A randomised controlled phase II trial of pre-operative celecoxib treatment reveals anti-tumour transcriptional response in primary breast cancer

Rita D Brandão, Jürgen Veeck, Koen K Van de Vijver, Patrick Lindsey, Bart de Vries, Catharina H M J van Elssen, Marinus J Blok, Kristien Keymeulen, Torik Ayoubi, Hubert J M Smeets, Vivianne C Tjan-Heijnen, Pierre S Hupperets, Rita D Brandão, Jürgen Veeck, Koen K Van de Vijver, Patrick Lindsey, Bart de Vries, Catharina H M J van Elssen, Marinus J Blok, Kristien Keymeulen, Torik Ayoubi, Hubert J M Smeets, Vivianne C Tjan-Heijnen, Pierre S Hupperets

Abstract

Introduction: Cyclooxygenase-2 (COX-2) is frequently over-expressed in primary breast cancer. In transgenic breast cancer models, over-expression of COX-2 leads to tumour formation while COX-2 inhibition exerts anti-tumour effects in breast cancer cell lines. To further determine the effect of COX-2 inhibition in primary breast cancer, we aimed to identify transcriptional changes in breast cancer tissues of patients treated with the selective COX-2 inhibitor celecoxib.

Methods: In a single-centre double-blind phase II study, thirty-seven breast cancer patients were randomised to receive either pre-operative celecoxib (400 mg) twice daily for two to three weeks (n = 22) or a placebo according to the same schedule (n = 15). Gene expression in fresh-frozen pre-surgical biopsies (before treatment) and surgical excision specimens (after treatment) was profiled by using Affymetrix arrays. Differentially expressed genes and altered pathways were bioinformatically identified. Expression of selected genes was validated by quantitative PCR (qPCR). Immunohistochemical protein expression analyses of the proliferation marker Ki-67, the apoptosis marker cleaved caspase-3 and the neo-angiogenesis marker CD34 served to evaluate biological response.

Results: We identified 972 and 586 significantly up- and down-regulated genes, respectively, in celecoxib-treated specimens. Significant expression changes in six out of eight genes could be validated by qPCR. Pathway analyses revealed over-representation of deregulated genes in the networks of proliferation, cell cycle, extracellular matrix biology, and inflammatory immune response. The Ki-67 mean change relative to baseline was -29.1% (P = 0.019) and -8.2% (P = 0.384) in the treatment and control arm, respectively. Between treatment groups, the change in Ki-67 was statistically significant (P = 0.029). Cleaved caspase-3 and CD34 expression were not significantly different between the celecoxib-treated and placebo-treated groups.

Conclusions: Short-term COX-2 inhibition by celecoxib induces transcriptional programs supporting anti-tumour activity in primary breast cancer tissue. The impact on proliferation-associated genes is reflected by a reduction of Ki-67 positive cells. Therefore, COX-2 inhibition should be considered as a treatment strategy for further clinical testing in primary breast cancer.

Trial registration: ClinicalTrials.gov NCT01695226.

Figures

Figure 1
Figure 1
Flow diagram of the presented study. The design is a double-blind, randomised, controlled phase II trial of pre-operative celecoxib versus placebo in early breast cancer. Note that eight patients had discontinued intervention in the treatment arm. Gene expression profiling (GEP) has been performed from samples where indicated.
Figure 2
Figure 2
qPCR validation of selected genes differentially expressed in celecoxib-treated samples as determined by microarray analysis. Fold-change and the 95% CI (error bars) are shown. Expression of six out of eight genes analysed (indicated by asterisks) was significantly changed in agreement with the microarray analysis.
Figure 3
Figure 3
Effects of celecoxib treatment on cell cycle and proliferation. Contributed map from GenMAPP software with an overview of the pathways and genes involved in cell cycle regulation. The expression fold-changes of each gene are indicated next to the gene box. Genes highlighted in red and green in the left box half represent genes with fold-changes increased and decreased, respectively. Red colour in the right box half indicates a significant change. Grey boxes correspond to genes that were not analysed in the arrays.
Figure 4
Figure 4
Effects of celecoxib treatment on extracellular matrix protein degradation. Map designed on GenMAPP software with an overview of the genes involved in the extracellular matrix protein degradation process. The expression fold-changes of each gene are indicated next to the gene box. Genes highlighted in red and green in the left box half represent genes with fold-changes increased and decreased, respectively. Red colour in the right box half indicates a significant change. Grey boxes correspond to genes that were not analysed in the arrays.
Figure 5
Figure 5
Effects of celecoxib treatment on Ki-67 protein expression. (A) Examples of immunohistochemical staining of nuclear Ki-67 protein on breast cancer tissues yielding a high score (left) and low score (right). Scale bar = 100 μm. (B) Shown are the Ki-67 scores from individual patients in the control arm (plot on left-hand side) and treatment arm (plot on right-hand side). Geometric means in the control group were statistically not different (P = 0.384), while the geometric mean after celecoxib treatment was significantly reduced (P = 0.019). Also, the change of the means between both groups was significantly greater in the celecoxib-treated group (P = 0.029).

References

    1. O'Byrne KJ, Dalgleish AG. Chronic immune activation and inflammation as the cause of malignancy. Br J Cancer. 2001;84:473–483.
    1. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025.
    1. Wang D, DuBois RN. Eicosanoids and cancer. Nat Rev Cancer. 2010;10:181–193. doi: 10.1038/nrc2809.
    1. Friedman G, Ury H. Initial screening for carcinogenicity of commonly used drugs. J Natl Cancer Inst. 1980;65:723–733.
    1. Harris RE, Namboodiri KK, Farrar WB. Nonsteroidal antiinflammatory drugs and breast cancer. Epidemiology. 1996;7:203–205. doi: 10.1097/00001648-199603000-00017.
    1. Thun MJ, Namboodiri MM, Heath CW. Aspirin use and reduced risk of fatal colon cancer. N Engl J Med. 1991;325:1593–1596. doi: 10.1056/NEJM199112053252301.
    1. Giardiello FM, Hamilton SR, Krush AJ, Piantadosi S, Hylind LM, Celano P, Booker SV, Robinson CR, Offerhaus GJ. Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N Engl J Med. 1993;328:1313–1316. doi: 10.1056/NEJM199305063281805.
    1. Giovannucci E, Egan KM, Hunter DJ, Stampfer MJ, Colditz GA, Willett WC, Speizer FE. Aspirin and the risk of colorectal cancer in women. N Engl J Med. 1995;333:609–614. doi: 10.1056/NEJM199509073331001.
    1. Schreinemachers DM, Everson RB. Aspirin use and lung, colon, and breast cancer incidence in a prospective study. Epidemiology. 1994;5:138–146. doi: 10.1097/00001648-199403000-00003.
    1. Sharpe CR, Collet JP, McNutt M, Belzile E, Boivin J-F, Hanley JA. Nested case-control study of the effects of non-steroidal anti-inflammatory drugs on breast cancer risk and stage. Br J Cancer. 2000;83:112–120.
    1. Baron JA, Sandler RS, Bresalier RS, Quan H, Riddell R, Lanas A, Bolognese JA, Oxenius B, Horgan K, Loftus S, Morton DG. APPROVe Trial Investigators. A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas. Gastroenterology. 2006;131:1674–1682. doi: 10.1053/j.gastro.2006.08.079.
    1. Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid biology. Science. 2001;294:1871–1875. doi: 10.1126/science.294.5548.1871.
    1. Bennett A, McDonald AM, Stamford IF, Charlier EM, Simpson JS, Zebro T. Prostaglandins and breast cancer. Lancet. 1977;310:624–626. doi: 10.1016/S0140-6736(77)92496-5.
    1. Smith CJ, Sun D, Hoegler C, Roth BS, Zhang X, Zhao G, Xu XB, Kobari Y, Pritchard K Jr, Sessa WC, Hintze TH. Reduced gene expression of vascular endothelial NO synthase and cyclooxygenase-1 in heart failure. Circ Res. 1996;78:58–64. doi: 10.1161/01.RES.78.1.58.
    1. Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem. 2000;69:145–182. doi: 10.1146/annurev.biochem.69.1.145.
    1. Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR. TIS10, a phorbol ester tumor promoter-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthase/cyclooxygenase homologue. J Biol Chem. 1991;266:12866–12872.
    1. Xie WL, Chipman JG, Robertson DL, Erikson RL, Simmons DL. Expression of a mitogen-responsive gene encoding prostaglendin synthase is regulated by mRNA splicing. Proc Natl Acad Sci USA. 1991;88:2692–2696. doi: 10.1073/pnas.88.7.2692.
    1. Soslow RA, Dannenberg AJ, Rush D, Woerner BM, Khan KN, Masferrer J, Koki AT. COX-2 is expressed in human pulmonary, colonic, and mammary tumors. Cancer. 2000;89:2637–2645. doi: 10.1002/1097-0142(20001215)89:12<2637::AID-CNCR17>;2-B.
    1. Liu CH, Chang SH, Narko K, Trifan OC, Wu MT, Smith E, Haudenschild C, Lane TF, Hla T. Overexpression of cyclooxygenase-2 Is sufficient to induce tumorigenesis in transgenic mice. J Biol Chem. 2001;276:18563–18569. doi: 10.1074/jbc.M010787200.
    1. Tavassoli FA, Devilee P. Pathology and Genetics of Tumours of the Breast and Female Genital Organs. Lyon: IARC Press; 2003.
    1. Sobin LH, Wittekind C. TNM Classification of Malignant Tumours. 6. New York: Wiley-Liss; 2002.
    1. Elston EW, Ellis IO. Method for grading breast cancer. J Clin Pathol. 1993;46:189–190.
    1. ArrayExpress.
    1. Dai M, Wang P, Boyd AD, Kostov G, Athey B, Jones EG, Bunney WE, Myers RM, Speed TP, Akil H, Watson SJ, Meng F. Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Res. 2005;33:e175. doi: 10.1093/nar/gni179.
    1. Akaike H. In: Second International Symposium of Inference Theory. Petrov B, Csàki F, editor. Budapest: Akadémiai Kiadó; 1973. Information theory and an extension of the maximum likelihood principle; pp. 267–281.
    1. Ihaka R, Gentleman R. R: a language for data analysis and graphics. Journal of Computational and Graphical Statistics. 1996;5:299–314.
    1. Lindsey J. Models for Repeated Measurements. 2. Oxford: Oxford University Press; 1999.
    1. Dennis G, Sherman B, Hosack D, Yang J, Gao W, Lane HC, Lempicki R. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003;4:3. doi: 10.1186/gb-2003-4-5-p3.
    1. van Iersel M, Kelder T, Pico A, Hanspers K, Coort S, Conklin B, Evelo C. Presenting and exploring biological pathways with PathVisio. BMC Bioinformatics. 2008;9:399. doi: 10.1186/1471-2105-9-399.
    1. Pico AR, Kelder T, van Iersel MP, Hanspers K, Conklin BR, Evelo C. WikiPathways: pathway editing for the people. PLoS Biol. 2008;6:e184. doi: 10.1371/journal.pbio.0060184.
    1. Harper-Wynne C, Ross G, Sacks N, Salter J, Nasiri N, Iqbal J, A'Hern R, Dowsett M. Effects of the aromatase inhibitor letrozole on normal breast epithelial cell proliferation and metabolic indices in postmenopausal women: a pilot study for breast cancer prevention. Cancer Epidemiol Biomarkers Prev. 2002;11:614–621.
    1. Kaiser CL, Chapman BJ, Guidi JL, Terry CE, Mangiardi DA, Cotanche DA. Comparison of activated caspase detection methods in the gentamicin-treated chick cochlea. Hear Res. 2008;240:1–11. doi: 10.1016/j.heares.2008.03.003.
    1. da Silva BB, Pires CG, dos Santos AR, Castro-Leão AHd, Alencar AP, Lopes-Costa PV. Effects of raloxifene on Ki-67 and CD34 antigen expression in breast cancer. Gynecol Obstet Invest. 2009;67:103–108. doi: 10.1159/000165512.
    1. Chalkley H. Method for the quantitative morphologic analysis of tissues. J Natl Cancer Inst. 1943;4:47–53.
    1. Sato H, Kinoshita T, Takino T, Nakayama K, Seiki M. Activation of a recombinant membrane type 1-matrix metalloproteinase (MT1-MMP) by furin and its interaction with tissue inhibitor of metalloproteinases (TIMP)-2. FEBS Lett. 1996;393:101–104. doi: 10.1016/0014-5793(96)00861-7.
    1. Talmadge JE. Pathways mediating the expansion and immunosuppressive activity of myeloid-derived suppressor cells and their relevance to cancer therapy. Clin Cancer Res. 2007;13:5243–5248. doi: 10.1158/1078-0432.CCR-07-0182.
    1. Callegari AJ, Kelly TJ. Shedding light on the DNA damage checkpoint. Cell Cycle. 2007;6:660–666. doi: 10.4161/cc.6.6.3984.
    1. Dvory-Sobol H, Cohen-Noyman E, Kazanov D, Figer A, Birkenfeld S, Madar-Shapiro L, Benamouzig R, Arber N. Celecoxib leads to G2/M arrest by induction of p21 and down-regulation of cyclin B1 expression in a p53-independent manner. Eur J Cancer. 2006;42:422–426. doi: 10.1016/j.ejca.2005.11.009.
    1. Liu W, Chen Y, Wang W, Keng P, Finkelstein J, Hu D, Liang L, Guo M, Fenton B, Okunieff P, Ding I. Combination of radiation and celebrex (celecoxib) reduce mammary and lung tumor growth. Am J Clin Oncol. 2003;26:103–109.
    1. Dittmann KH, Mayer C, Ohneseit PA, Raju U, Andratschke NH, Milas L, Rodemann HP. Celecoxib induced tumor cell radiosensitization by inhibiting radiation induced nuclear EGFR transport and DNA-repair: a COX-2 independent mechanism. Int J Radiat Oncol Biol Phys. 2008;70:203–212. doi: 10.1016/j.ijrobp.2007.08.065.
    1. Fatima N, Yi M, Ajaz S, Stephens RM, Stauffer S, Greenwald P, Munroe DJ, Ali IU. Altered gene expression profiles define pathways in colorectal cancer cell lines affected by celecoxib. Cancer Epidemiol Biomarkers Prev. 2008;17:3051–3061. doi: 10.1158/1055-9965.EPI-08-0261.
    1. Auman JT, Church R, Lee SY, Watson MA, Fleshman JW, McLeod HL. Celecoxib pre-treatment in human colorectal adenocarcinoma patients is associated with gene expression alterations suggestive of diminished cellular proliferation. Eur J Cancer. 2008;44:1754–1760. doi: 10.1016/j.ejca.2008.05.010.
    1. Orlichenko L, Radisky D. Matrix metalloproteinases stimulate epithelial-mesenchymal transition during tumor development. Clin Exp Metastasis. 2008;25:593–600. doi: 10.1007/s10585-008-9143-9.
    1. Wang S, Liu Q, Zhang Y, Liu K, Yu P, Liu K, Luan J, Duan H, Lu Z, Wang F, Wu E, Yagasaki K, Zhang G. Suppression of growth, migration and invasion of highly-metastatic human breast cancer cells by berbamine and its molecular mechanisms of action. Mol Cancer. 2009;8:81. doi: 10.1186/1476-4598-8-81.
    1. Wang L, Chen W, Xie X, He Y, Bai X. Celecoxib inhibits tumor growth and angiogenesis in an orthotopic implantation tumor model of human colon cancer. Exp Oncol. 2008;30:42–51.
    1. Lang S, Lauffer L, Clausen C, Lohr I, Schmitt B, Holzel D, Wollenberg B, Gires O, Kastenbauer E, Zeidler R. Impaired monocyte function in cancer patients: restoration with a cyclooxygenase-2 inhibitor. FASEB J. 2003;17:286–288.
    1. Lonnroth C, Andersson M, Arvidsson A, Nordgren S, Brevinge H, Lagerstedt K, Lundholm K. Preoperative treatment with a non-steroidal anti-inflammatory drug (NSAID) increases tumor tissue infiltration of seemingly activated immune cells in colorectal cancer. Cancer Immun. 2008;8:5.
    1. Harper-Wynne CL, Sacks NP, Shenton K, MacNeill FA, Sauven P, Laidlaw IJ, Rayter Z, Miall S, Howes A, Salter J, Hills MJ, Lowe FM, A'Hern R, Nasiri N, Doody D, Iqbal J, Dowsett M. Comparison of the systemic and intratumoral effects of tamoxifen and the aromatase inhibitor vorozole in postmenopausal patients with primary breast cancer. J Clin Oncol. 2002;20:1026–1035. doi: 10.1200/JCO.20.4.1026.
    1. Smith IE, Dowsett M, Ebbs SR, Dixon JM, Skene A, Blohmer JU, Ashley SE, Francis S, Boeddinghaus I, Walsh G. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anastrozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin Oncol. 2005;23:5108–5116. doi: 10.1200/JCO.2005.04.005.
    1. Brueggemeier R, Quinn A, Parrett M, Joarder F, Harris R, Robertson F. Correlation of aromatase and cyclooxygenase gene expression in human breast cancer specimens. Cancer Lett. 1999;140:27–35. doi: 10.1016/S0304-3835(99)00050-6.
    1. Dowsett M, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, Ashley S, Francis S, Walsh G, Smith IE. Biomarker changes during neoadjuvant anastrozole, tamoxifen, or the combination: influence of hormonal status and HER-2 in breast cancer - a study from the IMPACT trialists. J Clin Oncol. 2005;23:2477–2492. doi: 10.1200/JCO.2005.07.559.
    1. Dowsett M, Bundred NJ, Decensi A, Sainsbury RC, Lu Y, Hills MJ, Cohen FJ, Veronesi P, O'Brien ME, Scott T, Muchmore DB. Effect of raloxifene on breast cancer cell Ki67 and apoptosis: a double-blind, placebo-controlled, randomized clinical trial in postmenopausal patients. Cancer Epidemiol Biomarkers Prev. 2001;10:961–966.
    1. Martin LA, Davies G, Weigel M, Betambeau N, Hills M, Salter J, Walsh G, A'Hern R, Dowsett M. Pre-surgical study of the biological effects of the selective cyclo-oxygenase-2 inhibitor celecoxib in patients with primary breast cancer. Breast Cancer Res Treat. 2010;123:829–836. doi: 10.1007/s10549-010-1100-z.
    1. Subbaramaiah K, Norton L, Gerald W, Dannenberg AJ. Cyclooxygenase-2 is overexpressed in HER-2/neu-positive breast cancer: evidence for involvement of AP-1 and PEA3. J Biol Chem. 2002;277:18649–18657. doi: 10.1074/jbc.M111415200.
    1. Dang CT, Dannenberg AJ, Subbaramaiah K, Dickler MN, Moasser MM, Seidman AD, D'Andrea GM, Theodoulou M, Panageas KS, Norton L, Hudis CA. Phase II study of celecoxib and trastuzumab in metastatic breast cancer patients who have progressed after prior trastuzumab-based treatments. Clin Cancer Res. 2004;10:4062–4067. doi: 10.1158/1078-0432.CCR-03-0463.
    1. Dirix LY, Ignacio J, Nag S, Bapsy P, Gomez H, Raghunadharao D, Paridaens R, Jones S, Falcon S, Carpentieri M, Abbattista A, Lobelle JP. Treatment of advanced hormone-sensitive breast cancer in postmenopausal women with exemestane alone or in combination with celecoxib. J Clin Oncol. 2008;26:1253–1259. doi: 10.1200/JCO.2007.13.3744.
    1. Falandry C, Canney PA, Freyer G, Dirix LY. Role of combination therapy with aromatase and cyclooxygenase-2 inhibitors in patients with metastatic breast cancer. Ann Oncol. 2009;20:615–620. doi: 10.1093/annonc/mdn693.
    1. Algra AM, Rothwell PM. Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol. 2012;13:518–527. doi: 10.1016/S1470-2045(12)70112-2.
    1. Rothwell PM, Price JF, Fowkes FG, Zanchetti A, Roncaglioni MC, Tognoni G, Lee R, Belch JF, Wilson M, Mehta Z, Meade TW. Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials. Lancet. 2012;379:1602–1612. doi: 10.1016/S0140-6736(11)61720-0.
    1. Snead D, Bell J, Dixon A, Nicholson R, Elston C, Blamey R, Ellis I. Methodology of immunohistological detection of oestrogen receptor in human breast carcinoma in formalin-fixed, paraffin-embedded tissue: a comparison with frozen section methodology. Histopathology. 1993;23:233–238. doi: 10.1111/j.1365-2559.1993.tb01195.x.

Source: PubMed

3
購読する