Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
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 ; 87(10): 1166-72, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12402158

ABSTRACT

The anti-tumour effects of thalidomide have been associated with its anti-angiogenic properties. Second generation thalidomide analogues are distinct compounds with enhanced therapeutic potential. Although these compounds are beginning to enter trials for the treatment of cancer there is very little information regarding the anti-angiogenic activity of these clinically relevant compounds. Furthermore, it is not known how the various immunomodulatory activities of these compounds relate to anti-angiogenic activity. In this study we assessed the anti-angiogenic activity of compounds from both IMiD and SelCID classes of analogues using a novel in vitro multicellular human assay system and the established rat aorta assay. Our results show that both the IMiDs and SelCIDs tested are significantly more potent than thalidomide. The anti-angiogenic potency of the analogues was not related to inhibition of endothelial cell proliferation, nor their TNF-alpha/PDE type 4 inhibitory properties. However, anti-migratory effects in vitro and inhibition of tumour growth in vivo was observed with the analogue IMiD-1 (clinically known as REVIMID). Our results show that anti-angiogenic activity spans both currently defined classes of thalidomide analogue and is not related to their previously described immunomodulatory properties. Identification of the differential effects of these compounds will enable targeting of such compounds into the appropriate clinical setting.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Thalidomide/analogs & derivatives , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Animals , Cell Division/drug effects , Cell Movement/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4 , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Humans , Male , Mice , Rats , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
4.
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
5.
Org Lett ; 1(3): 379-81, 1999 Aug 12.
Article in English | MEDLINE | ID: mdl-10822580

ABSTRACT

[formula: see text] Deprotonation of enantiopure (R,R)-1,2-dicyclohexyl-1,2-ethanediol 1-chloro-4-cyanobutylboronates 5 with LDA followed by treatment with anhydrous magnesium bromide yields (R)-(trans-2-cyanocyclobutyl)boronic esters 7 in high diastereomeric and enantiomeric purity. No cyclobutane formation has been observed in the absence of at least a catalytic amount of magnesium halide.


Subject(s)
Boronic Acids/chemistry , Butyrates/chemical synthesis , Cyclobutanes/chemical synthesis , Nitriles/chemical synthesis , Catalysis , Cyclization , Esters , Magnesium/chemistry , Mass Spectrometry , Stereoisomerism
6.
J Med Chem ; 41(9): 1446-55, 1998 Apr 23.
Article in English | MEDLINE | ID: mdl-9554878

ABSTRACT

Comparison of the high-resolution X-ray structures of the native HIV-1 protease and its complexes with the inhibitors suggested that the enzyme flaps are flexible. The movement at the tip of the flaps could be as large as 7 A. On the basis of this observation, cyclic cyanoguanidines have been designed, synthesized, and evaluated as HIV-1 protease (PR) inhibitors. Cyclic cyanoguanidines were found to be very potent inhibitors of HIV-1 protease. The choice of cyclic cyanoguanidines over cyclic guanidines was based on the reduced basicity of the former. X-ray structure studies of the HIV PR complex with cyclic cyanoguanidine demonstrated that in analogy to cyclic urea, cyclic cyanoguanidines also displace the unique structural water molecule. The structure-activity relationship of the cyclic cyanoguanidines is compared with that of the corresponding cyclic urea analogues. The differences in binding constants of the two series of compounds have been rationalized using high-resolution X-ray structure information.


Subject(s)
Anti-HIV Agents , Guanidines , HIV Protease Inhibitors , HIV Protease/metabolism , HIV-1/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Cell Line , Crystallography, X-Ray , Guanidines/chemical synthesis , Guanidines/chemistry , Guanidines/metabolism , Guanidines/pharmacology , HIV Protease/chemistry , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/pharmacology , HIV-1/enzymology , Humans , Hydrogen Bonding , Models, Molecular , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL