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










Database
Language
Publication year range
1.
J Med Chem ; 65(7): 5300-5316, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35302767

ABSTRACT

Bruton's tyrosine kinase (BTK), a Tec family tyrosine kinase, is critical in immune pathways as an essential intracellular signaling element, participating in both adaptive and immune responses. Currently approved BTK inhibitors are irreversible covalent inhibitors and limited to oncology indications. Herein, we describe the design of covalent reversible BTK inhibitors and the discoveries of PRN473 (11) and rilzabrutinib (PRN1008, 12). These compounds have exhibited potent and durable inhibition of BTK, in vivo efficacy in rodent arthritis models, and clinical efficacy in canine pemphigus foliaceus. Compound 11 has completed phase 1 trials as a topical agent, and 12 is in phase 3 trials for pemphigus vulgaris and immune thrombocytopenia.


Subject(s)
Protein Kinase Inhibitors , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Animals , Dogs , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
J Invest Dermatol ; 138(4): 864-871, 2018 04.
Article in English | MEDLINE | ID: mdl-29129599

ABSTRACT

The chronic and highly prevalent skin disorder psoriasis vulgaris is characterized by a hyperproliferative epidermis and aberrant immune activity. Many studies have highlighted the role of differentiated T lymphocytes in psoriasis progression. Several biologics are currently available that target proinflammatory cytokines produced by T lymphocytes, but the need for improved therapies persists. The small molecule PRN694 covalently binds ITK and RLK, two Tec kinases activated downstream of T-lymphocyte activation, both of which are up-regulated in psoriatic skin. These Tec kinases are involved in signaling cascades mediating T-lymphocyte proliferation, differentiation, and migration and proinflammatory cytokine production. In vitro analysis showed that PRN694 effectively inhibited IL-17A production from murine T helper type 17-differentiated T lymphocytes. Additionally, PRN694 effectively reduced the psoriasis-like phenotype severity and reduced epidermal proliferation and thickness in both the Rac1V12 and imiquimod mouse models of psoriasis. PRN694 also inhibited CD3+ T-cell and γδ T-cell infiltration into skin regions. Inhibition of ITK and RLK attenuated psoriasis-associated signaling pathways, indicating that PRN694 is an effective psoriasis therapeutic.


Subject(s)
Benzimidazoles/pharmacology , Dermis/pathology , Gene Expression Regulation , Immunity, Cellular , Protein-Tyrosine Kinases/genetics , Psoriasis/genetics , Animals , Cells, Cultured , Dermis/metabolism , Disease Models, Animal , Humans , Mice , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/biosynthesis , Psoriasis/drug therapy , Psoriasis/immunology , RNA, Messenger/genetics , T-Lymphocytes/immunology
3.
J Clin Pharmacol ; 57(2): 194-210, 2017 02.
Article in English | MEDLINE | ID: mdl-27406873

ABSTRACT

The spleen tyrosine kinase (SYK) regulates immune cell activation in response to engagement of a variety of receptors, making it an intriguing target for the treatment of inflammatory and autoimmune disorders as well as certain B-cell malignancies. We have previously reported on the discovery and preclinical characterization of PRT062607, a potent and highly selective inhibitor of SYK that exhibits robust anti-inflammatory activity in a variety of animal models. Here we present data from our first human studies aimed at characterizing the pharmacokinetics (PK), pharmacodynamics (PD), and safety of PRT062607 in healthy volunteers following single and multiple oral administrations. PRT062607 demonstrated a favorable PK profile and the ability to completely inhibit SYK activity in multiple whole-blood assays. The PD half-life in the more sensitive assays was approximately 24 hours and returned to predose levels by 72 hours. Selectivity for SYK was observed at all dose levels tested. Analysis of the PK/PD relationship indicated an IC50 of 324 nM for inhibition of B-cell antigen receptor-mediated B-cell activation and 205 nM for inhibition of FcεRI-mediated basophil degranulation. PRT062607 was safe and well tolerated across the entire range of doses. Clinical PK/PD was related to in vivo anti-inflammatory activity of PRT062607 in the rat collagen-induced arthritis model, which predicts that therapeutic concentrations may be safely achieved in humans for the treatment of autoimmune disease. PRT062607 has a desirable PK profile and is capable of safely, potently, and selectively suppressing SYK kinase function in humans following once-daily oral dosing.


Subject(s)
Cyclohexylamines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Spleen/drug effects , Spleen/enzymology , Adult , Animals , Arthritis, Experimental/drug therapy , B-Lymphocytes/drug effects , Basophil Degranulation Test , Cyclohexylamines/pharmacokinetics , Dendritic Cells/drug effects , Half-Life , Healthy Volunteers , Humans , Macrophage Activation/drug effects , Male , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacokinetics , Rats , Receptors, Antigen, B-Cell/drug effects , Respiratory Burst/drug effects , Single-Blind Method
4.
J Immunol ; 195(10): 4822-31, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26466958

ABSTRACT

In T cells, the Tec kinases IL-2-inducible T cell kinase (ITK) and resting lymphocyte kinase (RLK) are activated by TCR stimulation and are required for optimal downstream signaling. Studies of CD4(+) T cells from Itk(-/-) and Itk(-/-)Rlk(-/-) mice have indicated differential roles of ITK and RLK in Th1, Th2, and Th17 differentiation and cytokine production. However, these findings are confounded by the complex T cell developmental defects in these mice. In this study, we examine the consequences of ITK and RLK inhibition using a highly selective and potent small molecule covalent inhibitor PRN694. In vitro Th polarization experiments indicate that PRN694 is a potent inhibitor of Th1 and Th17 differentiation and cytokine production. Using a T cell adoptive transfer model of colitis, we find that in vivo administration of PRN694 markedly reduces disease progression, T cell infiltration into the intestinal lamina propria, and IFN-γ production by colitogenic CD4(+) T cells. Consistent with these findings, Th1 and Th17 cells differentiated in the presence of PRN694 show reduced P-selectin binding and impaired migration to CXCL11 and CCL20, respectively. Taken together, these data indicate that ITK plus RLK inhibition may have therapeutic potential in Th1-mediated inflammatory diseases.


Subject(s)
Cell Differentiation/drug effects , Colitis/prevention & control , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/immunology , Th1 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Colitis/genetics , Colitis/immunology , Colitis/pathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Protein-Tyrosine Kinases/genetics , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
5.
J Pharmacol Exp Ther ; 339(1): 54-61, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21730013

ABSTRACT

It is known that hepatic metabolism limits the antiaggregatory activity of clopidogrel and, as a consequence, its clinical benefits. In this study, we investigated whether other factors exist that could account for clopidogrel's suboptimal antithrombotic activity. Using an in vivo murine FeCl(3) thrombosis model coupled with intravital microscopy, we found that at equivalent, maximal levels of inhibition of ADP-induced platelet aggregation, clopidogrel (50 mg/kg p.o.) failed to reproduce the phenotype associated with P2Y(12) deficiency. However, elinogrel (60 mg/kg p.o.), a direct-acting reversible P2Y(12) antagonist, achieved maximal levels of inhibition in vivo, and its administration (1 mg/kg i.v.) abolished residual thrombosis associated with clopidogrel dosing. Because elinogrel is constantly present in the plasma, whereas the active metabolite of clopidogrel exists for ∼2 h, we evaluated whether an intracellular pool of P2Y(12) exists that would be inaccessible to clopidogrel and contribute to its limited antithrombotic activity. Using saturation [(3)H]2-(methylthio)ADP ([(3)H]2MeSADP) binding studies, we first demonstrated that platelet stimulation with thrombin and convulxin (mouse) and thrombin receptor activating peptide (TRAP) (human) significantly increased surface expression of P2Y(12) relative to that of resting platelets. We next found that clopidogrel dose-dependently inhibited ADP-induced aggregation, signaling (cAMP), and surface P2Y(12) on resting mouse platelets, achieving complete inhibition at the highest dose (50 mg/kg), but failed to block this inducible pool. Thus, an inducible pool of P2Y(12) exists on platelets that can be exposed upon platelet activation by strong agonists. This inducible pool is not blocked completely by clopidogrel, contributes to thrombosis in vivo, and can be blocked by elinogrel.


Subject(s)
Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Quinazolinones/pharmacology , Receptors, Purinergic P2Y12/physiology , Sulfonamides/pharmacology , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Animals , Clopidogrel , Cyclic AMP/blood , Fibrinolytic Agents/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/metabolism , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Radioligand Assay , Receptors, Purinergic P2Y12/genetics , Thrombosis/chemically induced , Ticlopidine/pharmacology
6.
J Pharmacol Exp Ther ; 338(1): 22-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21447613

ABSTRACT

Clinical studies with clopidogrel or prasugrel show that although increased inhibition of P2Y(12) and platelet function improves efficacy, bleeding is also increased. Other preclinical and clinical studies have suggested a greater therapeutic index (TI) with reversible inhibitors and disproportionate effects of thienopyridines on bleeding at high doses. We used multiple in vivo (FeCl(3)-induced arterial thrombosis in mesenteric arteries, blood loss after tail transsection, and platelet deposition and wound closure time in a micropuncture model in mesenteric veins) and ex vivo (light transmittance aggregometry, prothrombin time, and activated partial thromboplastin time) mouse models to 1) compare the TI of clopidogrel, prasugrel, and elinogrel, a reversible, competitive antagonist, with that in P2Y(12)(-/-) mice and 2) determine whether the bleeding consequences of the thienopyridines are attributed only to the inhibition of P2Y(12). Data indicated greater (elinogrel) and decreased (thienopyridines) TI compared with that in P2Y(12)(-/-) mice. The impaired TI associated with the thienopyridines was not attributed to non-P2Y(12) activities on platelet function or coagulation but was related to a direct effect at the vessel wall (inhibition of vascular tone). Further analysis showed that the prasugrel off-target effect was dose- and time-dependent and of a reversible nature. In conclusion, the TI of thienopyridines in the mouse may be decreased by P2Y(12)-independent off-target effects at the vessel wall, whereas that of elinogrel may be enhanced by the reversible, competitive nature of the antiplatelet agent.


Subject(s)
Drug Delivery Systems , Endothelium, Vascular/drug effects , Hemorrhage/chemically induced , Quinazolinones/administration & dosage , Receptors, Purinergic P2Y12/deficiency , Sulfonamides/administration & dosage , Thienopyridines/administration & dosage , Animals , Dose-Response Relationship, Drug , Endothelium, Vascular/physiology , Hemorrhage/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Purinergic P2 Receptor Antagonists/administration & dosage , Purinergic P2 Receptor Antagonists/adverse effects , Quinazolinones/adverse effects , Sulfonamides/adverse effects , Thienopyridines/adverse effects , Thrombosis/drug therapy , Thrombosis/physiopathology
7.
J Neurosci ; 25(34): 7847-57, 2005 Aug 24.
Article in English | MEDLINE | ID: mdl-16120787

ABSTRACT

Morphine activates mu-opioid receptors (MORs) without promoting their rapid endocytosis in a number of cell types. A previous study suggested that morphine can drive rapid redistribution of MORs in the nucleus accumbens, but it was not possible in this in vivo study to identify a specific membrane trafficking pathway affected by morphine, to exclude possible indirect actions of morphine via opiate-regulated neural circuitry, or to define the mechanism of this morphine-dependent regulation. In the present study, we addressed these questions using dissociated primary cultures of rat striatal neurons as a model system. Morphine promoted a rapid redistribution of both endogenous and recombinant MORs within 30 min after drug addition to the culture medium. This effect was mediated by rapid endocytosis and occurred in a cell-autonomous manner, as indicated by its detection in cells plated at low density and in cultures in which depolarization was blocked by tetrodotoxin. Morphine-induced endocytosis of MORs was quantitatively similar to that induced by the enkephalin analog D-Ala2-N-Me-Phe4-Glycol5-enkephalin, and endocytosis induced by both ligands was inhibited by a dominant-negative mutant version of arrestin-3 (beta-arrestin-2). These results extend previous in vivo results and indicate that morphine is indeed capable of driving rapid endocytosis of mu-opioid receptors in an important subset of opiate-responsive CNS neurons. They also suggest a cellular mechanism by which beta-arrestins may modulate the physiological effects of morphine in vivo.


Subject(s)
Arrestin/physiology , Corpus Striatum/drug effects , Endocytosis/drug effects , Morphine/pharmacology , Neurons/drug effects , Receptors, Opioid, mu/metabolism , Animals , Arrestin/metabolism , Cells, Cultured , Corpus Striatum/metabolism , Endocytosis/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , Neurons/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors
8.
Curr Opin Neurobiol ; 13(3): 348-53, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12850220

ABSTRACT

Opiate drugs such as morphine and heroin are among the most effective analgesics known. Prolonged or repeated administration of opiates produces adaptive changes in the nervous system that lead to reduced drug potency or efficacy (tolerance), as well as physiological withdrawal symptoms and behavioral manifestations such as craving when drug use is terminated (dependence). These adaptations limit the therapeutic utility of opiate drugs, particularly in the treatment of chronically painful conditions, and are thought to contribute to the highly addictive nature of opiates. For many years it has been proposed that physiological tolerance to opiate drugs is associated with a modification of the number or functional activity of opioid receptors in specific neurons. We now understand certain mechanisms of opioid receptor desensitization and endocytosis in considerable detail. However, the functional roles that these mechanisms play in the complex physiological adaptation of the intact nervous system to opiates are only beginning to be explored.


Subject(s)
Adaptation, Physiological/drug effects , Analgesics, Opioid/pharmacology , Endocytosis/drug effects , Receptors, Opioid/agonists , Adaptation, Physiological/physiology , Animals , Drug Tolerance/physiology , Endocytosis/physiology , Humans , Receptors, Opioid/chemistry , Receptors, Opioid/physiology
9.
J Neurosci ; 23(10): 4324-32, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12764121

ABSTRACT

Morphine stimulates the internalization of mu-opioid receptors (MORs) in transfected cell models to a lesser degree than opioid peptides and other analgesic drugs, such as methadone, and previous studies have reported that morphine does not produce a detectable redistribution of MORs in neural tissue after either acute or chronic administration. Nevertheless, morphine produces profound physiological effects, raising the question of whether receptor trafficking plays any role in the in vivo actions of morphine. We investigated the effects of opiate drugs on recombinant and native opioid receptors in the nucleus accumbens, which plays an important role in mediating the behavioral effects of opiate drugs. Morphine and methadone differed in their effects on the internalization of epitope-tagged MORs in cell bodies, introduced by viral gene transfer and imaged by fluorescence microscopy. A mutation of the cytoplasmic tail that confers morphine-induced internalization in cultured cells had a similar effect on receptor trafficking in nucleus accumbens cell bodies. Surprisingly, in contrast to its failure to affect MOR distribution detectably in cell bodies, acute morphine administration produced a pronounced change in MOR distribution visualized in the processes of the same neurons. A similar effect of acute morphine administration was observed for endogenously expressed MORs by immunoelectron microscopy; the acute administration of morphine increased the density of MORs associated with internal membrane structures specifically in dendrites. These results provide the first evidence that morphine regulates the distribution of MORs in neuronal processes, suggesting that "compartment-selective" membrane trafficking represents a previously unanticipated type of opioid receptor regulation contributing to the in vivo effects of opiate drugs on a physiologically relevant population of CNS neurons.


Subject(s)
Dendrites/drug effects , Dendrites/metabolism , Morphine/pharmacology , Nucleus Accumbens/drug effects , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Dendrites/physiology , Dendrites/ultrastructure , Endocytosis/drug effects , Endocytosis/physiology , Genetic Vectors/genetics , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Membrane Proteins/metabolism , Methadone/administration & dosage , Methadone/pharmacology , Morphine/administration & dosage , Mutation , Neurons/chemistry , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/chemistry , Nucleus Accumbens/ultrastructure , Nucleus Accumbens/virology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/ultrastructure , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/ultrastructure , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Simplexvirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...