Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunother ; 43(4): 107-120, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31899702

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment for patients with CD19 B-cell malignancies. Combination strategies that improve CAR T-cell potency, limit tumor environment-mediated immune dysfunction, and directly reduce tumor burden may increase the potential for durable clinical benefit of CAR T-cell therapy. Lisocabtagene maraleucel (liso-cel) is a product therapy candidate being tested in patients with relapsed/refractory non-Hodgkin lymphoma or chronic lymphocytic leukemia. This study assessed the in vitro and in vivo functionality of CAR T cells transduced to express the anti-CD19 CAR of liso-cel in combination with ibrutinib or acalabrutinib. In prolonged stimulation assays, the presence of ibrutinib or acalabrutinib improved the CAR T-cell effector function. RNA-Seq analysis and surface marker profiling of these CAR T cells treated with ibrutinib but not acalabrutinib revealed gene expression changes consistent with skewing toward a memory-like, type 1 T-helper, Bruton tyrosine kinase phenotype. Ibrutinib or acalabrutinib improved CD19 tumor clearance and prolonged survival of tumor-bearing mice when used in combination with CAR T cells. A combination of the defined cell product therapy candidate, liso-cel, with ibrutinib or acalabrutinib is an attractive approach that may potentiate the promising clinical responses already achieved in CD19 B-cell malignancies with each of these single agents.


Assuntos
Antígenos CD19/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia Adotiva , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Adenina/administração & dosagem , Adenina/análogos & derivados , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Benzamidas/administração & dosagem , Biomarcadores , Terapia Combinada , Citocinas/metabolismo , Citotoxicidade Imunológica , Modelos Animais de Doenças , Humanos , Imunoterapia Adotiva/métodos , Ativação Linfocitária/imunologia , Camundongos , Neoplasias/etiologia , Neoplasias/metabolismo , Piperidinas/administração & dosagem , Pirazinas/administração & dosagem , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Eur J Pharm Biopharm ; 142: 435-448, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31306750

RESUMO

Acalabrutinib (Calquence®) 100 mg (bid) has received accelerated approval by FDA for the treatment of adult patients with mantle cell lymphoma (MCL) who have received at least one prior therapy. Acalabrutinib is a substrate of PgP and CYP3A4, with a significant fraction of drug metabolized by first pass gut extraction and 25% absolute bioavailability. The absorption of acalabrutinib is affected by stomach pH, with lower pharmacokinetic exposure observed following co-administration with proton pump inhibitors. During dissolution at pH values below its highest basic pKa, the two basic moieties of acalabrutinib react with protons from the aqueous solution, leading to a higher pH at the drug surface than in the bulk solution. A batch-specific product particle size distribution (P-PSD), was derived from dissolution data using a mechanistic model that was based on the understanding of surface pH and the contribution of micelles to the dissolution rate. P-PSD values obtained for various batches of acalabrutinib products in simple buffers, or in complex fluids such as fruit juices, were successfully integrated into a physiologically based pharmacokinetic (PBPK) model developed using GastroPlus v9.0™. The integrated model allowed the prediction of clinical pharmacokinetics under normal physiological stomach pH conditions as well as following treatment with proton pump inhibitors. The model also accounted for lower pharmacokinetic exposure that was observed when acalabrutinib was co-administered with the acidic beverages, grapefruit juice, (which contains CYP3A inhibitors), and orange drink (which does not contain CYP3A inhibitors), relative to administration with water. The integration of dissolution data in the PBPK model enables mechanistic understanding and the establishment of more robust in vitro-in vivo correlations (IVIVC) under a variety of conditions. The model can then distinguish the interplay between dissolution and first pass extraction and how in vivo stomach pH, saturation of gut PgP, and saturation or inhibition of gut CYP3A4, will impact the pharmacokinetics of acalabrutinib.


Assuntos
Benzamidas/química , Benzamidas/farmacocinética , Interações Medicamentosas/fisiologia , Sucos de Frutas e Vegetais/efeitos adversos , Inibidores da Bomba de Prótons/química , Inibidores da Bomba de Prótons/farmacocinética , Pirazinas/química , Pirazinas/farmacocinética , Solubilidade/efeitos dos fármacos , Disponibilidade Biológica , Química Farmacêutica/métodos , Humanos , Modelos Biológicos
3.
PLoS One ; 8(6): e67256, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825648

RESUMO

Rituximab, a monoclonal antibody targeting CD20 on B cells, is currently used to treat many subtypes of B cell lymphomas. However, treatment is not curative and response rates are variable. Recombinant interleukin-21 (rIL-21) is a cytokine that enhances immune effector function and affects both primary and transformed B cell differentiation. We hypothesized that the combination of rIL-21 plus rituximab would be a more efficacious treatment for B cell malignancies than rituximab alone. We cultured human and cynomolgus monkey NK cells with rIL-21 and found that their activity was increased and proteins associated with antibody dependent cytotoxicity were up-regulated. Studies in cynomolgus monkeys modeled the effects of rIL-21 on rituximab activity against CD20 B cells. In these studies, rIL-21 activated innate immune effectors, increased ADCC and mobilized B cells into peripheral blood. When rIL-21 was combined with rituximab, deeper and more durable B cell depletion was observed. In another series of experiments, IL-21 was shown to have direct antiproliferative activity against a subset of human lymphoma cell lines, and combination of murine IL-21 with rituximab yielded significant survival benefits over either agent alone in xenogeneic mouse tumor models of disseminated lymphoma. Therefore, our results do suggest that the therapeutic efficacy of rituximab may be improved when used in combination with rIL-21.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Antineoplásicos/farmacologia , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Interleucinas/farmacologia , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/imunologia , Animais , Anticorpos Monoclonais Murinos/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Linfócitos B/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Linfoma de Células B/patologia , Macaca fascicularis , Masculino , Camundongos , Rituximab , Análise de Sobrevida
4.
Arthritis Res Ther ; 12(2): R48, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20302641

RESUMO

INTRODUCTION: B-lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) family that regulate B-cell maturation, survival, and function. They are overexpressed in a variety of autoimmune diseases and reportedly exist in vivo not only as homotrimers, but also as BLyS/APRIL heterotrimers. METHODS: A proprietary N-terminal trimerization domain was used to produce recombinant BLyS/APRIL heterotrimers. Heterotrimer biologic activity was compared with that of BLyS and APRIL in a 4-hour signaling assay by using transmembrane activator and CAML interactor (TACI)-transfected Jurkat cells and in a 4-day primary human B-cell proliferation assay. A bead-based immunoassay was developed to quantify native heterotrimers in human sera from healthy donors (n = 89) and patients with systemic lupus erythematosus (SLE; n = 89) or rheumatoid arthritis (RA; n = 30). Heterotrimer levels were compared with BLyS and APRIL homotrimer levels in a subset of these samples. RESULTS: The recombinant heterotrimers consisted mostly of one BLyS and two APRIL molecules. Heterotrimer signaling did not show any significant difference compared with APRIL in the TACI-Jurkat assay. Heterotrimers were less-potent inducers of B-cell proliferation than were homotrimeric BLyS or APRIL (EC(50), nMol/L: BLyS, 0.02; APRIL, 0.17; heterotrimers, 4.06). The soluble receptor fusion proteins atacicept and B-cell maturation antigen (BCMA)-immunoglobulin (Ig) neutralized the activity of BLyS, APRIL, and heterotrimers in both cellular assays, whereas B-cell activating factor belonging to the TNF family receptor (BAFF-R)-Ig neutralized only the activity of BLyS. In human sera, significantly more patients with SLE had detectable BLyS (67% versus 18%; P < 0.0001), APRIL (38% versus 3%; P < 0.0002), and heterotrimer (27% versus 8%; P = 0.0013) levels compared with healthy donors. Significantly more patients with RA had detectable APRIL, but not BLyS or heterotrimer, levels compared with healthy donors (83% versus 3%; P < 0.0001). Heterotrimer levels weakly correlated with BLyS, but not APRIL, levels. CONCLUSIONS: Recombinant BLyS/APRIL heterotrimers have biologic activity and are inhibited by atacicept and BCMA-Ig, but not by BAFF-R-Ig. A novel immunoassay demonstrated that native BLyS/APRIL heterotrimers, as well as BLyS and APRIL homotrimers, are elevated in patients with autoimmune diseases.


Assuntos
Doenças Autoimunes/sangue , Fator Ativador de Células B/sangue , Antígeno de Maturação de Linfócitos B/farmacologia , Proliferação de Células/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Humanos , Interleucina-4/farmacologia , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , Multimerização Proteica , Proteínas Recombinantes
5.
J Biol Chem ; 285(21): 16116-24, 2010 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-20332089

RESUMO

Glutamate cysteine ligase (GCL) catalyzes the rate-limiting step in the formation of the cellular antioxidant glutathione (GSH). The GCL holoenzyme consists of two separately coded proteins, a catalytic subunit (GCLC) and a modifier subunit (GCLM). Both GCLC and GLCM are controlled transcriptionally by a variety of cellular stimuli, including oxidative stress. This study addresses post-translational control of GCL activity, which increased rapidly in human lymphocytes following oxidative stress. Activation of GCL occurred within minutes of treatment and without any change in GCL protein levels and coincided with an increase in the proportion of GCLC in the holoenzyme form. Likewise, GCLM shifted from the monomeric form to holoenzyme and higher molecular weight species. Normal rat tissues also showed a distribution of monomeric and higher molecular weight forms. Neither GCL activation, nor the formation of holoenzyme, required a covalent intermolecular disulfide bridge between GCLC and GCLM. However, in immunoprecipitation studies, a neutralizing epitope associated with enzymatic activity was protected following cellular oxidative stress. Thus, the N-terminal portion of GCLC may undergo a change that stabilizes the GCL holoenzyme. Our results suggest that a dynamic equilibrium exists between low and high activity forms of GCL and is altered by transient oxidative stress. This provides a mechanism for the rapid post-translational activation of GCL and maintenance of cellular GSH homeostasis.


Assuntos
Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Homeostase/fisiologia , Estresse Oxidativo/fisiologia , Transcrição Gênica/fisiologia , Animais , Dissulfetos/metabolismo , Ativação Enzimática/fisiologia , Holoenzimas/metabolismo , Humanos , Células Jurkat , Linfócitos/enzimologia , Camundongos , Especificidade de Órgãos/fisiologia , Estrutura Terciária de Proteína , Ratos
6.
Reprod Toxicol ; 19(1): 117-29, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15336720

RESUMO

The placenta and the yolk sac play critical roles in fetal development, including protection from oxidative stress through the presence of detoxifying enzymes. Glutathione (GSH; gamma-glutamylcysteinylglycine), a crucial molecule in the maintenance of cellular redox status, plays a critical role in development, and it is also protective against methylmercury toxicity. Glutamate-cysteine ligase (GCL), the enzyme that catalyzes the rate-limiting step in GSH synthesis, is widely expressed in the mouse embryo and extraembryonic membranes throughout development. The aim of this study was to investigate the effect of low-level subchronic methylmercury exposure on GCL expression in the mouse placenta and yolk sac, after describing the basal developmental expression of the enzyme in these tissues. We found that basal mRNA expression levels increased dramatically in the placenta and the yolk sac at gd 18, whereas protein levels did not increase in parallel with the mRNA. We also found that methylmercury induced GCLc mRNA expression in the placenta at gd 18 in a dose-dependent manner, suggesting an important role for this enzyme in the response of the placenta to toxicants. These changes in expression may be useful as a biomarker of MeHg exposure during development.


Assuntos
Poluentes Ambientais/toxicidade , Glutamato-Cisteína Ligase/biossíntese , Compostos de Metilmercúrio/toxicidade , Placenta/efeitos dos fármacos , Saco Vitelino/efeitos dos fármacos , Administração Oral , Animais , Relação Dose-Resposta a Droga , Poluentes Ambientais/administração & dosagem , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Hibridização In Situ , Masculino , Compostos de Metilmercúrio/administração & dosagem , Camundongos , Placenta/enzimologia , Gravidez , RNA Mensageiro/metabolismo , Abastecimento de Água , Saco Vitelino/enzimologia
7.
Free Radic Biol Med ; 37(5): 632-42, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15288121

RESUMO

Glutathione (GSH) is important in free radical scavenging, maintaining cellular redox status, and regulating cell survival in response to a wide variety of toxicants. The rate-limiting enzyme in GSH synthesis is glutamate-cysteine ligase (GCL), which is composed of catalytic (GCLC) and modifier (GCLM) subunits. To determine whether increased GSH biosynthetic capacity enhances cellular resistance to tumor necrosis factor-alpha- (TNF-alpha-) induced apoptotic cell death, we have established several mouse liver hepatoma (Hepa-1) cell lines overexpressing GCLC and/or GCLM. Cells overexpressing GCLC alone exhibit modest increases in GCL activity, while cells overexpressing both subunits have large increases in GCL activity. Importantly, cells overexpressing both GCL subunits exhibit increased resistance to TNF-induced apoptosis as judged by a loss of redox potential; mitochondrial membrane potential; translocation of cytochrome c to the cytoplasm; and activation of caspase-3, caspase-8, and caspase-9. Analysis of the effects of TNF on these parameters indicates that maintaining mitochondrial integrity mediates this protective effect in GCL-overexpressing cells.


Assuntos
Apoptose/efeitos dos fármacos , Glutamato-Cisteína Ligase/metabolismo , Mitocôndrias/patologia , Fator de Necrose Tumoral alfa/toxicidade , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Glutationa/metabolismo , Humanos , Neoplasias Hepáticas , Camundongos , Mitocôndrias/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Transfecção
8.
Anal Biochem ; 318(2): 175-80, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12814619

RESUMO

Glutamate-cysteine ligase (GCL; also known as gamma-glutamylcysteine synthetase) is the rate-limiting enzyme in glutathione (GSH) synthesis. Traditional assays for the activity of this enzyme are based either on coupled reactions with other enzymes or on high-performance liquid chromatography (HPLC) assessment of gamma-glutamylcysteine (gamma-GC) product formation. We took advantage of the reaction of naphthalene dicarboxaldehyde (NDA) with GSH or gamma-GC to form cyclized products that are highly fluorescent. Hepa-1 cells which were designed to overexpress mouse GCL and mouse liver homogenates were used to evaluate and compare the utility of the NDA method with an assay based on monobromobimane derivatization and HPLC analysis with fluorescence detection. Excellent agreement was found between GCL activities measured by HPLC and NDA-microtiter plate analyses. This assay should be useful for high-throughput GCL activity analyses.


Assuntos
Glutamato-Cisteína Ligase/análise , Glutamato-Cisteína Ligase/metabolismo , Animais , Células Cultivadas , Fluorescência , Concentração de Íons de Hidrogênio , Fígado/citologia , Fígado/enzimologia , Extratos Hepáticos , Camundongos , Microquímica/métodos , Fatores de Tempo
9.
Am J Pathol ; 160(5): 1887-94, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12000740

RESUMO

Apoptotic cell death is usually accompanied by activation of a family of cysteine proteases termed caspases. Caspases mediate the selective proteolysis of multiple cellular targets often resulting in the disruption of survival pathways. Intracellular levels of the antioxidant glutathione (GSH) are an important determinant of cellular susceptibility to apoptosis. The rate-limiting step in GSH biosynthesis is mediated by glutamate-L-cysteine ligase (GCL), a heterodimeric enzyme consisting of a catalytic (GCLC) and a modifier (GCLM) subunit. In this report we demonstrate that GCLC is a direct target for caspase-mediated cleavage in multiple models of apoptotic cell death. Mutational analysis revealed that caspase-mediated cleavage of GCLC occurs at Asp(499) within the sequence AVVD(499)G. GCLC cleavage occurs upstream of Cys(553), which is thought to be important for association with GCLM. GCLC cleavage is accompanied by a rapid loss of intracellular GSH due to caspase-mediated extrusion of GSH from the cell. However, while GCLC cleavage is dependent on caspase-3, GSH extrusion occurs by a caspase-3-independent mechanism. Our identification of GCLC as a target for caspase-3-dependent cleavage during apoptotic cell death suggests that this post-translational modification may represent a novel mechanism for regulating GSH biosynthesis during apoptosis.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Glutamato-Cisteína Ligase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Sítios de Ligação/genética , Caspase 3 , Domínio Catalítico , Relação Dose-Resposta a Droga , Ativação Enzimática , Glutamato-Cisteína Ligase/genética , Glutationa/metabolismo , Células HeLa , Humanos , Células Jurkat , Camundongos , Receptores de Superfície Celular/fisiologia , Fatores de Tempo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
10.
Mol Reprod Dev ; 62(1): 83-91, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11933164

RESUMO

The tripeptide glutathione (GSH), which plays a crucial role in protecting cells against oxidative stress, is synthesized in a two-step process. The rate-limiting step is the binding of glutamate and cysteine, which is catalyzed by the enzyme glutamate-cysteine ligase (GCL). This enzyme is composed of two subunits: a large catalytic subunit (GCLc) and a smaller modifying subunit (GCLm), originating from different genes. Control of cellular GSH levels is essential for normal development. In the current study, we investigated the tissue distribution of Gclc and Gclm transcripts, as well as GCLc protein, in the developing mouse embryo. We found that both mRNAs were highly expressed in the liver and CNS at gestational day 10 (gd 10) and gd 12, with Gclm being more abundant than Gclc in the liver relative to other tissues. Also, the expression of the two subunit mRNAs was not always parallel in the embryo, in that some tissues expressed one of the subunits preferentially, suggesting that the two genes are differentially expressed during mouse development. The GCLc protein was also widely expressed throughout the embryo, and, in general, it co-localized with the Gclc mRNA.


Assuntos
Expressão Gênica , Glutamato-Cisteína Ligase/genética , Tecido Adiposo Marrom/enzimologia , Animais , Domínio Catalítico , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/enzimologia , Desenvolvimento Embrionário e Fetal , Feminino , Fígado/embriologia , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , RNA Mensageiro , Glândulas Salivares/embriologia , Glândulas Salivares/enzimologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA