A novel mechanism of PPAR gamma induction via EGFR signalling constitutes rational for combination therapy in bladder cancer

Jose Joao Mansure, Roland Nassim, Simone Chevalier, Konrad Szymanski, Joice Rocha, Saad Aldousari, Wassim Kassouf, Jose Joao Mansure, Roland Nassim, Simone Chevalier, Konrad Szymanski, Joice Rocha, Saad Aldousari, Wassim Kassouf

Abstract

Background: Two signalling molecules that are attractive for targeted therapy are the epidermal growth factor receptor (EGFR) and the peroxisome proliferator-activated receptor gamma (PPARγ). We investigated possible crosstalk between these 2 pathways, particularly in light of the recent evidence implicating PPARγ for anticancer therapy.

Principal findings: As evaluated by MTT assays, gefitinib (EGFR inhibitor) and DIM-C (PPARγ agonist) inhibited growth of 9 bladder cancer cell lines in a dose-dependent manner but with variable sensitivity. In addition, combination of gefitinib and DIM-C demonstrated maximal inhibition of cell proliferation compared to each drug alone. These findings were confirmed in vivo, where combination therapy maximally inhibited tumor growth in contrast to each treatment alone when compared to control (p<0.04). Induction of PPARγ expression along with nuclear accumulation was observed in response to increasing concentrations of gefitinib via activation of the transcription factor CCAT/enhancer-binding protein-β (CEBP-β). In these cell lines, DIM-C significantly sensitized bladder cancer cell lines that were resistant to EGFR inhibition in a schedule-specific manner.

Conclusion: These results suggest that PPARγ agonist DIM-C can be an excellent alternative to bladder tumors resistant to EGFR inhibition and combination efficacy might be achieved in a schedule-specific manner.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Baseline expression of PPARγ and…
Figure 1. Baseline expression of PPARγ and EGFR.
(A) Expression of PPARγ and EGFR relative to endogenous levels of β-actin and tubulin, respectively, and represented in units among 9 bladder cancer cell lines reflecting different stages of the disease (from Superficial to Invasive & no Metastasis, Invasive and Lymphatic Metastasis). (B) Dose-response of bladder cancer cell lines to PPARγ agonist (DIM-C) and EGFR inhibitor (gefitinib). The GI50 value was defined as the mean concentration of drug that generates 50% of growth inhibition as compared to controls.
Figure 2. Antiproliferative effects of combined therapy.
Figure 2. Antiproliferative effects of combined therapy.
Growth was monitored by MTT assays. Cells were treated with gefitinib 5 µM and DIM-C 3 µM and compared to each drug alone. Red: gefitinib; Yellow: DIM-C; Blue: gefitinib+DIM-C.
Figure 3. Effect of combination therapy on…
Figure 3. Effect of combination therapy on EGFR downstream signaling.
(A) Phosphorylation pattern of p42/44 MAPK (Erk1/2) in cells treated with gefitinib 5 µM and DIM-C 3 µM for 5, 15 and 30 minutes. T0 is the untreated control. Whole-cell lysates were immunoblotted with phosho-p42/44 MAPK and p42/44 MAP. GAPDH was used as loading control on the Western blotting. (B) Comparison of PTEN expression in cell lines treated with gefitinib 5 µM and DIM-C 3 µM for 24 hs. T0 is the untreated control.
Figure 4. Effects of combination therapy in…
Figure 4. Effects of combination therapy in vivo.
Bladder tumor growth of combination treatment arm compared to control arm (P

Figure 5. Effects of combination therapy on…

Figure 5. Effects of combination therapy on p21 expression in vivo and in vitro .

(A)…

Figure 5. Effects of combination therapy on p21 expression in vivo and in vitro.
(A) Immunohistochemistry (IHC) staining for p21 in tumor xenograft tissues. Mice were treated with placebo or combination therapy (Gefitinib 2 mg/day, 5 times per week and DIM-C 60 mg/Kg, 3 times per week). Graphic on the right side represents quantification of positive staining cells. (B) In vitro expression of p21 in Western blot of lysate cells treated with gefitinib 5 µM and DIM-C 3 µM for 24 hs. Graphic on the right side, represents quantification of p21 expression related to GAPDH.

Figure 6. Induction of PPARγ expression in…

Figure 6. Induction of PPARγ expression in response to different concentrations of gefitinib.

(A) Fold…

Figure 6. Induction of PPARγ expression in response to different concentrations of gefitinib.
(A) Fold increase relative to control was determined after normalization with β-actin as external loading control. (B) Upregulation and nuclear accumulation of PPARγ following treatment with gefitinib (24 hs).

Figure 7. Immunohistochemistry (IHC) staining for PPARγ…

Figure 7. Immunohistochemistry (IHC) staining for PPARγ in tumor xenograft tissues Mice were treated with…

Figure 7. Immunohistochemistry (IHC) staining for PPARγ in tumor xenograft tissues Mice were treated with placebo or gefitinib (Gefitinib 2 mg/day, 5 times per week).
Graphic on the lower level represents quantification of positive staining cells. Black arrows indicate nuclear staining.

Figure 8. Schedule-specific efficacy of combination therapy.

Figure 8. Schedule-specific efficacy of combination therapy.

Three relatively resistant cell lines to gefitinib (UM-UC3,…

Figure 8. Schedule-specific efficacy of combination therapy.
Three relatively resistant cell lines to gefitinib (UM-UC3, UM-UC13, and KU-7) and one sensitive (UM-UC6). Growth (MTT assays) after 48 hs treatment. Gefitinib: 2 µM. DIM-C: 2 µM.

Figure 9. mRNA expression of CEBPβ after…

Figure 9. mRNA expression of CEBPβ after treatment with gefitinib.

(A) RT-PCR of cells treated…

Figure 9. mRNA expression of CEBPβ after treatment with gefitinib.
(A) RT-PCR of cells treated with gefitinib (B) Western blot of CEBPβ expression in KU-7 and UM-UC-13 cell lines in response to different concentrations of gefitinib.
All figures (9)
Figure 5. Effects of combination therapy on…
Figure 5. Effects of combination therapy on p21 expression in vivo and in vitro.
(A) Immunohistochemistry (IHC) staining for p21 in tumor xenograft tissues. Mice were treated with placebo or combination therapy (Gefitinib 2 mg/day, 5 times per week and DIM-C 60 mg/Kg, 3 times per week). Graphic on the right side represents quantification of positive staining cells. (B) In vitro expression of p21 in Western blot of lysate cells treated with gefitinib 5 µM and DIM-C 3 µM for 24 hs. Graphic on the right side, represents quantification of p21 expression related to GAPDH.
Figure 6. Induction of PPARγ expression in…
Figure 6. Induction of PPARγ expression in response to different concentrations of gefitinib.
(A) Fold increase relative to control was determined after normalization with β-actin as external loading control. (B) Upregulation and nuclear accumulation of PPARγ following treatment with gefitinib (24 hs).
Figure 7. Immunohistochemistry (IHC) staining for PPARγ…
Figure 7. Immunohistochemistry (IHC) staining for PPARγ in tumor xenograft tissues Mice were treated with placebo or gefitinib (Gefitinib 2 mg/day, 5 times per week).
Graphic on the lower level represents quantification of positive staining cells. Black arrows indicate nuclear staining.
Figure 8. Schedule-specific efficacy of combination therapy.
Figure 8. Schedule-specific efficacy of combination therapy.
Three relatively resistant cell lines to gefitinib (UM-UC3, UM-UC13, and KU-7) and one sensitive (UM-UC6). Growth (MTT assays) after 48 hs treatment. Gefitinib: 2 µM. DIM-C: 2 µM.
Figure 9. mRNA expression of CEBPβ after…
Figure 9. mRNA expression of CEBPβ after treatment with gefitinib.
(A) RT-PCR of cells treated with gefitinib (B) Western blot of CEBPβ expression in KU-7 and UM-UC-13 cell lines in response to different concentrations of gefitinib.

References

    1. Ciardiello F (2000) Epidermal growth factor receptor tyrosine kinase inhibitors as anticancer agents. Drugs 60 Suppl 1: 25–32; discussion 41–22.
    1. Mendelsohn J, Dinney CP (2001) The Willet F. Whitmore, Jr., Lectureship: blockade of epidermal growth factor receptors as anticancer therapy. J Urol 165: 1152–1157.
    1. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, et al. (2004) EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304: 1497–1500.
    1. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, et al. (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. Blehm KN, Spiess PE, Bondaruk JE, Dujka ME, Villares GJ, et al. (2006) Mutations within the kinase domain and truncations of the epidermal growth factor receptor are rare events in bladder cancer: implications for therapy. Clin Cancer Res 12: 4671–4677.
    1. Kassouf W, Black PC, Tuziak T, Bondaruk J, Lee S, et al. (2008) Distinctive expression pattern of ErbB family receptors signifies an aggressive variant of bladder cancer. J Urol 179: 353–358.
    1. Kassouf W, Dinney CP, Brown G, McConkey DJ, Diehl AJ, et al. (2005) Uncoupling between epidermal growth factor receptor and downstream signals defines resistance to the antiproliferative effect of Gefitinib in bladder cancer cells. Cancer Res 65: 10524–10535.
    1. Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, et al. (1997) The organization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem 272: 18779–18789.
    1. Motomura W, Okumura T, Takahashi N, Obara T, Kohgo Y (2000) Activation of peroxisome proliferator-activated receptor gamma by troglitazone inhibits cell growth through the increase of p27KiP1 in human. Pancreatic carcinoma cells. Cancer Res 60: 5558–5564.
    1. Mansure JJ, Nassim R, Kassouf W (2009) Peroxisome proliferator-activated receptor gamma in bladder cancer: a promising therapeutic target. Cancer Biol Ther 8: 6–15.
    1. Guan YF, Zhang YH, Breyer RM, Davis L, Breyer MD (1999) Expression of peroxisome proliferator-activated receptor gamma (PPARgamma) in human transitional bladder cancer and its role in inducing cell death. Neoplasia 1: 330–339.
    1. Suzuki T, Nakagawa T, Endo H, Mitsudomi T, Masuda A, et al. (2003) The sensitivity of lung cancer cell lines to the EGFR-selective tyrosine kinase inhibitor ZD1839 (‘Iressa’) is not related to the expression of EGFR or HER-2 or to K-ras gene status. Lung Cancer 42: 35–41.
    1. Burgermeister E, Tencer L, Liscovitch M (2003) Peroxisome proliferator-activated receptor-gamma upregulates caveolin-1 and caveolin-2 expression in human carcinoma cells. Oncogene 22: 3888–3900.
    1. Xin X, Yang S, Kowalski J, Gerritsen ME (1999) Peroxisome proliferator-activated receptor gamma ligands are potent inhibitors of angiogenesis in vitro and in vivo. J Biol Chem 274: 9116–9121.
    1. Bishop-Bailey D, Hla T (1999) Endothelial cell apoptosis induced by the peroxisome proliferator-activated receptor (PPAR) ligand 15-deoxy-Delta12, 14-prostaglandin J2. J Biol Chem 274: 17042–17048.
    1. Kassouf W, Chintharlapalli S, Abdelrahim M, Nelkin G, Safe S, et al. (2006) Inhibition of bladder tumor growth by 1,1-bis(3′-indolyl)-1-(p-substitutedphenyl)methanes: a new class of peroxisome proliferator-activated receptor gamma agonists. Cancer Res 66: 412–418.
    1. Lee SY, Hur GY, Jung KH, Jung HC, Kim JH, et al. (2006) PPAR-gamma agonist increase gefitinib's antitumor activity through PTEN expression. Lung Cancer 51: 297–301.
    1. Zhou Y, Zheng S, Lin J, Zhang QJ, Chen A (2007) The interruption of the PDGF and EGF signaling pathways by curcumin stimulates gene expression of PPARgamma in rat activated hepatic stellate cell in vitro. Lab Invest 87: 488–498.
    1. Dinney CP, Fishbeck R, Singh RK, Eve B, Pathak S, et al. (1995) Isolation and characterization of metastatic variants from human transitional cell carcinoma passaged by orthotopic implantation in athymic nude mice. J Urol 154: 1532–1538.
    1. Sabichi A, Keyhani A, Tanaka N, Delacerda J, Lee IL, et al. (2006) Characterization of a panel of cell lines derived from urothelial neoplasms: genetic alterations, growth in vivo and the relationship of adenoviral mediated gene transfer to coxsackie adenovirus receptor expression. J Urol 175: 1133–1137.
    1. Ono M, Kuwano M (2006) Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs. Clin Cancer Res 12: 7242–7251.
    1. Morrison RF, Farmer SR (1999) Role of PPARgamma in regulating a cascade expression of cyclin-dependent kinase inhibitors, p18(INK4c) and p21(Waf1/Cip1), during adipogenesis. J Biol Chem 274: 17088–17097.
    1. Zhu Y, Qi C, Korenberg JR, Chen XN, Noya D, et al. (1995) Structural organization of mouse peroxisome proliferator-activated receptor gamma (mPPAR gamma) gene: alternative promoter use and different splicing yield two mPPAR gamma isoforms. Proc Natl Acad Sci U S A 92: 7921–7925.
    1. Pettaway CA, Pisters LL, Dinney CP, Jularbal F, Swanson DA, et al. (1995) Sentinel lymph node dissection for penile carcinoma: the M. D. Anderson Cancer Center experience. J Urol 154: 1999–2003.
    1. Emmans VC, Rodway HA, Hunt AN, Lillycrop KA (2004) Regulation of cellular processes by PPARgamma ligands in neuroblastoma cells is modulated by the level of retinoblastoma protein expression. Biochem Soc Trans 32: 840–842.
    1. Hau P, Kunz-Schughart L, Bogdahn U, Baumgart U, Hirschmann B, et al. (2007) Low-dose chemotherapy in combination with COX-2 inhibitors and PPAR-gamma agonists in recurrent high-grade gliomas - a phase II study. Oncology 73: 21–25.
    1. Shrader M, Pino MS, Brown G, Black P, Adam L, et al. (2007) Molecular correlates of gefitinib responsiveness in human bladder cancer cells. Mol Cancer Ther 6: 277–285.
    1. Lin J, Chen A (2008) Activation of peroxisome proliferator-activated receptor-gamma by curcumin blocks the signaling pathways for PDGF and EGF in hepatic stellate cells. Lab Invest 88: 529–540.
    1. Tang QQ, Lane MD (1999) Activation and centromeric localization of CCAAT/enhancer-binding proteins during the mitotic clonal expansion of adipocyte differentiation. Genes Dev 13: 2231–2241.
    1. Lewis JD, Ferrara A, Peng T, Hedderson M, Bilker WB, et al. (2011) Risk of bladder cancer among diabetic patients treated with pioglitazone: interim report of a longitudinal cohort study. Diabetes Care 34: 916–922.
    1. Dormandy J, Bhattacharya M, van Troostenburg de Bruyn AR (2009) Safety and tolerability of pioglitazone in high-risk patients with type 2 diabetes: an overview of data from PROactive. Drug Saf 32: 187–202.

Source: PubMed

3
Abonnere