Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Transl Sci ; 14(3): 820-828, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33650758

RESUMEN

Bruton's tyrosine kinase (BTK) is a target for treatment of hematologic malignancies and autoimmune diseases. TAK-020 is a highly selective covalent BTK inhibitor that inhibits both B cell receptor and fragment crystallizable receptor signaling. We assessed the safety/tolerability and pharmacokinetics/pharmacodynamics (PDs) of TAK-020 in healthy subjects. Each cohort of the single-rising dose (n = 72; 9 cohorts) and the multiple-rising dose (n = 48; 6 cohorts) portions of the study comprised six TAK-020-treated and two placebo-treated, subjects aged 18-55 years (inclusive). The PD effects were assessed by measuring BTK occupancy and the inhibition of fragment crystallizable epsilon receptor 1 (FcεRI)-mediated activation of basophils. Overall, treatment-emergent adverse events (TEAEs) were similar to placebo; there were no serious TEAEs or no TEAEs leading to discontinuation. TAK-020 was rapidly absorbed (median time to maximum plasma concentration (Tmax ) 45-60 minutes) with a half-life of ~ 3-9 hours at doses ≥ 2.5 mg. TAK-020 exposure was generally dose proportional for single doses ≤ 70 mg and after multiple doses of ≤ 60 mg once daily. Target occupancy was dose dependent, with doses ≥ 2.5 mg yielding maximum and sustained occupancy > 70% for > 96 hours. Single doses ≥ 4.4 mg reduced FcεRI-mediated activation of basophils by > 80% and comparable inhibition was observed with daily dosing ≥3.75 mg for 9 days. Inhibition persisted for 24-72 hours postdose and the duration generally increased with dose. TAK-020 was generally well-tolerated in healthy subjects after single and multiple doses and demonstrated target engagement and pathway modulation. The PD effects outlasted drug exposures, as expected for covalent inhibition of BTK.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Basófilos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/efectos adversos , Administración Oral , Adolescente , Adulto , Agammaglobulinemia Tirosina Quinasa/metabolismo , Basófilos/inmunología , Basófilos/metabolismo , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Miastenia Gravis/tratamiento farmacológico , Miastenia Gravis/inmunología , Placebos/administración & dosificación , Placebos/efectos adversos , Inhibidores de Proteínas Quinasas/administración & dosificación , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Púrpura Trombocitopénica Idiopática/inmunología , Adulto Joven
2.
Br J Clin Pharmacol ; 86(7): 1314-1325, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32045493

RESUMEN

AIMS: This investigation characterised tolerability, pharmacokinetics and pharmacodynamics of the anti-CD38 antibody TAK-079. METHODS: A randomised, double-blind, placebo-controlled trial of a single intravenous (i.v.) infusion or subcutaneous (s.c.) injection of TAK-079 at escalating doses in healthy subjects (n = 74), who were followed for 92 days postexposure. RESULTS: TAK-079 was well tolerated. All adverse events were mild or moderate. There were no withdrawals, infusion, or injection site reactions over the tested i.v. and s.c. doses up to 0.06 and 0.6 mg kg-1 , respectively. At higher doses, transient cytokine level increases, following i.v. administration, coincided with reduction in CD38-expressing cells; clinical symptoms included mild pyrexia, headache, and postural hypotension. Following an i.v. infusion of 0.06 mg kg-1 TAK-079, maximum observed serum concentration (Cmax ) was 100.4 (%CV: 52) ng mL-1 , time to Cmax was the end of infusion and natural killer (NK_ cells were reduced 93.8 (±8.5) % from baseline levels. Following a s.c. injection of 0.6 mg kg-1 TAK-079, Cmax was 23.0 (%CV: 67) ng mL-1 with time to Cmax of 24 (range 7.98-96.02) hours, and plasmablasts were subsequently reduced 93.4 (±8.8) % from predose levels. Serum immunoglobulin (Ig)M, IgA and IgG levels were reduced by 15-60% and had not returned to baseline levels within 78 days after administration at ≥0.3 mg kg-1 s.c. Reductions in NK cells at 0.6 mg kg-1 s.c. were approximately 2-3 times more durable than at 0.06 mg kg-1 i.v. CONCLUSIONS: TAK-079 was well tolerated and s.c. administration elicited more durable reductions in plasmablasts and NK cells. This plasmacytolytic profile could be useful for treating disorders caused by plasma or NK cells, malignant counterparts, and/or pathogenic antibodies.


Asunto(s)
Anticuerpos Monoclonales , Antineoplásicos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Voluntarios Sanos , Humanos , Células Asesinas Naturales
3.
J Pharmacol Exp Ther ; 370(2): 182-196, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31085699

RESUMEN

Ectoenzyme CD38 is increased on lymphocytes in response to an antigenic challenge and it is hypothesized that targeting these activated lymphocytes could ameliorate pathologic activities in autoimmune diseases. The cynomolgus monkey is an appropriate model for assessing potential effects of targeting CD38 in humans because these species exhibit similar expression profiles. TAK-079 is a human monoclonal antibody (IgG1 λ ) that binds to CD38 and lyses bound cells by complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity. TAK-079 binds to monkey CD38 with an affinity at EC50 4.5 nM, and the potential activity of TAK-079 was investigated in a monkey collagen-induced arthritis model of autoimmune disease. Prophylactic administration of TAK-079 (3 mg/kg i.v. weekly) was well tolerated and prevented arthritis development compared with vehicle-treated control animals, which exhibited progressive disease with radiographic damage and worsening clinical scores over the study course. Therapeutic treatment of arthritic monkeys with TAK-079 (3 mg/kg i.v. weekly) was also well tolerated and reduced disease progression and symptoms. Arthritis scores and joint swelling were significantly lower than the vehicle control, accompanied by decreases in blood levels of C-reactive protein, alkaline phosphatase, and natural killer, B, and T cells. Histopathology, morphometry, and radiology revealed significantly less joint damage in animals exposed prophylactically to TAK-079 treatment compared with vehicle-treated animals and significantly less damage in animals treated therapeutically with TAK-079 or dexamethasone (0.1 mg/kg oral gavage daily), illustrating potential disease-modifying activity. In conclusion, these data indicate that depletion of CD38-expressing cells could be a therapeutic mechanism for treating autoimmune diseases. SIGNIFICANCE STATEMENT: This study demonstrates that targeting CD38-expressing leukocytes with a cytolytic antibody can ameliorate autoimmune disease in cynomolgus monkeys. The study gives a unique perspective into this therapeutic strategy because the three other anti-CD38 cytolytic antibodies in clinical development (daratumumab, isatuximab, and MOR202) cannot be tested in similar models because they do not crossreact with CD38 expressed by new world primates.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Anticuerpos Monoclonales/inmunología , Artritis Experimental/inmunología , Linfocitos B/metabolismo , Regulación de la Expresión Génica/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos T/metabolismo , ADP-Ribosil Ciclasa 1/inmunología , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Linfocitos B/inmunología , Células CHO , Cricetulus , Progresión de la Enfermedad , Células Asesinas Naturales/inmunología , Macaca fascicularis , Linfocitos T/inmunología
4.
Pharmacol Res Perspect ; 6(3): e00402, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29864242

RESUMEN

We are studying the fully human, IgG1λ cytolytic monoclonal antibody TAK-079, which binds CD38. CD38 is expressed on plasma and natural killer (NK) cells constitutively and upregulated on subsets of B and T lymphocytes upon activation. TAK-079 cross-reacts with CD38 expressed by cynomolgus monkeys and depletes subsets of NK, B, and T cells. Therefore, safety and function of TAK-079 was evaluated in this species, prior to clinical development, using bioanalytical, and flow cytometry assays. We pooled the data from eight studies in healthy monkeys (dose range 0.03-100 mg/kg) and developed mathematical models that describe the pharmacokinetics and the exposure-effect relationship for NK cells, B cells, and T cells. NK cell depletion was identified as the most sensitive pharmacodynamic effect of TAK-079. It was adequately described with a turnover model (C50  = 27.5 µg/mL on depletion rate) and complete depletion was achieved with an IV dose of 0.3 mg/kg. Intermediate effects on T-cell counts were described with a direct response model (C50  = 11.9 µg/mL) and on B-cell counts with a 4-transit-compartment model (C50  = 19.8 µg/mL on depletion rate). Our analyses substantiate the observation that NK, B and T cells are cleared by TAK-079 at different rates and required different time spans to replete the blood compartment. The models were used to simulate pharmacokinetic and cell depletion profiles in humans after applying a straightforward scaling approach for monoclonal antibodies in preparation for the first-in-human clinical trial.


Asunto(s)
ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacocinética , Linfocitos B/inmunología , Células Asesinas Naturales/inmunología , Glicoproteínas de Membrana/antagonistas & inhibidores , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Linfocitos B/efectos de los fármacos , Esquema de Medicación , Femenino , Humanos , Células Asesinas Naturales/efectos de los fármacos , Depleción Linfocítica , Macaca fascicularis , Masculino , Modelos Teóricos , Linfocitos T/efectos de los fármacos
5.
Int Immunopharmacol ; 60: 160-169, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29730559

RESUMEN

C-C chemokine receptor 9 (CCR9) is the homing receptor for C-C motif chemokine ligand 25 (CCL25), and contributes to the maintenance of mucosal immunity and pathogenesis of inflammatory bowel disease (IBD) through the recruitment of T cells into the gut mucosa. Recent reports suggest that the interaction of CCR9 and CCL25 in the large intestine correlate with disease severity of colonic IBD. MLN3126 is an orally available small molecular compound with potent and selective CCR9 antagonist activity. MLN3126 inhibited CCL25-induced calcium mobilization in human CCR9 transfected cells and CCL25-induced chemotaxis of mouse primary thymocytes in a dose-dependent manner. The potential effect of MLN3126 in an activated T cell transfer mouse colitis model was compared with that of an anti-tumor necrosis factor (TNF)-α antibody. CCL25 protein was detected in the colon of mucosal epithelial cells and CCR9+ CD4+ T cells were observed in the lamina propria of the colon of mice with colitis. Dietary administration of MLN3126 to the mice maintained sufficient concentration of the compound in the plasma and dose-dependently inhibited progression of colitis compared to the vehicle control group. Anti-TNF-α antibody, a surrogate for a standard of care for IBD treatment, was also efficacious in the colitis model. These results suggest that MLN3126 would be a promising orally available CCR9 antagonist to treat colonic IBD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Receptores CCR/antagonistas & inhibidores , Animales , Células CHO , Movimiento Celular/efectos de los fármacos , Quimiocinas CC/inmunología , Colitis/inmunología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Cricetulus , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Ratones SCID , Linfocitos T/inmunología
6.
Clin Pharmacokinet ; 56(11): 1287-1301, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28523450

RESUMEN

Vedolizumab is a humanized anti-α4ß7 integrin monoclonal antibody that selectively blocks trafficking of memory T cells to inflamed gut tissue by inhibiting the α4ß7-mucosal addressin cell adhesion molecule-1 (MAdCAM-1) interaction. Approved for treating patients with moderately to severely active ulcerative colitis (UC) or Crohn's disease (CD), vedolizumab is administered as a 300 mg intravenous infusion. Vedolizumab undergoes a rapid, saturable, non-linear, target-mediated elimination process at low concentrations and a slower, linear, non-specific elimination process at higher concentrations. At therapeutic concentrations, vedolizumab primarily undergoes linear elimination. From population pharmacokinetic modeling, the vedolizumab terminal elimination half-life (t ½ ß) was estimated to be 25.5 days; linear clearance (CLL) was similar for patients with UC (0.159 L/day) and CD (0.155 L/day). Extreme low albumin concentrations and extreme high body weight values were potentially clinically important predictors of vedolizumab CLL. Other factors, including concomitant therapy use (methotrexate, azathioprine, mercaptopurine, or aminosalicylates) or prior tumor necrosis factor-α (TNF-α) antagonist use, had no clinically relevant effects on CLL. A positive exposure-efficacy relationship for clinical remission and clinical response was apparent for vedolizumab induction therapy in patients with UC or CD. On average, patients with higher albumin, lower fecal calprotectin (UC only), lower C-reactive protein (CD only), and no prior TNF-α antagonist use had a higher probability of remission. Off drug, 10% of patients with UC or CD were positive for anti-drug antibodies. This article provides a comprehensive review of the clinical pharmacokinetics, pharmacodynamics, exposure-efficacy relationships, and immunogenicity of vedolizumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacología , Colitis Ulcerosa/sangre , Colitis Ulcerosa/tratamiento farmacológico , Enfermedad de Crohn/sangre , Enfermedad de Crohn/tratamiento farmacológico , Interacciones Farmacológicas , Fármacos Gastrointestinales/sangre , Fármacos Gastrointestinales/inmunología , Fármacos Gastrointestinales/farmacocinética , Fármacos Gastrointestinales/farmacología , Humanos , Inducción de Remisión
7.
J Crohns Colitis ; 10(12): 1437-1444, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27252400

RESUMEN

Vedolizumab is a novel therapeutic monoclonal antibody recently approved for the treatment of moderately to severely active ulcerative colitis and Crohn's disease in adults who have failed at least one conventional therapy. An integrin antagonist, vedolizumab binds to the α4ß7 integrin which is expressed specifically by a subset of gastrointestinal-homing T lymphocytes. The binding of α4ß7 integrin to mucosal addressin cell adhesion molecule-1 expressed on the surface of mucosal endothelial cells is a crucial component of the gut-selective homing mechanism for lymphocytes.In contrast, other monoclonal antibodies approved for the treatment of inflammatory bowel diseases, such as tumour necrosis factor α antagonists and the integrin antagonist natalizumab, act systemically or on multiple targets to reduce inflammation.The unique gut selectivity of vedolizumab may contribute to the favourable benefit-risk profile observed in vedolizumab clinical trials. In this review, we summarise data from the preclinical development of vedolizumab and describe the current understanding of the mechanism of action as it relates to other biological therapies for inflammatory bowel disease.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Colitis Ulcerosa/tratamiento farmacológico , Enfermedad de Crohn/tratamiento farmacológico , Factores Inmunológicos/farmacología , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Humanos , Factores Inmunológicos/uso terapéutico , Mucosa Intestinal/efectos de los fármacos , Leucocitos/efectos de los fármacos
8.
J Immunol ; 190(5): 1961-73, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23365083

RESUMEN

The immune system is characterized by the preferential migration of lymphocytes through specific tissues (i.e., tissue tropism). Tissue tropism is mediated, in part, by the α(4) integrins expressed by T lymphocytes. The α(4)ß(1) integrin mediates migration of memory T lymphocytes into the CNS, whereas the α(4)ß(7) integrin mediates migration preferentially into gastrointestinal tissue. This paradigm was established primarily from investigations in rodents; thus, the objective of this investigation was to determine if blocking the α(4)ß(7) integrin exclusively would affect migration of T lymphocytes into the CNS of primates. The effects of the dual α(4)ß(1) and α(4)ß(7) antagonist natalizumab were compared with those of the α(4)ß(7) antagonist vedolizumab on experimental autoimmune encephalomyelitis in the rhesus monkey. Animals received an initial i.v. bolus of placebo, natalizumab (30 mg/kg), or vedolizumab (30 mg/kg) before intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein and then Ab once weekly thereafter. Natalizumab prevented CNS inflammation and demyelination significantly (p < 0.05), compared with time-matched placebo control animals, whereas vedolizumab did not inhibit these effects, despite saturating the α(4)ß(7) integrin in each animal for the duration of the investigation. These results demonstrate that blocking α(4)ß(7) exclusively does not inhibit immune surveillance of the CNS in primates.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Integrina alfa4beta1/antagonistas & inhibidores , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Humanos , Vigilancia Inmunológica/efectos de los fármacos , Inyecciones Intravenosas , Integrina alfa4beta1/inmunología , Macaca mulatta , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Natalizumab , Especificidad de Órganos , Placebos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología
9.
MAbs ; 5(6): 842-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24492340

RESUMEN

Vedolizumab (VDZ) is a humanized monoclonal antibody in development for the treatment of inflammatory bowel disease. VDZ binds to the α4ß7 integrin complex and inhibits its binding to mucosal addressin cell adhesion molecule-1 (MAdCAM-1), thus preventing lymphocyte extravasation to gut mucosal tissues. To understand whether VDZ has additional effects that may affect its overall safety as a therapeutic molecule, we examined other potential actions of VDZ. In vitro assays with human peripheral blood lymphocytes demonstrated that VDZ fails to elicit cytotoxicity, lymphocyte activation, and cytokine production from memory T lymphocytes and does not interfere with the suppressive ability of regulatory T cells. Furthermore, we demonstrated that VDZ induces internalization of α4ß7 and that the integrin is rapidly re-expressed and fully functional after VDZ withdrawal. These studies provide insight into the mechanisms underlying the observed safety profile of VDZ in clinical trials.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Anticuerpos Monoclonales Humanizados/metabolismo , Citometría de Flujo , Humanos , Técnicas In Vitro , Integrinas/metabolismo , Ratones , Unión Proteica
10.
Inflamm Bowel Dis ; 18(11): 2107-19, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22419649

RESUMEN

BACKGROUND: Biological therapies that antagonize specific molecules have demonstrated efficacy in inflammatory bowel diseases, but infections resulting from systemic immunosuppression underscore the need for safer therapies. The objective of this investigation was to determine if antagonism of the α(4) ß(7) integrin would exclusively yield gut-selective antiinflammatory activity in primates. METHODS: A series of experiments were conducted to investigate potential intra- and extraintestinal effects in healthy nonhuman primates dosed repeatedly with the α(4) ß(7) -exclusive antagonist vedolizumab (former versions: MLN0002, MLN02, LDP-02) for 4, 13, and 26 weeks. RESULTS: No adverse clinical effects of vedolizumab were observed in healthy cynomolgus monkeys up to the highest doses tested (100 mg/kg). Histomorphologic analyses indicated a reduction in the frequency of leukocytes in gastrointestinal tissue, but not other organs. A significant (P < 0.05) decrease in the frequency of ß 7+ lymphocytes in gastrointestinal tissues corresponded to a significant (P < 0.05) increase in α(4) ß 7+ memory helper T lymphocytes in peripheral blood. This elevation was specific to α(4) ß 7+ memory helper T lymphocytes; levels of other leukocyte subsets remained unaffected. Systemic opportunistic infections were not observed, and vedolizumab did not inhibit adaptive or innate immune responses systemically. CONCLUSIONS: These data demonstrate that blocking the α(4) ß(7) integrin exclusively yields gut-selective antiinflammatory activity in primates.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Tracto Gastrointestinal/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Integrinas/antagonistas & inhibidores , Mucosa Intestinal/efectos de los fármacos , Leucocitos/efectos de los fármacos , Animales , Femenino , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Macaca fascicularis , Masculino , Natalizumab , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
11.
J Pharmacol Exp Ther ; 330(3): 864-75, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19509315

RESUMEN

Vedolizumab is a humanized monoclonal antibody that targets the alpha(4)beta(7) integrin exclusively, and modulates inflammation in the gastrointestinal tract without inducing the systemic immunosuppression that characterizes anti-alpha(4) chain monoclonal antibodies, such as natalizumab. This unique pharmacologic profile is largely attributable to four determinants. The first determinant is the restriction of the expression of the alpha(4)beta(7) integrin to subsets of leukocytes. Vedolizumab does not bind to the majority of memory CD4(+) T lymphocytes (60%), neutrophils, and most monocytes. The highest level of vedolizumab binding is to a subset (approximately 25%) of human peripheral blood memory CD4(+) T lymphocytes that include gut-homing interleukin 17 T-helper lymphocytes. Vedolizumab also binds to eosinophils at high levels, and to naive T-helper lymphocytes, naive and memory cytotoxic T lymphocytes, B lymphocytes, natural killer cells, and basophils at lower levels; vedolizumab binds to memory CD4(+) T and B lymphocytes with subnanomolar potency (EC(50) = 0.3-0.4 nM). The second determinant is binding specificity; vedolizumab binds exclusively to the alpha(4)beta(7) integrin, and not to the alpha(4)beta(1) and alpha(E)beta(7) integrins. The third determinant is selective antagonism; vedolizumab selectively inhibits adhesion of alpha(4)beta(7)-expressing cells to mucosal addressin cell adhesion molecule 1 (median inhibition concentration [IC(50)] = 0.02-0.06 microg/ml) and fibronectin (IC(50) = 0.02 microg/ml), but not vascular cell adhesion molecule 1. The fourth determinant is the gastrointestinal-specific tropism of the alpha(4)beta(7) integrin function. These pharmacologic properties of vedolizumab, in conjunction with the gastrointestinal tropism of alpha(4)beta(7) integrin function, may ultimately confer an improved risk-to-benefit profile for patients with inflammatory bowel diseases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Integrinas/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Basófilos/efectos de los fármacos , Basófilos/metabolismo , Unión Competitiva/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular , Fibronectinas/metabolismo , Humanos , Inmunohistoquímica , Integrina alfa4beta1/efectos de los fármacos , Integrina alfa4beta1/metabolismo , Integrinas/metabolismo , Interleucina-17/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Unión Proteica , Linfocitos T Colaboradores-Inductores/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Neurotoxicology ; 29(2): 232-43, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18155769

RESUMEN

GT-1 murine neuronal cells exposed to an experimental proteasome inhibitor (EPI) for 24h showed increased cell death via a non-apoptotic mechanism, as assessed by TUNEL and DNA fragmentation assays. Immunofluorescence staining demonstrated that EPI induced reorganization and relocation of non-ubiquinated actin microfilaments and microtubules to the perinuclear region in EPI treated cells. Immunohistochemistry analysis also demonstrated that other non-cytoskeletal proteins became ubiquitinated and/or upregulated including ubiquitin and other stress proteins. Perinuclear-centrosomal accumulation of gamma-tubulin and vimentin, key components of aggresomes, was observed in the EPI treated cells. Biochemical analysis indicated that EPI-induced accumulation of ubiquitinated protein aggregates in GT-1 cells was detergent - and mechanical - disruption resistant, a feature of aggresomes. Similar results were observed in GT-1 cells treated with lactacystin, a prototypical proteasome inhibitor, which is structurally dissimilar to EPI indicating a pharmacologic effect. In conclusion, EPI causes cytoskeletal reorganization and accumulation of diverse ubiquitinated and non-ubiquitinated proteins in the perinuclear region and potentially overloads the endoplasmic reticulum-dependent quality control mechanism. These processes acting alone, or in combination, are hypothesized to affect axonal transport or other aspects of cellular homeostasis and thus, represent events potentially relevant to the development of peripheral neuropathy associated with administration of proteasome inhibitors in nonclinical studies.


Asunto(s)
Acetilcisteína/análogos & derivados , Inhibidores de Cisteína Proteinasa/toxicidad , Citoesqueleto/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Inhibidores de Proteasoma , Acetilcisteína/toxicidad , Actinas/metabolismo , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citoesqueleto/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Proteínas de Choque Térmico/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Microscopía Fluorescente , Neuronas/enzimología , Neuronas/metabolismo , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Ubiquitina/metabolismo
13.
J Immunol ; 177(10): 6940-51, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17082609

RESUMEN

CD4+ Th2 cells are important regulators of allergic inflammation. CCR8 is thought to play a role in Th2-mediated responses, however, expression of CCR8 in peripheral blood has not been fully characterized. Using a fluorescent form of the ligand selective for CCR8 (F-CCL1), we identified the leukocytes expressing CCR8 in human, monkey, and mouse peripheral blood. CCR8 expression is primarily restricted to a subset of human CD4 memory T lymphocytes (15%). Approximately 40% of CCR8+CD4+ T cells express Th2 cytokines IL-4 or IL-13 while 13% express the Th1 cytokine IFN-gamma. In fact, 50% of all Th2, but only 5% of Th1, cells express CCR8. Upon anti-CD3/anti-CD28 mAb-mediated activation, CCR8+CD4+ T cells secrete 3- to 7-fold higher levels of IL-4, IL-5, IL-9, and IL-13 and 10- to 20-fold lower levels of IFN-gamma or IL-17, compared with CCR8-CD4+ memory T cells. Two-thirds of CCR8+CD4 T cells express cutaneous lymphocyte-associated Ag while the majority lack gut-homing receptors. CCR8+CD4+ cells express CCR7 and CD62L and are present in spleen and lymph nodes of mice. Approximately 25% of CCR8+CD4 T cells express CD25high while 20% of CCR8+CD4+ express the T regulatory cell transcription factor FOXP3 accounting for 60% of all FOXP3-expressing CD4+ T cells. In conclusion, CCR8 marks a diverse subset of CD4 memory T cells enriched for T regulatory and Th2 cells which have the potential for recruitment into sites of allergic inflammation where they could participate in the induction and regulation of the allergic response.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Memoria Inmunológica , Receptores de Quimiocina/biosíntesis , Linfocitos T Reguladores/metabolismo , Células Th2/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Quimiocina CCL1 , Quimiocinas CC/fisiología , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Humanos , Memoria Inmunológica/genética , Inmunofenotipificación , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR8 , Receptores de Quimiocina/sangre , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/fisiología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células Th2/citología , Células Th2/metabolismo
14.
Genomics ; 88(2): 173-84, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16713170

RESUMEN

Imbalanced protease activity has long been recognized in the progression of disease states such as cancer and inflammation. Serpins, the largest family of endogenous protease inhibitors, target a wide variety of serine and cysteine proteases and play a role in a number of physiological and pathological states. The expression profiles of 20 serpins and 105 serine and cysteine proteases were determined across a panel of normal and diseased human tissues. In general, expression of serpins was highly restricted in both normal and diseased tissues, suggesting defined physiological roles for these protease inhibitors. A high correlation in expression for a particular serpin-protease pair in healthy tissues was often predictive of a biological interaction. The most striking finding was the dramatic change observed in the regulation of expression between proteases and their cognate inhibitors in diseased tissues. The loss of regulated serpin-protease matched expression may underlie the imbalanced protease activity observed in pathological states.


Asunto(s)
Cisteína Endopeptidasas/genética , Perfilación de la Expresión Génica/métodos , Regulación Enzimológica de la Expresión Génica , Reacción en Cadena de la Polimerasa/métodos , Serina Endopeptidasas/genética , Serpinas/genética , Secuencia de Aminoácidos , Línea Celular , Línea Celular Transformada , Cisteína Endopeptidasas/metabolismo , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/metabolismo , Serina Endopeptidasas/metabolismo , Serpinas/metabolismo , Especificidad de la Especie
15.
Prostaglandins Other Lipid Mediat ; 76(1-4): 133-47, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15967168

RESUMEN

Chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2), a G protein-coupled receptor activated by prostaglandin D(2) (PGD(2)), has been identified as a receptor expressed on cell types critical to the pathogenesis of asthma. The cDNA encoding guinea pig CRTH2 was cloned and mRNA expression examined in selected tissues. Transcript profiling of guinea pig CRTH2 indicated relatively high levels of expression in bone marrow, intermediate levels in brain and relatively low levels in lung, spleen, thymus, lymph node, etc. Characterization of the molecular pharmacology of guinea pig CRTH2 revealed that guinea pig CRTH2 exhibited a greater affinity for Delta(12)-PGJ(2), a stable PGD(2) metabolite relative to human CRTH2. The CRTH2 selective agonists 13,14-dihydro-15-keto PGD(2) and Delta(12)-PGJ(2) induced the recruitment of eosinophils following intradermal administration of these ligands in guinea pigs. Chemotaxis of guinea pig eosinophils was elicited by either PGD(2) or Delta(12)-PGJ(2), and was abolished by a CRTH2-specific antagonist. These results indicate that PGD(2) and the stable metabolite, Delta(12)-PGJ(2), play important roles in CRTH2 activation in the guinea pig.


Asunto(s)
Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Animales , Secuencia de Bases , Calcio/metabolismo , Línea Celular , Quimiotaxis de Leucocito , Clonación Molecular , Cartilla de ADN , ADN Complementario , Eosinófilos/citología , Cobayas , Humanos , Pulmón/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética , Bazo/metabolismo , Timo/metabolismo
16.
J Immunol ; 171(3): 1542-55, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12874248

RESUMEN

Chemokine-induced T lymphocyte recruitment to the lung is critical for allergic inflammation, but chemokine signaling pathways are incompletely understood. Regulator of G protein signaling (RGS)16, a GTPase accelerator (GTPase-activating protein) for Galpha subunits, attenuates signaling by chemokine receptors in T lymphocytes, suggesting a role in the regulation of lymphocyte trafficking. To explore the role of RGS16 in T lymphocyte-dependent immune responses in a whole-organism model, we generated transgenic (Tg) mice expressing RGS16 in CD4(+) and CD8(+) cells. rgs16 Tg T lymphocytes migrated to CC chemokine ligand 21 or CC chemokine ligand 12 injection sites in the peritoneum, but not to CXC chemokine ligand 12. In a Th2-dependent model of allergic pulmonary inflammation, CD4(+) lymphocytes bearing CCR3, CCR5, and CXCR4 trafficked in reduced numbers to the lung after acute inhalation challenge with allergen (OVA). In contrast, spleens of sensitized and challenged Tg mice contained increased numbers of CD4(+)CCR3(+) cells producing more Th2-type cytokines (IL-4, IL-5, and IL-13), which were associated with increased airway hyperreactivity. Migration of Tg lymphocytes to the lung parenchyma after adoptive transfer was significantly reduced compared with wild-type lymphocytes. Naive lymphocytes displayed normal CCR3 and CXCR4 expression and cytokine responses, and compartmentation in secondary lymphoid organs was normal without allergen challenge. These results suggest that RGS16 may regulate T lymphocyte activation in response to inflammatory stimuli and migration induced by CXCR4, CCR3, and CCR5, but not CCR2 or CCR7.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Inflamación/inmunología , Pulmón/patología , Activación de Linfocitos/inmunología , Proteínas/fisiología , Proteínas RGS/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Enfermedad Aguda , Traslado Adoptivo , Alérgenos/administración & dosificación , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Quimiotaxis de Leucocito/genética , Cruzamientos Genéticos , Citocinas/biosíntesis , Citocinas/metabolismo , Femenino , Homeostasis/genética , Homeostasis/inmunología , Humanos , Inmunización , Inflamación/genética , Inflamación/patología , Pulmón/inmunología , Activación de Linfocitos/genética , Tejido Linfoide/crecimiento & desarrollo , Tejido Linfoide/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Biosíntesis de Proteínas , Proteínas/genética , Proteínas RGS/biosíntesis , Proteínas RGS/genética , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/fisiología , Transducción de Señal/genética , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...