Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 305(3): 1222-32, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12649301

ABSTRACT

CC-4047 (Actimid) and CC-5013 (Revimid) belong to a class of thalidomide analogs collectively known as the immunomodulatory drugs (IMiDs), which are currently being assessed in the treatment of patients with multiple myeloma and other cancers. IMiDs potently enhance T cell and natural killer cell responses and inhibit tumor necrosis factor-alpha, interleukin (IL)-1 beta, and IL-12 production from LPS-stimulated peripheral blood mononuclear cells. However, the molecular mechanism of action for these compounds is unknown. Herein, we report on the ability of the IMiDs to up-regulate production of IL-2 from activated human CD4+ and CD8+ peripheral blood T cells, production of IL-2 and IFN-gamma from T helper (Th)1-type cells, and production of IL-5 and IL-10 from Th2-type cells. Elevation of IL-2 production from Jurkat T cells was observed as early as 6 h poststimulation and correlated with an increase in IL-2 promoter activity that was dependent upon the proximal but not the distal AP-1 binding site. The IMiDs enhanced AP-1-driven transcriptional activity 2- to 4-fold after 6 h of T cell stimulation, and their relative potencies for AP-1 activation correlated with their potencies for increased IL-2 production in Jurkat T cells and in CD4+ or CD8+ human peripheral blood T cells. The most potent of these IMiDs, CC-4047, had no effect on nuclear factor of activated T cells transcriptional activity, calcium signaling, or phosphorylation of extracellular signal-regulated kinase 1/2, c-Jun NH2-terminal kinase 1/2, p38 mitogen-activated protein kinase, or c-Jun/Jun D in Jurkat T cells. These data suggest that IMiDs increase T cell cytokine production by potentiating AP-1 transcriptional activity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cytokines/biosynthesis , Proto-Oncogene Proteins , T-Lymphocytes/drug effects , Thalidomide/pharmacology , Transcription Factor AP-1/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcineurin/metabolism , Calcium Signaling , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Interleukin-2/biosynthesis , JNK Mitogen-Activated Protein Kinases , Janus Kinase 2 , Jurkat Cells , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Thalidomide/analogs & derivatives , p38 Mitogen-Activated Protein Kinases
2.
Cancer Res ; 63(3): 593-9, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12566301

ABSTRACT

Thalidomide is clinically useful in a number of cancers. Antitumor activity may be related to a number of known properties, including anti-tumor necrosis factor (TNF)-alpha and T-cell costimulatory and antiangiogenic activities. However, it may also involve direct antitumor effects. A series of second generation thalidomide analogues have been separated into two distinct groups of compounds, each with enhanced therapeutic potential, i.e., SelCIDs, which are phosphodiesterase (PDE) type IV inhibitors, and IMiDs, which have unknown mechanism(s) of action. We report here our efforts to determine direct antitumor effects of thalidomide and compounds from both groups. We found that one of the SelCID analogues (SelCID-3) was consistently effective at reducing tumor cell viability in a variety of solid tumor lines but had no effect on non-neoplastic cells. The antitumor activity was independent of known PDE4 inhibitory activity and did not involve cAMP elevation. Growth arrest was preceded by the early induction of G(2)-M cell cycle arrest, which led to caspase 3 mediated apoptosis. This was associated with increased expression of pro-apoptotic proteins and decreased expression of antiapoptotic bcl-2. Furthermore, extensive apoptosis in vivo was detected during SelCID-3-mediated inhibition of tumor growth in a murine xenotransplantation cancer model. Our results suggest that SelCID-3 represents a novel antitumor agent distinct from thalidomide and from previously characterized analogues with therapeutic potential against a range of solid tumors. This effect appears to be mediated via alterations in the expression of bcl-2 family proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Animals , Apoptosis/physiology , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Cycle/drug effects , Cell Division/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclic AMP/biosynthesis , Female , Humans , Male , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/biosynthesis , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...