Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Mol Cancer Ther ; 6(1): 112-21, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237271

ABSTRACT

Atiprimod is a novel anticancer and antiangiogenic drug candidate which is currently being evaluated in patients with liver carcinoid and multiple myeloma. In this study, we report that atiprimod selectively inhibited proliferation and induced apoptosis in HCC cells that expressed either hepatitis B virus (HBV) or hepatitis C virus, through deactivation of protein kinase B (Akt) and signal transducers and activators of transcription 3 (STAT3) signaling. In HepG2 AD38 cells, which express HBV genome under the control of a tetracycline-off promoter, both Akt and STAT3 were constitutively activated in response to HBV expression. However, this constitutive activation was not sensitive to lamivudine, a drug that inhibits HBV replication without affecting its gene expression, suggesting that HBV replication per se might not be responsible for the activation. Interestingly, the electrophoretic mobility of p-STAT3 protein bands on immunoblot was slower when AD38 cells were cultured in the absence of tetracycline, suggesting a differential phosphorylation in response to HBV expression. In HCC cells, interleukin 6 stimulates the phosphorylation of STAT3 both at serine 727 and at tyrosine 705 positions. The interleukin 6-stimulated activation of STAT3 and Akt was inhibited not only by atiprimod but also by LY294002, a phosphoinositide-3-kinase-specific inhibitor, and by NS398, a cyclooxygenase-2-selective inhibitor. The combination of these compounds did not produce any additive effect, implying that the mechanisms by which HBV activates Akt and STAT3 might also involve phosphoinositide-3-kinase and cyclooxygenase-2. Collectively, these results suggest that atiprimod could be useful as a multifunctional drug candidate for the treatment of HCC in humans.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Spiro Compounds/pharmacology , Carcinoma, Hepatocellular/virology , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation, Viral/drug effects , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , Interleukin-6/pharmacology , Liver Neoplasms/virology , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Virus Replication/drug effects
2.
J Exp Ther Oncol ; 4(4): 267-79, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15844657

ABSTRACT

Atiprimod, a novel compound belonging to the azaspirane class of cationic amphiphilic drugs, exhibits both anti-proliferative and anti-angiogenic activities. Atiprimod inhibited proliferation of all human cancer cell lines included in the National Cancer Institute panel with IC50 values in the low micromolar range. Notably, metastatic cell lines were more sensitive to the compound compared to the non-metastatic cell lines derived from the same tumor tissue types. Atiprimod also induced apoptosis and activated both caspase-9 and caspase-3 in T84 colon carcinoma cells. Hence, the anti-proliferative activity could partly be due to its pro-apoptotic activity. Regarding angiogenesis in vitro, atiprimod inhibited both bFGF and VEGF induced proliferation and migration of human umbilical vein endothelial cells (HUVECs), resulting in disruption of cord formation. In addition, atiprimod also suppressed formation of new blood vessels in a chorioallantoic membrane assay. Previous studies have also shown that atiprimod treatment reduced production of IL-6, VEGF and inhibited activation of Stat3, a constitutively activated protein in majority of human cancers. Together these findings suggest that atiprimod acts on several molecules that are essential for tumor growth, invasion and metastasis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic , Spiro Compounds/pharmacology , Apoptosis , Caspase 3 , Caspase 9 , Caspases/metabolism , Cations , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Collagen/pharmacology , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Drug Combinations , Endothelium, Vascular/cytology , Enzyme Activation , Fibroblast Growth Factor 2/metabolism , Humans , Inhibitory Concentration 50 , Laminin/pharmacology , Models, Chemical , Neoplasm Invasiveness , Neoplasm Metastasis , Proteoglycans/pharmacology , STAT3 Transcription Factor , Trans-Activators/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Blood ; 105(11): 4470-6, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15705788

ABSTRACT

Azaspirane (N-N-diethyl-8,8-dipropyl-2-azaspiro [4.5] decane-2-propanamine; trade name, Atiprimod) is an orally bioavailable cationic amphiphilic compound that significantly inhibits production of interleukin 6 (IL-6) and inflammation in rat arthritis and autoimmune animal models. We here characterize the effect of atiprimod on human multiple myeloma (MM) cells. Azaspirane significantly inhibited growth and induced caspase-mediated apoptosis in drug-sensitive and drug-resistant MM cell lines, as well as patient MM cells. IL-6, insulin-like growth factor 1 (IGF-1), or adherence of MM cells to bone marrow stromal cells (BMSCs) did not protect against atiprimod-induced apoptosis. Both conventional (dexamethasone, doxorubicin, melphalan) and novel (arsenic trioxide) agents augment apoptosis induced by atiprimod. Azaspirane inhibits signal transducer activator of transcription 3 (STAT3) and a PI3-K (phosphatidylinositol 3-kinase) target (Akt), but not extracellular signal-regulated kinase 1 and 2 (ERK1/2), inhibits phosphorylation triggered by IL-6, and also inhibits inhibitorkappaBalpha (IkappaBalpha) and nuclear factor kappaB (NFkappaB) p65 phosphorylation triggered by tumor necrosis factor alpha (TNF-alpha). Of importance, azaspirane inhibits both IL-6 and vascular endothelial growth factor (VEGF) secretion in BMSCs triggered by MM cell binding and also inhibits angiogenesis on human umbilical vein cells (HUVECs). Finally, azaspirane demonstrates in vivo antitumor activity against human MM cell growth in severe combined immunodeficient (SCID) mice. These results, therefore, show that azaspirane both induces MM cell apoptosis and inhibits cytokine secretion in the BM milieu, providing the framework for clinical trials to improve patient outcome in MM.


Subject(s)
Bone Marrow/pathology , Multiple Myeloma/drug therapy , Spiro Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Communication , Cell Proliferation/drug effects , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Mice , Mice, SCID , Multiple Myeloma/pathology , Neovascularization, Physiologic/drug effects , Signal Transduction/drug effects , Spiro Compounds/therapeutic use , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL