The PI discovered the first metastasis suppressor gene, nm23. Basic and translational research has investigated the role of Nm23 in the regulation of tumor metastasis. Eleven transfection studies have documented that overexpression of Nm23 in various tumor cell lines resulted in a 50-90% decrease in tumor metastatic potential in vivo. The biochemical mechanism whereby Nm23 suppresses metastatic potential is under investigation, based on microarray analysis of control- and Nm23-H1 transfectants. We have identified several genes that were down-regulated in nm23 transfectants, but not in transfectants of mutant forms of nm23 which failed to suppress in vitro motility, including c-met, CTGF, EDG2, FZD1, L1CAM, NETO2, PTN and SMOH. Confirmatory experiments included the down-regulation of the gene set in: (a) wild type as opposed to nm23-M1 knockout mouse hepatocellular carcinoma tissues; (b)independent MDA-MB-435 breast carcinoma transfectants; (c) nm23-H1 transfectants of another breast carcinoma cell line, MDA-MB-231; (d) an cohort of breast carcinomas, stratified by nm23 expression level. Transfection of each of the genes into nm23-suppressed MDA-MB-435 cells revealed that only EDG2,and to a lesser extent c-met, overcame nm23-H1 inhibition of motility. The data suggest that Nm23 inhibits tumor motility by down regulating the receptors to widely available factors such as LPA (EDG2) and HGF (Met). Metastasis experiments using EDG2 transfectants are underway. Translational research on Nm23 proposed that elevation of Nm23 expression in micrometastatic or overtly metastatic breast or other carcinomas may limit colonization, motility and de-differentiation, with a clinical benefit. Analysis of the nm23-H1 promoter revealed a 400 bp region which controlled expression, and contained a cassette of transcription factors regulated by a glucocorticoid response element (GRE). Deletion studies showed that these sites were functional in regulating nm23-H1 transcription. Medroxyprogesterone acetate (MPA), an unusual agonist for GR, as well as the androgen receptor and progesterone receptor, elevated Nm23-H1 expression of breast carcinoma cell lines in vitro. MPA acted via a post-transcriptional mechanism using the GR, at pharmacologic doses. We have reported preclinical experiments to determine if MPA can halt metastatic colonization. Mice were injected iv with metastatic human MDA-MB-231 breast carcinoma cells, and permitted to develop micrometastases for one month. Mice were then randomized to vehicle or MPA, the latter given in a one month induction and subsequent bimonthly maintenance dose. Mice receiving MPA had significantly fewer gross metastases in the lung, a smaller proportion of mice with metastases and smaller metastases. Immunohistochemistry revealed that MPA treated mice had a greater proportion of pulmonary metastases with high Nm23 expression. Side effects included weight gain, but no effects on bone mineral density or mammary histology. The data indicate that agents elevating metastsis suppressor gene expression may be effective against metastatic colonization. A Phase I trial of MPA will open at Indiana University, (PI Kathy Miller) in 2006, funded by an Avon-NCI grant to Dr. Miller. Our laboratory and that of Dr. Merino will participate in the trial.

Agency
National Institute of Health (NIH)
Institute
Division of Clinical Sciences - NCI (NCI)
Type
Intramural Research (Z01)
Project #
1Z01SC000892-23
Application #
7331352
Study Section
(LMP)
Project Start
Project End
Budget Start
Budget End
Support Year
23
Fiscal Year
2006
Total Cost
Indirect Cost
Name
Clinical Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Marino, Natascia; Collins, Joshua W; Shen, Changyu et al. (2014) Identification and validation of genes with expression patterns inverse to multiple metastasis suppressor genes in breast cancer cell lines. Clin Exp Metastasis 31:771-86
Steeg, Patricia S; Anderson, Robin L; Bar-Eli, Menashe et al. (2009) An open letter to the FDA and other regulatory agencies: Preclinical drug development must consider the impact on metastasis. Clin Cancer Res 15:4529
Lu, Jing; Steeg, Patricia S; Price, Janet E et al. (2009) Breast cancer metastasis: challenges and opportunities. Cancer Res 69:4951-3
Horak, Christine E; Lee, Jong Heun; Elkahloun, Abdel G et al. (2007) Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2. Cancer Res 67:7238-46
Athauda, Gagani; Giubellino, Alessio; Coleman, Jonathan A et al. (2006) c-Met ectodomain shedding rate correlates with malignant potential. Clin Cancer Res 12:4154-62
Palmieri, Diane; Horak, Christine E; Lee, Jong-Heun et al. (2006) Translational approaches using metastasis suppressor genes. J Bioenerg Biomembr 38:151-61
Steeg, Patricia S (2006) Tumor metastasis: mechanistic insights and clinical challenges. Nat Med 12:895-904
Salerno, Massimiliano; Palmieri, Diane; Bouadis, Amina et al. (2005) Nm23-H1 metastasis suppressor expression level influences the binding properties, stability, and function of the kinase suppressor of Ras1 (KSR1) Erk scaffold in breast carcinoma cells. Mol Cell Biol 25:1379-88
Steeg, Patricia S (2005) New insights into the tumor metastatic process revealed by gene expression profiling. Am J Pathol 166:1291-4
Palmieri, Diane; Halverson, Douglas O; Ouatas, Taoufik et al. (2005) Medroxyprogesterone acetate elevation of Nm23-H1 metastasis suppressor expression in hormone receptor-negative breast cancer. J Natl Cancer Inst 97:632-42

Showing the most recent 10 out of 19 publications