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








Base de dados
Intervalo de ano de publicação
1.
Clin Transl Sci ; 16(8): 1431-1444, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37154518

RESUMO

Growth Differentiation Factor-15 (GDF15) is a circulating polypeptide linked to cellular stress and metabolic adaptation. GDF15's half-life is ~3 h and activates the glial cell line-derived neurotrophic factor family receptor alpha-like (GFRAL) receptor expressed in the area postrema. To characterize sustained GFRAL agonism on food intake (FI) and body weight (BW), we tested a half-life extended analog of GDF15 (Compound H [CpdH]) suitable for reduced dosing frequency in obese cynomolgus monkeys. Animals were chronically treated once weekly (q.w.) with CpdH or long-acting GLP-1 analog dulaglutide. Mechanism-based longitudinal exposure-response modeling characterized effects of CpdH and dulaglutide on FI and BW. The novel model accounts for both acute, exposure-dependent effects reducing FI and compensatory changes in energy expenditure (EE) and FI occurring over time with weight loss. CpdH had linear, dose-proportional pharmacokinetics (terminal half-life ~8 days) and treatment caused exposure-dependent reductions in FI and BW. The 1.6 mg/kg CpdH reduced mean FI by 57.5% at 1 week and sustained FI reductions of 31.5% from weeks 9-12, resulting in peak reduction in BW of 16 ± 5%. Dulaglutide had more modest effects on FI and peak BW loss was 3.8 ± 4.0%. Longitudinal modeling of both the FI and BW profiles suggested reductions in BW observed with both CpdH and dulaglutide were fully explained by exposure-dependent reductions in FI without increase in EE. Upon verification of the pharmacokinetic/pharmacodynamic relationship established in monkeys and humans for dulaglutide, we predicted that CpdH could reach double digit BW loss in humans. In summary, a long-acting GDF15 analog led to sustained reductions in FI in overweight monkeys and holds potential for effective clinical obesity pharmacotherapy.


Assuntos
Ingestão de Alimentos , Obesidade , Humanos , Animais , Obesidade/metabolismo , Redução de Peso , Peso Corporal/fisiologia , Primatas , Fator 15 de Diferenciação de Crescimento/farmacologia , Fator 15 de Diferenciação de Crescimento/uso terapêutico
2.
MAbs ; 12(1): 1708030, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31906797

RESUMO

Mucosal immunity is dominated by secretory IgA and IgM, although these are less favorable compared to IgG molecules for therapeutic development. Polymeric IgA and IgM are actively transported across the epithelial barrier via engagement of the polymeric Ig receptor (pIgR), but IgG molecules lack a lumen-targeted active transport mechanism, resulting in poor biodistribution of IgG therapeutics in mucosal tissues. In this work, we describe the discovery and characterization of single-domain antibodies (VHH) that engage pIgR and undergo transepithelial transport across the mucosal epithelium. The anti-pIgR VHH panel displayed a broad range of biophysical characteristics, epitope diversity, IgA competition profiles and transcytosis activity in cell and human primary lung tissue models. Making use of this diverse VHH panel, we studied the relationship between biophysical and functional properties of anti-pIgR binders targeting different domains and epitopes of pIgR. These VHH molecules will serve as excellent tools for studying pIgR-mediated transport of biologics and for delivering multispecific IgG antibodies into mucosal lumen, where they can target and neutralize mucosal antigens.


Assuntos
Produtos Biológicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Receptores de Imunoglobulina Polimérica , Anticorpos de Domínio Único , Transcitose/fisiologia , Animais , Descoberta de Drogas , Humanos , Imunoglobulina G , Mucosa
3.
MAbs ; 12(1): 1688616, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31852337

RESUMO

The development of mechanism-based, multiscale pharmacokinetic-pharmacodynamic (PK-PD) models for chimeric antigen receptor (CAR)-T cells is needed to enable investigation of in vitro and in vivo correlation of CAR-T cell responses and to facilitate preclinical-to-clinical translation. Toward this goal, we first developed a cell-level in vitro PD model that quantitatively characterized CAR-T cell-induced target cell depletion, CAR-T cell expansion and cytokine release. The model accounted for key drug-specific (CAR-affinity, CAR-densities) and system-specific (antigen densities, E:T ratios) variables and was able to characterize comprehensive in vitro datasets from multiple affinity variants of anti-EGFR and anti-HER2 CAR-T cells. Next, a physiologically based PK (PBPK) model was developed to simultaneously characterize the biodistribution of untransduced T-cells, anti-EGFR CAR-T and anti-CD19 CAR-T cells in xenograft -mouse models. The proposed model accounted for the engagement of CAR-T cells with tumor cells at the site of action. Finally, an integrated PBPK-PD relationship was established to simultaneously characterize expansion of CAR-T cells and tumor growth inhibition (TGI) in xenograft mouse model, using datasets from anti-BCMA, anti-HER2, anti-CD19 and anti-EGFR CAR-T cells. Model simulations provided potential mechanistic insights toward the commonly observed multiphasic PK profile (i.e., rapid distribution, expansion, contraction and persistence) of CAR-T cells in the clinic. Model simulations suggested that CAR-T cells may have a steep dose-exposure relationship, and the apparent Cmax upon CAR-T cell expansion in blood may be more sensitive to patient tumor-burden than CAR-T dose levels. Global sensitivity analysis described the effect of other drug-specific parameters toward CAR-T cell expansion and TGI. The proposed modeling framework will be further examined with the clinical PK and PD data, and the learnings can be used to inform design and development of future CAR-T therapies.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Animais , Movimento Celular , Proliferação de Células , Simulação por Computador , Receptores ErbB/imunologia , Xenoenxertos , Humanos , Camundongos , Modelos Teóricos , Neoplasias/terapia , Ligação Proteica , Receptor ErbB-2/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia
4.
Nat Med ; 23(10): 1150-1157, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28846097

RESUMO

Growth differentiation factor 15 (GDF15), a distant member of the transforming growth factor (TGF)-ß family, is a secreted protein that circulates as a 25-kDa dimer. In humans, elevated GDF15 correlates with weight loss, and the administration of GDF15 to mice with obesity reduces body weight, at least in part, by decreasing food intake. The mechanisms through which GDF15 reduces body weight remain poorly understood, because the cognate receptor for GDF15 is unknown. Here we show that recombinant GDF15 induces weight loss in mice fed a high-fat diet and in nonhuman primates with spontaneous obesity. Furthermore, we find that GDF15 binds with high affinity to GDNF family receptor α-like (GFRAL), a distant relative of receptors for a distinct class of the TGF-ß superfamily ligands. Gfral is expressed in neurons of the area postrema and nucleus of the solitary tract in mice and humans, and genetic deletion of the receptor abrogates the ability of GDF15 to decrease food intake and body weight in mice. In addition, diet-induced obesity and insulin resistance are exacerbated in GFRAL-deficient mice, suggesting a homeostatic role for this receptor in metabolism. Finally, we demonstrate that GDF15-induced cell signaling requires the interaction of GFRAL with the coreceptor RET. Our data identify GFRAL as a new regulator of body weight and as the bona fide receptor mediating the metabolic effects of GDF15, enabling a more comprehensive assessment of GDF15 as a potential pharmacotherapy for the treatment of obesity.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator 15 de Diferenciação de Crescimento/genética , Obesidade/metabolismo , Redução de Peso/efeitos dos fármacos , Animais , Dieta Hiperlipídica , Ingestão de Alimentos/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 15 de Diferenciação de Crescimento/metabolismo , Fator 15 de Diferenciação de Crescimento/farmacologia , Humanos , Macaca fascicularis , Camundongos , Camundongos Knockout , Redução de Peso/genética
5.
Thromb Res ; 138: 96-102, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26688324

RESUMO

BACKGROUND: Impaired platelet function may underlie bleeding associated with cardiopulmonary bypass (CPB) and at present is incompletely evaluated with existing diagnostic technologies. Sonorheometry (SR) is a recently developed ultrasound-based technology that quantifies hemostasis and platelet activity from a blood sample by measuring ex vivo clot stiffness (S). We hypothesized that impaired platelet-fibrin interactions as assessed by SR would correlate with transfusion during CPB and history of prior aspirin therapy. METHODS: Thirty-nine patients undergoing elective cardiopulmonary bypass (CPB) were enrolled following informed consent (University of Virginia IRB#14050) in a prospective observational pilot study to assess pre-operative platelet function and transfusion frequency. To assess platelet activity, abciximab was added to blood prior to SR and native S versus abciximab treated S created a differential test for platelet activity. Patient blood samples were activated with kaolin and SR was then used to measure clot stiffness. Patients were transfused with blood products as directed by clinical practice, with the surgical team blinded to SR results. RESULTS: Blood clot stiffness with and without abciximab, was compared in a ratio test (S/Sabciximab) named the Platelet Function Index (PFI). PFI was hypothesized to be positively correlated with platelet contributions through integrin αIIbß3 to clot stiffness. PFI for CPB subjects was lower for those receiving transfusions than those not receiving transfusions (p<0.006). A receiver-operator characteristics (ROC) analysis correlating the PFI with the blinded surgical team's decision on transfusions that included platelet concentrates generated an area under the curve (AUC) of 0.79 (p<0.001). Additionally, the mean value of PFI for subjects on aspirin therapy was lower than for those not on aspirin therapy (p<0.02) and correlated with a 1.73-fold enhanced risk of receiving a peri-operative transfusion. CONCLUSION: Evaluation of platelet function with SR may help in the specification of blood transfusion needs in cardiac surgery and in the assessment of aspirin effects on risk of surgical bleeding.


Assuntos
Aspirina/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Ponte Cardiopulmonar , Inibidores da Agregação Plaquetária/uso terapêutico , Testes de Função Plaquetária/métodos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Idoso , Aspirina/farmacologia , Testes de Coagulação Sanguínea/métodos , Perda Sanguínea Cirúrgica , Plaquetas/citologia , Plaquetas/metabolismo , Transfusão de Sangue , Ponte Cardiopulmonar/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Adesividade Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Estudos Prospectivos , Ultrassonografia/métodos
6.
Biotechnol J ; 5(7): 660-70, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20665641

RESUMO

Algal fuel sources promise unsurpassed yields in a carbon neutral manner that minimizes resource competition between agriculture and fuel crops. Many challenges must be addressed before algal biofuels can be accepted as a component of the fossil fuel replacement strategy. One significant challenge is that the cost of algal fuel production must become competitive with existing fuel alternatives. Algal biofuel production presents the opportunity to fine-tune microbial metabolic machinery for an optimal blend of biomass constituents and desired fuel molecules. Genome-scale model-driven algal metabolic design promises to facilitate both goals by directing the utilization of metabolites in the complex, interconnected metabolic networks to optimize production of the compounds of interest. Network analysis can direct microbial development efforts towards successful strategies and enable quantitative fine-tuning of the network for optimal product yields while maintaining the robustness of the production microbe. Metabolic modeling yields insights into microbial function, guides experiments by generating testable hypotheses, and enables the refinement of knowledge on the specific organism. While the application of such analytical approaches to algal systems is limited to date, metabolic network analysis can improve understanding of algal metabolic systems and play an important role in expediting the adoption of new biofuel technologies.


Assuntos
Biocombustíveis , Microbiologia Industrial/métodos , Chlamydomonas reinhardtii/isolamento & purificação , Chlamydomonas reinhardtii/metabolismo , Modelos Biológicos , Análise de Sistemas
7.
Clin Chim Acta ; 411(1-2): 106-13, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19861121

RESUMO

BACKGROUND: Unregulated hemostasis represents a leading cause of mortality and morbidity in the developed world. Being able to recognize and quantify defects of the hemostatic process is critical to reduce mortality and implement appropriate treatment. METHODS: We describe a novel ultrasound-based technology, named sonorheometry, which can assess hemostasis function from a small sample of blood. Sonorheometry uses the phenomenon of acoustic radiation force to measure the dynamic changes in blood viscoelasticity during clot formation and clot dissolution. We performed in vitro experiments using whole blood samples of 1 ml to demonstrate that sonorheometry is indicative of hemostatic functions that depend on plasma coagulation factors, platelets, and plasma fibrinolytic factors. RESULTS: Sonorheometry measurements show titration effects to compounds known to alter the coagulation factors (GPRP peptide, 0 to 8 mmol/l), platelets (abciximab, 0 to 12 microg/ml), and fibrinolytic factors (urokinase, 0 to 200 U). Repeated measurements of blood samples from the same subjects yielded reproducibility errors on the order of 5%. CONCLUSIONS: These data indicate that sonorheometry accurately quantifies the functional role of the components of hemostasis in vitro.


Assuntos
Sangue , Hemostasia , Ultrassom , Fatores de Coagulação Sanguínea/análise , Plaquetas/citologia , Fibrinólise , Humanos , Reprodutibilidade dos Testes , Reologia , Viscosidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA