Elsevier

Gynecologic Oncology

Volume 94, Issue 3, September 2004, Pages 685-692
Gynecologic Oncology

Methylation of tumor suppressor gene p16 and prognosis of epithelial ovarian cancer

https://doi.org/10.1016/j.ygyno.2004.06.018Get rights and content

Abstract

Objective. Methylation of p16 promoter was evaluated in ovarian cancer to determine the role of p16 methylation in ovarian cancer prognosis.

Methods. Two hundred and forty-nine patients with primary epithelial ovarian cancer were selected for the study; these patients were followed for a median of 31 months. Genomic DNA extracted from fresh frozen tumor tissues were treated with sodium bisulfite and were analyzed for p16 methylation using methylation-specific PCR (MSP). Cox regression survival analysis was performed to examine the associations of p16 methylation with progression-free and overall survivals.

Results. Of the 249 patients, 100 (40%) were tested positive for p16 promoter methylation. The status of p16 methylation did not change significantly with patient age, disease stage, histological grade, residual tumor size, and debulking results, although p16 methylation seemed to occur more often in patients with advanced diseases or aggressive tumors. Compared to those without p16 methylation, patients with p16 methylation had significantly higher risk for disease progression (P = 0.01). The relative risk for progression was 1.69 (95% CI: 1.12–2.54), and the association remained statistically significant (RR = 1.54, 95% CI: 1.01–2.34) after adjusting for clinical and pathological variables. The risk for death was also higher in methylation positive patients than in methylation negative patients (RR = 1.33, 95% CI: 0.88–2.00), but the difference was not statistically significant.

Conclusion. The study suggests that promoter methylation in the p16 gene is associated with ovarian cancer progression, and evaluation of p16 methylation may have values in predicting ovarian cancer prognosis.

Introduction

Tumor suppressor gene p16 (INK4A/MTS1/CDKN2) encodes a 15.8-kDa protein that interacts with cyclin-dependent kinase CDK4 and CDK6. CDK4 and CDK6 bind to cylcin D regulating cell cycle progression from G1 to S phases. Binding of p16 to CDKs inhibits the formation of CDK/cyclin D complex, resulting in cell cycle arrest at G1 phase. The role of p16 in cell cycle control is believed to be involved in cancer development and progression [1], [2], [3], [4], [5]. Loss of p16 expression has been observed in many types of cancer [2]. Genetic and epigenetic alterations, including homozygous deletion, point mutation, and promoter methylation, are considered to be responsible for the loss of p16 activity [5], [6].

Promoter methylation has been recognized as an important mechanism in regulation of gene expression [7], [8]. Aberrant methylation of p16 promoter has been found in many forms of cancer, including ovarian cancer [5]. Clinical studies indicate that p16 expression is undetectable in about a third of ovarian cancer cases [9], [10], [11], [12] and that patients with low p16 expression have poor responses to chemotherapy and unfavorable survival outcome [11], [13]. Cell culture experiments demonstrate that reintroducing functional p16 into p16-null ovarian cancer cells results in inhibition of cell growth and increases in apoptosis, suggesting that p16 plays a role in ovarian cancer progression [14].

Epithelial ovarian cancer is one of the most lethal malignancies; 5-year survival of patients with the disease is only 50% [15], [16]. To improve our understanding of ovarian cancer progression and to search for molecular markers for prediction of ovarian cancer prognosis, we analyzed p16 promoter methylation in epithelial ovarian cancer tissue and examined its association with patient progression-free and overall survival.

Section snippets

Study subjects

Between October 1991 and February 2000, 270 patients underwent surgery for ovarian cancer in the Department of Gynecology, Gynecologic Oncology Unit, at University of Turin in Italy. Of the 270 patients, 18 had borderline tumors (histological grade 0) and 2 patients had metastatic cancer to the ovary. One patient did not have sufficient tissue specimen for analysis. The final number of patients included in this study was 249, all of who had primary epithelial ovarian cancer. The age of these

Results

Of the 249 tumor samples analyzed, 100 were found to be positive for p16 promoter methylation, which accounted for 40% of the samples. Patient age at diagnosis was not significantly different between patients with and without p16 methylation. The mean age was 57.8 years for patients with methylation and 56.9 years for those without methylation (P = 0.576). Table 1 shows the distributions of p16 methylation in relation to clinical and pathological variables. Compared to patients without p16

Discussion

In this study, we found p16 promoter methylation in 40% of epithelial ovarian cancer. Compared to those without p16 methylation, patients with p16 methylation had a 50% increase in risk for disease progression. Moreover, the increased risk for disease progression was independent from clinical and pathological factors, suggesting that p16 methylation may have values in predicting ovarian cancer prognosis. The study findings seem to support the notion that p16 methylation in ovarian cancer is

Acknowledgements

Drs. D. Katsaros, S. Fracchioli, and I.A. Rigault de la Longrais are partially supported by AIRC (Italian Association for Cancer Research).

References (39)

  • T.U. Ha et al.

    Mullerian inhibiting substance inhibits ovarian cell growth through an Rb-independent mechanism

    J. Biol. Chem.

    (2000 (Nov. 24))
  • W.H. Liggett et al.

    Role of the p16 tumor suppressor gene in cancer

    J. Clin. Oncol.

    (1998 (Mar.))
  • M. Serrano

    The INK4a/ARF locus in murine tumorigenesis

    Carcinogenesis

    (2000 (May))
  • M. Serrano et al.

    A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4

    Nature

    (1993 (Dec. 16))
  • G.I. Shapiro et al.

    Multiple mechanisms of p16INK4A inactivation in non-small cell lung cancer cell lines

    Cancer Res.

    (1995 Dec 15)
  • A.R. Karpf et al.

    Reactivating the expression of methylation silenced genes in human cancer

    Oncogene

    (2002 Aug 12)
  • S. Marchini et al.

    Absence of deletions but frequent loss of expression of p16INK4 in human ovarian tumours

    Br. J. Cancer

    (1997)
  • T. Kusume et al.

    The p16-cyclin D1/CDK4-pRb pathway and clinical outcome in epithelial ovarian cancer

    Clin. Cancer Res.

    (1999 (Dec.))
  • K. Kudoh et al.

    Inactivation of p16/CDKN2 and p15/MTS2 is associated with prognosis and response to chemotherapy in ovarian cancer

    Int. J. Cancer

    (2002 (Jun. 1))
  • Cited by (56)

    • Use of Chromatin Changes as Biomarkers

      2016, Chromatin Signaling and Diseases
    • Investigating the effects of the presence of foreign DNA on DNA methylation and DNA repair events in cultured eukaryotic cells

      2013, Gene
      Citation Excerpt :

      The gene product associates with cyclin-dependant kinases CDK4 and CDK6, causing arrest in the cell cycle in the G1 phase. This would encourage growth of cells instead of dividing into daughter cells (Katsaros, et al., 2004). As this promoter region shows no methylation it would be expected that the gene would be over-expressed.

    • Frequent promoter hypermethylation of TGFBI in epithelial ovarian cancer

      2010, Gynecologic Oncology
      Citation Excerpt :

      In the field of ovarian cancer research, there has been continuous effort to explore the relevant hypermethylated genes as biomarkers for prediction of cancer risk or prognosis. Many researchers have reported the methylation frequency of several genes (14-3-3 sigma, 73.5%; RASSF1A, 41%; p16, 40%; HIC1, 35%), which are significantly more methylated in cancer tissue compared to normal tissues [18,33–35]. Others have reported that methylated genes such as hMLH1 and FANCF may be useful to predict platinum-based chemosensitivity in ovarian cancer [36,37].

    • Could cannabinoids provide a new hope for ovarian cancer patients?

      2023, Pharmacology Research and Perspectives
    View all citing articles on Scopus
    View full text