Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Pharmacol Exp Ther ; 364(3): 474-484, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29311111

RESUMEN

Despite the efficacy of biologics for treatment of rheumatoid arthritis (RA), many patients show inadequate responses and likely require neutralization of multiple mediators. Neutralization of both interleukin (IL)-1ß and IL-17A with monoclonal antibodies showed greater efficacy than either agent alone in a mouse arthritis model with cooperative inhibition of key inflammatory factors, IL-6, granulocyte colony-stimulating factor (G-CSF), and CXC chemokine ligand (CXCL)1. Given the potential clinical benefit in RA, we generated a human dual variable domain antibody Ig, ABBV-615, capable of simultaneous binding and neutralization of IL-1ß and IL-17A. ABBV-615 was characterized and evaluated in cynomolgus monkeys for pharmacokinetics and toxicity to enable clinical development. ABBV-615 exhibited affinities (KD) of 12 and 3 pM on human IL-1ß and IL-17A, respectively, and potencies (IC50) of 3 and 58 pM, respectively, as well as excellent drug-like properties. ABBV-615 pharmacokinetics in cynomolgus monkeys was dose proportional from 20 to 100 mg/kg with a mean half-life of 16 days. However, a 13-week repeat-dose toxicity study in cynomolgus monkeys revealed time-dependent spontaneous infections exclusively in skin at all doses tested and not historically seen with single-agent anti-IL-1α/ß or anti-IL-17A. Consistent with reduced resistance to skin infections, IL-1ß- and IL-17A-stimulated human keratinocytes demonstrate cooperative or compensatory production of key antibacterial and inflammatory mediators such as lipocalin-2, G-CSF, CXCL1, IL-8, tumor necrosis factor, and IL-6, which aid in defense against skin bacterial infections. These results illustrate the skin-specific antimicrobial mechanisms of IL-1ß and IL-17A and highlight the importance of understanding unique combinatorial effects of biologic agents.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Interleucina-17/inmunología , Interleucina-1beta/inmunología , Piel/efectos de los fármacos , Piel/microbiología , Animales , Artritis Experimental/inmunología , Humanos , Macaca fascicularis , Masculino , Ratones
2.
RMD Open ; 10(2)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38942592

RESUMEN

OBJECTIVES: To investigate the efficacy, safety, pharmacokinetics and pharmacodynamics of nipocalimab in participants with moderate to severe active rheumatoid arthritis (RA) and inadequate response or intolerance to ≥1 antitumour necrosis factor agent. METHODS: In this phase 2a study, participants with RA seropositive for anticitrullinated protein antibodies (ACPA) or rheumatoid factors were randomised 3:2 to nipocalimab (15 mg/kg intravenously every 2 weeks) or placebo from Weeks 0 to 10. Efficacy endpoints (primary endpoint: change from baseline in Disease Activity Score 28 using C reactive protein (DAS28-CRP) at Week 12) and patient-reported outcomes (PROs) were assessed through Week 12. Safety, pharmacokinetics and pharmacodynamics were assessed through Week 18. RESULTS: 53 participants were enrolled (nipocalimab/placebo, n=33/20). Although the primary endpoint did not reach statistical significance for nipocalimab versus placebo, a numerically higher change from baseline in DAS28-CRP at Week 12 was observed (least squares mean (95% CI): -1.03 (-1.66 to -0.40) vs -0.58 (-1.24 to 0.07)), with numerically higher improvements in all secondary efficacy outcomes and PROs. Serious adverse events were reported in three participants (burn infection, infusion-related reaction and deep vein thrombosis). Nipocalimab significantly and reversibly reduced serum immunoglobulin G, ACPA and circulating immune complex levels but not serum inflammatory markers, including CRP. ACPA reduction was associated with DAS28-CRP remission and 50% response rate in American College of Rheumatology (ACR) criteria; participants with a higher baseline ACPA had greater clinical improvement. CONCLUSIONS: Despite not achieving statistical significance in the primary endpoint, nipocalimab showed consistent, numerical efficacy benefits in participants with moderate to severe active RA, with greater benefit observed for participants with a higher baseline ACPA. TRIAL REGISTRATION NUMBER: NCT04991753.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antirreumáticos , Artritis Reumatoide , Índice de Severidad de la Enfermedad , Humanos , Artritis Reumatoide/tratamiento farmacológico , Masculino , Femenino , Persona de Mediana Edad , Resultado del Tratamiento , Antirreumáticos/uso terapéutico , Antirreumáticos/administración & dosificación , Antirreumáticos/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anciano , Adulto , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Método Doble Ciego , Medición de Resultados Informados por el Paciente , Anticuerpos Antiproteína Citrulinada/sangre
3.
Protein Expr Purif ; 75(1): 55-62, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20826216

RESUMEN

Acidic mammalian chitinase (AMCase) is an enzyme that selectively degrades the biopolymer chitin. Several chitinase enzymes are utilized by mammals to hydrolyze chitin encountered by inhalation and ingestion. AMCase is distinct from other mammalian chitinases as its activity is retained in strongly acidic conditions (pH <2.0). AMCase expression is induced by antigen-induced mouse models of allergic lung inflammation. This protein has also been implicated in the pathogenesis of asthma although its precise role is poorly defined. We describe a novel way to express and purify active murine AMCase. This material retains properties observed in mouse bronchoalveolar lavage (BAL) fluid with regard to pH preference of activity and its inhibition by cyclic peptide inhibitors argifin and argadin. We found that chitinase in BAL from both antigen-challenged and control animals have similar properties in this regard. This strongly supports the notion the same enzyme (AMCase) gives rise to chitinase activity in both challenged and unchallenged animals. We also describe expression of active human AMCase. The methods described in this paper provide a reliable source of recombinant AMCase that can be utilized to expand understanding of AMCase's role in regulating allergic inflammation.


Asunto(s)
Quitinasas/genética , Quitinasas/metabolismo , Clonación Molecular/métodos , Secuencia de Aminoácidos , Animales , Líquido del Lavado Bronquioalveolar , Células COS , Línea Celular , Quitinasas/aislamiento & purificación , Chlorocebus aethiops , Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Péptidos Cíclicos/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
4.
J Transl Autoimmun ; 4: 100079, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33490940

RESUMEN

Nuclear factor (erythroid-derived 2) like 2 (NRF2) is a nuclear transcription factor activated in response to oxidative stress that induces a gene program that dampens inflammation and can limit cell damage that perpetuates the inflammatory response. We have identified A-1396076, a potent and selective NRF2 activator with demonstrated KEAP1 binding and modulation of cellular NRF2 mediated effects. In vivo administration of A-1396076 inhibits inflammation across several rodent models of autoimmunity when administered at or before the time of antigen challenge while also inducing NRF2 modulated gene transcription in the liver of the animals. It was not effective when administered after the time of antigen challenge or in a T cell independent model of arthritis induced by passive transfer of anti-collagen antibodies. A-1396076 inhibited antigen dependent T cell activation as measured by IFN-γ production in an ex vivo re-stimulation assay and following anti-CD3 challenge of MOG-sensitized mice. A-1396076 reduced costimulatory molecule expression on dendritic cells in the lungs of OVA LPS challenged mice suggesting that the mechanism of T cell inhibition was mediated at least partially by interfering with antigen presentation. These data suggest that NRF2 activation may be an effective strategy to dampen inflammation for treatment of autoimmune disease.

5.
J Pharmacol Exp Ther ; 324(2): 576-86, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17986646

RESUMEN

Statins, because of their excellent efficacy and manageable safety profile, represent a key component in the current armamentarium for the treatment of hypercholesterolemia. Nonetheless, myopathy remains a safety concern for this important drug class. Cerivastatin was withdrawn from the market for myotoxicity safety concerns. BMS-423526 [{(3R,5S)-7-[4-(4-fluorophenyl)-6,7-dihydro-2-(1-methylethyl)-5H-benzo[6,7]cyclohepta[1,2-b]pyridin-3-yl]-3,5-dihydroxy-heptenoic acid} sodium salt], similar to cerivastatin in potency and lipophilicity, was terminated in early clinical development due to an unacceptable myotoxicity profile. In this report, we describe the guinea pig as a model of statin-induced cholesterol lowering and myotoxicity and show that this model can distinguish statins with unacceptable myotoxicity profiles from statins with acceptable safety profiles. In our guinea pig model, both cerivastatin and BMS-423526 induced myotoxicity at doses near the ED(50) for total cholesterol (TC) lowering in plasma. In contrast, wide differences between myotoxic and TC-lowering doses were established for the currently marketed, more hydrophilic statins, pravastatin, rosuvastatin, and atorvastatin. This in vivo model compared favorably to an in vitro model, which used statin inhibition of cholesterol synthesis in rat hepatocytes and L6 myoblasts as surrogates of potential efficacy and toxicity, respectively. Our conclusion is that the guinea pig is a useful preclinical in vivo model for demonstrating whether a statin is likely to have an acceptable therapeutic safety margin.


Asunto(s)
Cobayas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Modelos Animales , Animales , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Cobayas/sangre , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ratas , Ratas Sprague-Dawley
6.
J Med Chem ; 51(9): 2722-33, 2008 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-18412317

RESUMEN

3-hydroxy-3-methylglutaryl coenzyme-A reductase (HMGR) inhibitors, more commonly known as statins, represent the gold standard in treating hypercholesterolemia. Although statins are regarded as generally safe, they are known to cause myopathy and, in rare cases, rhabdomyolysis. Statin-dependent effects on plasma lipids are mediated through the inhibition of HMGR in the hepatocyte, whereas evidence suggests that myotoxicity is due to inhibition of HMGR within the myocyte. Thus, an inhibitor with increased selectivity for hepatocytes could potentially result in an improved therapeutic window. Implementation of a strategy that focused on in vitro potency, compound polarity, cell selectivity, and oral absorption, followed by extensive efficacy and safety modeling in guinea pig and rat, resulted in the identification of compound 1b (BMS-644950). Using this discovery pathway, we compared 1b to other marketed statins to demonstrate its outstanding efficacy and safety profile. With the potential to generate an excellent therapeutic window, 1b was advanced into clinical development.


Asunto(s)
Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/síntesis química , Pirimidinas/síntesis química , Triazoles/síntesis química , Administración Oral , Animales , Disponibilidad Biológica , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Colesterol/biosíntesis , Colesterol/sangre , Cristalografía por Rayos X , Perros , Femenino , Cobayas , Haplorrinos , Humanos , Hidroximetilglutaril-CoA Reductasas/química , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Técnicas In Vitro , Hígado/efectos de los fármacos , Hígado/metabolismo , Modelos Moleculares , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Pirimidinas/farmacología , Pirimidinas/toxicidad , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad , Triazoles/farmacología , Triazoles/toxicidad
7.
J Biol Chem ; 277(38): 35350-6, 2002 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-12122008

RESUMEN

Targeted gene disruption or overexpression of 12/15-lipoxygenase in mice on the genetic background of apolipoprotein E or low density lipoprotein-receptor (LDL-R) deficiency has implicated 12/15-lipoxygenase in atherogenesis. The data support indirectly a role for 12/15-lipoxygenase in the oxidative modification of low density lipoprotein. In this study we set out to explore other potential mechanisms for 12/15-lipoxygenase in atherosclerosis using apolipoprotein B mRNA editing catalytic polypeptide-1/LDL-R double-deficient mice, a model highly related to the human condition of familial hypercholesterolemia. 12/15-Lipoxygenase deficiency in this strain led to approximately 50% decrease in aortic lesions in male and female mice at 8 months on a chow diet in the absence of cholesterol differences. While studying 12/15-lipoxygenase-deficient macrophages in culture, we discovered a remarkable selective defect (75-90% decrease) in interleukin-12 production but not in tumor necrosis factor-alpha or nitric oxide release, in response to lipopolysaccharide in the presence or absence of interferon-gamma priming. The lipopolysaccharide/interferon-gamma response was associated with a 33-50% decrease in nuclear interferon consensus sequence-binding protein, which is consistent with interferon consensus sequence-binding protein containing protein complex-dependent regulation of the interleukin-12 p40 gene. The decrease in interleukin-12 production was recapitulated in vivo in mouse aortas of the triple knockout group and was reflected in a marked decrease in interferon-gamma expression. The data provide support for a novel mechanism linking the 12/15-lipoxygenase pathway to a known immunomodulatory Th1 cytokine in atherogenesis.


Asunto(s)
Araquidonato 12-Lipooxigenasa/fisiología , Araquidonato 15-Lipooxigenasa/fisiología , Arteriosclerosis/metabolismo , Modelos Animales de Enfermedad , Hiperlipoproteinemia Tipo II/metabolismo , Interleucina-12/biosíntesis , Macrófagos Peritoneales/metabolismo , Desaminasas APOBEC-1 , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Arteriosclerosis/enzimología , Arteriosclerosis/patología , Citidina Desaminasa/genética , Hiperlipoproteinemia Tipo II/enzimología , Hiperlipoproteinemia Tipo II/patología , Macrófagos Peritoneales/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA