Disclosure of erlotinib as a multikinase inhibitor in pancreatic ductal adenocarcinoma

Laura Conradt, Klaus Godl, Christoph Schaab, Andreas Tebbe, Stefan Eser, Sandra Diersch, Christoph W Michalski, Jörg Kleeff, Angelika Schnieke, Roland M Schmid, Dieter Saur, Günter Schneider, Laura Conradt, Klaus Godl, Christoph Schaab, Andreas Tebbe, Stefan Eser, Sandra Diersch, Christoph W Michalski, Jörg Kleeff, Angelika Schnieke, Roland M Schmid, Dieter Saur, Günter Schneider

Abstract

A placebo-controlled phase 3 trial demonstrated that the epidermal growth factor receptor (EGFR) inhibitor erlotinib in combination with gemcitabine was especially efficient in a pancreatic ductal adenocarcinoma (PDAC) subgroup of patients developing skin toxicity. However, EGFR expression was not predictive for response, and markers to characterize an erlotinib-responding PDAC group are currently missing. In this work, we observed high erlotinib IC50 values in a panel of human and murine PDAC cell lines. Using EGFR small interfering RNA, we detected that the erlotinib response was marginally influenced by EGFR. To find novel EGFR targets, we used an unbiased chemical proteomics approach for target identification and quality-controlled target affinity determination combined with quantitative mass spectrometry based on stable isotope labeling by amino acids in cell culture. In contrast to gefitinib, we observed a broad target profile of erlotinib in PDAC cells by quantitative proteomics. Six protein kinases bind to erlotinib with similar or higher affinity (K(d) = 0.09-0.358 µM) than the EGFR (K(d) 0.434 µM). We provide evidence that one of the novel erlotinib targets, ARG, contributes in part to the erlotinib response in a PDAC cell line. Our data show that erlotinib is a multikinase inhibitor, which can act independent of EGFR in PDAC. These findings may help to monitor future erlotinib trials in the clinic.

Figures

Figure 1
Figure 1
Erlotinib IC50 values of murine and human PDAC cells. (A) Viability of PDAC cells after treatment with 6, 12, and 18 µM erlotinib was measured by MTT assay and compared to vehicle-treated controls. IC50 values of murine (black) and human (red) PDAC cell lines were calculated using a nonlinear regression model. Statistics can be found in Table 1. (B) Mean IC50 values of murine (black triangle) and human (red square) PDAC cell lines show no statistically significant differences (Student's t test, P = .15). (C) Western blot analysis of EGFR expression levels in the indicated PDAC cell lines. β-Actin controls equal protein loading. (D) Correlation of the erlotinib IC50 values with EGFR protein expression levels in 22 PDAC cell lines. The Spearman correlation coefficient and the P value (two-tailed) are indicated.
Figure 2
Figure 2
Erlotinib response of PDAC cells is EGFR independent. MiaPaCa2 (A), BxPc3 (B), T3M4 (C), and PPT6554 (D) cells were transfected with a control siRNA or an EGFR-specific siRNA. Left panel: Knockdown of the EGFR 48 hours after the transfection by Western blot analysis. β-Actin controls equal protein loading. Twenty-four hours after transfection, cells were treated with increasing doses of erlotinib as indicated for 48 hours or were left as an untreated control. Viability was determined using MTT assays. Middle panel: Viability of control siRNA-transfected cells was arbitrarily set to 100% and compared to (I) control siRNA-transfected and erlotinib-treated cells, to (II) EGFR siRNA-transfected cells, and to (III) EGFR siRNA-transfected and erlotinib-treated cells. Right panel: Erlotinib-induced therapeutic response in control and EGFR siRNA-transfected cells. Note that the right panels rely on an extrapolation of the data presented in the middle panels to better visualize the erlotinib-induced loss of viability (=therapeutic response) in control and EGFR siRNA-transfected cells. Student's t test, *P < .05 versus controls.
Figure 3
Figure 3
Target profiles of gefitinib and erlotinib. Shown are the target proteins of erlotinib (A) and gefitinib (B) in dispersed primary murine pancreatic cancer cells PPT6554. Kd values are plotted on a logarithmic scale, and target proteins are ranked from low (top) to high (bottom) Kd values. Protein kinases are depicted in blue; other kinases, purple; associated proteins, orange; and other proteins, green. Target proteins with Kd values lower than 10 µM are shown. In addition, target proteins with Kd values greater than 10 µM are indicated if they were identified as targets for both compounds. A complete list of all target proteins is provided in Table W2.
Figure 4
Figure 4
Role of ARG in the erlotinib response of PDAC cells. MiaPaCa2 (A), BxPc3 (B), T3M4 (C), and PPT6554 (D) cells were transfected with a control siRNA or an ARG-specific siRNA. Left panel: Knockdown of ARG 48 hours after the transfection by Western blot analysis. β-Actin controls equal protein loading. Twenty-four hours after transfection, cells were treated with increasing doses of erlotinib as indicated for 48 hours or were left as an untreated control. Viability was determined using MTT assays. Middle panel: Viability of control siRNA-transfected cells was arbitrarily set to 100% and compared to (I) control siRNA-transfected and erlotinib-treated cells, to (II) ARG siRNA-transfected cells, and to (III) ARG siRNA-transfected and erlotinib-treated cells. Right panel: Erlotinib-induced therapeutic response in control and ARG siRNA-transfected cells. Note that the right panels rely on an extrapolation of the data presented in the middle panels to better visualize the erlotinib-induced loss of viability (=therapeutic response) in control and ARG siRNA-transfected cells. Student's t test, *P < .05 versus controls.
Figure 5
Figure 5
Tyrosine phosphorylation of ARG is reduced on erlotinib treatment. (A) T3M4 cells were treated with erlotinib as indicated or were left as an untreated control. After 20 minutes, whole-cell extracts were prepared, and ARG was immunoprecipitated. Pre-immune IgGs were used as precipitation controls. Western blots were performed with ARG and anti-phosphotyrosine antibodies. The input (5%) was probed with ARG and anti-phosphotyrosine antibodies. (B) Western blot analysis of ARG protein expression levels in the indicated PDAC cell lines. β-Actin controls equal protein loading. (C) Correlation of the erlotinib IC50 values with the ARG protein expression in 22 PDAC cell lines. The Spearman correlation coefficient and the P value (two-tailed) are indicated. T3M4 cells are depicted as a red box. MiaPaCa2 (D), BxPc3 (E), T3M4 (F), and PPT6554 (G) were treated with imatinib as indicated or with vehicle as control. Viability was determined after 48 hours using MTT assays. Imatinib IC50 values and the 95% confidence interval (95% CI) are indicated.

References

    1. Brehmer D, Greff Z, Godl K, Blencke S, Kurtenbach A, Weber M, Muller S, Klebl B, Cotten M, Keri G, et al. Cellular targets of gefitinib. Cancer Res. 2005;65:379–382.
    1. Knesl P, Roseling D, Jordis U. Improved synthesis of substituted 6,7-dihydroxy-4-quinazolineamines: tandutinib, erlotinib and gefitinib. Molecules. 2006;11:286–297.
    1. Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, Pandey A, Mann M. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1:376–386.
    1. Sharma K, Weber C, Bairlein M, Greff Z, Keri G, Cox J, Olsen JV, Daub H. Proteomics strategy for quantitative protein interaction profiling in cell extracts. Nat Methods. 2009;6:741–744.
    1. Olsen JV, de Godoy LM, Li G, Macek B, Mortensen P, Pesch R, Makarov A, Lange O, Horning S, Mann M. Parts per million mass accuracy on an Orbitrap mass spectrometer via lock mass injection into a C-trap. Mol Cell Proteomics. 2005;4:2010–2021.
    1. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26:1367–1372.
    1. Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. 1973;22:3099–3108.
    1. Ray D, Ahsan A, Helman A, Chen G, Hegde A, Gurjar SR, Zhao L, Kiyokawa H, Beer DG, Lawrence TS, et al. Regulation of EGFR protein stability by the HECT-type ubiquitin ligase SMURF2. Neoplasia. 2011;13:570–578.
    1. Wakasaki T, Masuda M, Niiro H, Jabbarzadeh-Tabrizi S, Noda K, Taniyama T, Komune S, Akashi K. A critical role of c-Cbl-interacting protein of 85 kDa in the development and progression of head and neck squamous cell carcinomas through the ras-ERK pathway. Neoplasia. 2010;12:789–796.
    1. Avraham R, Yarden Y. Feedback regulation of EGFR signalling: decision making by early and delayed loops. Nat Rev Mol Cell Biol. 2011;12:104–117.
    1. Bloomston M, Bhardwaj A, Ellison EC, Frankel WL. Epidermal growth factor receptor expression in pancreatic carcinoma using tissue microarray technique. Dig Surg. 2006;23:74–79.
    1. Grunwald V, Hidalgo M. Developing inhibitors of the epidermal growth factor receptor for cancer treatment. J Natl Cancer Inst. 2003;95:851–867.
    1. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–1966.
    1. von Burstin J, Eser S, Paul MC, Seidler B, Brandl M, Messer M, von Werder A, Schmidt A, Mages J, Pagel P, et al. E-cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex. Gastroenterology. 2009;137:361–371. 371.e1–371.e5.
    1. Schneider G, Henrich A, Greiner G, Wolf V, Lovas A, Wieczorek M, Wagner T, Reichardt S, von Werder A, Schmid RM, et al. Cross talk between stimulated NF-κB and the tumor suppressor p53. Oncogene. 2010;29:2795–2806.
    1. Fritsche P, Seidler B, Schüler S, Schnieke A, Göttlicher M, Schmid RM, Saur D, Schneider G. HDAC2 mediates therapeutic resistance of pancreatic cancer cells via the BH3-only protein NOXA. Gut. 2009;58:1399–1409.
    1. Retzer-Lidl M, Schmid RM, Schneider G. Inhibition of CDK4 impairs proliferation of pancreatic cancer cells and sensitizes towards TRAIL-induced apoptosis via downregulation of survivin. Int J Cancer. 2007;121:66–75.
    1. Schneider G, Saur D, Siveke JT, Fritsch R, Greten FR, Schmid RM. IKKα controls p52/RelB at the skp2 gene promoter to regulate G1- to S-phase progression. EMBO J. 2006;25:3801–3812.
    1. Wirth M, Fritsche P, Stojanovic N, Brandl M, Jaeckel S, Schmid RM, Saur D, Schneider G. A simple and cost-effective method to transfect small interfering RNAs into pancreatic cancer cell lines using polyethylenimine. Pancreas. 2011;40:144–150.
    1. Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, Pandey A, Mann M. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1:376–386.
    1. Sharma K, Weber C, Bairlein M, Greff Z, Keri G, Cox J, Olsen JV, Daub H. Proteomics strategy for quantitative protein interaction profiling in cell extracts. Nat Methods. 2009;6:741–744.
    1. Brehmer D, Greff Z, Godl K, Blencke S, Kurtenbach A, Weber M, Muller S, Klebl B, Cotten M, Keri G, et al. Cellular targets of gefitinib. Cancer Res. 2005;65:379–382.
    1. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26:1367–1372.
    1. Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. 1973;22:3099–3108.
    1. Buck E, Eyzaguirre A, Haley JD, Gibson NW, Cagnoni P, Iwata KK. Inactivation of Akt by the epidermal growth factor receptor inhibitor erlotinib is mediated by HER-3 in pancreatic and colorectal tumor cell lines and contributes to erlotinib sensitivity. Mol Cancer Ther. 2006;5:2051–2059.
    1. Jimeno A, Tan AC, Coffa J, Rajeshkumar NV, Kulesza P, Rubio-Viqueira B, Wheelhouse J, Diosdado B, Messersmith WA, Iacobuzio-Donahue C, et al. Coordinated epidermal growth factor receptor pathway gene overexpression predicts epidermal growth factor receptor inhibitor sensitivity in pancreatic cancer. Cancer Res. 2008;68:2841–2849.
    1. Tzeng CW, Frolov A, Frolova N, Jhala NC, Howard JH, Vickers SM, Buchsbaum DJ, Heslin MJ, Arnoletti JP. EGFR genomic gain and aberrant pathway signaling in pancreatic cancer patients. J Surg Res. 2007;143:20–26.
    1. Frolov A, Schuller K, Tzeng CW, Cannon EE, Ku BC, Howard JH, Vickers SM, Heslin MJ, Buchsbaum DJ, Arnoletti JP. ErbB3 expression and dimerization with EGFR influence pancreatic cancer cell sensitivity to erlotinib. Cancer Biol Ther. 2007;6:548–554.
    1. Tan AR, Yang X, Hewitt SM, Berman A, Lepper ER, Sparreboom A, Parr AL, Figg WD, Chow C, Steinberg SM, et al. Evaluation of biologic end points and pharmacokinetics in patients with metastatic breast cancer after treatment with erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor. J Clin Oncol. 2004;22:3080–3090.
    1. Hidalgo M, Siu LL, Nemunaitis J, Rizzo J, Hammond LA, Takimoto C, Eckhardt SG, Tolcher A, Britten CD, Denis L, et al. Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J Clin Oncol. 2001;19:3267–3279.
    1. Tanis KQ, Veach D, Duewel HS, Bornmann WG, Koleske AJ. Two distinct phosphorylation pathways have additive effects on Abl family kinase activation. Mol Cell Biol. 2003;23:3884–3896.
    1. Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, Cooc J, Weinkle J, Kim GE, Jakkula L, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17:500–503.
    1. Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–1806.
    1. da Cunha Santos G, Dhani N, Tu D, Chin K, Ludkovski O, Kamel-Reid S, Squire J, Parulekar W, Moore MJ, Tsao MS. Molecular predictors of outcome in a phase 3 study of gemcitabine and erlotinib therapy in patients with advanced pancreatic cancer: National Cancer Institute of Canada Clinical Trials Group Study PA.3. Cancer. 2010;116:5599–5607.
    1. Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–132.
    1. Tigno-Aranjuez JT, Asara JM, Abbott DW. Inhibition of RIP2's tyrosine kinase activity limits NOD2-driven cytokine responses. Genes Dev. 2010;24:2666–2677.
    1. Sebens S, Arlt A, Schäfer H. NF-κB as a molecular target in the therapy of pancreatic carcinoma. Recent Results Cancer Res. 2008;177:151–164.
    1. Su F, Li H, Yan C, Jia B, Zhang Y, Chen X. Depleting MEKK1 expression inhibits the ability of invasion and migration of human pancreatic cancer cells. J Cancer Res Clin Oncol. 2009;135:1655–1663.
    1. Sawai H, Okada Y, Funahashi H, Matsuo Y, Takahashi H, Takeyama H, Manabe T. Integrin-linked kinase activity is associated with interleukin-1α-induced progressive behavior of pancreatic cancer and poor patient survival. Oncogene. 2006;25:3237–3246.
    1. Duxbury MS, Ito H, Benoit E, Waseem T, Ashley SW, Whang EE. RNA interference demonstrates a novel role for integrin-linked kinase as a determinant of pancreatic adenocarcinoma cell gemcitabine chemoresistance. Clin Cancer Res. 2005;11:3433–3438.
    1. Yau CY, Wheeler JJ, Sutton KL, Hedley DW. Inhibition of integrin-linked kinase by a selective small molecule inhibitor, QLT0254, inhibits the PI3K/PKB/mTOR, Stat3, and FKHR pathways and tumor growth, and enhances gemcitabine-induced apoptosis in human orthotopic primary pancreatic cancer xenografts. Cancer Res. 2005;65:1497–1504.
    1. Crnogorac-Jurcevic T, Efthimiou E, Nielsen T, Loader J, Terris B, Stamp G, Baron A, Scarpa A, Lemoine NR. Expression profiling of micro-dissected pancreatic adenocarcinomas. Oncogene. 2002;21:4587–4594.
    1. Hatanpaa KJ, Burma S, Zhao D, Habib AA. Epidermal growth factor receptor in glioma: signal transduction, neuropathology, imaging, and radioresistance. Neoplasia. 2010;12:675–684.
    1. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321:1807–1812.
    1. Stommel JM, Kimmelman AC, Ying H, Nabioullin R, Ponugoti AH, Wiedemeyer R, Stegh AH, Bradner JE, Ligon KL, Brennan C, et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science. 2007;318:287–290.
    1. Moore PS, Sipos B, Orlandini S, Sorio C, Real FX, Lemoine NR, Gress T, Bassi C, Kloppel G, Kalthoff H, et al. Genetic profile of 22 pancreatic carcinoma cell lines. Analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch. 2001;439:798–802.
    1. Cabodi S, del Pilar Camacho-Leal M, Di Stefano P, Defilippi P. Integrin signalling adaptors: not only figurants in the cancer story. Nat Rev Cancer. 2010;10:858–870.
    1. Philip PA, Benedetti J, Corless CL, Wong R, O'Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol. 2010;28:3605–3610.
    1. Yamamoto N, Honma M, Suzuki H. Off-target serine/threonine kinase 10 inhibition by erlotinib enhances lymphocytic activity leading to severe skin disorders. Mol Pharmacol. 2011;80:466–475.

Source: PubMed

3
購読する