Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Signal ; 26(9): 2016-29, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24882690

ABSTRACT

Apremilast, an oral small molecule inhibitor of phosphodiesterase 4 (PDE4), is in development for chronic inflammatory disorders, and has shown efficacy in psoriasis, psoriatic arthropathies, and Behçet's syndrome. In March 2014, the US Food and Drug Administration approved apremilast for the treatment of adult patients with active psoriatic arthritis. The properties of apremilast were evaluated to determine its specificity, effects on intracellular signaling, gene and protein expression, and in vivo pharmacology using models of innate and adaptive immunity. Apremilast inhibited PDE4 isoforms from all four sub-families (A1A, B1, B2, C1, and D2), with IC50 values in the range of 10 to 100 nM. Apremilast did not significantly inhibit other PDEs, kinases, enzymes, or receptors. While both apremilast and thalidomide share a phthalimide ring structure, apremilast lacks the glutarimide ring and thus fails to bind to cereblon, the target of thalidomide action. In monocytes and T cells, apremilast elevated intracellular cAMP and induced phosphorylation of the protein kinase A substrates CREB and activating transcription factor-1 while inhibiting NF-κB transcriptional activity, resulting in both up- and down-regulation of several genes induced via TLR4. Apremilast reduced interferon-α production by plasmacytoid dendritic cells and inhibited T-cell cytokine production, but had little effect on B-cell immunoglobulin secretion. In a transgenic T-cell and B-cell transfer murine model, apremilast (5mg/kg/day p.o.) did not affect clonal expansion of either T or B cells and had little or no effect on their expression of activation markers. The effect of apremilast on innate immunity was tested in the ferret lung neutrophilia model, which allows monitoring of the known PDE4 inhibitor gastrointestinal side effects (nausea and vomiting). Apremilast significantly inhibited lung neutrophilia at 1mg/kg, but did not induce significant emetic reflexes at doses <30 mg/kg. Overall, the pharmacological effects of apremilast are consistent with those of a targeted PDE4 inhibitor, with selective effects on innate immune responses and a wide therapeutic index compared to its gastrointestinal side effects.


Subject(s)
Immunity, Innate/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Thalidomide/analogs & derivatives , Adaptive Immunity/drug effects , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Ferrets , Humans , Jurkat Cells , Lung Diseases/drug therapy , Male , Mice , Mice, Transgenic , Phosphodiesterase 4 Inhibitors/metabolism , Phosphodiesterase 4 Inhibitors/therapeutic use , Protein Binding , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thalidomide/metabolism , Thalidomide/pharmacology , Thalidomide/therapeutic use , Vomiting/prevention & control
2.
Br J Pharmacol ; 159(4): 842-55, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20050849

ABSTRACT

BACKGROUND AND PURPOSE: Apremilast is an orally administered phosphodiesterase-4 inhibitor, currently in phase 2 clinical studies of psoriasis and other chronic inflammatory diseases. The inhibitory effects of apremilast on pro-inflammatory responses of human primary peripheral blood mononuclear cells (PBMC), polymorphonuclear cells, natural killer (NK) cells and epidermal keratinocytes were explored in vitro, and in a preclinical model of psoriasis. EXPERIMENTAL APPROACH: Apremilast was tested in vitro against endotoxin- and superantigen-stimulated PBMC, bacterial peptide and zymosan-stimulated polymorphonuclear cells, immunonoglobulin and cytokine-stimulated NK cells, and ultraviolet B light-activated keratinocytes. Apremilast was orally administered to beige-severe combined immunodeficient mice, xenotransplanted with normal human skin and triggered with human psoriatic NK cells. Epidermal skin thickness, proliferation index and inflammation markers were analysed. KEY RESULTS: Apremilast inhibited PBMC production of the chemokines CXCL9 and CXCL10, cytokines interferon-gamma and tumour necrosis factor (TNF)-alpha, and interleukins (IL)-2, IL-12 and IL-23. Production of TNF-alpha by NK cells and keratinocytes was also inhibited. In vivo, apremilast significantly reduced epidermal thickness and proliferation, decreased the general histopathological appearance of psoriasiform features and reduced expression of TNF-alpha, human leukocyte antigen-DR and intercellular adhesion molecule-1 in the lesioned skin. CONCLUSIONS AND IMPLICATIONS: Apremilast displayed a broad pattern of anti-inflammatory activity in a variety of cell types and decreased the incidence and severity of a psoriasiform response in vivo. Inhibition of TNF-alpha, IL-12 and IL-23 production, as well as NK and keratinocyte responses by this phosphodiesterase-4 inhibitor suggests a novel approach to the treatment of psoriasis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Psoriasis/drug therapy , Skin/drug effects , Thalidomide/analogs & derivatives , Administration, Oral , Adult , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enterotoxins/immunology , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, SCID , Middle Aged , Phosphodiesterase Inhibitors/administration & dosage , Psoriasis/enzymology , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , RNA, Messenger/metabolism , Severity of Illness Index , Skin/enzymology , Skin/immunology , Skin/pathology , Skin/radiation effects , Skin Transplantation , Thalidomide/administration & dosage , Thalidomide/pharmacology , Time Factors , Transplantation, Heterologous , U937 Cells , Ultraviolet Rays , Zymosan/metabolism
3.
Br J Cancer ; 90(5): 955-61, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-14997189

ABSTRACT

We assessed the safety, tolerability and efficacy of the immunomodulatory drug, CC-5013 (REVIMID trade mark ), in the treatment of patients with metastatic malignant melanoma and other advanced cancers. A total of 20 heavily pretreated patients received a dose-escalating regimen of oral CC-5013. Maximal tolerated dose, toxicity and clinical responses were evaluated and analysis of peripheral T-cell surface markers and serum for cytokines and proangiogenic factors were performed. CC-5013 was well tolerated. In all, 87% of adverse effects were classified as grade 1 or grade 2 according to Common Toxicity Criteria and there were no serious adverse events attributable to CC-5013 treatment. Six patients failed to complete the study, three because of disease progression, two withdrew consent and one was entered inappropriately and withdrawn from the study. The remaining 14 patients completed treatment without dose reduction, with one patient achieving partial remission. Evidence of T-cell activation was indicated by significantly increased serum levels of sIL-2 receptor, granulocyte-macrophage colony-stimulating factor, interleukin-12 (IL-12), tumour necrosis factor-alpha and IL-8 in nine patients from whom serum was available. However, levels of proangiogenic factors vascular endothelial growth factor and basic foetal growth factor were not consistently affected. This study demonstrates the safety, tolerability and suggests the clinical activity of CC-5013 in the treatment of refractory malignant melanoma. Furthermore, this is the first report demonstrating T-cell stimulatory activity of this class of compound in patients with advanced cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Cytokines/blood , Female , Fibroblast Growth Factor 2/metabolism , Humans , Immunization , Lenalidomide , Male , Maximum Tolerated Dose , Melanoma/secondary , Middle Aged , Neoplasms/drug therapy , Neoplasms/pathology , Safety , Skin Neoplasms/pathology , T-Lymphocytes/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Leukemia ; 17(1): 41-4, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12529658

ABSTRACT

We have previously shown that thalidomide and its potent immunomodulatory derivatives (IMiDs) inhibit the in vitro growth of multiple myeloma (MM) cell lines and patient MM cells that are resistant to conventional therapy. In this study, we further characterize the effect of these drugs on growth of B cell malignancies and angiogenesis. We established a beige-nude-xid (BNX) mouse model to allow for simultaneous in vivo measurement of both anti-tumor and anti-angiogenic effects of thalidomide and its analogs. Daily treatment (50 mg/kg/d) with thalidomide or IMiDs was nontoxic. The IMiDs were significantly more potent than thalidomide in vivo in suppressing tumor growth, evidenced by decreased tumor volume and prolonged survival, as well as mediating anti-angiogenic effects, as determined by decreased microvessel density. Our results therefore show that the IMiDs have more potent direct anti-tumor and anti-angiogenic effects than thalidomide in vivo, providing the framework for clinical protocols evaluating these agents in MM and other B cell neoplasms.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Myeloma/drug therapy , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/prevention & control , Neovascularization, Pathologic/drug therapy , Thalidomide/therapeutic use , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Female , Humans , Injections, Intraperitoneal , Mice , Mice, Nude , Microcirculation/drug effects , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms, Experimental/drug therapy , Survival Rate , Thalidomide/analogs & derivatives , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
5.
Leukemia ; 15(12): 1950-61, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753617

ABSTRACT

Increased angiogenesis has recently been recognized in active multiple myeloma (MM). Since vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are two key mediators of angiogenesis, we characterized the production of VEGF, b-FGF and interleukin-6 (IL-6) (a MM growth and survival factor) in MM cell lines and Epstein-Barr virus (EBV) transformed B cell lines from MM patients, patient MM cells, as well as bone marrow stromal cells (BMSCs) from normal healthy donors and MM patients. We detected secretion of VEGF, but no bFGF and IL-6, in MM cell lines (MM.1S, RPMI 8226 and U266); EBV transformed B cell lines from MM patients (IM-9, HS-Sultan and ARH77); MM cell lines resistant to doxorubicin (RPMI-DOX40), mitoxantrone (RPMI-MR20), melphalan (RPMI-LR5) and dexamethasone (MM.1R); and patient MM cells (MM1 and MM2). BMSCs from MM patients and normal donors secreted VEGF, b-FGF and IL-6. Importantly, when MM cells were adhered to BMSCs, there was a significant increase in VEGF (1.5- to 3.1-fold) and IL-6 (1.9- to 56-fold) secretion. In contrast, the bFGF decreased in co-cultures of BMSCs and MM cells. Paraformaldehyde fixation of BMSCs or MM cells prior to adhesion revealed that VEGF was produced both from BMSCs and MM cells, though it may come primarily from BMSCs in some cultures. IL-6 was produced exclusively in BMSCs, rather than MM cells. Moreover, when MM cells were placed in Transwell insert chambers to allow their juxtaposition to BMSCs without cell to cell contact, induction of VEGF and IL-6 secretion persisted, suggesting the importance of humoral factors. Addition of exogenous IL-6 (10 ng/ml) increased VEGF secretion by BMSCs. Conversely, VEGF (100 ng/ml) significantly increased IL-6 secretion by BMSCs. Moreover, anti-human VEGF (1 microg/ml) and anti-human IL-6 (10 microg/ml) neutralizing antibodies reduced IL-6 and VEGF secretion, respectively, in cultures of BMSCs alone and co-cultures of BMSCs and MM cells. Finally, thalidomide (100 microM) and its immunomodulatory analog IMiD1-CC4047 (1 microM) decreased the upregulation of IL-6 and VEGF secretion in cultures of BMSCs, MM cells and co-cultures of BMSCs with MM cells. These data demonstrate the importance of stromal-MM cell interactions in regulating VEGF and IL-6 secretion, and suggest additional mechanisms whereby thalidomide and IMiD1-CC4047 act against MM cells in the BM millieu.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Multiple Myeloma/pathology , Stromal Cells/cytology , Angiogenesis Inhibitors/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Adhesion , Cell Communication/physiology , Coculture Techniques , Drug Interactions , Fibroblast Growth Factor 2/metabolism , Humans , Interleukin-6/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/physiopathology , Stromal Cells/metabolism , Stromal Cells/physiology , Thalidomide/pharmacology , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
J Clin Immunol ; 21(5): 357-64, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11720008

ABSTRACT

In Crohn's disease, intestinal lamina propria (LP) T cells overproduce TNF-alpha and IFN-gamma, and clinical and animal studies indicate that this is pathogenic. Thalidomide influences cytokine production by leukocytes, inhibiting macrophage TNF-alpha, and is beneficial in treating Crohn's disease. Chemical analogues have been synthesized that may lack teratogenic and other side effects of thalidomide. We tested three analogues [selective cytokine inhibitory drugs (SelCIDs) A, B, and C, all potent PDE4 inhibitors] for effect on TNF-alpha, IFN-gamma, and IL-10 production by and on proliferation of intestinal LP mononuclear cells after T-cell stimulation and results were compared with those for peripheral blood leukocytes (PBL). While thalidomide itself had little effect, the SelCIDs were potent inhibitors, with relative inhibitory potencies: A> or =B>>C. The LP T cells were less sensitive to inhibition by the SelCIDs than were PBL. Since highly pre-activated PBL were even less sensitive, activation state alone can account for the responsiveness of intestinal LP T cells. Thalidomide analogues could play a role in treating Crohn's disease and other inflammatory disorders.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Intestines/immunology , T-Lymphocytes/drug effects , Thalidomide/analogs & derivatives , Tumor Necrosis Factor-alpha/biosynthesis , Basement Membrane , Cell Division , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4 , Humans , Intestines/cytology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
7.
Drug Metab Dispos ; 29(10): 1355-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11560881

ABSTRACT

As part of a double-blind placebo-controlled study of the effect of thalidomide on body weight and the viral load of human immunodeficiency virus-seropositive patients, plasma and semen samples were analyzed for the presence of thalidomide. Patients were orally dosed with 100 mg of thalidomide/day for 8 weeks. Blood samples were obtained at baseline and weeks 4, 8, and 12, and semen was obtained at baseline and weeks 4 and 8. Samples were extracted with solid-phase cartridges and analyzed by liquid chromatography/tandem mass spectrometry using atmospheric pressure chemical ionization in the negative ion mode. Two of four patients taking thalidomide were able to provide semen samples. Both had detectable levels of thalidomide in their plasma (10-350 ng/ml) and semen (10-250 ng/g) at weeks 4 and 8. There was an apparent correlation between plasma and semen levels. Semen levels could be significantly greater for therapeutic doses of more than 100 mg/day. Since the threshold dose for birth defects and thalidomide exposure is not known, male patients are advised to use barrier contraception.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , HIV Infections/metabolism , Semen/metabolism , Thalidomide/pharmacokinetics , Administration, Oral , Angiogenesis Inhibitors/therapeutic use , Double-Blind Method , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Thalidomide/therapeutic use
8.
J Clin Pharmacol ; 41(6): 662-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11402635

ABSTRACT

Thalidomide is approved in the United States for treating erythema nodosum leprosum, a complication of leprosy. The present study determined the single-dose oral pharmacokinetics and dose proportionality from 50 to 400 mg of Celgene's commercial Thalomid thalidomide formulation in an open-label, single-dose, three-way crossover study. Fifteen healthy subjects were given 50, 200, and 400 mg of thalidomide on three occasions, and blood samples were collected over 48 hours. Pharmacokinetic parameters were determined using noncompartmental methods, and dose proportionality was assessed by linear regression of dose-normalized Cmax and AUC0-infinity. No serious or unexpected adverse events occurred. The most common adverse events were dizziness, somnolence, headache, and nausea. One patient was discontinued because of pharyngitis. There was a significant deviation from proportionality for Cmax with increases being less than proportional than changes in dose. AUC0-infinity increased proportionally with dose, suggesting that the overall amount of thalidomide absorbed, as well as its clearance, is independent of dose over the range used. V/F was found to increase with dose. This was most likely due to the terminal rate constant, which is used to calculate V/F, actually representing the absorption process rather than elimination (i.e., flip-flop phenomenon). The terminal rate constant (absorption rate constant) for the highest dose was 50% less than for the other two lower doses. The less than proportional increases in Cmax were most likely due to thalidomide's low aqueous solubility. Thalidomide shows reasonable dose proportionality with respect to AUC from 50 to 400 mg.


Subject(s)
Leprostatic Agents/pharmacokinetics , Thalidomide/pharmacokinetics , Adult , Area Under Curve , Body Weight , Female , Humans , Leprostatic Agents/administration & dosage , Leprostatic Agents/adverse effects , Leprostatic Agents/blood , Male , Middle Aged , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/blood
9.
Blood ; 98(1): 210-6, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418482

ABSTRACT

The antiangiogenic activity of thalidomide (Thal), coupled with an increase in bone marrow angiogenesis in multiple myeloma (MM), provided the rationale for the use of Thal in MM. Previously, the direct anti-MM activity of Thal and its analogues (immunomodulatory drugs, IMiDs) on MM cells was demonstrated, suggesting multiple mechanisms of action. In this study, the potential immunomodulatory effects of Thal/IMiDs in MM were examined. It was demonstrated that Thal/IMiDs do not induce T-cell proliferation alone but act as costimulators to trigger proliferation of anti-CD3-stimulated T cells from patients with MM, accompanied by an increase in interferon-gamma and IL-2 secretion. However, an increase in autologous T-cell killing of patient MM cells could not be demonstrated. A role for natural killer (NK)- and LAK-cell-mediated killing is suggested because IL-2-primed peripheral blood mononuclear cells (PBMCs) treated with Thal/IMiDs demonstrated significantly increased lysis of MM cell lines. Cold target inhibition assays suggested NK- rather than LAK-cell-mediated killing. Furthermore, this killing was not major histocompatibility complex-class restricted, and the depletion of CD56(+) cells blocked the drug-induced MM cell lysis. It was significant that increased killing of patient MM cells by autologous PBMCs treated with Thal/IMiDs was also observed. Although the in vivo relevance of NK-cell-mediated MM cell killing is unknown, phenotypic analysis performed in MM patients receiving Thal therapy demonstrated an increase in CD3(-)CD56(+) cells in patients responding to therapy. Thus in vitro and in vivo data support the hypothesis that Thal may mediate its anti-MM effect, at least in part, by modulating NK cell number and function.


Subject(s)
Adjuvants, Immunologic/pharmacology , Killer Cells, Natural/drug effects , Multiple Myeloma/drug therapy , Thalidomide/pharmacology , Adjuvants, Immunologic/administration & dosage , Case-Control Studies , Cytotoxicity, Immunologic/drug effects , Humans , Immunophenotyping , Killer Cells, Natural/immunology , Lymphocyte Count , Multiple Myeloma/blood , Multiple Myeloma/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Thalidomide/administration & dosage , Tumor Cells, Cultured/drug effects
10.
Toxicol Sci ; 59(1): 160-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134555

ABSTRACT

Fifty-six adult beagle dogs (28 male, 28 female) were orally administered thalidomide at 43, 200, or 1000 mg/kg/day for 53 weeks. Sixteen (2/sex/dose group) and 32 (4/sex/dose group) dogs were euthanized and necropsied after 26 and 53 weeks of dosing, respectively. The remaining 8 animals (2/sex/group; high-dose and control groups) were dosed for 53 weeks, euthanized, and necropsied at 58 weeks after a 5-week recovery period. There were no deaths during the study. The only observed clinical signs attributable to thalidomide administration were green-colored urine, white-colored fecal residue presumed to be unchanged thalidomide, enlarged and/or blue coloration of female mammary tissue, and prolonged estrus. There were no thalidomide-related changes in body weights, food consumption, electrocardiography, ophthalmoscopy, neurological function, and endocrine function. The mostly slight and/or transient variations observed in some hematology and blood chemistry values of dosed dogs were considered to be toxicologically insignificant and were supported by the lack of histopathologic correlates. The only gross finding attributable to thalidomide was a yellow-green discoloration of the femur, rib, and/or calvarium that was observed at each euthanization interval including recovery. There was no microscopic correlate for this finding. No thalidomide-related microscopic changes were seen in any of the organs and tissues at 26 weeks. Mammary duct dilatation and/or glandular hyperplasia observed in females at 53 and 58 weeks and hepatic bile pigment exhibited by high-dose males at 53 weeks were microscopic changes considered to be thalidomide-related. There was no gross and histopathologic evidence of any tumors. In summary, thalidomide at up to 1000 mg/kg/day for 53 weeks did not induce any major systemic toxicity or tumors in dogs. The NOAEL was 200 mg/kg/day.


Subject(s)
Hypnotics and Sedatives/toxicity , Thalidomide/toxicity , Animals , Body Weight/drug effects , Bone and Bones/drug effects , Bone and Bones/pathology , Clinical Chemistry Tests , Color , Dogs , Eating/drug effects , Electrocardiography , Female , Hematologic Tests , Male , Organ Size/drug effects , Pilot Projects , Toxicity Tests
11.
Blood ; 96(9): 2943-50, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11049970

ABSTRACT

Although thalidomide (Thal) was initially used to treat multiple myeloma (MM) because of its known antiangiogenic effects, the mechanism of its anti-MM activity is unclear. These studies demonstrate clinical activity of Thal against MM that is refractory to conventional therapy and delineate mechanisms of anti-tumor activity of Thal and its potent analogs (immunomodulatory drugs [IMiDs]). Importantly, these agents act directly, by inducing apoptosis or G1 growth arrest, in MM cell lines and in patient MM cells that are resistant to melphalan, doxorubicin, and dexamethasone (Dex). Moreover, Thal and the IMiDs enhance the anti-MM activity of Dex and, conversely, are inhibited by interleukin 6. As for Dex, apoptotic signaling triggered by Thal and the IMiDs is associated with activation of related adhesion focal tyrosine kinase. These studies establish the framework for the development and testing of Thal and the IMiDs in a new treatment paradigm to target both the tumor cell and the microenvironment, overcome classical drug resistance, and achieve improved outcome in this presently incurable disease.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Drug Resistance, Multiple , Immunosuppressive Agents/toxicity , Multiple Myeloma/pathology , Thalidomide/toxicity , Thalidomide/therapeutic use , Cell Cycle/drug effects , DNA Replication/drug effects , Dexamethasone/toxicity , Doxorubicin/toxicity , Female , G1 Phase/drug effects , Humans , Male , Melphalan/toxicity , Thalidomide/analogs & derivatives , Tumor Cells, Cultured
12.
Biopharm Drug Dispos ; 21(1): 33-40, 2000 Jan.
Article in English | MEDLINE | ID: mdl-11038436

ABSTRACT

The effect of food on the oral pharmacokinetics of thalidomide and the relative bioavailability of two oral thalidomide formulations were determined in an open label, single dose, randomized, three-way crossover study. Five male and eight female healthy volunteers received a single oral dose of 200 mg Celgene thalidomide capsules under fasted and non-fasted conditions, and a single dose of 200 mg tablets of Serral thalidomide under fasted conditions. The high-fat meal resulted in a 0.5-1.5 h absorption lag time, an increased mean C(max), a decreased mean AUC and a delay in mean T(max). The Serral tablet formulation resulted in a lower mean C(max), and slower terminal decline in plasma thalidomide concentrations compared with both Celgene treatments. Mean C(max) concentrations were 1.99+/-0.41 microg/mL (range 1.28-2.76) within 4.00+/-1.13 h (2-5) for the Celgene formulation fasted, 2.17+/-0.51 microg/mL (1.43-3.01) within 6.08+/-2.33 h (3-12) for the Celgene formulation with food, and 1. 05+/-0.31 microg/mL (0.62-1.65) within 6.23+/-1.88 h (5-10) for the Serral formulation fasted. Mean terminal half-lives were 13.50+/-6. 77 h for the Serral product, compared with 5.80+/-1.72 h and 5. 09+/-1.03 h for Celgene fasted and fed, respectively. Celgene's formulation exhibited slightly greater bioavailability than Serral's formulation, with mean ratios of 122% and 110% for Ln-transformed AUC(0-t) and AUC(0-infinity), respectively. The mean C(max) for the Celgene formulation was approximately two times greater than Serral's. Food delayed the onset of absorption of by 0.5-1.5 h, but had little effect on the extent of absorption from the Celgene capsule. Under fasted conditions, the Celgene thalidomide resulted in a two-fold greater C(max) and 10% greater AUC(0-infinity) than the Serral formulation.


Subject(s)
Dietary Fats/pharmacology , Hypnotics and Sedatives/pharmacokinetics , Intestinal Absorption/drug effects , Thalidomide/pharmacokinetics , Administration, Oral , Adult , Analysis of Variance , Area Under Curve , Biological Availability , Capsules , Chemistry, Pharmaceutical , Fasting , Female , Half-Life , Humans , Hypnotics and Sedatives/blood , Male , Middle Aged , Thalidomide/blood
13.
Bioorg Med Chem Lett ; 9(11): 1625-30, 1999 Jun 07.
Article in English | MEDLINE | ID: mdl-10386948

ABSTRACT

Thalidomide, (1), is a known inhibitor of TNF-alpha release in LPS stimulated human PBMC. Herein we describe the TNF-alpha inhibitory activity of amino substituted analogs of thalidomide (1) and its isoindolin-1-one analog, EM-12 (2). The 4-amino substituted analogs were found to be potent inhibitors of TNF-alpha release in LPS stimulated human PBMC.


Subject(s)
Thalidomide/analogs & derivatives , Thalidomide/chemical synthesis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Blood/drug effects , Humans , Inhibitory Concentration 50 , Models, Chemical , Structure-Activity Relationship , U937 Cells
14.
J Immunol ; 163(1): 380-6, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10384139

ABSTRACT

TNF-alpha mediates both protective and detrimental manifestations of the host immune response. Our previous work has shown thalidomide to be a relatively selective inhibitor of TNF-alpha production in vivo and in vitro. Additionally, we have recently reported that thalidomide exerts a costimulatory effect on T cell responses. To develop thalidomide analogues with increased anti-TNF-alpha activity and reduced or absent toxicities, novel TNF-alpha inhibitors were designed and synthesized. When a selected group of these compounds was examined for their immunomodulatory activities, different patterns of cytokine modulation were revealed. The tested compounds segregated into two distinct classes: one class of compounds, shown to be potent phosphodiesterase 4 inhibitors, inhibited TNF-alpha production, increased IL-10 production by LPS-induced PBMC, and had little effect on T cell activation; the other class of compounds, similar to thalidomide, were not phosphodiesterase 4 inhibitors and markedly stimulated T cell proliferation and IL-2 and IFN-gamma production. These compounds inhibited TNF-alpha, IL-1beta, and IL-6 and greatly increased IL-10 production by LPS-induced PBMC. Similar to thalidomide, the effect of these agents on IL-12 production was dichotomous; IL-12 was inhibited when PBMC were stimulated with LPS but increased when cells were stimulated by cross-linking the TCR. The latter effect was associated with increased T cell CD40 ligand expression. The distinct immunomodulatory activities of these classes of thalidomide analogues may potentially allow them to be used in the clinic for the treatment of different immunopathological disorders.


Subject(s)
Cytokines/metabolism , Lymphocyte Activation/drug effects , T-Lymphocytes/immunology , Thalidomide/classification , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , CD40 Antigens/metabolism , CD40 Ligand , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cytokines/biosynthesis , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Isoenzymes/metabolism , Ligands , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Membrane Glycoproteins/biosynthesis , T-Lymphocytes/metabolism , Thalidomide/analogs & derivatives , Tumor Necrosis Factor-alpha/biosynthesis
15.
Semin Cutan Med Surg ; 17(4): 231-42, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9859910

ABSTRACT

Thalidomide, originally marketed as a sedative, was introduced in West Germany in 1956 and in numerous other countries soon thereafter. In part because it did not impair coordination or respiratory function, the drug rapidly became extremely popular. By 1961, however, there were mounting reports of phocomelia and other severe congenital abnormalities associated with maternal use of thalidomide, and the drug was withdrawn from the market and its availability highly restricted. A few years later, thalidomide would find use in dermatology after it was reported that leprosy patients with erythema nodosum leprosum (ENL) experienced rapid and dramatic improvement after taking the drug as a sedative. Additional data quickly confirmed thalidomide's efficacy in ENL, and today it is the drug of choice in the condition. In subsequent decades, the drug has been successfully tried in treatment of a variety of apparently unrelated dermatologic disorders. Meanwhile, thalidomide has been shown to possess a range of biologic actions, including inhibition of tumor necrosis factor alpha, possibly relevant to its clinical efficacy. Dermatologic disorders in addition to ENL in which thalidomide's effectiveness is well documented include aphthous stomatitis, discoid lupus erythematosus, actinic prurigo, Behçet's disease, and prurigo nodularis. More recently, the drug has been employed in dermatologic conditions associated with HIV infection. When used with safeguards to prevent teratogenicity and the drug's other major adverse effect, peripheral neuropathy, thalidomide may offer a good therapeutic option for many patients in whom other drug therapies have proven inadequate.


Subject(s)
Dermatologic Agents/therapeutic use , Skin Diseases/drug therapy , Thalidomide/therapeutic use , Dermatologic Agents/adverse effects , Dermatologic Agents/pharmacology , Humans , Thalidomide/adverse effects , Thalidomide/pharmacology
19.
Philos Trans R Soc Lond B Biol Sci ; 297(1088): 481-96, 1982 Jun 11.
Article in English | MEDLINE | ID: mdl-6125958

ABSTRACT

There have been numerous instances reported when potentially recalcitrant compounds have been modified by microorganisms or completely mineralized by mixed communities or organisms; an example is pesticide biodegradation. Both situations rely upon the ability of microorganisms to transform compounds that they cannot utilize as sole sources of carbon and energy. This phenomenon of co-oxidation or co-metabolism has been fraught with confusion for many years as a result of the ambiguous use of terms and definitions. A redefinition of co-metabolism is proposed in an attempt to alleviate the problem: Co-metabolism--the transformation of a non-growth substrate in the obligate presence of a growth substrate or another transformable compound. The term 'non-growth substrate' describes compounds that are unable to support cell replication as opposed to an increase in biomass. This definition was devised primarily as a result of non-growth substrate metabolism studies with methane-utilizing bacteria. These studies are described in the text. The possible impact of endogenous polymer reserves on co-metabolic events is discussed. A number of examples where non-growth substrate metabolism is of environmental importance are presented, in particular the potential role of methane-oxidizing bacteria in the removal of CO from the environment. The evolutionary significance, if any, of fortuitous metabolism or co-metabolism is discussed, as are potential applications of these phenomena.


Subject(s)
Bacteria/metabolism , Biodegradation, Environmental , Methylococcaceae/metabolism , Pesticides/metabolism , Bacteria/enzymology , Biological Evolution , Kinetics , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...