Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cell ; 168(5): 856-866.e12, 2017 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-28215707

RESUMEN

HSP90 acts as a protein-folding buffer that shapes the manifestations of genetic variation in model organisms. Whether HSP90 influences the consequences of mutations in humans, potentially modifying the clinical course of genetic diseases, remains unknown. By mining data for >1,500 disease-causing mutants, we found a strong correlation between reduced phenotypic severity and a dominant (HSP90 ≥ HSP70) increase in mutant engagement by HSP90. Examining the cancer predisposition syndrome Fanconi anemia in depth revealed that mutant FANCA proteins engaged predominantly by HSP70 had severely compromised function. In contrast, the function of less severe mutants was preserved by a dominant increase in HSP90 binding. Reducing HSP90's buffering capacity with inhibitors or febrile temperatures destabilized HSP90-buffered mutants, exacerbating FA-related chemosensitivities. Strikingly, a compensatory FANCA somatic mutation from an "experiment of nature" in monozygotic twins both prevented anemia and reduced HSP90 binding. These findings provide one plausible mechanism for the variable expressivity and environmental sensitivity of genetic diseases.


Asunto(s)
Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteínas HSP90 de Choque Térmico/genética , Pliegue de Proteína , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/química , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Mutación Missense , Dominios y Motivos de Interacción de Proteínas , Estrés Fisiológico , Gemelos Monocigóticos
2.
J Immunol ; 207(1): 34-43, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34108258

RESUMEN

Systemic lupus erythematosus (SLE) is associated with an IL-2-deficient state, with regulatory T cells (Tregs) showing diminished immune regulatory capacity. A low dose of IL-2 has shown encouraging clinical benefits in SLE patients; however, its clinical utility is limited because of the requirement of daily injections and the observation of increase in proinflammatory cytokines and in non-Tregs. We recently showed that a fusion protein of mouse IL-2 and mouse IL-2Rα (CD25), joined by a noncleavable linker, was effective in treating diabetes in NOD mice by selectively inducing Treg expansion. In this report, we show that mouse IL-2 (mIL-2)/CD25 at doses up to 0.5 mg/kg twice a week induced a robust Treg expansion without showing signs of increase in the numbers of NK, CD4+Foxp3-, or CD8+ T cells or significant increase in proinflammatory cytokines. In both NZB × NZW and MRL/lpr mice, mIL-2/CD25 at 0.2-0.4 mg/kg twice a week demonstrated efficacy in inducing Treg expansion, CD25 upregulation, and inhibiting lupus nephritis based on the levels of proteinuria, autoantibody titers, and kidney histology scores. mIL-2/CD25 was effective even when treatment was initiated at the time when NZB × NZW mice already showed signs of advanced disease. Furthermore, we show coadministration of prednisolone, which SLE patients commonly take, did not interfere with the ability of mIL-2/CD25 to expand Tregs. The prednisolone and mIL-2/CD25 combination treatment results in improvements in most of the efficacy readouts relative to either monotherapy alone. Taken together, our results support further evaluation of IL-2/CD25 in the clinic for treating immune-mediated diseases such as SLE.


Asunto(s)
Lupus Eritematoso Sistémico , Linfocitos T Reguladores , Animales , Linfocitos T CD8-positivos , Factores de Transcripción Forkhead , Humanos , Interleucina-2 , Subunidad alfa del Receptor de Interleucina-2 , Lupus Eritematoso Sistémico/tratamiento farmacológico , Ratones , Ratones Endogámicos MRL lpr , Ratones Endogámicos NOD
3.
Bioorg Med Chem Lett ; 35: 127778, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33422603

RESUMEN

The discovery of a series of substituted diarylether compounds as retinoic acid related orphan receptor γt (RORγt) agonists is described. Compound 1 was identified from deck mining as a RORγt agonist. Hit-to-lead optimization led to the identification of lead compound 5, which possesses improved potency (10x). Extensive SAR exploration led to the identification of a potent and selective compound 22, that demonstrated an improved pharmacokinetic profile and a dose-dependent pharmacodynamic response. However, when dosed in a MC38 syngeneic tumor model, no evidence of efficacy was observed. ©2020 Elsevier Science Ltd. All rights reserved.


Asunto(s)
Éteres/farmacología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Tretinoina/farmacología , Animales , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Éteres/síntesis química , Éteres/química , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Células Th17 , Tretinoina/síntesis química , Tretinoina/química
4.
Bioorg Med Chem Lett ; 30(23): 127521, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32882417

RESUMEN

In order to rapidly develop C6 and C8 SAR of our reported tricyclic sulfone series of RORγt inverse agonists, a late-stage bromination was employed. Although not regioselective, the bromination protocol allowed us to explore new substitution patterns/vectors that otherwise would have to be incorporated at the very beginning of the synthesis. Based on the SAR obtained from this exercise, compound 15 bearing a C8 fluorine was developed as a very potent and selective RORγt inverse agonist. This analog's in vitro profile, pharmacokinetic (PK) data and efficacy in an IL-23 induced mouse acanthosis model will be discussed.


Asunto(s)
Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Melanosis/tratamiento farmacológico , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Sulfonas/uso terapéutico , Animales , Cristalografía por Rayos X , Agonismo Inverso de Drogas , Femenino , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Compuestos Heterocíclicos con 3 Anillos/farmacocinética , Interleucina-18 , Masculino , Melanosis/inducido químicamente , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Estructura Molecular , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Unión Proteica , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/farmacocinética
5.
EMBO J ; 33(15): 1698-712, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24966277

RESUMEN

Several proteins in the BRCA-Fanconi anemia (FA) pathway, such as FANCJ, BRCA1, and FANCD2, interact with mismatch repair (MMR) pathway factors, but the significance of this link remains unknown. Unlike the BRCA-FA pathway, the MMR pathway is not essential for cells to survive toxic DNA interstrand crosslinks (ICLs), although MMR proteins bind ICLs and other DNA structures that form at stalled replication forks. We hypothesized that MMR proteins corrupt ICL repair in cells that lack crosstalk between BRCA-FA and MMR pathways. Here, we show that ICL sensitivity of cells lacking the interaction between FANCJ and the MMR protein MLH1 is suppressed by depletion of the upstream mismatch recognition factor MSH2. MSH2 depletion suppresses an aberrant DNA damage response, restores cell cycle progression, and promotes ICL resistance through a Rad18-dependent mechanism. MSH2 depletion also suppresses ICL sensitivity in cells deficient for BRCA1 or FANCD2, but not FANCA. Rescue by Msh2 loss was confirmed in Fancd2-null primary mouse cells. Thus, we propose that regulation of MSH2-dependent DNA damage response underlies the importance of interactions between BRCA-FA and MMR pathways.


Asunto(s)
Proteína BRCA1/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Daño del ADN , Reparación de la Incompatibilidad de ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteína BRCA1/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Línea Celular/efectos de los fármacos , Aberraciones Cromosómicas , Daño del ADN/efectos de los fármacos , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Humanos , Ratones , Ratones Mutantes , Mitomicina/farmacología , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas
6.
Bioorg Med Chem Lett ; 28(2): 85-93, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29233651

RESUMEN

We disclose the optimization of a high throughput screening hit to yield benzothiazine and tetrahydroquinoline sulfonamides as potent RORγt inverse agonists. However, a majority of these compounds showed potent activity against pregnane X receptor (PXR) and modest activity against liver X receptor α (LXRα). Structure-based drug design (SBDD) led to the identification of benzothiazine and tetrahydroquinoline sulfonamide analogs which completely dialed out LXRα activity and were less potent at PXR. Pharmacodynamic (PD) data for compound 35 in an IL-23 induced IL-17 mouse model is discussed along with the implications of a high Ymax in the PXR assay for long term preclinical pharmacokinetic (PK) studies.


Asunto(s)
Compuestos Bicíclicos con Puentes/farmacología , Diseño de Fármacos , Propanoles/farmacología , Receptores de Ácido Retinoico/agonistas , Receptores de Esteroides/agonistas , Sulfonamidas/farmacología , Animales , Compuestos Bicíclicos con Puentes/síntesis química , Compuestos Bicíclicos con Puentes/química , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Receptores X del Hígado/agonistas , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Estructura Molecular , Receptor X de Pregnano , Propanoles/síntesis química , Propanoles/química , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Receptor de Ácido Retinoico gamma
7.
Bioorg Med Chem Lett ; 27(4): 855-861, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28108251

RESUMEN

As demonstrated in preclinical animal models, the disruption of PI3Kδ expression or its activity leads to a decrease in inflammatory and immune responses. Therefore, inhibition of PI3Kδ may provide an alternative treatment for autoimmune diseases, such as RA, SLE, and respiratory ailments. Herein, we disclose the identification of 7-(3-(piperazin-1-yl)phenyl)pyrrolo[2,1-f][1,2,4]triazin-4-amine derivatives as highly potent, selective and orally bioavailable PI3Kδ inhibitors. The lead compound demonstrated efficacy in an in vivo mouse KLH model.


Asunto(s)
Aminas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/química , Aminas/metabolismo , Aminas/uso terapéutico , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Sitios de Unión , Fosfatidilinositol 3-Quinasa Clase I , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Piperazina , Piperazinas/química , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Relación Estructura-Actividad , Triazinas/química
8.
Bioorg Med Chem Lett ; 26(17): 4256-60, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27476421

RESUMEN

Aberrant Class I PI3K signaling is a key factor contributing to many immunological disorders and cancers. We have identified 4-amino pyrrolotriazine as a novel chemotype that selectively inhibits PI3Kδ signaling despite not binding to the specificity pocket of PI3Kδ isoform. Structure activity relationship (SAR) led to the identification of compound 30 that demonstrated efficacy in mouse Keyhole Limpet Hemocyanin (KLH) and collagen induced arthritis (CIA) models.


Asunto(s)
Aminas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/química , Triazinas/química , Aminas/metabolismo , Aminas/uso terapéutico , Animales , Artritis/tratamiento farmacológico , Artritis/metabolismo , Artritis/patología , Sitios de Unión , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Concentración 50 Inhibidora , Ratones , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Estructura Terciaria de Proteína , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 26(10): 2470-2474, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27055941

RESUMEN

The synthesis and structure-activity relationship (SAR) of a series of pyridyl-isoxazole based agonists of S1P1 are discussed. Compound 5b provided potent in vitro activity with selectivity, had an acceptable pharmacokinetic profile, and demonstrated efficacy in a dose dependent manner when administered orally in a rodent model of arthritis.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Lisofosfolípidos/agonistas , Esfingosina/análogos & derivados , Relación Estructura-Actividad , Administración Oral , Animales , Técnicas de Química Sintética , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Humanos , Isoxazoles/química , Isoxazoles/farmacología , Recuento de Linfocitos , Masculino , Ratas Endogámicas Lew , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/agonistas
10.
J Immunol ; 192(9): 4083-92, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24670803

RESUMEN

CD40-CD40L interactions play a critical role in regulating immune responses. Blockade of CD40L by Abs, such as the anti-CD40L Ab 5c8, demonstrated positive clinical effects in patients with autoimmune diseases; however, incidents of thromboembolism (TE) precluded further development of these molecules. In this study, we examined the role of the Fc domain interaction with FcγRs in modulating platelet activation and potential for TE. Our results show that the interaction of the 5c8 wild-type IgG1 Fc domain with FcγRs is responsible for platelet activation, as measured by induction of PAC-1 and CD62P. A version of 5c8 with a mutated IgG1 tail was identified that showed minimal FcγR binding and platelet activation while maintaining full binding to CD40L. To address whether Fc effector function is required for immunosuppression, a potent Ab fragment, termed a "domain Ab" (dAb), against murine CD40L was identified and fused to a murine IgG1 Fc domain containing a D265A mutation that lacks Fc effector function. In vitro, this dAb-Fc demonstrated comparable potency to the benchmark mAb MR-1 in inhibiting B cell and dendritic cell activation. Furthermore, the anti-CD40L dAb-Fc exhibited a notable efficacy comparable to MR-1 in various preclinical models, such as keyhole limpet hemocyanin-induced Ab responses, alloantigen-induced T cell proliferation, "heart-to-ear" transplantation, and NZB × NZW F1 spontaneous lupus. Thus, our data show that immunosuppression and TE can be uncoupled and that a CD40L dAb with an inert Fc tail is expected to be efficacious for treating autoimmune diseases, with reduced risk for TE.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Ligando de CD40/inmunología , Activación Plaquetaria/efectos de los fármacos , Anticuerpos de Dominio Único/farmacología , Animales , Anticuerpos Monoclonales/efectos adversos , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Activación Plaquetaria/inmunología , Receptores de IgG/inmunología , Anticuerpos de Dominio Único/inmunología , Resonancia por Plasmón de Superficie , Tromboembolia/etiología , Tromboembolia/prevención & control , Transfección
11.
J Pharmacol Exp Ther ; 355(3): 506-15, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26442523

RESUMEN

BMS-931699 (lulizumab pegol), a domain antibody (dAb) conjugated with 40-kDa branched polyethylene glycol, is a human anti-CD28 receptor antagonist under development for the treatment of inflammatory and autoimmune diseases. In the present work, the minimal anticipated biologic effect level (MABEL) was determined for BMS-931699 by integrating all the available preclinical data. The relevance of the in vitro mixed lymphocyte reaction (MLR) assay to a whole blood CD28 receptor occupancy (RO) assessment, as well as the relationship between the CD28 RO and the inhibition of T-cell-dependent antibody response to keyhole limpet hemocyanin in vivo, was demonstrated through an integrated pharmacokinetic/pharmacodynamic analysis using anti-hCD28 dAb-001 (differing from BMS-931699 by two additional amino acids at the N-terminus) and a mouse surrogate. Based on this analysis, the EC10 value (0.32 nM) from the human MLR assay and the human plasma volume (0.04 l/kg) were employed to calculate the MABEL (0.01 mg) of BMS-931699 in humans, with a CD28 RO predicted to be ≤10%. The estimated MABEL dose was threefold higher than the value derived from the binding constant and twofold less than the MABEL converted from animal efficacy studies based on the body surface area. Furthermore, it was 2900-fold lower than the human equivalent dose derived from the no observed adverse effect level in monkeys (15 mg/kg/week for 5 doses, intravenous dosing) with a 10-fold safety factor applied. Therefore, the MABEL dose represented a sound approach to mitigate any potential risk in targeting CD28 and was successfully used as the first-in-human starting dose for BMS-931699.


Asunto(s)
Anticuerpos/farmacología , Antígenos CD28/antagonistas & inhibidores , Polietilenglicoles/farmacología , Polietilenglicoles/farmacocinética , Algoritmos , Animales , Superficie Corporal , Relación Dosis-Respuesta a Droga , Femenino , Hemocianinas/antagonistas & inhibidores , Humanos , Prueba de Cultivo Mixto de Linfocitos , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Pruebas de Sensibilidad Microbiana , Monocitos/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 354(2): 152-65, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26015463

RESUMEN

Therapies targeting either interleukin (IL)-23 or IL-17 have shown promise in treating T helper 17 (Th17)-driven autoimmune diseases. Although IL-23 is a critical driver of IL-17, recognition of nonredundant and independent functions of IL-23 and IL-17 has prompted the notion that dual inhibition of both IL-23 and IL-17 could offer even greater efficacy for treating autoimmune diseases relative to targeting either cytokine alone. To test this hypothesis, we generated selective inhibitors of IL-23 and IL-17 and tested the effect of either treatment alone compared with their combination in vitro and in vivo. In vitro, using a novel culture system of murine Th17 cells and NIH/3T3 fibroblasts, we showed that inhibition of both IL-23 and IL-17 completely suppressed IL-23-dependent IL-22 production from Th17 cells and cooperatively blocked IL-17-dependent IL-6 secretion from the NIH/3T3 cells to levels below either inhibitor alone. In vivo, in the imiquimod induced skin inflammation model, and in the myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis model, we demonstrated that dual inhibition of IL-17 and IL-23 was more efficacious in reducing disease than targeting either cytokine alone. Together, these data support the hypothesis that neutralization of both IL-23 and IL-17 may provide enhanced benefit against Th17 mediated autoimmunity and provide a basis for a therapeutic strategy aimed at dual targeting IL-23 and IL-17.


Asunto(s)
Autoinmunidad/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Interleucina-23/antagonistas & inhibidores , Interleucina-23/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Autoinmunidad/efectos de los fármacos , Técnicas de Cocultivo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células 3T3 NIH , Distribución Aleatoria
13.
PLoS Genet ; 8(7): e1002786, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22792074

RESUMEN

BRCA1 promotes DNA repair through interactions with multiple proteins, including CtIP and FANCJ (also known as BRIP1/BACH1). While CtIP facilitates DNA end resection when de-acetylated, the function of FANCJ in repair processing is less well defined. Here, we report that FANCJ is also acetylated. Preventing FANCJ acetylation at lysine 1249 does not interfere with the ability of cells to survive DNA interstrand crosslinks (ICLs). However, resistance is achieved with reduced reliance on recombination. Mechanistically, FANCJ acetylation facilitates DNA end processing required for repair and checkpoint signaling. This conclusion was based on the finding that FANCJ and its acetylation were required for robust RPA foci formation, RPA phosphorylation, and Rad51 foci formation in response to camptothecin (CPT). Furthermore, both preventing and mimicking FANCJ acetylation at lysine 1249 disrupts FANCJ function in checkpoint maintenance. Thus, we propose that the dynamic regulation of FANCJ acetylation is critical for robust DNA damage response, recombination-based processing, and ultimately checkpoint maintenance.


Asunto(s)
Acetilación , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Daño del ADN/genética , ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Lisina/metabolismo , Proteína BRCA1/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , ADN/genética , ADN/metabolismo , Reparación del ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Recombinación Homóloga , Humanos , Mutación
14.
Sci China Life Sci ; 65(6): 1075-1090, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35258743

RESUMEN

T follicular helper (Tfh) cells are critical in providing help for B cells in the germinal center reaction. Tfh cell plasticity, especially with regard to their expression of effector Th cytokines, has been described, but lacks in-depth analysis with genetic approaches. In this study, we systemically compared transcriptomic profiles of Tfh cells derived from various types of immune responses and found gene clusters corresponding to effector Th cells were differentially induced in response to pathogens or immune responses. Of special interest, a subset of Tfh cells producing IFN-γ was generated in an influenza virus infection, partially dependent on the innate cytokine IL-12. Lineage-tracing mouse model revealed unique developmental regulation of IFN-γ+ Tfh cells, while selective ablation of these cells impaired the induction of IgG2c+ germinal center B cells and the control of influenza infection. These results indicate that pathogen-associated Tfh cell plasticity is necessary for host immunity, which has implications in vaccine design.


Asunto(s)
Gripe Humana , Células T Auxiliares Foliculares , Animales , Antivirales , Diferenciación Celular , Citocinas , Centro Germinal , Humanos , Ratones , Linfocitos T Colaboradores-Inductores/fisiología
15.
Oncol Rep ; 47(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34958112

RESUMEN

Hepatocellular carcinoma (HCC) is an immunogenic malignancy, which exhibits low responsiveness to programmed cell death protein­1 (PD­1)/programmed death ligand­1 (PD­L1) antibodies. Therefore, the identification of novel immunotherapeutic targets to treat HCC is imperative. Systematic characterization of the HCC tumor microenvironment (TME) can provide novel insight into additional therapeutic approaches. In the present study, the RNA­sequencing (RNA­seq) data of 360 patients with HCC were integrated from The Cancer Genome Atlas to assess the expression of membrane spanning 4­domains A1 (MS4A1; encoding CD20) in tumors and normal liver tissues. Immunofluorescence and multiplex tissue fluorescence analyses were performed and combined with flow cytometry staining to measure CD20/CD19 expression at the protein level. In addition, published single cell RNA­seq data of CD45+ cells were derived from 16 treatment­naïve patients from Beijing Shijitan Hospital with HCC to illustrate the characteristics of CD19+ B cells. The results indicated that the HCC TME included nuclear receptor subfamily 4 group A member 2+ (NR4A2) B cells. Patients with HCC and high density of intratumoral B cells demonstrated compromised antitumor immunity manifested by low percentages of cytotoxic CD8+ T cells and high density of regulatory T cells. Furthermore, PD­L1 expression was significantly correlated with the B cell signature marker CD20. The present study indicated that tumor­infiltrating B cells may play a negative role in antitumor immunity and serve as a promising target for HCC immunotherapy, alone or in combination with anti­PD­L1/PD­1 antibodies.


Asunto(s)
Linfocitos B/inmunología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/inmunología , Inmunoterapia/métodos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/efectos de los fármacos , China , Femenino , Humanos , Masculino , Persona de Mediana Edad , Microambiente Tumoral/efectos de los fármacos
16.
Clin Pharmacol Drug Dev ; 10(1): 8-21, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33090733

RESUMEN

Sphingosine-1-phosphate (S1P) binding to the S1P-1 receptor (S1P1R) controls the egress of lymphocytes from lymphoid organs and targets modulation of immune responses in autoimmune diseases. Pharmacologic modulation of S1P receptors has been linked to heart rate reduction. BMS-986166, a prodrug of the active phosphorylated metabolite BMS-986166-P, presents an improved cardiac safety profile in preclinical studies compared to other S1P1R modulators. The pharmacokinetics, safety, and pharmacodynamics of BMS-986166 versus placebo after single (0.75-5.0 mg) and repeated (0.25-1.5 mg/day) oral administration were assessed in healthy participants after a 1-day lead-in placebo period. A population model was developed to jointly describe BMS-986166 and BMS-986166-P pharmacokinetics and predict individual exposures. Inhibitory sigmoid models described the relationships between average daily BMS-986166-P concentrations and nadir of time-matched (day -1) placebo-corrected heart rate on day 1 (nDDHR, where DD represents ∆∆) and nadir of absolute lymphocyte count (nALC). Predicted decreases in nDDHR and nALC were 9 bpm and 20% following placebo, with maximum decreases of 10 bpm in nDDHR due to drug effect, and approximately 80% in nALC due to drug and placebo. A 0.5-mg/day dose regimen achieves the target 65% reduction in nALC associated with a 2-bpm decrease in nDDHR over placebo.


Asunto(s)
Modelos Biológicos , Tetrahidronaftalenos/farmacocinética , Adulto , Simulación por Computador , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Voluntarios Sanos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Receptores de Esfingosina-1-Fosfato , Tetrahidronaftalenos/administración & dosificación , Adulto Joven
17.
J Med Chem ; 64(3): 1454-1480, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33492963

RESUMEN

Sphingosine-1-phosphate (S1P) binds to a family of sphingosine-1-phosphate G-protein-coupled receptors (S1P1-5). The interaction of S1P with these S1P receptors has a fundamental role in many physiological processes in the vascular and immune systems. Agonist-induced functional antagonism of S1P1 has been shown to result in lymphopenia. As a result, agonists of this type hold promise as therapeutics for autoimmune disorders. The previously disclosed differentiated S1P1 modulator BMS-986104 (1) exhibited improved preclinical cardiovascular and pulmonary safety profiles as compared to earlier full agonists of S1P1; however, it demonstrated a long pharmacokinetic half-life (T1/2 18 days) in the clinic and limited formation of the desired active phosphate metabolite. Optimization of this series through incorporation of olefins, ethers, thioethers, and glycols into the alkyl side chain afforded an opportunity to reduce the projected human T1/2 and improve the formation of the active phosphate metabolite while maintaining efficacy as well as the improved safety profile. These efforts led to the discovery of 12 and 24, each of which are highly potent, biased agonists of S1P1. These compounds not only exhibited shorter in vivo T1/2 in multiple species but are also projected to have significantly shorter T1/2 values in humans when compared to our first clinical candidate. In models of arthritis, treatment with 12 and 24 demonstrated robust efficacy.


Asunto(s)
Compuestos Bicíclicos con Puentes/síntesis química , Compuestos Bicíclicos con Puentes/farmacología , Proproteína Convertasas/efectos de los fármacos , Serina Endopeptidasas/efectos de los fármacos , Animales , Artritis Experimental/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Biotransformación , Compuestos Bicíclicos con Puentes/efectos adversos , Líquido del Lavado Bronquioalveolar , Quimiotaxis de Leucocito/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/patología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Fosforilación , Ratas , Ratas Endogámicas Lew , Relación Estructura-Actividad
18.
ACS Med Chem Lett ; 12(5): 827-835, 2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34055233

RESUMEN

Structure-activity relationship studies directed toward the replacement of the fused phenyl ring of the lead hexahydrobenzoindole RORγt inverse agonist series represented by 1 with heterocyclic moieties led to the identification of three novel aza analogs 5-7. The hexahydropyrrolo[3,2-f]quinoline series 5 (X = N, Y = Z=CH) showed potency and metabolic stability comparable to series 1 but with improved in vitro membrane permeability and serum free fraction. This structural modification was applied to the hexahydrocyclopentanaphthalene series 3, culminating in the discovery of 8e as a potent and selective RORγt inverse agonist with an excellent in vitro profile, good pharmacokinetic properties, and biologic-like in vivo efficacy in preclinical models of rheumatoid arthritis and psoriasis.

19.
J Med Chem ; 64(5): 2714-2724, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33591748

RESUMEN

SAR efforts directed at identifying RORγt inverse agonists structurally different from our clinical compound 1 (BMS-986251) led to tricyclic-carbocyclic analogues represented by 3-7 and culminated in the identification of 3d (BMS-986313), with structural differences distinct from 1. The X-ray co-crystal structure of 3d with the ligand binding domain of RORγt revealed several key interactions, which are different from 1. The in vitro and in vivo PK profiles of 3d are described. In addition, we demonstrate robust efficacy of 3d in two preclinical models of psoriasis-the IMQ-induced skin lesion model and the IL-23-induced acanthosis model. The efficacy seen with 3d in these models is comparable to the results observed with 1.


Asunto(s)
Amidas/uso terapéutico , Hidrocarburos Cíclicos/uso terapéutico , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Psoriasis/tratamiento farmacológico , Amidas/química , Amidas/farmacocinética , Animales , Agonismo Inverso de Drogas , Femenino , Humanos , Hidrocarburos Cíclicos/química , Hidrocarburos Cíclicos/farmacocinética , Interleucina-23 , Ratones Endogámicos C57BL , Microsomas Hepáticos/metabolismo , Estructura Molecular , Psoriasis/inducido químicamente , Ratas , Relación Estructura-Actividad
20.
Expert Opin Investig Drugs ; 29(4): 411-422, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32306792

RESUMEN

Background: Safety, pharmacokinetics and pharmacodynamics of BMS-986166, a novel sphingosine-1-phosphate receptor 1 modulator, were assessed.Research design and methods: Two double-blind, placebo-controlled, randomized Phase l studies were conducted in healthy participants. In the single ascending dose study (N = 70), BMS-986166 was administered as a single dose, upwardly titrated daily doses or a single dose in participants who were fed, fasted or administered famotidine. In the multiple ascending dose study (N = 32), BMS-986166 was administered daily for 28 days. Safety, pharmacokinetics and pharmacodynamics (absolute lymphocyte count [ALC]) were assessed.Results: BMS-986166 was generally well tolerated; treatment-related adverse events were mild. Dose-related, clinically insignificant reductions in time-matched heart rate were recorded versus placebo. Pharmacokinetics were linear and stationary with approximately dose-related increases in blood exposure of BMS-986166. Decreases in ALC percent change from baseline with multiple doses of BMS-986166 versus placebo were dose-related. Between Day 0 and 35, median nadir lymphocyte reductions were 53.7%, 75.9% and 81.9% with 0.25-, 0.75- and 1.5-mg BMS-986166 doses. ALC recovery began 14, 14-21 and 7 days after last dose of 0.25, 0.75 and 1.5 mg.Conclusions: BMS-986166 was generally well tolerated in this population and warrants further investigation.Trial registration: ClinicalTrials.gov: NCT02790125, NCT03038711.


Asunto(s)
Receptores de Esfingosina-1-Fosfato , Tetrahidronaftalenos/administración & dosificación , Adulto , Grasas de la Dieta/administración & dosificación , Método Doble Ciego , Famotidina/administración & dosificación , Ayuno/metabolismo , Voluntarios Sanos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Recuento de Linfocitos , Persona de Mediana Edad , Tetrahidronaftalenos/efectos adversos , Tetrahidronaftalenos/farmacocinética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA