo, S.L., Padalino, D.J., McGillis, S. et al. Bay846, a new irreversible small molecule inhibitor of EGFR and Her2, is highly effective against malignant brain tumor models. Invest New Drugs 30, 2161–2172 (2012). https://doi.org/10.1007/s10637-011-9784-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10637-011-9784-4

Keywords

Navigation