TY - JOUR T1 - Raloxifene-Induced Myeloma Cell Apoptosis: A Study of Nuclear Factor-κB Inhibition and Gene Expression Signature JF - Molecular Pharmacology JO - Mol Pharmacol SP - 1615 LP - 1623 DO - 10.1124/mol.105.020479 VL - 69 IS - 5 AU - Sabine Olivier AU - Pierre Close AU - Emilie Castermans AU - Laurence de Leval AU - Sebastien Tabruyn AU - Alain Chariot AU - Michel Malaise AU - Marie-Paule Merville AU - Vincent Bours AU - Nathalie Franchimont Y1 - 2006/05/01 UR - http://molpharm.aspetjournals.org/content/69/5/1615.abstract N2 - Because multiple myeloma remains associated with a poor prognosis, novel drugs targeting specific signaling pathways are needed. The efficacy of selective estrogen receptor modulators for the treatment of multiple myeloma is not well documented. In the present report, we studied the antitumor activity of raloxifene, a selective estrogen receptor modulator, on multiple myeloma cell lines. Raloxifene effects were assessed by tetrazolium salt reduction assay, cell cycle analysis, and Western blotting. Mobility shift assay, immunoprecipitation, chromatin immunoprecipitation assay, and gene expression profiling were performed to characterize the mechanisms of raloxifene-induced activity. Indeed, raloxifene, as well as tamoxifen, decreased JJN-3 and U266 myeloma cell viability and induced caspase-dependent apoptosis. Raloxifene and tamoxifen also increased the cytotoxic response to vincristine and arsenic trioxide. Moreover, raloxifene inhibited constitutive nuclear factor-κB (NF-κB) activity in myeloma cells by removing p65 from its binding sites through estrogen receptor α interaction with p65. It is noteworthy that microarray analysis showed that raloxifene treatment decreased the expression of known NF-κB-regulated genes involved in myeloma cell survival and myeloma-induced bone lesions (e.g., c-myc, mip-1α, hgf, pac1,...) and induced the expression of a subset of genes regulating cellular cycle (e.g., p21, gadd34, cyclin G2,...). In conclusion, raloxifene induces myeloma cell cycle arrest and apoptosis partly through NF-κB-dependent mechanisms. These findings also provide a transcriptional profile of raloxifene treatment on multiple myeloma cells, offering the framework for future studies of selective estrogen receptor modulators therapy in multiple myeloma. ER -