Synergistic growth inhibition by Iressa and Rapamycin is modulated by VHL mutations in renal cell carcinoma

R M Gemmill, M Zhou, L Costa, C Korch, R M Bukowski, H A Drabkin, R M Gemmill, M Zhou, L Costa, C Korch, R M Bukowski, H A Drabkin

Abstract

Epidermal growth factor receptor (EGFR) and tumour growth factor alpha (TGFalpha) are frequently overexpressed in renal cell carcinoma (RCC) yet responses to single-agent EGFR inhibitors are uncommon. Although von Hippel-Lindau (VHL) mutations are predominant, RCC also develops in individuals with tuberous sclerosis (TSC). Tuberous sclerosis mutations activate mammalian target of rapamycin (mTOR) and biochemically resemble VHL alterations. We found that RCC cell lines expressed EGFR mRNA in the near-absence of other ErbB family members. Combined EGFR and mTOR inhibition synergistically impaired growth in a VHL-dependent manner. Iressa blocked ERK1/2 phosphorylation specifically in wt-VHL cells, whereas rapamycin inhibited phospho-RPS6 and 4E-BP1 irrespective of VHL. In contrast, phospho-AKT was resistant to these agents and MYC translation initiation (polysome binding) was similarly unaffected unless AKT was inhibited. Primary RCCs vs cell lines contained similar amounts of phospho-ERK1/2, much higher levels of ErbB-3, less phospho-AKT, and no evidence of phospho-RPS6, suggesting that mTOR activity was reduced. A subset of tumours and cell lines expressed elevated eIF4E in the absence of upstream activation. Despite similar amounts of EGFR mRNA, cell lines (vs tumours) overexpressed EGFR protein. In the paired cell lines, PRC3 and WT8, EGFR protein was elevated post-transcriptionally in the VHL mutant and EGF-stimulated phosphorylation was prolonged. We propose that combined EGFR and mTOR inhibitors may be useful in the subset of RCCs with wt-VHL. However, apparent differences between primary tumours and cell lines require further investigation.

Figures

Figure 1
Figure 1
Differential EGFR levels and ERK phosphorylation in RCC cells. (A) Aliquots (10 μg) of cell lysates were analysed by Western blot for levels of EGFR protein. PRC3, MPR6 and WT8 are derived from 786-O cells by transfection of empty vector (PRC3) or independent wild-type VHL expression constructs (MPR6 and WT8). Tubulin served as a loading control. (B) RCC cell lines WT8VHL-wt and PRC3VHL-mut were grown until 50% confluent, then treated for 2 h with DMSO (lanes 1 and 5), rapamycin (10 nM; lanes 2 and 6), Iressa (10 μM; lanes 3 and 7) or both (lanes 4 and 8). Aliquots were analysed for the indicated total and phospho-proteins. (C) WT8VHL-wt and PRC3VHL-mut cells at 50% confluency were treated for 2 h with decreasing doses of Iressa (10–0.1 μM) and analysed as above. (D) WT8VHL-wt cells at 50% confluency were treated for 20 h with 10 or 100 μM CoCl2, or not, as indicated (Co:). For the last 2 h, selected cultures were treated with rapamycin (10 nM), Iressa (10 μM) or both; control cells were treated with DMSO. Cells were washed twice with PBS, harvested and analysed with the indicated antibodies. One lane of PRC3VHL-mut lysate was included as a positive control for the HIF2α antibody (arrow). This lane was not analysed with the other four antibodies.
Figure 2
Figure 2
Growth inhibition by Iressa and rapamycin in derivatives of 786-O cells. The parental RCC line 786-O and three stably transfected derivatives were tested for the effects of varying doses of Iressa and rapamycin on growth using MTT assays. MTT absorbance values from single and combined agents were converted to the combination index (see Methods).
Figure 3
Figure 3
Phospho-protein analysis in additional RCC cell lines. Six RCC cell lines were grown to 50% confluency, then treated for 2 h with DMSO (lanes 1, 5, 9, 13, 17 and 21), rapamycin (10 nM; lanes 2, 6, 10, 14, 18 and 22), Iressa (10 μM; lanes 3, 7, 11, 15, 19 and 23) or both (lanes 4, 8, 12, 16, 20 and 24). Lysates were analysed as in Figure 1. VHL mutational status is indicated along the bottom.
Figure 4
Figure 4
Total and phospho-EGFR levels in PRC3 and WT8. (A) Protein lysates, as described in Figure 1, were analysed for total EGFR protein. The blot was stripped and reprobed with anti-VHL and anti-HA antibodies. (B) PRC3 and WT8 cultures were grown to 50% confluency, washed with PBS, then starved in 0.1% serum for 2 h. Cells were harvested at the indicated time points (min) after the addition of 10% serum and 50 ng ml−1 EGF. More total protein was loaded in the WT8 lanes to compensate for the lower amounts of EGFR in this cell line. Otherwise, equal protein aliquots were analysed for the phosphorylation levels of tyrosine 845 and 1068 using phospho-specific antibodies.
Figure 5
Figure 5
Polysome analysis of WT8 cells. WT8 cells were grown to 70% confluency, then treated for 2 h with DMSO, rapamycin (10 nM), Iressa (10 μM) or both, and harvested for polysome fractionation. A representative A280 fractionation profile is shown along with the fractions pooled and the results from quantitative RT–PCR analysis.
Figure 6
Figure 6
Analysis of primary tumours. (A) Protein (10 μg) from 12 matched pairs of renal tumours (T) and normal kidney (N) were analysed with the indicated antibodies on two separate sets of blots. Equivalent gels were stained with Coomassie blue (CBB) for loading. The low protein content of tumour #10 indicates that relative Erk phosphorylation was even higher than the band intensity suggests. VHL mutational status is summarised by m=mutant; w=wild type. (B) In total, 30 μg of tumour and normal lysates were analysed for levels of EGFR, phospho-RPS6 and total RPS6. The four cell line lysates were treated with Iressa and rapamycin (I/R) for 2 h or not (−). Densitometric analysis for the left panel was normalised to the tumour with the lowest EGFR signal. Densitometry for the right panel utilised a shorter exposure to remain within the linear response range. Even the longest exposures failed to detect EGFR in T-12. The asterisk denotes a background band. (C) Direct comparisons of phospho-Erk and phospho-Akt levels in a subset of tumours and cell lines on the same filter. (D) WT8VHL-wt cultures starting at 70% confluency (time zero) were grown under standard culture conditions over 48 h and periodically sampled. Phospho-RPS6 levels were downregulated at 100% confluency.

References

    1. Al-Saleem T, Wessner LL, Scheithauer BW, Patterson K, Roach ES, Dreyer SJ, Fujikawa K, Bjornsson J, Bernstein J, Henske EP (1998) Malignant tumors of the kidney, brain, and soft tissues in children and young adults with the tuberous sclerosis complex. Cancer 83: 2208–2216
    1. Amato RJ (2005) Renal cell carcinoma: review of novel single-agent therapeutics and combination regimens. Ann Oncol 16: 7–15
    1. Atkins MB, Hidalgo M, Stadler WM, Logan TF, Dutcher JP, Hudes GR, Park Y, Liou SH, Marshall B, Boni JP, Dukart G, Sherman ML (2004) Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 22: 909–918
    1. Baulida J, Kraus MH, Alimandi M, Di Fiore PP, Carpenter G (1996) All ErbB receptors other than the epidermal growth factor receptor are endocytosis impaired. J Biol Chem 271: 525–5257
    1. Beerli RR, Hynes NE (1996) Epidermal growth factor-related peptides activate distinct subsets of ErbB receptors and differ in their biological activities. J Biol Chem 271: 6071–6076
    1. Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin Jr WG (2003) TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell 4: 147–158
    1. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ, Arden KC, Blenis J, Greenberg ME (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857–868
    1. Burgess AW, Cho HS, Eigenbrot C, Ferguson KM, Garrett TP, Leahy DJ, Lemmon MA, Sliwkowski MX, Ward CW, Yokoyama S (2003) An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol Cell 12: 541–552
    1. Castagnino P, Lorenzi MV, Yeh J, Breckenridge D, Sakata H, Munz B, Werner S, Bottaro DP (2000) Neu differentiation factor/heregulin induction by hepatocyte and keratinocyte growth factors. Oncogene 19: 640–648
    1. Cheville JC, Lohse CM, Zincke H, Weaver AL, Leibovich BC, Frank I, Blute ML (2004) Sarcomatoid renal cell carcinoma: an examination of underlying histologic subtype and an analysis of associations with patient outcome. Am J Surg Pathol 28: 435–441
    1. Chou TC, Talalay P (1984) Quantitative analysis of dose–effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regulat 22: 27–55
    1. de Paulsen N, Brychzy A, Fournier MC, Klausner RD, Gnarra JR, Pause A, Lee S (2001) Role of transforming growth factor-alpha in von Hippel–Lindau (VHL)(−/−) clear cell renal carcinoma cell proliferation: a possible mechanism coupling VHL tumor suppressor inactivation and tumorigenesis. Proc Natl Acad Sci USA 98: 1387–1392
    1. Dilling MB, Germain GS, Dudkin L, Jayaraman AL, Zhang X, Harwood FC, Houghton PJ (2002) 4E-binding proteins, the suppressors of eukaryotic initiation factor 4E, are down-regulated in cells with acquired or intrinsic resistance to rapamycin. J Biol Chem 277: 13907–13917
    1. Drabkin HA, Parsy C, Ferguson K, Guilhot F, Lacotte L, Roy L, Zeng C, Baron A, Hunger SP, Varella-Garcia M, Gemmill R, Brizard F, Brizard A, Roche J (2002) Quantitative HOX expression in chromosomally defined subsets of acute myelogenous leukemia. Leukemia 16: 186–195
    1. Drucker B, Bacik J, Ginsberg M, Marion S, Russo P, Mazumdar M, Motzer R (2003) Phase II trial of ZD1839 (IRESSA) in patients with advanced renal cell carcinoma. Invest New Drugs 21: 341–345
    1. Fedi P, Pierce JH, di Fiore PP, Kraus MH (1994) Efficient coupling with phosphatidylinositol 3-kinase, but not phospholipase C gamma or GTPase-activating protein, distinguishes ErbB-3 signaling from that of other ErbB/EGFR family members. Mol Cell Biol 14: 492–500
    1. Foon KA, Yang XD, Weiner LM, Belldegrun AS, Figlin RA, Crawford J, Rowinsky EK, Dutcher JP, Vogelzang NJ, Gollub J, Thompson JA, Schwartz G, Bukowski RM, Roskos LK, Schwab GM (2004) Preclinical and clinical evaluations of ABX-EGF, a fully human anti-epidermal growth factor receptor antibody. Int J Radiat Oncol Biol Phys 58: 984–990
    1. Freeman MR, Washecka R, Chung LW (1989) Aberrant expression of epidermal growth factor receptor and HER-2 (erbB-2) messenger RNAs in human renal cancers. Cancer Res 49: 6221–6225
    1. Galban S, Fan J, Martindale JL, Cheadle C, Hoffman B, Woods MP, Temeles G, Brieger J, Decker J, Gorospe M (2003) von Hippel–Lindau protein-mediated repression of tumor necrosis factor alpha translation revealed through use of cDNA arrays. Mol Cell Biol 23: 2316–2328
    1. Gera JF, Mellinghoff IK, Shi Y, Rettig MB, Tran C, Hsu JH, Sawyers CL, Lichtenstein AK (2004) AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem 279: 2737–2746
    1. Gomella LG, Sargent ER, Wade TP, Anglard P, Linehan WM, Kasid A (1989) Expression of transforming growth factor alpha in normal human adult kidney and enhanced expression of transforming growth factors alpha and beta 1 in renal cell carcinoma. Cancer Res 49: 6972–6975
    1. Gunaratnam L, Morley M, Franovic A, de Paulsen N, Mekhail K, Parolin DA, Nakamura E, Lorimer IA, Lee S (2003) Hypoxia inducible factor activates the transforming growth factor-alpha/epidermal growth factor receptor growth stimulatory pathway in VHL(−/−) renal cell carcinoma cells. J Biol Chem 278: 44966–44974
    1. Hay N, Sonenberg N (2004) Upstream and downstream of mTOR. Genes Dev 18: 1926–1945
    1. Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, Samid D, Duan DS, Gnarra JR, Linehan WM (1994) Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc Natl Acad Sci USA 91: 9700–9704
    1. Hidalgo M (2003) Erlotinib: preclinical investigations. Oncology (Huntingt) 17(11 Suppl 12): 11–16
    1. Hosoi H, Dilling MB, Shikata T, Liu LN, Shu L, Ashmun RA, Germain GS, Abraham RT, Houghton PJ (1999) Rapamycin causes poorly reversible inhibition of mTOR and induces p53-independent apoptosis in human rhabdomyosarcoma cells. Cancer Res 59: 886–894
    1. Humes HD, Beals TF, Cieslinski DA, Sanchez IO, Page TP (1991) Effects of transforming growth factor-beta, transforming growth factor-alpha, and other growth factors on renal proximal tubule cells. Lab Invest 64: 538–545
    1. Iliopoulos O, Kibel A, Gray S, Kaelin Jr WG (1995) Tumour suppression by the human von Hippel–Lindau gene product. Nat Med 1: 822–826
    1. Kaelin Jr WG (2003) The von Hippel–Lindau gene, kidney cancer, and oxygen sensing. J Am Soc Nephrol 14: 2703–2711
    1. Kau TR, Schroeder F, Ramaswamy S, Wojciechowski CL, Zhao JJ, Roberts TM, Clardy J, Sellers WR, Silver PA (2003) A chemical genetic screen identifies inhibitors of regulated nuclear export of a Forkhead transcription factor in PTEN-deficient tumor cells. Cancer Cell 4: 463–476
    1. Kenerson HL, Aicher LD, True LD, Yeung RS (2002) Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors. Cancer Res 62: 5645–5650
    1. Knebelmann B, Ananth S, Cohen HT, Sukhatme VP (1998) Transforming growth factor alpha is a target for the von Hippel–Lindau tumor suppressor. Cancer Res 58: 226–231
    1. Kondo K, Klco J, Nakamura E, Lechpammer M, Kaelin Jr WG (2002) Inhibition of HIF is necessary for tumor suppression by the von Hippel–Lindau protein. Cancer Cell 1: 237–246
    1. Kondo K, Yao M, Kobayashi K, Ota S, Yoshida M, Kaneko S, Baba M, Sakai N, Kishida T, Kawakami S, Uemura H, Nagashima Y, Nakatani Y, Hosaka M (2001) PTEN/MMAC1/TEP1 mutations in human primary renal-cell carcinomas and renal carcinoma cell lines. Int J Cancer 91: 219–224
    1. Koochekpour S, Jeffers M, Wang PH, Gong C, Taylor GA, Roessler LM, Stearman R, Vasselli JR, Stetler-Stevenson WG, Kaelin Jr WG, Linehan WM, Klausner RD, Gnarra JR, Vande Woude GF (1999) The von Hippel–Lindau tumor suppressor gene inhibits hepatocyte growth factor/scatter factor-induced invasion and branching morphogenesis in renal carcinoma cells. Mol Cell Biol 19: 5902–5912
    1. Latif F, Tory K, Gnarra J (1993) Identification of the von Hippel–Lindau disease tumor suppressor gene. Science 260: 1317–1320
    1. Lazaris-Karatzas A, Montine KS, Sonenberg N (1990) Malignant transformation by a eukaryotic initiation factor subunit that binds to mRNA 5′ cap. Nature 345: 544–547
    1. Li Z, Wang D, Na X, Schoen SR, Messing EM, Wu G (2002) Identification of a deubiquitinating enzyme subfamily as substrates of the von Hippel–Lindau tumor suppressor. Biochem Biophys Res Commun 294: 700–709
    1. Louro ID, McKie-Bell P, Gosnell H, Brindley BC, Bucy RP, Ruppert JM (1999) The zinc finger protein GLI induces cellular sensitivity to the mTOR inhibitor rapamycin. Cell Growth Differ 10: 503–516
    1. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA (2004) Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350: 2129–2139
    1. Maranchie JK, Vasselli JR, Riss J, Bonifacino JS, Linehan WM, Klausner RD (2002) The contribution of VHL substrate binding and HIF1-alpha to the phenotype of VHL loss in renal cell carcinoma. Cancer Cell 1: 247–255
    1. Meyuhas O (2000) Synthesis of the translational apparatus is regulated at the translational level. Eur J Biochem 267: 6321–6330
    1. Mohi MG, Boulton C, Gu TL, Sternberg DW, Neuberg D, Griffin JD, Gilliland DG, Neel BG (2004) Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci USA 101: 3130–3135
    1. Mondesire WH, Jian W, Zhang H, Ensor J, Hung MC, Mills GB, Meric-Bernstam F (2004) Targeting mammalian target of rapamycin synergistically enhances chemotherapy-induced cytotoxicity in breast cancer cells. Clin Cancer Res 10: 7031–7042
    1. Motzer RJ, Amato R, Todd M, Hwu WJ, Cohen R, Baselga J, Muss H, Cooper M, Yu R, Ginsberg MS, Needle M (2003) Phase II trial of antiepidermal growth factor receptor antibody C225 in patients with advanced renal cell carcinoma. Invest New Drugs 21: 99–101
    1. Neshat MS, Mellinghoff IK, Tran C, Stiles B, Thomas G, Petersen R, Frost P, Gibbons JJ, Wu H, Sawyers CL (2001) Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci USA 98: 10314–10319
    1. Oh RR, Park JY, Lee JH, Shin MS, Kim HS, Lee SK, Kim YS, Lee SH, Lee SN, Yang YM, Yoo NJ, Lee JY, Park WS (2002) Expression of HGF/SF and Met protein is associated with genetic alterations of VHL gene in primary renal cell carcinomas. APMIS 110: 229–238
    1. Ohh M, Yauch RL, Lonergan KM, Whaley JM, Stemmer-Rachamimov AO, Louis DN, Gavin BJ, Kley N, Kaelin Jr WG, Iliopoulos O (1998) The von Hippel–Lindau tumor suppressor protein is required for proper assembly of an extracellular fibronectin matrix. Mol Cell 1: 959–968
    1. Perera AD, Kleymenova EV, Walker CL (2000) Requirement for the von Hippel–Lindau tumor suppressor gene for functional epidermal growth factor receptor blockade by monoclonal antibody C225 in renal cell carcinoma. Clin Cancer Res 6: 1518–1523
    1. Pioli PA, Rigby WF (2001) The von Hippel–Lindau protein interacts with hnRNP A2 and regulates its expression. J Biol Chem 276: 40346–40352
    1. Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 68: 320–344
    1. Rowinsky EK, Schwartz GH, Gollob JA, Thompson JA, Vogelzang NJ, Figlin R, Bukowski R, Haas N, Lockbaum P, Li YP, Arends R, Foon KA, Schwab G, Dutcher J (2004) Safety pharmacokinetics and activity of ABX-EGF a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J Clin Oncol 22: 3003–3015
    1. Ruggero D, Montanaro L, Ma L, Xu W, Londei P, Cordon-Cardo C, Pandolfi PP (2004) The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis. Nat Med 10: 484–486
    1. Schoenfeld A, Davidowitz EJ, Burk RD (1998) A second major native von Hippel–Lindau gene product initiated from an internal translation start site functions as a tumor suppressor. Proc Natl Acad Sci USA 95: 8817–8822
    1. Shi Y, Sharma A, Wu H, Lichtenstein A, Gera J (2005) Cyclin D1 and c-Myc internal ribosome entry site (IRES)-dependent translation is regulated by AKT activity and enhanced by rapamycin through a p38 MAPK and ERK-dependent pathway. J Biol Chem 280: 10964–10973
    1. Shin Lee J, Seok Kim H, Bok Kim Y, Cheol Lee M, Soo Park C (2003) Expression of PTEN in renal cell carcinoma and its relation to tumor behavior and growth. J Surg Oncol 84: 166–172
    1. Stoneley M, Willis AE (2004) Cellular internal ribosome entry segments: structures trans-acting factors and regulation of gene expression. Oncogene 23: 3200–3207
    1. Stumm G, Eberwein S, Rostock-Wolf S, Stein H, Pomer S, Schlegel J, Waldherr R (1996) Concomitant overexpression of the EGFR and erbB-2 genes in renal cell carcinoma (RCC) is correlated with dedifferentiation and metastasis. Int J Cancer 69: 17–22
    1. Thomasson M, Hedman H, Junttila TT, Elenius K, Ljungberg B, Henriksson R (2004) ErbB4 is downregulated in renal cell carcinoma – a quantitative RT–PCR and immunohistochemical analysis of the epidermal growth factor receptor family. Acta Oncol 43: 453–459
    1. Thompson JE, Thompson CB (2004) Putting the rap on Akt. J Clin Oncol 22: 4217–4226
    1. Uhlman DL, Nguyen P, Manivel JC, Zhang G, Hagen K, Fraley E, Aeppli D, Niehans GA (1995) Epidermal growth factor receptor and transforming growth factor alpha expression in papillary and nonpapillary renal cell carcinoma: correlation with metastatic behavior and prognosis. Clin Cancer Res 1: 913–920
    1. Xu G, Zhang W, Bertram P, Zheng XF, McLeod H (2004) Pharmacogenomic profiling of the PI3K/PTEN-AKT-mTOR pathway in common human tumors. Int J Oncol 24: 893–900
    1. Yao M, Shuin T, Misaki H, Kubota Y (1988) Enhanced expression of c-myc and epidermal growth factor receptor (C-erbB-1) genes in primary human renal cancer. Cancer Res 48: 6753–6757

Source: PubMed

3
Abonnieren