Death-Associated Protein Kinase Loss of Expression Is a New Marker for Breast Cancer Prognosis

Purpose: Death-associated protein (DAP)-kinase is a new Ser/Thr kinase involved in cell apoptosis and tumor suppression, the expression of which has been correlated to invasive potential and metastasis in several human neoplastic tissues. We analyzed the level of DAP-kinase expression in breast cancer specimens and its correlation with survival. Experimental Design: One hundred twenty-eight breast cancer specimens were analyzed by immunohistochemistry. Patient records were studied retrospectively for demographic characteristics, clinical data, hormonal treatment, outcome, and survival. DAP-kinase protein expression was also studied in normal breast cells primary cultures under estrogen and antiestrogen treatment. Results: Among the 128 patients, 30 showed a DAP-kinase staining ≤ 20%, whereas 98 had a staining over 20%. Mean follow-up time was 62 months. The association between tumor Scarff-Bloom and Richardson grade (P = 0.009), estrogen receptor and progesterone receptor expression (P = 0.002 and 0.001, respectively), tumor size (P = 0.05), Bcl-2 expression (P = 0.004), and DAP-kinase immunostaining in the ductal carcinoma group was highly significant. Overall (64 months) and disease-free (63 months) survival in the high DAP-kinase expression group were significantly longer compared with the women whose tumors showed a loss of DAP-kinase expression (51 and 43 months, respectively). DAP-kinase protein was strongly expressed in normal breast tissue and in human breast epithelial cells primary cultures. Estradiol decreased DAP-kinase expression in these cells, arguing for hormonal regulation of the protein. Conclusions: Loss of DAP-kinase expression negatively correlates to survival and positively correlates to the probability of recurrence in a very significant manner. DAP-kinase thus constitutes a novel and independent prognosis marker for breast cancer.

[1]  William Rostène,et al.  Antiestrogens are pro‐apoptotic in normal human breast epithelial cells , 2003, International journal of cancer.

[2]  Michael J Thun,et al.  Long‐term trends in cancer mortality in the United States, 1930–1998 , 2003, Cancer.

[3]  U. Lehmann,et al.  Promoter hypermethylation of the death-associated protein kinase gene in breast cancer is associated with the invasive lobular subtype. , 2002, Cancer research.

[4]  Ruey-Hwa Chen,et al.  DAP-kinase induces apoptosis by suppressing integrin activity and disrupting matrix survival signals , 2002, The Journal of cell biology.

[5]  W. Farrell,et al.  Preferential loss of Death Associated Protein kinase expression in invasive pituitary tumours is associated with either CpG island methylation or homozygous deletion , 2002, Oncogene.

[6]  H. Ng,et al.  Frequent death-associated protein kinase promoter hypermethylation in multiple myeloma. , 2001, Clinical cancer research : an official journal of the American Association for Cancer Research.

[7]  A. Kimchi,et al.  DAP-kinase: from functional gene cloning to establishment of its role in apoptosis and cancer , 2001, Cell Death and Differentiation.

[8]  William Rostène,et al.  Hormonal regulation of apoptosis in breast cells and tissues , 2000, Steroids.

[9]  F. Khuri,et al.  Hypermethylation of the death-associated protein (DAP) kinase promoter and aggressiveness in stage I non-small-cell lung cancer. , 2000, Journal of the National Cancer Institute.

[10]  Gustavo Droguett,et al.  DAP kinase activates a p19ARF/p53-mediated apoptotic checkpoint to suppress oncogenic transformation , 2000, Nature Cell Biology.

[11]  J. Herman,et al.  Hypermethylation of the DAP-kinase CpG island is a common alteration in B-cell malignancies. , 1999, Blood.

[12]  M. Inganäs,et al.  Prognostic value of P53 gene mutations in a large series of node-negative breast cancer patients. , 1998, Cancer research.

[13]  S. Polak‐Charcon,et al.  DAP kinase links the control of apoptosis to metastasis , 1997, Nature.

[14]  Peter A. Jones,et al.  DAP-kinase loss of expression in various carcinoma and B-cell lymphoma cell lines: possible implications for role as tumor suppressor gene , 1997, Oncogene.

[15]  A. Kimchi,et al.  DAP‐kinase is a Ca2+/calmodulin‐dependent, cytoskeletal‐associated protein kinase, with cell death‐inducing functions that depend on its catalytic activity , 1997, The EMBO journal.

[16]  A. Gompel,et al.  Antagonism between estradiol and progestin on Bcl‐2 expression in breast‐cancer cells , 1996, International journal of cancer.

[17]  Z. Darżynkiewicz Apoptosis in anititumor strategies: Modulation of cell cycle or differentiation , 1995, Journal of cellular biochemistry.

[18]  A. Kimchi,et al.  Identification of a novel serine/threonine kinase and a novel 15-kD protein as potential mediators of the gamma interferon-induced cell death. , 1995, Genes & development.

[19]  K. Keyomarsi,et al.  Synchronization of tumor and normal cells from G1 to multiple cell cycles by lovastatin. , 1991, Cancer research.

[20]  I. Mowszowicz,et al.  Estradiol and progesterone receptors in cultured normal human breast epithelial cells and fibroblasts: immunocytochemical studies. , 1991, The Journal of clinical endocrinology and metabolism.

[21]  P. Spritzer,et al.  Progestin effect on cell proliferation and 17 beta-hydroxysteroid dehydrogenase activity in normal human breast cells in culture. , 1986, The Journal of clinical endocrinology and metabolism.

[22]  Ruth P. Mack,et al.  Long-Term Trends , 2011 .