Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Mol Med (Berl) ; 98(8): 1161-1173, 2020 08.
Article in English | MEDLINE | ID: mdl-32632752

ABSTRACT

Small molecules targeting the cereblon-containing E3 ubiquitin ligase including thalidomide, lenalidomide, and pomalidomide modulate turnover of downstream client proteins and demonstrate pre-clinical and clinical anti-myeloma activity. Different drugs that engage with cereblon hold the potential of unique phenotypic effects, and we therefore studied the novel protein homeostatic modulator (PHM™) BTX306 with a unique thiophene-fused scaffold bearing a substituted phenylurea and glutarimide. This agent much more potently reduced human-derived myeloma cell line viability, with median inhibitory concentrations in the single nanomolar range versus micromolar values for lenalidomide or pomalidomide, and more potently activated caspases 3/8/9. While lenalidomide and pomalidomide induced greater degradation of Ikaros and Aiolos in myeloma cells, BTX306 more potently reduced levels of GSPT1, eRF1, CK1α, MCL-1, and c-MYC. Suppression of cereblon or overexpression of Aiolos or Ikaros induced relative resistance to BTX306, and this agent did not impact viability of murine hematopoietic cells in an in vivo model, demonstrating its specificity for human cereblon. Interestingly, BTX306 did show some reduced activity in lenalidomide-resistant cell line models but nonetheless retained its nanomolar potency in vitro, overcame bortezomib resistance, and was equipotent against otherwise isogenic cell line models with either wild-type or knockout TP53. Finally, BTX306 demonstrated strong activity against primary CD138-positive plasma cells, showed enhanced anti-proliferative activity in combination with bortezomib and dexamethasone, and was effective in an in vivo systemic model of multiple myeloma. Taken together, the data support further translational studies of BTX306 and its derivatives to the clinic for patients with relapsed and/or refractory myeloma. KEY MESSAGES: BTX306 has a unique thiophene-fused scaffold bearing phenylurea and glutarimide. BTX306 is more potent against myeloma cells than lenalidomide or pomalidomide. BTX306 overcomes myeloma cell resistance to lenalidomide or bortezomib in vitro. BTX306 is active against primary myeloma cells, and shows efficacy in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Drug Resistance, Neoplasm/drug effects , Lenalidomide/pharmacology , Proteostasis/drug effects , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Humans , Mice , Multiple Myeloma , Ubiquitin-Protein Ligases/antagonists & inhibitors
2.
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
3.
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
4.
J Transl Med ; 5: 38, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17640369

ABSTRACT

BACKGROUND: Ocular melanoma is the leading intraocular malignancy. There is no effective treatment for metastatic ocular melanoma. We sought a treatment targeting the tumor microenvironment as well as the tumor cells. METHODS: Migration of HUVEC cells, the ability of HUVEC cells to form tubes, and proliferative capacity of a human ocular melanoma cell line were tested in the presence of lenalidomide and sorafenib alone and in combination. The compounds were also tested in a rat aortic ring assay and were tested in a highly aggressive human ocular melanoma xenograft model. RESULTS: Lenalidomide and Sorafenib inhibit HUVEC ability to migrate and form tubes and when used in combination the inhibition is increased. The agents alone and in combination inhibit outgrowth in the rat aortic ring model. The combination of the agents improved the inhibition over either single agent. In a xenograft model, combination therapy inhibited tumor growth over inhibition by single agent alone in a significant fashion (p < 0.004: lenalidomide and p < 0.0035: sorafenib). Furthermore, spontaneous lung metastasis development was completely inhibited in the combination treated animals. Sixty percent of vehicle treated animals developed lung metastases compared to 50% of lenalidomide treated animals, and 33% of sorafenib treated animals. CONCLUSION: Lenalidomide and sorafenib are effective at targeting endothelial cells, inhibiting growth of ocular melanoma cells and can inhibit growth of tumors in a xenograft model as well as inhibit development of metastases. Combining these agents works in an additive to synergistic way to inhibit the growth of tumors and development of metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Eye Neoplasms/drug therapy , Melanoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/therapeutic use , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Eye Neoplasms/blood supply , Eye Neoplasms/pathology , Humans , In Vitro Techniques , Lenalidomide , Melanoma/blood supply , Melanoma/pathology , Neovascularization, Pathologic/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Sorafenib , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Thalidomide/therapeutic use , Xenograft Model Antitumor Assays
5.
Birth Defects Res B Dev Reprod Toxicol ; 80(3): 188-207, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17570132

ABSTRACT

BACKGROUND: Lenalidomide, a thalidomide analog, is indicated for treatment of patients with deletion-5q myelodysplastic syndromes or multiple myeloma. NZW rabbits were used because of sensitivity to thalidomide's teratogenicity. METHODS: Range-finding and pulse-dosing studies preceded a full developmental toxicity study in New Zealand white (NZW) rabbits (25/group) given lenalidomide (0, 3, 10, or 20 mg/kg/day) or thalidomide (180 mg/kg/day) by stomach tube on gestation days (GD) 7-19. Clinical signs, body weights, and feed consumption were recorded daily from GD 7. On GD 29, standard maternal necropsy, uterine content, and fetal evaluations were carried out. RESULTS: In all studies, thalidomide was selectively toxic to development. In the pulse-dosing study, lenalidomide did not affect development at 100 mg/kg/day. Increases in C(max) and AUC(0-24 hr) values for lenalidomide were slightly less than dose-proportional; lenalidomide occurred in the fetuses. At 10 and 20 mg/kg/day, lenalidomide was maternally toxic (reduced body weight gain and feed consumption; at 20 mg/kg/day, weight loss and one abortion). Developmental toxicity at 10 and 20 mg/kg/day included reduced fetal body weights and increased postimplantation losses and fetal variations (morbidity/purple-discolored skin, undeveloped intermediate lung lobe, irregular nasal-frontal suture, and delayed metacarpal ossification). Thalidomide selectively reduced fetal body weight, increased postimplantation loss and caused characteristic limb and other dysmorphology. CONCLUSIONS: The maternal and developmental NOAELs for lenalidomide are 3 mg/kg/day. Unlike thalidomide, lenalidomide affected embryo-fetal development only at maternally toxic dosages, confirming that structure-activity relationships may not predict maternal or developmental effects. No fetal malformations were attributable to lenalidomide.


Subject(s)
Abnormalities, Drug-Induced/etiology , Embryonic Development/drug effects , Fetal Death/chemically induced , Thalidomide/analogs & derivatives , Animals , Female , Fetus/drug effects , Lenalidomide , Pregnancy , Rabbits , Teratogens/toxicity , Thalidomide/administration & dosage , Thalidomide/toxicity
6.
Leuk Res ; 30(7): 849-58, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16494942

ABSTRACT

Lenalidomide (Revlimid, CC-5013) belongs to a line of compounds known as immunomodulatory drugs (IMiDs) that are under clinical investigation in hematopoietic and solid tumor cancers. Lenalidomide efficacy has been reported in clinical trials of multiple myeloma and myelodysplastic syndromes (MDS), particularly in MDS patients with a del 5q cytogenetic abnormality, with or without other cytogenetic abnormalities. Here we report that lenalidomide inhibits proliferation of chromosome 5 deleted hematopoietic tumor cell lines in vitro, whether from the B cell, T cell, or myeloid lineage. There was diversity in the responses of the various cell lines to lenalidomide, with one undergoing cell cycle arrest, and others undergoing apoptosis. In the most lenalidomide-sensitive chromosome 5 deleted cell line, Namalwa CSN.70, the compound induced G0/G1 cell cycle arrest, inhibited Akt and Gab1 phosphorylation, and inhibited the ability of Gab1 to associate with a receptor tyrosine kinase. Lenalidomide also enhanced AP-1 transcriptional activity in Namalwa, but not in the other cell lines tested. These studies provide evidence for the mechanism of action of lenalidomide in chromosome 5 deleted hematopoietic tumors in vitro, and may provide a better understanding of the drug's activity in clinical applications.


Subject(s)
Adaptor Protein Complex 1/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Thalidomide/analogs & derivatives , Adaptor Protein Complex 1/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , G1 Phase/drug effects , Humans , Lenalidomide , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/drug effects , Receptors, Erythropoietin/metabolism , Resting Phase, Cell Cycle/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Thalidomide/pharmacology
7.
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
8.
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
9.
Drug Discov Today ; 10(2): 107-14, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15718159

ABSTRACT

Thalidomide and its immunomodulatory analogues have numerous effects on the body's immune system, including potential anti-cancer and anti-inflammatory activities. Thalidomide is currently used experimentally to treat various cancers, dermatological, neurological and inflammatory diseases. This drug is approved in the USA for cutaneous manifestations of lepromatous leprosy and is in Phase III trials for multiple myeloma. Thalidomide and its analogues modulate the immune system in various ways. Some of these immunomodulatory activities, together with the anti-angiogenic, anti-proliferative and pro-apoptotic properties, are believed to mediate anti-tumor responses as observed in multiple myeloma and some solid tumors. The analogue lenalidomide has shown potential in treating the bone marrow disorders multiple myeloma and myelodysplastic syndrome, and is presently in Phase II and III trials, respectively.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Dermatologic Agents/therapeutic use , Thalidomide/therapeutic use , Clinical Trials as Topic , Humans
10.
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
11.
Blood ; 104(13): 4188-93, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15319277

ABSTRACT

Previous studies have demonstrated the in vitro and in vivo activity of CC-5013 (Revlimid), an immunomodulatory analog (IMiD) of thalidomide, in multiple myeloma (MM). In the present study, we have examined the anti-MM activity of rapamycin (Rapamune), a specific mTOR inhibitor, combined with CC-5013. Based on the Chou-Talalay method, combination indices of less than 1 were obtained for all dose ranges of CC-5013 when combined with rapamycin, suggesting strong synergism. Importantly, this combination was able to overcome drug resistance when tested against MM cell lines resistant to conventional chemotherapy. Moreover, the combination, but not rapamycin alone, was able to overcome the growth advantage conferred on MM cells by interleukin-6 (IL-6), insulin-like growth factor-1 (IGF-1), or adherence to bone marrow stromal cells (BMSCs). Combining rapamycin and CC-5013 induced apoptosis of MM cells. Differential signaling cascades, including the mitogen-activated protein kinase (MAPK) and the phosphatidylinositol 3'-kinase/Akt kinase (PI3K/Akt) pathways, were targeted by these drugs individually and in combination, suggesting the molecular mechanism by which they interfere with MM growth and survival. These studies, therefore, provide the framework for clinical evaluation of mTOR inhibitors combined with IMiDs to improve patient outcome in MM.


Subject(s)
Antineoplastic Agents/toxicity , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Protein Kinases/metabolism , Sirolimus/toxicity , Thalidomide/analogs & derivatives , Thalidomide/toxicity , Apoptosis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Division/drug effects , Cell Line, Tumor , Drug Synergism , Humans , Lenalidomide , Stromal Cells/cytology , Stromal Cells/drug effects , TOR Serine-Threonine Kinases
12.
Toxicol Sci ; 81(2): 379-89, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15229368

ABSTRACT

The present study determined effects of thalidomide on three successive generations of New Zealand White rabbits after oral dosing to F0 maternal rabbits during the later third of gestation (post major organogenesis) and lactation. One hundred and twenty four time-mated F0 rabbits (31/dose) were gavaged with 0, 30, 150, or 500 mg/kg thalidomide from gestation day 18 (DG 18) to lactation day 28 (DP or day postpartum 28) for approximately 42 days. At 6 months, 12 F1 males and 12 F1 females were randomly paired within each dose group and mated. Reproductive evaluation and/or gross necropsy of the thoracic, abdominal, and pelvic viscera was performed on day 29 postpartum (DP 29) for F0 rabbits, on DP 49 for F1 pups not selected for continued evaluation, after completion of mating for F1 rabbits, and on DG 29 for F1 rabbits on continued evaluation of F2 litter. There was no thalidomide-related mortality in F0 and F1 rabbits. One F0 doe at 30 and 150 mg/kg and 2 at 500 mg/kg aborted. Maternal F0 rabbits had reductions in feed consumption but not body weight gain during the gestation and lactation periods for 150 and 500 mg/kg. The numbers of does with stillborn and all pups dying from DP 1-4 was increased at 150 and 500 mg/kg. Mean number of liveborn (litter size) and percentage of live pups were decreased at 500 mg/kg. A significantly increased number of pups died at 150 and 500 mg/kg, resulting in a reduced viability index and decreased litter size. There were some F1 male and female body weight reductions at 150 and 500 mg/kg postweaning with no change in feed consumption. F1 Caesarean-sectioning and litter observations were normal. Fertility of F1 offspring was not affected by maternal doses of thalidomide, but the pregnancy index may have been reduced by the 500 mg/kg maternal thalidomide dose. There was an apparent dose-related increase in splayed limbs in F1 pups. Splaying has been reported in New Zealand White rabbits and may be a recessive trait. The splay could be caused by the nerve and muscle fiber degeneration and skeletal muscle atrophy observed in some pups. It could also be due to the decrease in litter size, resulting in fewer pups per litter for nursing, leading to rapid weight gain and a failure of the pups to support this weight. No F2 fetal gross external alterations were observed. In summary, pregnant rabbits orally dosed with up to 500 mg/kg thalidomide from gestation day 18 to lactation day 28 had increased abortion, changes in some natural delivery and litter parameters, and limb splay in some F1 pups. No gross external changes were observed in F1 and F2 pups.


Subject(s)
Animals, Newborn/physiology , Embryo, Mammalian/physiology , Pregnancy, Animal/drug effects , Teratogens/toxicity , Thalidomide/toxicity , Abnormalities, Drug-Induced/pathology , Abortifacient Agents/toxicity , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Eating/drug effects , Embryo, Mammalian/pathology , Female , Fertility/drug effects , Fetus/pathology , Limb Deformities, Congenital/chemically induced , Limb Deformities, Congenital/pathology , Milk/chemistry , Nervous System Diseases/chemically induced , Nervous System Diseases/pathology , Parturition/drug effects , Pregnancy , Rabbits , Reproduction/drug effects , Sexual Behavior, Animal/drug effects , Sexual Maturation/drug effects , Temperature , Teratogens/pharmacokinetics , Thalidomide/pharmacokinetics
13.
Clin Pharmacokinet ; 43(5): 311-27, 2004.
Article in English | MEDLINE | ID: mdl-15080764

ABSTRACT

Thalidomide is a racemic glutamic acid derivative approved in the US for erythema nodosum leprosum, a complication of leprosy. In addition, its use in various inflammatory and oncologic conditions is being investigated. Thalidomide interconverts between the (R)- and (S)-enantiomers in plasma, with protein binding of 55% and 65%, respectively. More than 90% of the absorbed drug is excreted in the urine and faeces within 48 hours. Thalidomide is minimally metabolised by the liver, but is spontaneously hydrolysed into numerous renally excreted products. After a single oral dose of thalidomide 200 mg (as the US-approved capsule formulation) in healthy volunteers, absorption is slow and extensive, resulting in a peak concentration (C(max)) of 1-2 mg/L at 3-4 hours after administration, absorption lag time of 30 minutes, total exposure (AUC( infinity )) of 18 mg. h/L, apparent elimination half-life of 6 hours and apparent systemic clearance of 10 L/h. Thalidomide pharmacokinetics are best described by a one-compartment model with first-order absorption and elimination. Because of the low solubility of the drug in the gastrointestinal tract, thalidomide exhibits absorption rate-limited pharmacokinetics (the 'flip-flop' phenomenon), with its elimination rate being faster than its absorption rate. The apparent elimination half-life of 6 hours therefore represents absorption, not elimination. The 'true' apparent volume of distribution was estimated to be 16L by use of the faster elimination-rate half-life. Multiple doses of thalidomide 200 mg/day over 21 days cause no change in the pharmacokinetics, with a steady-state C(max) (C(ss)(max)) of 1.2 mg/L. Simulation of 400 and 800 mg/day also shows no accumulation, with C(ss)(max) of 3.5 and 6.0 mg/L, respectively. Multiple-dose studies in cancer patients show pharmacokinetics comparable with those in healthy populations at similar dosages. Thalidomide exhibits a dose-proportional increase in AUC at doses from 50 to 400 mg. Because of the low solubility of thalidomide, C(max) is less than proportional to dose, and t(max) is prolonged with increasing dose. Age, sex and smoking have no effect on the pharmacokinetics of thalidomide, and the effect of food is minimal. Thalidomide does not alter the pharmacokinetics of oral contraceptives, and is also unlikely to interact with warfarin and grapefruit juice. Since thalidomide is mainly hydrolysed and passively excreted, its pharmacokinetics are not expected to change in patients with impaired liver or kidney function.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Thalidomide/pharmacokinetics , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Area Under Curve , Half-Life , Humans , Kidney Diseases/physiopathology , Liver Diseases/physiopathology , Stereoisomerism , Thalidomide/administration & dosage , Thalidomide/chemistry
14.
Br J Pharmacol ; 142(2): 339-51, 2004 May.
Article in English | MEDLINE | ID: mdl-15066910

ABSTRACT

Monocytes and macrophages provide key targets for the action of novel anti-inflammatory therapeutics targeted at inhibition of PDE4 cAMP-specific phosphodiesterases. PDE4 enzymes provide the dominant cAMP phosphodiesterase activity in U937 human monocytic cells. Differentiation of U937 monocytic cells to a macrophage-like phenotype causes a marked reduction in total cellular PDE4 activity. Monocytic U937 cells express the long PDE4A4, PDE4D5 and PDE4D3 isoforms plus the short PDE4B2 isoform. Differentiation of U937 cells to a macrophage-like phenotype causes a marked downregulation of PDE4D3 and PDE4D5, elicits a marked upregulation of PDE4B2 and induces the novel PDE4A10 long isoform. Comparable patterns are found in human peripheral blood monocytes and macrophages differentiated from them. Immunopurification of PDE4 subfamilies identifies long PDE4D isoforms as providing the major PDE4 activity in U937 monocytic cells. In U937 macrophage-like cells, the activity of the short PDE4B2 isoform predominates. No indication of either the expression or induction of PDE4C was evident. Activation of ERK exerts an inhibitory effect on total PDE4 activity in monocytic U937 cells, where the activity of long PDE4 isoforms predominates. The effect of ERK activation is switched to one of overall stimulation of total PDE4 activity in macrophage U937 cells, where the activity of the short PDE4B2 isoform predominates.10 The profound differentiation-induced changes in PDE4 isoform profile identified here suggests that the development of inhibitors specific for particular PDE4 isoforms may allow for selective effects on monocytes and macrophages to be achieved.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , Cell Differentiation/physiology , Macrophages/cytology , Macrophages/enzymology , Monocytes/cytology , Monocytes/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Cell Differentiation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Macrophages/drug effects , Monocytes/drug effects , U937 Cells
15.
Article in English | MEDLINE | ID: mdl-14991906

ABSTRACT

BACKGROUND: The present work was performed to determine the effect of thalidomide exposure on reproductive function and early embryonic development. METHODS: Twenty-five female New Zealand White rabbits were orally gavaged with 0, 10, 50, or 100 mg/kg/day thalidomide 14 days prior to mating through to gestation day 7 for a total of 22 days. Treated females were Caesarean-sectioned approximately 29 days after the date of attempted mating. Following mating with treated females, male rabbits (25/dose) were gavaged with 0, 30, 150, or 500 mg/kg/day beginning 14 days prior to mating with a group of untreated females (25/dose). Doses were administered through mating until the day before sacrifice for a minimum of 56 days. Untreated females were Caesarean-sectioned 29 days after the last attempted mating. Comprehensive necropsy and histopathology of the reproductive system were performed. RESULTS: Treated females had reduction in body weight gain during gestation. Mating and pregnancy parameters were unaffected by thalidomide. At 100 m/kg, litter averages for corpora lutea, implantations, litter sizes, does with viable fetuses and live fetuses decreased and the number of early resorptions, does with any resorptions, does with all conceptuses resorbed, and the percent resorbed conceptuses per litter increased. The number of early resorptions, the average number of early resorptions per litter, and the percent resorbed conceptuses per litter increased at 10 and 50 mg/kg. There were no thalidomide-related external fetal malformations. Mating and fertility in male rabbits were unaffected by thalidomide. There was an increased incidence of flaccid testes at 150 and 500 mg/kg and of bilateral small testes in all treated groups. At 500 mg/kg, there was degeneration of the germinal epithelium of the testicles with an increase in multinucleated giant cells in seminiferous tubule and a loss of round and elongating spermatids. CONCLUSIONS: Thalidomide had no adverse effects on mating and fertility in male and female rabbits dosed up to 500 and 100 mg/kg/day, respectively, for 14 days prior to mating. After 56 day of dosing, histopathologic changes with no associated sperm abnormalities were observed in the testicles. Embryonic development NOAEL for treated females mated to untreated males was <10 mg/kg. Corresponding fertility NOAEL for treated males mated to untreated females was 500 mg/kg.


Subject(s)
Embryonic and Fetal Development/drug effects , Reproduction/drug effects , Thalidomide/toxicity , Aborted Fetus , Abortion, Spontaneous/chemically induced , Animals , Body Weight/drug effects , Cesarean Section , Feeding Behavior/drug effects , Female , Fertility/drug effects , Fetus/drug effects , Fetus/embryology , Male , Molecular Structure , Organ Size/drug effects , Rabbits , Sexual Behavior, Animal/drug effects , Sperm Motility/drug effects , Testis/cytology , Thalidomide/chemistry
16.
J Clin Pharmacol ; 44(2): 173-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14747426

ABSTRACT

Attention deficit hyperactivity disorder (ADHD) in children is effectively treated by racemic oral methylphenidate (dl-MPH). The d-isomer (d-MPH) has been developed as an improved treatment for ADHD since only half the racemic dose is used. This study, performed in healthy subjects, assessed the effect of food on the pharmacokinetics of dexmethylphenidate hydrochloride (d-MPH HCl) in a single dose (2 x 10-mg tablets), two-way crossover with d-MPH administered to subjects in both a fasting state or after a high-fat breakfast. There were no serious or unexpected adverse events during the course of this study, with most events reported in comparable numbers of fed and fasted subjects. The bioequivalence of d-MPH was similar with or without food, with 90% confidence intervals of 88.2% to 104.6% and 105.9% to 118.2% for ln(C(max)) and ln[(AUC(0-infinity))], respectively. There was a marginal but statistically significant 1-hour increase in t(max) in the fed versus fasted state, reflecting an absorption delay. The rate of formation of the major metabolite, d-ritalinic acid (d-RA), was marginally decreased ( approximately 14%) after food. The extent of exposure to d-RA was similar (within 1.2%) between both treatments. There was a marginal but statistically significant difference in mean t(max) for d-RA between fed and fasted conditions, with peak concentration occurring 1.5 hours later after d-MPH administration with food. There was no measurable in vivo chiral inversion of d-MPH to l-MPH in plasma. In addition, the metabolism of d-MPH was stereospecific as d-MPH only produced d-RA. In summary, food had no substantial effect on the bioavailability of d-MPH, with an equivalent rate and extent of exposure obtained. Therefore, d-MPH can be administered without regard to food intake.


Subject(s)
Central Nervous System Stimulants/pharmacokinetics , Dexmethylphenidate Hydrochloride , Food-Drug Interactions , Methylphenidate/analogs & derivatives , Methylphenidate/pharmacokinetics , Adult , Area Under Curve , Biological Availability , Central Nervous System Stimulants/blood , Central Nervous System Stimulants/chemistry , Cross-Over Studies , Fasting , Female , Half-Life , Humans , Intestinal Absorption , Male , Methylphenidate/blood , Methylphenidate/chemistry , Postprandial Period , Stereoisomerism , Therapeutic Equivalency
17.
s.l; s.n; 2004. 17 p. ilus, tab, graf.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1242666

ABSTRACT

Thalidomide is a racemic glutamic acid derivative approved in the US for erythema nodosum leprosum, a complication of leprosy. In addition, its use in various inflammatory and oncologic conditions in being investigated. Thalidomide interconverts between the (R)- and (S)-enantiomers in plasma, with protein binding of 55% and 65%, respectively. More than 90% of the absorbed drug is excreted in the urine and faeces within 48 hours. Thalidomide is minimally metabolised by the liver, but is spontaneously hydrolysed into numerous renally excreted products. After a single oral dose of thalidomide 200mg (as the US-approved capsule formulation) in healthy volunteers, absorption is slow and extensive, resulting in a peak concentration (Cmax) of 1-2mg/L at 3-4 hours after administration, absorption lag time of 30 minutes, total exposure (AUCoo) of 18mg - h/L, apparent elimination half-life of 6 hours and apparent systemic clearence of 10 L/H. Thalidomide pharmacokinetics are best described by a one-comportment model with first-order absorption and elimination. Because of the low solubility of the drug in the gastrointestinal tract, thalidomide exhibits absorption rate-limited pharmacolinetics (the 'flip-flop' phenomenon), with its elimination rate being faster than in absorption rate. The apparent elimination half-life of 6 hours therefore represents absorption, not elimination. The 'true' apparent volume of distribution was estimated to be 16L by use of the faster elimination-rate half-life. Multiple doses of thalidomide 200 mg/day over 21 days cause no change in the pharmacokinetics, with a steady-state Cmax (Cssmax) of 1.2 mg/L. Simulation of 400 and 800 mg/day also shows no accululation, with Css of 3.5 and 6.0 mg/L, respectively. Multiple-dose studies in cancer patients show pharmacokinetics comparable with those in healthy populations at similar dosages. Thalidomide exhibits a dose-proportional increase in AUC at doses from 50 to 400mg. Because of the low solubility of thalidomide Cmax is less than proportional to dose, and tmax is prolonged with increasing dose. Age, sex and smoking have no effect on the pharmacokinetics of thalidomide, and the effect of food is minimal. Thalidomide does not alter the pharmacokinetics of oral contraceptives, and is also unlikely to interact with warfarin and grapefruit juice. Since thalidomide is mainly hydrolysed and passively excreted, its pharmacokonetics are not expected to change in patients with impaired liver...


Subject(s)
Humans , Thalidomide , Thalidomide/administration & dosage , Thalidomide/pharmacokinetics , Thalidomide/history , Thalidomide/isolation & purification , Thalidomide/metabolism , Thalidomide/standards , Thalidomide/chemical synthesis , Thalidomide/toxicity , Thalidomide/therapeutic use , Administration, Oral , Cimetidine/antagonists & inhibitors , Diltiazem/antagonists & inhibitors , Erythema Nodosum/etiology , Phenobarbital/antagonists & inhibitors , Drug Interactions/physiology , Rifampin/antagonists & inhibitors , Feline Acquired Immunodeficiency Syndrome/therapy , Warfarin/antagonists & inhibitors
18.
s.l; s.n; 2004. 11 p. tab.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1242671

ABSTRACT

The present study determined effects of thalidomide on three successive generations of New Zeland Whith rabbits after oral dosing to FO maternal rabbits during the later third of gestation (post major organogenesis) and lactation. On hundred and twenty four time-mated FO rabbits (31/dose) were gaveged with 0,030,150, or 500mg/kg thalidomide from gestation day 18 (DG 18) to lactation day 28 (DP or day postpartum 28) for approximately 42 days. At 6 months, 12 F1 females were randomly paired witthin each dose group and mated. Reproductive evaluation and/or gross necropsy of the thoracic, abodominal, and pelvic viscera was perfomed on day 29 postpartum (DP 29) for FO rabbits, on DP49 for F1 pups not selected for continued evaluation, sfter completion of mating for F1 rabbits, and on DG 29 for F1 rabbits on continued evaluation of F2 litter. There was no thalidomide-related mortality in FO and F1 rabbits. One FO doe at 30 and 150 mg/kg and 2 at 500 mg/kg aborted. Maternal FO rabbits had reductions in feed consumption but no body weight gain during the gestation and lactation periods for 150 and 500 mg/kg. The numbers of does with stillborn and all pups dyving from DP 1-4 was increased at 150 and 500 mg/kg. Mean number of liverborn (litter size) and percentage of live pups were decreased at 500 mg/kg. A significantly increased number of pups died at 150 and 500 mg/kg, resulting in a reduced viability index and decreased litter size. There were some F1 male and female bodyweight reductions at 150 and 500 mg/kg postweaning with no changein feed consumption. F1 Caesarean-sectioning and litter observations were normal. Fertility of F1 offspring was not affected by maternal doses of thalidomide, but the pregnancy index may have been reduced by the 500 mg/kg maternal thalidomide dose. There was an apparent dose-related increased in splayed limbs in F1 pups. Splaving has been reported in New Zealand Whith rabbits and may be a recessive trait. The splay could be caused by the nerve and muscle fiber degeneration and skeletal muscle atrophy observed in some pups. It could also be due to the decreased in litter size, resulting in fewer pups per litter for nursing, leading to rapid weight gain and a failure of the pups to support this weight. No F@ fetal gross external alterations were observed.


Subject(s)
Rabbits , Rabbits/abnormalities , Rabbits/growth & development , Rabbits/embryology , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/pharmacology , Abortion, Veterinary/etiology , Birth Weight
19.
Bioorg Med Chem Lett ; 13(20): 3415-7, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-14505639

ABSTRACT

Thalidomide, (1), has made a remarkable comeback from its days of a sedative with teratogenic properties due to its ability to selectively inhibit TNF-alpha, a key pro-inflammatory cytokine and its clinical benefit in the treatment of cancer. Thalidomide contains one chiral center and is known to be chirally unstable under in vitro and in vivo conditions. It has been hypothesized that different biological properties are associated with each isomer. Thus, chirally stable analogues of thalidomide, alpha-fluorothalidomide, (3) and alpha-fluoro-4-aminothalidomide (4) were prepared by electrophilic fluorination. Analogue 3 was found to be cytotoxic and did not inhibit TNF-alpha production in LPS stimulated hPBMC below toxic concentrations. On the other hand, 4 was non-cytotoxic at the tested concentrations and was found to be 830-fold more potent than thalidomide as TNF-alpha inhibitor.


Subject(s)
Fluorine/chemistry , Thalidomide/chemistry , Cells, Cultured , Humans , Monocytes/drug effects , Monocytes/metabolism , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
20.
Int J Toxicol ; 22(3): 215-26, 2003.
Article in English | MEDLINE | ID: mdl-12851154

ABSTRACT

d-Methylphenidate (d-MPH) was approved as a treatment for attention deficit hyperactivity disorder (ADHD) in children. The repeated-dose toxicity of the d enantiomer of d,l-methylphenidate (d,l-MPH) was assessed in male and female Beagle dogs. Dogs were orally dosed twice a day in equally divided doses 6 hours apart for total daily doses of 1, 3, and 10 mg/kg/day d-MPH or 20 mg/kg/day d,l-MPH for 90 days, followed by a 30-day recovery period. The top d-MPH dose of 10 mg/kg was equimolar to 20 mg/kg d,l-MPH in d-MPH content. The 10-mg/kg d-MPH and d,l-MPH doses were at least 13 times the maximum therapeutic dose giving rise to systemic exposures that were equivalent to or at least 2 times greater than those at the maximum therapeutic doses in children. The 10-mg/kg d-MPH and 20-mg/kg d,l-MPH doses had systemic exposures that were equivalent to or two to five times greater than the maximum therapeutic plasma levels in children respectively. There was no treatment-related mortality in all doses tested. Reversible salivation, hyperactivity, and diarrhea were seen in the high-dose d-MPH and d,l-MPH groups. Significant body weight loss and reduction in food consumption were observed in males for both high-dose groups with weights comparable to control values by the end of the recovery period. There were no abnormal clinical pathology or macroscopic or microscopic findings. Based on body weight changes, the no-observed-adverse-effect level (NOAEL) of d-MPH in beagle dogs was 3 mg/kg/day.


Subject(s)
Central Nervous System Stimulants/toxicity , Methylphenidate/toxicity , Administration, Oral , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Body Weight/drug effects , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacokinetics , Dogs , Dose-Response Relationship, Drug , Drug Administration Schedule , Eating/drug effects , Female , Male , Methylphenidate/administration & dosage , Methylphenidate/pharmacokinetics , No-Observed-Adverse-Effect Level , Recovery of Function , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...