Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Surg Res ; 164(1): 116-25, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19726061

ABSTRACT

INTRODUCTION: CC-5079, a small molecule inhibitor of tubulin polymerization and phosphodiesterase-4 activity, was evaluated for antiangiogenic and antitumor activities. MATERIALS AND METHODS: First, CC-5079 in vitro activity on human umbilical vein endothelial cells (HUVECs), fibroblasts, and MC38 were evaluated by proliferation, migration, and invasion assays. Second, CC-5079 effect on microvessel formation was evaluated ex vivo by chick chorioallantoic membrane (CAM), rat aortic rings assays, and with directed in vivo angiogenesis assay (DIVAA). Third, CC-5079 antitumor effect was determined in treatment of C57BL/6 mice with MC38 tumors. Finally, CC-5079 modulation of MKP1 in HUVECs, human fibroblast, and MC38 were determined by RNA isolation for qRT-PCR. RESULTS: At the 0.1 µM concentration CC-5079 significantly inhibited HUVEC, fibroblast, and MC38 proliferation and migration (all P < 0.001). At the 0.1 µM concentration, CC-5079 also inhibited HUVEC invasion (P < 0.05) but not fibroblast. In the CAM and rat aortic ring assays, CC-5079 at 0.1 µM inhibited microvessel formation (P < 0.05). By DIVAA, CC-5079 at 1 mg/kg/d continuous delivered inhibited microvessel formation (P < 0.05). Intraperitoneal CC-5079 was well tolerated and inhibited the growth of subcutaneous MC38 at 100 mg/kg/d (P < 0.01). By qRT-PCR, CC-5079 stimulated MKP1 expression in HUVEC and fibroblast. CONCLUSION: CC-5079 demonstrated stimulation of MKP1, antiangiogenic, and antitumor properties.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Dual Specificity Phosphatase 1/metabolism , Nitriles/pharmacology , Angiogenesis Inhibitors/chemistry , Animals , Antineoplastic Agents/chemistry , Aorta/cytology , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chickens , Chorioallantoic Membrane/cytology , Colonic Neoplasms/metabolism , Dual Specificity Phosphatase 1/genetics , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Nitriles/chemistry , Rats , Umbilical Veins/cytology
2.
Microvasc Res ; 77(2): 78-86, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18805433

ABSTRACT

Lenalidomide (Revlimid) is approved for the treatment of transfusion-dependent patients with anemia due to low- or intermediate-1-risk Myelodysplastic Syndromes (MDS) associated with a del 5q cytogenetic abnormality with or without additional cytogenetic abnormalities, and in combination with dexamethasone for the treatment of multiple myeloma patients who have received at least one prior therapy. Previous reports suggest that lenalidomide is anti-angiogenic and this property appears to be related to efficacy in patients with MDS. We have investigated the effect of lenalidomide on the formation of microvessels in a novel in vitro angiogenesis assay utilizing human umbilical arterial rings and in a capillary-like cord formation assay using cultured primary endothelial cells. We found that lenalidomide consistently inhibits both sprout formation by arterial rings and cord formation by endothelial cells in a dose-dependent manner. We also found an inhibitory effect of lenalidomide on the associations between cadherin 5, beta-catenin and CD31, adherens junction proteins whose interaction is critical for endothelial cell cord formation. Furthermore, lenalidomide inhibited VEGF-induced PI3K-Akt pathway signaling, which is known to regulate adherens junction formation. We also found a strong inhibitory effect of lenalidomide on hypoxia-induced endothelial cell formation of cords and HIF-1 alpha expression, the main mediator of hypoxia-mediated effects and a key driver of angiogenesis and metastasis. Anti-metastatic activity of lenalidomide in vivo was confirmed in the B16-F10 mouse melanoma model by a >40% reduction in melanoma lung colony counts versus untreated mice. Our results suggest that inhibitory effects on microvessel formation, in particular adherens junction formation and inhibition of hypoxia-induced processes support a potential anti-angiogenic and anti-metastatic mechanism for this clinically active drug.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/prevention & control , Thalidomide/analogs & derivatives , Adherens Junctions/drug effects , Animals , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Lenalidomide , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Mice , Microcirculation/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Thalidomide/pharmacology , Umbilical Arteries/drug effects , Umbilical Arteries/growth & development , beta Catenin/metabolism
3.
Cancer Res ; 66(2): 951-9, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424030

ABSTRACT

We have found that the synthetic compound CC-5079 potently inhibits cancer cell growth in vitro and in vivo by a novel combination of molecular mechanisms. CC-5079 inhibits proliferation of cancer cell lines from various organs and tissues at nanomolar concentrations. Its IC(50) value ranges from 4.1 to 50 nmol/L. The effect of CC-5079 on cell growth is associated with cell cycle arrest in G(2)-M phase, increased phosphorylation of G(2)-M checkpoint proteins, and apoptosis. CC-5079 prevents polymerization of purified tubulin in a concentration-dependent manner in vitro and depolymerizes microtubules in cultured cancer cells. In competitive binding assays, CC-5079 competes with [(3)H]colchicine for binding to tubulin; however, it does not compete with [(3)H]paclitaxel (Taxol) or [(3)H]vinblastine. Our data indicate that CC-5079 inhibits cancer cell growth with a mechanism of action similar to that of other tubulin inhibitors. However, CC-5079 remains active against multidrug-resistant cancer cells unlike other tubulin-interacting drugs, such as Taxol and colchicine. Interestingly, CC-5079 also inhibits tumor necrosis factor-alpha (TNF-alpha) secretion from lipopolysaccharide-stimulated human peripheral blood mononuclear cells (IC(50), 270 nmol/L). This inhibitory effect on TNF-alpha production is related to its inhibition of phosphodiesterase type 4 enzymatic activity. Moreover, in a mouse xenograft model using HCT-116 human colorectal tumor cells, CC-5079 significantly inhibits tumor growth in vivo. In conclusion, our data indicate that CC-5079 represents a new chemotype with novel mechanisms of action and that it has the potential to be developed for neoplastic and inflammatory disease therapy.


Subject(s)
Nitriles/pharmacology , Tubulin/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Neoplasms/pathology , Transplantation, Heterologous , Tubulin Modulators/pharmacology , Tumor Cells, Cultured
4.
J Interferon Cytokine Res ; 25(10): 604-16, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16241859

ABSTRACT

Immunomodulatory drugs (IMiDs) are orally available small molecules that potently inhibit tumor necrosis factor-alpha (TNF-alpha) production by lipopolysaccharide (LPS)-stimulated human peripheral blood mononuclear cells (HuPBMCs) but enhance secretion of such cytokines as interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) by stimulated T cells. The mechanism of cytokine regulation by IMiDs has not yet been determined. In the present study, we investigated the effects of one of the IMiDs, CC-4047 (Actimid, Celgene, Warren, NJ), on synthesis of IL-2 protein and mRNA and on the activity and expression of transcription factors. Treatment with CC-4047 enhances the secretion of IL-2 protein and the expression of IL-2 mRNA in a dose-dependent and time-dependent manner. In T cells stimulated with phorbol myristate acetate (PMA)/ionomycin, CC-4047 enhanced the DNA-binding activity of activated protein-1 (AP-1) but not NF-kappaB, Octomer-1 (OCT-1), or NFAT by 2-fold and 4-fold after an incubation time of 1 and 3 h, respectively. Luciferase reporter assays in Jurkat cells showed similar effects on transcription factor activity. Using in vitro kinase activity assays, we also showed that CC-4047 enhances the activity of protein kinase C-theta (PKC-theta) in stimulated T cells. The secreted IL-2 from HuPBMCs was shown to activate natural killer (NK) cells to lyse their target cell line K562. Taken together, our results demonstrate that the IMiDs exert their effects at least in part by activating PKC-theta and acting on AP-1 DNA-binding activity in T cells, resulting in augmented IL-2 synthesis and activation of IL- 2-dependent downstream effectors, such as NK cells.


Subject(s)
Immunologic Factors/pharmacology , Interleukin-2/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , T-Lymphocytes/enzymology , Thalidomide/analogs & derivatives , Transcription Factors/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , K562 Cells , Killer Cells, Natural/enzymology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Protein Kinase C-theta , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thalidomide/pharmacology
5.
Cell Immunol ; 230(2): 81-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15598423

ABSTRACT

Immunomodulatory drugs (IMiDs) are potent inhibitors of TNF-alpha and IL-1beta and elevators of IL-10 production in LPS-stimulated human PBMC. They are currently in clinical trials for various diseases, including multiple myeloma, myelodysplastic syndrome, and melanoma. In the present study, we have investigated the effects of thalidomide, CC-5013 and CC-4047 on the expression of COX-2 by stimulated PBMC. Our results show that thalidomide and IMiDs inhibited the expression of COX-2 but not the COX-1 protein in LPS-TNF-alpha and IL-1beta stimulated PBMC and shortened the half-life of COX-2 mRNA in a dose-dependent manner. They also inhibited the synthesis of prostaglandin E2 from LPS-stimulated PBMC. While anti-TNF-alpha or IL-1beta neutralizing antibodies had no effect on COX-2 expression, anti-IL-10 neutralizing antibody elevated the expression of COX-2 mRNA, and protein from treated PBMC. These data suggest that the anti-inflammatory and anti-tumor effects of IMiDs may be due in part to elevation of IL-10 production and its subsequent inhibition of COX-2 expression.


Subject(s)
Immunologic Factors/pharmacology , Interleukin-10/immunology , Isoenzymes/antagonists & inhibitors , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Thalidomide/analogs & derivatives , Blotting, Northern , Blotting, Western , Cyclooxygenase 2 , Dinoprostone/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-10/genetics , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/immunology , Lenalidomide , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Lymphotoxin-alpha/immunology , Lymphotoxin-alpha/pharmacology , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/immunology , RNA/chemistry , RNA/genetics , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
6.
J Transl Med ; 2(1): 4, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14754458

ABSTRACT

Reliable quantitative evaluation of molecular pathways is critical for both drug discovery and treatment monitoring. We have modified the CAM assay to quantitatively measure vascular density, endothelial proliferation, and changes in protein expression in response to anti-angiogenic and pro-angiogenic agents. This improved CAM assay can correlate changes in vascular density with changes seen on a molecular level. We expect that these described modifications will result in a single in vivo assay system, which will improve the ability to investigate molecular mechanisms underlying the angiogenic response.

7.
Cancer Res ; 63(23): 8073-8, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678955

ABSTRACT

Selective cytokine inhibitory drugs (SelCIDs) are a novel class of phosphodiesterase 4 inhibitors discovered during a thalidomide analog discovery program. These analogs were evaluated for their ability to inhibit tumor angiogenesis, vascularity, and growth. Two analogs (CC-7034 and CC-9088) were identified that had enhanced antiangiogenic activity in Matrigel assays compared with parental thalidomide. These analogs also inhibited the growth of established K1735 and RENCA murine tumors. Tumors whose growth was suppressed by SelCID treatment exhibited decreased vessel density together with increased tumor cell hypoxia and death. The decrease in vascularity produced by SelCID treatment is attributed to a selective loss of vessels devoid of pericyte coverage, suggesting that these agents target immature tumor vessels. That tumor cell death was localized to relatively avascular or hypoxic areas, coupled with the fact that none of the analogs was cytotoxic in vitro against the tumor cells, demonstrates that these analogs are novel antivascular agents with potent antitumor activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cytokines/antagonists & inhibitors , Endothelium, Vascular/drug effects , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/drug therapy , Thalidomide/analogs & derivatives , Animals , Cell Division/drug effects , Cell Hypoxia/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Thalidomide/pharmacology
8.
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
9.
Prostate ; 52(3): 183-200, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12111695

ABSTRACT

BACKGROUND: Activation of the NF-kappaB transcription factor has been previously demonstrated in two androgen receptor negative prostate cancer cell lines. We wished to extend this work to additional prostate cancer cells and to characterize the mechanisms responsible for constitutive NF-kappaB activation. METHODS: Electrophoretic mobility shift assays were performed to measure NF-kappaB DNA-binding activity in prostate cancer cell lines, and immunohistochemistry was performed to detect nuclear localization of NF-kappaB in prostate cancer tissues. Western blot analysis was used to study the status of IkappaBalpha. Transient transfection assays were employed to characterize the contributions of IkappaB kinase (IKK), MAPK kinase kinases (MAPKKKs), androgen receptor (AR), and tyrosine phosphorylation to the constitutive activation of NF-kappaB in the prostate cancer cell lines. RESULTS: Constitutive NF-kappaB activity was observed in AR-negative cell lines as well as in the prostate cancer patient samples, but was not present in AR positive cells. A "super-repressor" IkappaBalpha, as well as dominant negative forms of IKKbeta and NF-kappaB-inducing kinase (NIK), and tyrosine kinase inhibition were able to suppress NF-kappaB activity in the cells with constitutive activation. CONCLUSIONS: The constitutive activation of NF-kappaB observed in prostate cancer cells is likely due to a signal transduction pathway involving tyrosine kinases, NIK, and IKK activation.


Subject(s)
I-kappa B Proteins , NF-kappa B/physiology , Prostatic Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Humans , I-kappa B Kinase , Male , NF-KappaB Inhibitor alpha , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...