Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38695469

RESUMEN

Interleukin receptor associated kinase 4 (IRAK4) plays an important role in innate immune signaling through Toll-like and interleukin-1 receptors and represents an attractive target for the treatment of inflammatory diseases and cancer. We previously reported the development of a potent, selective, and brain-penetrant imidazopyrimidine series of IRAK4 inhibitors. However, lead molecule BIO-7488 (1) suffered from low solubility which led to variable PK, compound accumulation, and poor in vivo tolerability. Herein, we describe the discovery of a series of pyridone analogs with improved solubility which are highly potent, selective and demonstrate desirable PK profiles including good oral bioavailability and excellent brain penetration. BIO-8169 (2) reduced the in vivo production of pro-inflammatory cytokines, was well tolerated in safety studies in rodents and dog at margins well above the predicted efficacious exposure and showed promising results in a mouse model for multiple sclerosis.

2.
J Med Chem ; 67(6): 4676-4690, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38467640

RESUMEN

Interleukin receptor-associated kinase 4 (IRAK4) is a key node of signaling within the innate immune system that regulates the production of inflammatory cytokines and chemokines. The presence of damage-associated molecular patterns (DAMPs) after tissue damage such as stroke or traumatic brain injury (TBI) initiates signaling through the IRAK4 pathway that can lead to a feed-forward inflammatory loop that can ultimately hinder patient recovery. Herein, we describe the first potent, selective, and CNS-penetrant IRAK4 inhibitors for the treatment of neuroinflammation. Lead compounds from the series were evaluated in CNS PK/PD models of inflammation, as well as a mouse model of ischemic stroke. The SAR optimization detailed within culminates in the discovery of BIO-7488, a highly selective and potent IRAK4 inhibitor that is CNS penetrant and has excellent ADME properties.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1 , Accidente Cerebrovascular Isquémico , Ratones , Animales , Humanos , Transducción de Señal , Citocinas , Pirimidinas/farmacología , Pirimidinas/uso terapéutico
3.
Blood Adv ; 2(21): 2904-2916, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30396910

RESUMEN

The main complication of replacement therapy with factor in hemophilia A (HemA) is the formation of inhibitors (neutralizing anti-factor VIII [FVIII] antibodies) in ∼30% of severe HemA patients. Because these inhibitors render replacement FVIII treatment essentially ineffective, preventing or eliminating them is of top priority in disease management. The extended half-life recombinant FVIII Fc fusion protein (rFVIIIFc) is an approved therapy for HemA patients. In addition, it has been reported that rFVIIIFc may induce tolerance to FVIII more readily than FVIII alone in HemA patients that have developed inhibitors. Given that the immunoglobulin G1 Fc region has the potential to interact with immune cells expressing Fc receptors (FcRs) and thereby affect the immune response to rFVIII, we investigated how human macrophages, expressing both FcRs and receptors reported to bind FVIII, respond to rFVIIIFc. We show herein that rFVIIIFc, but not rFVIII, uniquely skews macrophages toward an alternatively activated regulatory phenotype. rFVIIIFc initiates signaling events that result in morphological changes, as well as a specific gene expression and metabolic profile that is characteristic of the regulatory type Mox/M2-like macrophages. Further, these changes are dependent on rFVIIIFc-FcR interactions. Our findings elucidate mechanisms of potential immunomodulatory properties of rFVIIIFc.


Asunto(s)
Factor VIII/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Células Cultivadas , Factor VIII/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Hemofilia A/tratamiento farmacológico , Hemofilia A/patología , Humanos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Leucocitos Mononucleares/citología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/efectos de los fármacos
4.
Sci Signal ; 11(541)2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30065029

RESUMEN

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/metabolismo , Regulación Neoplásica de la Expresión Génica , FN-kappa B/metabolismo , Osteosarcoma/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Ciclinas/metabolismo , Perfilación de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Indoles/farmacología , Receptor beta de Linfotoxina/antagonistas & inhibidores , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Propionatos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoma , Transducción de Señal , Receptor de TWEAK/antagonistas & inhibidores , Receptor de TWEAK/genética , Receptor de TWEAK/metabolismo , Células Tumorales Cultivadas , Quinasa de Factor Nuclear kappa B
5.
Transfusion ; 54(1): 179-89, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23621760

RESUMEN

BACKGROUND: Red blood cell (RBC) alloantibodies to nonself antigens may develop after transfusion or pregnancy, leading to morbidity and mortality in the form of hemolytic transfusion reactions or hemolytic disease of the newborn. A better understanding of the mechanisms of RBC alloantibody induction, or strategies to mitigate the consequences of such antibodies, may ultimately improve transfusion safety. However, such studies are inherently difficult in humans. STUDY DESIGN AND METHODS: We recently generated transgenic mice with RBC-specific expression of the human KEL glycoprotein, specifically the KEL2 or KEL1 antigens. Herein, we investigate recipient alloimmune responses to transfused RBCs in this system. RESULTS: Transfusion of RBCs from KEL2 donors into wild-type recipients (lacking the human KEL protein but expressing the murine KEL ortholog) resulted in dose-dependent anti-KEL glycoprotein immunoglobulin (Ig)M and IgG antibody responses, enhanced by recipient inflammation with poly(I:C). Boostable responses were evident upon repeat transfusion, with morbid-appearing alloimmunized recipients experiencing rapid clearance of transfused KEL2 but not control RBCs. Although KEL1 RBCs were also immunogenic after transfusion into wild-type recipients, transfusion of KEL1 RBCs into KEL2 recipients or vice versa failed to lead to detectable anti-KEL1 or anti-KEL2 responses. CONCLUSIONS: This murine model, with reproducible and clinically significant KEL glycoprotein alloantibody responses, provides a platform for future mechanistic studies of RBC alloantibody induction and consequences. Long-term translational goals of these studies include improving transfusion safety for at-risk patients.


Asunto(s)
Transfusión de Eritrocitos/métodos , Eritrocitos/inmunología , Isoanticuerpos/biosíntesis , Sistema del Grupo Sanguíneo de Kell/inmunología , Anemia Hemolítica/genética , Anemia Hemolítica/inmunología , Animales , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/genética , Incompatibilidad de Grupos Sanguíneos/genética , Incompatibilidad de Grupos Sanguíneos/inmunología , Eritrocitos/metabolismo , Humanos , Inflamación/inmunología , Sistema del Grupo Sanguíneo de Kell/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Poli I-C
6.
Blood ; 122(8): 1494-504, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23801629

RESUMEN

Exposure to nonself red blood cell (RBC) antigens, either from transfusion or pregnancy, may result in alloimmunization and incompatible RBC clearance. First described as a pregnancy complication 80 years ago, hemolytic disease of the fetus and newborn (HDFN) is caused by alloimmunization to paternally derived RBC antigens. Despite the morbidity/mortality of HDFN, women at risk for RBC alloimmunization have few therapeutic options. Given that alloantibodies to antigens in the KEL family are among the most clinically significant, we developed a murine model with RBC-specific expression of the human KEL antigen to evaluate the impact of maternal/fetal KEL incompatibility. After exposure to fetal KEL RBCs during successive pregnancies with KEL-positive males, 21 of 21 wild-type female mice developed anti-KEL alloantibodies; intrauterine fetal anemia and/or demise occurred in a subset of KEL-positive pups born to wild type, but not agammaglobulinemic mothers. Similar to previous observations in humans, pregnancy-associated alloantibodies were detrimental in a transfusion setting, and transfusion-associated alloantibodies were detrimental in a pregnancy setting. This is the first pregnancy-associated HDFN model described to date, which will serve as a platform to develop targeted therapies to prevent and/or mitigate the dangers of RBC alloantibodies to fetuses and newborns.


Asunto(s)
Anemia Hemolítica/inmunología , Eritrocitos/citología , Isoanticuerpos/inmunología , Sistema del Grupo Sanguíneo de Kell/inmunología , Modelos Animales , Anemia Hemolítica/genética , Animales , Transfusión Sanguínea , Citocinas/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Inmunoglobulina G/inmunología , Sistema del Grupo Sanguíneo de Kell/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Preñez
7.
Transfusion ; 52(12): 2620-30, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22494154

RESUMEN

BACKGROUND: KEL1, also known as "K", is one of the most immunogenic red blood cell (RBC) antigens. KEL2, also known as "k," differs from KEL1 by a single amino acid. Anti-Kell system antibodies can lead to significant adverse clinical outcomes in humans, including hemolytic complications in alloimmunized transfusion recipients or in infants of alloimmunized mothers. To provide a platform for in-depth immunologic studies of alloimmunization and subsequent sequelae, we generated transgenic mice expressing the human KEL1 or KEL2 antigens. STUDY DESIGN AND METHODS: Vectors were created in which cDNAs encoding either KEL1 or KEL2 were regulated by an erythroid specific ß-globin promoter and enhancer. Pronuclear microinjections were carried out into a C57BL6 background, and founder pups were identified by polymerase chain reaction and screened for expression by flow cytometry. RBC life span and antigen stability were assessed by dye labeling RBCs, transfusing into agammaglobulinemic (µMT) recipients, and tracking by flow cytometry. RESULTS: The expression of either KEL1 or KEL2 is RBC specific and first occurs on early RBC precursors. Both KEL1 and KEL2 RBCs have a normal circulatory life span and stable antigen expression. Expression of KEL1 or KEL2 does not result in altered levels of murine Kell, and resulting RBCs have normal hematologic variables. CONCLUSION: The KEL1 and KEL2 mice represent the first murine system of RBC immunity with antithetical antigens, allowing a more precise modeling of human RBC immunology in general and also a platform for development of novel therapeutics to prevent or minimize the dangers of RBC alloimmunization to the KEL1 and KEL2 antigens in particular.


Asunto(s)
Eritrocitos/inmunología , Sistema del Grupo Sanguíneo de Kell/genética , Sistema del Grupo Sanguíneo de Kell/inmunología , Ratones Transgénicos , Modelos Animales , Animales , Transfusión Sanguínea , Epítopos/genética , Epítopos/inmunología , Eritrocitos/citología , Biblioteca de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...