Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38734778

RESUMO

Hereditary angioedema (HAE) due to C1-inhibitor deficiency is a rare, debilitating, genetic disorder characterized by recurrent, unpredictable, attacks of edema. The clinical symptoms of HAE arise from excess bradykinin generation due to dysregulation of the plasma kallikrein-kinin system (KKS). A quantitative systems pharmacology (QSP) model that mechanistically describes the KKS and its role in HAE pathophysiology was developed based on HAE attacks being triggered by autoactivation of factor XII (FXII) to activated FXII (FXIIa), resulting in kallikrein production from prekallikrein. A base pharmacodynamic model was constructed and parameterized from literature data and ex vivo assays measuring inhibition of kallikrein activity in plasma of HAE patients or healthy volunteers who received lanadelumab. HAE attacks were simulated using a virtual patient population, with attacks recorded when systemic bradykinin levels exceeded 20 pM. The model was validated by comparing the simulations to observations from lanadelumab and plasma-derived C1-inhibitor clinical trials. The model was then applied to analyze the impact of nonadherence to a daily oral preventive therapy; simulations showed a correlation between the number of missed doses per month and reduced drug effectiveness. The impact of reducing lanadelumab dosing frequency from 300 mg every 2 weeks (Q2W) to every 4 weeks (Q4W) was also examined and showed that while attack rates with Q4W dosing were substantially reduced, the extent of reduction was greater with Q2W dosing. Overall, the QSP model showed good agreement with clinical data and could be used for hypothesis testing and outcome predictions.

2.
Surg Endosc ; 37(6): 4658-4672, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36879167

RESUMO

BACKGROUND: Consensus on the best surgical strategy for the management of synchronous colorectal liver metastases (sCRLM) has not been achieved. This study aimed to assess the attitudes of surgeons involved in the treatment of sCRLM. METHODS: Surveys designed for colorectal, hepato-pancreato-biliary (HPB), and general surgeons were disseminated through representative societies. Subgroup analyses were performed to compare responses between specialties and continents. RESULTS: Overall, 270 surgeons (57 colorectal, 100 HPB and 113 general surgeons) responded. Specialist surgeons more frequently utilized minimally invasive surgery (MIS) than general surgeons for colon (94.8% vs. 71.7%, p < 0.001), rectal (91.2% vs. 64.6%, p < 0.001), and liver resections (53% vs. 34.5%, p = 0.005). In patients with an asymptomatic primary, the liver-first two-stage approach was preferred in most respondents' centres (59.3%), while the colorectal-first approach was preferred in Oceania (83.3%) and Asia (63.4%). A substantial proportion of the respondents (72.6%) had personal experience with minimally invasive simultaneous resections, and an expanding role for this procedure was foreseen (92.6%), while more evidence was desired (89.6%). Respondents were more reluctant to combine a hepatectomy with low anterior (76.3%) and abdominoperineal resections (73.3%), compared to right (94.4%) and left hemicolectomies (90.7%). Colorectal surgeons were less inclined to combine right or left hemicolectomies with a major hepatectomy than HPB and general surgeons (right: 22.8% vs. 50% and 44.2%, p = 0.008; left: 14% vs. 34% and 35.4%, p = 0.002, respectively). CONCLUSION: The clinical practices and viewpoints on the management of sCRLM differ between continents, and between and within surgical specialties. However, there appears to be consensus on a growing role for MIS and a need for evidence-based input.


Assuntos
Neoplasias Colorretais , Neoplasias Hepáticas , Especialidades Cirúrgicas , Humanos , Neoplasias Colorretais/cirurgia , Neoplasias Colorretais/patologia , Reto/patologia , Inquéritos e Questionários , Hepatectomia/métodos , Neoplasias Hepáticas/cirurgia , Neoplasias Hepáticas/secundário
3.
BMC Nutr ; 8(1): 13, 2022 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-35152906

RESUMO

BACKGROUND: Severe acute malnutrition (SAM) is the most life-threatening form of malnutrition, and in 2019, approximately 14.3 million children under the age of 5 were considered to have SAM. The prevalence of child malnutrition is recorded through large-scale household surveys run at multi-year intervals. However, these surveys are expensive, yield estimates with high levels of aggregation, are run over large time intervals, and may show gaps in area coverage. Geospatial modelling approaches could address some of these challenges by combining geo-located survey data with geospatial data to produce mapped estimates that predict malnutrition risk in both surveyed and non-surveyed areas. METHODS: A secondary analysis of cluster-level program evaluation data (n = 123 primary sampling units) was performed to map severe acute malnutrition (SAM) in Papuan children under 2 years (0-23 months) of age with a spatial resolution of 1 × 1 km in Papua, Indonesia. The approach used Bayesian geostatistical modelling techniques and publicly available geospatial data layers. RESULTS: In Papua, Indonesia, SAM was predicted in geostatistical models by using six geospatial covariates related primarily to conditions of remoteness and inaccessibility. The predicted 1-km spatial resolution maps of SAM showed substantial spatial variation across the province. By combining the predicted rates of SAM with estimates of the population under 2 years of age, the prevalence of SAM in late 2018 was estimated to be around 15,000 children (95% CI 10,209-26,252). Further tests of the predicted levels suggested that in most areas of Papua, more than 5% of Papuan children under 2 years of age had SAM, while three districts likely had more than 15% of children with SAM. CONCLUSIONS: Eradication of hunger and malnutrition remains a key development goal, and more spatially detailed data can guide efficient intervention strategies. The application of additional household survey datasets in geostatistical models is one way to improve the monitoring and timely estimation of populations at risk of malnutrition. Importantly, geospatial mapping can yield insights for both surveyed and non-surveyed areas and can be applied in low-income country contexts where data is scarce and data collection is expensive or regions are inaccessible.

4.
Matern Child Nutr ; 17(1): e13070, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32761792

RESUMO

Severe acute malnutrition (SAM) among children in Nigeria is tackled through the outpatient therapeutic programme (OTP) of the Community-based Management of Acute Malnutrition (CMAM) programme. CMAM is evidently effective in resolving SAM, but little evidence exists on the remaining risk of SAM relapse for children discharged as cured from the OTP. We aimed to measure and compare the 6-month incidence of SAM among OTP-cured and community control children and identify factors associated with SAM relapse. We conducted a prospective matched cohort study that tracked 553 OTP-cured and 526 control children in Sokoto State, Northern Nigeria. Outcomes and covariates were measured fortnightly in up to 12 home visits. We used multivariate Cox and accelerated failure time models to identify significant risk correlates, where the covariates to be tested for correlation with relapse were selected using domain knowledge and automatic feature selection methods. SAM incidence rates were 52 times higher in the OTP-cured cohort (0.204/100 child-days) than in the community control cohort (0.004/100 child-days). Children with lower mid-upper arm circumference at OTP admission, with lower height/length-for-age z-scores, whose household head did not work over the full year, who lived in an area previously affected by environmental shocks, who were female and who had diarrhoea before the visit had a significantly higher relapse risk. Our study shows that OTP-cured children remain at a significantly excess risk of SAM. To improve long-term health outcomes of these children, programmes adopting a CMAM approach should strengthen follow-up care and be integrated with other preventive services.


Assuntos
Desnutrição , Desnutrição Aguda Grave , Estudos de Coortes , Feminino , Humanos , Incidência , Lactente , Desnutrição/epidemiologia , Nigéria/epidemiologia , Estudos Prospectivos , Desnutrição Aguda Grave/epidemiologia , Desnutrição Aguda Grave/terapia
5.
CPT Pharmacometrics Syst Pharmacol ; 9(9): 534-541, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32697437

RESUMO

Monoclonal antibodies (mAbs) can be engineered to have "extended half-life" and "catch and release" properties to improve target coverage. We have developed a mAb physiologically-based pharmacokinetic model that describes intracellular trafficking, neonatal Fc receptor (FcRn) recycling, and nonspecific clearance of mAbs. We extended this model to capture target binding as a function of target affinity, expression, and turnover. For mAbs engineered to have an extended half-life, the model was able to accurately predict the terminal half-life (82% within 2-fold error of the observed value) in the human FcRn transgenic (Tg32) homozygous mouse and human. The model also accurately captures the trend in pharmacokinetic and target coverage data for a set of mAbs with differing catch and release properties in the Tg32 mouse. The mechanistic nature of this model allows us to explore different engineering techniques early in drug discovery, potentially expanding the number of "druggable" targets.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Antígenos de Histocompatibilidade Classe I/genética , Receptores Fc/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Amplamente Neutralizantes/imunologia , Anticorpos Amplamente Neutralizantes/metabolismo , Simulação por Computador , Desenvolvimento de Medicamentos , Descoberta de Drogas , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/metabolismo , Meia-Vida , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Homozigoto , Humanos , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Mutação , Ligação Proteica/imunologia , Engenharia de Proteínas/métodos , Receptores Fc/efeitos dos fármacos
6.
J Pharmacokinet Pharmacodyn ; 47(5): 513-526, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32710210

RESUMO

A modeling and simulation approach was used for quantitative comparison of a new generation HER2 antibody drug conjugate (ADC, PF-06804103) with trastuzumab-DM1 (T-DM1). To compare preclinical efficacy, the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of PF-06804103 and T-DM1 was determined across a range of mouse tumor xenograft models, using a tumor growth inhibition model. The tumor static concentration was assigned as the minimal efficacious concentration. PF-06804103 was concluded to be more potent than T-DM1 across cell lines studied. TSCs ranged from 1.0 to 9.8 µg/mL (n = 7) for PF-06804103 and from 4.7 to 29 µg/mL (n = 5) for T-DM1. Two experimental models which were resistant to T-DM1, responded to PF-06804103 treatment. A mechanism-based target mediated drug disposition (TMDD) model was used to predict the human PK of PF-06804103. This model was constructed and validated based on T-DM1 which has non-linear PK at doses administered in the clinic, driven by binding to shed HER2. Non-linear PK is predicted for PF-06804103 in the clinic and is dependent upon circulating HER2 extracellular domain (ECD) concentrations. The models were translated to human and suggested greater efficacy for PF-06804103 compared to T-DM1. In conclusion, a fit-for-purpose translational PK/PD strategy for ADCs is presented and used to compare a new generation HER2 ADC with T-DM1.


Assuntos
Ado-Trastuzumab Emtansina/farmacocinética , Antineoplásicos Imunológicos/farmacocinética , Imunoconjugados/farmacocinética , Neoplasias/tratamento farmacológico , Receptor ErbB-2/antagonistas & inibidores , Administração Intravenosa , Ado-Trastuzumab Emtansina/administração & dosagem , Animais , Antineoplásicos Imunológicos/administração & dosagem , Linhagem Celular Tumoral , Simulação por Computador , Relação Dose-Resposta a Droga , Feminino , Humanos , Imunoconjugados/administração & dosagem , Macaca fascicularis , Masculino , Camundongos , Modelos Biológicos , Neoplasias/patologia , Receptor ErbB-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
CPT Pharmacometrics Syst Pharmacol ; 9(7): 374-383, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32558397

RESUMO

Gaucher's disease type 1 (GD1) leads to significant morbidity and mortality through clinical manifestations, such as splenomegaly, hematological complications, and bone disease. Two types of therapies are currently approved for GD1: enzyme replacement therapy (ERT), and substrate reduction therapy (SRT). In this study, we have developed a quantitative systems pharmacology (QSP) model, which recapitulates the effects of eliglustat, the only first-line SRT approved for GD1, on treatment-naïve or patients with ERT-stabilized adult GD1. This multiscale model represents the mechanism of action of eliglustat that leads toward reduction of spleen volume. Model capabilities were illustrated through the application of the model to predict ERT and eliglustat responses in virtual populations of adult patients with GD1, representing patients across a spectrum of disease severity as defined by genotype-phenotype relationships. In summary, the QSP model provides a mechanistic computational platform for predicting treatment response via different modalities within the heterogeneous GD1 patient population.


Assuntos
Doença de Gaucher/tratamento farmacológico , Modelos Biológicos , Pirrolidinas/farmacologia , Biologia de Sistemas , Adulto , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/fisiopatologia , Humanos , Índice de Gravidade de Doença , Esplenomegalia/tratamento farmacológico , Esplenomegalia/etiologia , Resultado do Tratamento
8.
CPT Pharmacometrics Syst Pharmacol ; 9(6): 342-352, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32419339

RESUMO

Quantitative understanding about the dynamics of drug-target interactions in biological systems is essential, especially in rare disease programs with small patient populations. Follistatin, by antagonism of myostatin and activin, which are negative regulators of skeletal muscle and inflammatory response, is a promising therapeutic target for Duchenne Muscular Dystrophy. In this study, we constructed a quantitative systems pharmacology model for FS-EEE-Fc, a follistatin recombinant protein to investigate its efficacy from dual target binding, and, subsequently, to project its human efficacious dose. Based on model simulations, with an assumed efficacy threshold of 7-10% muscle volume increase, 3-5 mg/kg weekly dosing of FS-EEE-Fc is predicted to achieve meaningful clinical outcome. In conclusion, the study demonstrated an application of mechanism driven approach at early stage of a rare disease drug development to support lead compound optimization, enable human dose, pharmacokinetics, and efficacy predictions.


Assuntos
Folistatina/farmacocinética , Modelos Biológicos , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Fármacos Neuromusculares/farmacocinética , Biologia de Sistemas , Receptores de Activinas Tipo II/antagonistas & inibidores , Receptores de Activinas Tipo II/metabolismo , Animais , Cálculos da Dosagem de Medicamento , Folistatina/administração & dosagem , Humanos , Ligantes , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Miostatina/antagonistas & inibidores , Miostatina/metabolismo , Fármacos Neuromusculares/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Transdução de Sinais
9.
CPT Pharmacometrics Syst Pharmacol ; 8(10): 738-747, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31464379

RESUMO

Monoclonal antibody (mAb) pharmacokinetics (PK) have largely been predicted via allometric scaling with little consideration for cross-species differences in neonatal Fc receptor (FcRn) affinity or clearance/distribution mechanisms. To address this, we developed a mAb physiologically-based PK model that describes the intracellular trafficking and FcRn recycling of mAbs in a human FcRn transgenic homozygous mouse and human. This model uses mAb-specific in vitro data together with species-specific FcRn tissue expression, tissue volume, and blood-flow physiology to predict mAb in vivo linear PK a priori. The model accurately predicts the terminal half-life of 90% of the mAbs investigated within a twofold error. The mechanistic nature of this model allows us to not only predict linear PK from in vitro data but also explore the PK and target binding of mAbs engineered to have pH-dependent binding to its target or FcRn and could aid in the selection of mAbs with optimal PK and pharmacodynamic properties.


Assuntos
Anticorpos Monoclonais/farmacocinética , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Homozigoto , Humanos , Técnicas In Vitro , Modelos Lineares , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Especificidade de Órgãos , Especificidade da Espécie
10.
CPT Pharmacometrics Syst Pharmacol ; 7(7): 442-452, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29920993

RESUMO

Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder with heterogeneous clinical manifestations, including hepatosplenomegaly and infiltrative pulmonary disease, and is associated with significant morbidity and mortality. Olipudase alfa (recombinant human acid sphingomyelinase) is an enzyme replacement therapy under development for the non-neurological manifestations of ASMD. We present a quantitative systems pharmacology (QSP) model supporting the clinical development of olipudase alfa. The model is multiscale and mechanistic, linking the enzymatic deficiency driving the disease to molecular-level, cellular-level, and organ-level effects. Model development was informed by natural history, and preclinical and clinical studies. By considering patient-specific pharmacokinetic (PK) profiles and indicators of disease severity, the model describes pharmacodynamic (PD) and clinical end points for individual patients. The ASMD QSP model provides a platform for quantitatively assessing systemic pharmacological effects in adult and pediatric patients, and explaining variability within and across these patient populations, thereby supporting the extrapolation of treatment response from adults to pediatrics.


Assuntos
Terapia de Reposição de Enzimas/métodos , Modelos Biológicos , Doenças de Niemann-Pick/terapia , Proteínas Recombinantes/uso terapêutico , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/uso terapêutico , Animais , Calibragem , Humanos , Camundongos , Camundongos Knockout , Proteínas Recombinantes/farmacocinética , Esfingomielina Fosfodiesterase/farmacocinética
11.
CPT Pharmacometrics Syst Pharmacol ; 7(1): 26-33, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28941225

RESUMO

Gastrointestinal (GI) adverse events (AEs) are frequently dose limiting for oncology agents, requiring extensive clinical testing of alternative schedules to identify optimal dosing regimens. Here, we develop a translational mathematical model to predict these clinical AEs starting from preclinical GI toxicity data. The model structure incorporates known biology and includes stem cells, daughter cells, and enterocytes. Published data, including cellular numbers and division times, informed the system parameters for humans and rats. The drug-specific parameters were informed with preclinical histopathology data from rats treated with irinotecan. The model fit the rodent irinotecan-induced pathology changes well. The predicted time course of enterocyte loss in patients treated with weekly doses matched observed AE profiles. The model also correctly predicts a lower level of AEs for every 3 weeks (Q3W), as compared to the weekly schedule.


Assuntos
Antineoplásicos/administração & dosagem , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Trato Gastrointestinal/efeitos dos fármacos , Irinotecano/administração & dosagem , Modelos Biológicos , Biologia de Sistemas/métodos , Animais , Antineoplásicos/toxicidade , Relação Dose-Resposta a Droga , Esquema de Medicação , Trato Gastrointestinal/fisiologia , Humanos , Irinotecano/toxicidade , Valor Preditivo dos Testes , Especificidade da Espécie , Pesquisa Translacional Biomédica
12.
AAPS J ; 18(5): 1101-1116, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27198897

RESUMO

A mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) model was used for preclinical to clinical translation of inotuzumab ozogamicin, a CD22-targeting antibody-drug conjugate (ADC) for B cell malignancies including non-Hodgkin's lymphoma (NHL) and acute lymphocytic leukemia (ALL). Preclinical data was integrated in a PK/PD model which included (1) a plasma PK model characterizing disposition and clearance of inotuzumab ozogamicin and its released payload N-Ac-γ-calicheamicin DMH, (2) a tumor disposition model describing ADC diffusion into the tumor extracellular environment, (3) a cellular model describing inotuzumab ozogamicin binding to CD22, internalization, intracellular N-Ac-γ-calicheamicin DMH release, binding to DNA, or efflux from the tumor cell, and (4) tumor growth and inhibition in mouse xenograft models. The preclinical model was translated to the clinic by incorporating human PK for inotuzumab ozogamicin and clinically relevant tumor volumes, tumor growth rates, and values for CD22 expression in the relevant patient populations. The resulting stochastic models predicted progression-free survival (PFS) rates for inotuzumab ozogamicin in patients comparable to the observed clinical results. The model suggested that a fractionated dosing regimen is superior to a conventional dosing regimen for ALL but not for NHL. Simulations indicated that tumor growth is a highly sensitive parameter and predictive of successful outcome. Inotuzumab ozogamicin PK and N-Ac-γ-calicheamicin DMH efflux are also sensitive parameters and would be considered more useful predictors of outcome than CD22 receptor expression. In summary, a multiscale, mechanism-based model has been developed for inotuzumab ozogamicin, which can integrate preclinical biomeasures and PK/PD data to predict clinical response.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Simulação por Computador , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/farmacocinética , Pesquisa Translacional Biomédica/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/uso terapêutico , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Humanos , Imunoglobulina G/metabolismo , Inotuzumab Ozogamicina , Camundongos , Camundongos Nus , Estudos Retrospectivos , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/uso terapêutico
14.
Drug Metab Dispos ; 44(3): 356-64, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26700958

RESUMO

Because of the importance of intracellular unbound drug concentrations in the prediction of in vivo concentrations that are determinants of drug efficacy and toxicity, a number of assays have been developed to assess in vitro unbound concentrations of drugs. Here we present a rapid method to determine the intracellular unbound drug concentrations in cultured cells, and we apply the method along with a mechanistic model to predict concentrations of metformin in subcellular compartments of stably transfected human embryonic kidney 293 (HEK293) cells. Intracellular space (ICS) was calculated by subtracting the [(3)H]-inulin distribution volume (extracellular space, ECS) from the [(14)C]-urea distribution volume (total water space, TWS). Values obtained for intracellular space (mean ± S.E.M.; µl/10(6) cells) of monolayers of HEK cells (HEK-empty vector [EV]) and cells overexpressing human organic cation transporter 1 (HEK-OCT1), 1.21± 0.07 and 1.25±0.06, respectively, were used to determine the intracellular metformin concentrations. After incubation of the cells with 5 µM metformin, the intracellular concentrations were 26.4 ± 7.8 µM and 268 ± 11.0 µM, respectively, in HEK-EV and HEK-OCT1. In addition, intracellular metformin concentrations were lower in high K(+) buffer (140 mM KCl) compared with normal K(+) buffer (5.4 mM KCl) in HEK-OCT1 cells (54.8 ± 3.8 µM and 198.1 ± 11.2 µM, respectively; P < 0.05). Our mechanistic model suggests that, depending on the credible range of assumed physiologic values, the positively charged metformin accumulates to particularly high levels in endoplasmic reticulum and/or mitochondria. This method together with the computational model can be used to determine intracellular unbound concentrations and to predict subcellular accumulation of drugs in other complex systems such as primary cells.


Assuntos
Metformina/metabolismo , Transportador 1 de Cátions Orgânicos/metabolismo , Transporte Biológico/fisiologia , Linhagem Celular , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Mitocôndrias/metabolismo , Transfecção/métodos
15.
Sci Rep ; 5: 13545, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26310312

RESUMO

Understanding the therapeutic effect of drug dose and scheduling is critical to inform the design and implementation of clinical trials. The increasing complexity of both mono, and particularly combination therapies presents a substantial challenge in the clinical stages of drug development for oncology. Using a systems pharmacology approach, we have extended an existing PK-PD model of tumor growth with a mechanistic model of the cell cycle, enabling simulation of mono and combination treatment with the ATR inhibitor AZD6738 and ionizing radiation. Using AZD6738, we have developed multi-parametric cell based assays measuring DNA damage and cell cycle transition, providing quantitative data suitable for model calibration. Our in vitro calibrated cell cycle model is predictive of tumor growth observed in in vivo mouse xenograft studies. The model is being used for phase I clinical trial designs for AZD6738, with the aim of improving patient care through quantitative dose and scheduling prediction.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Biomarcadores/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Humanos , Cinética , Camundongos , Modelos Biológicos , Radiação Ionizante , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Artigo em Inglês | MEDLINE | ID: mdl-22255295

RESUMO

Pharmacokinetic models of antibody distribution and dynamics are useful for predicting and optimizing therapeutic behavior. Targeted antigens are produced and distributed in various tissues in specific patterns in disease phenotypes. Existing models leave out significant mechanistic detail which would enable an understanding of how to modify therapeutics in an optimal manner to allow appropriate tissue penetration in either a healthy or diseased state. The model presented here incorporates additional complexity such as diffusion through endothelial barriers, differential transcytosis properties, FcRn-mediated recycling, and incorporates these properties in an organ-specific manner. This creates a platform which can be expanded upon to include understanding of the effect of target on therapeutic distribution and clearance, differences in dynamics during a diseased versus healthy state, differential dose strategies, and mechanistic translation between animal models and human disease state. This model represents a superior alternative to typical and potentially over-simplified scaling strategies utilized in most existing physiologically-based pharmacokinetic models. Ultimately, this will enable better therapeutic design and greater pharmacological effects.


Assuntos
Anticorpos/metabolismo , Modelos Teóricos , Farmacocinética
17.
Cancer Res ; 70(9): 3843-50, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20406974

RESUMO

Most non-small cell lung cancers (NSCLC) display elevated expression of epidermal growth factor receptor (EGFR), but response to EGFR kinase inhibitors is predominantly limited to NSCLC harboring EGFR-activating mutations. These mutations are associated with increased activity of survival pathways, including phosphatidylinositol 3-kinase/AKT and signal transducer and activator of transcription 3/5. We report that EGFR-activating mutations also surprisingly lead to decreased ability to activate extracellular signal-regulated kinase (ERK) compared with wild-type EGFR. In NSCLC cells and mouse embryonic fibroblasts expressing mutant EGFR, this effect on ERK correlates with decreased EGFR internalization and reduced phosphorylation of SHP2, a tyrosine phosphatase required for the full activation of ERK. We further show that ERK activation levels affect cellular response to gefitinib. NSCLC cells with EGFR mutation display reduced gefitinib sensitivity when ERK activation is augmented by expression of constitutively active mutants of mitogen-activated protein kinase/ERK kinase (MEK). Conversely, in a NSCLC cell line expressing wild-type EGFR, gefitinib treatment along with or following MEK inhibition increases death response compared with treatment with gefitinib alone. Our results show that EGFR-activating mutations may promote some survival pathways but simultaneously impair others. This multivariate alteration of the network governing cellular response to gefitinib, which we term "oncogene imbalance," portends a potentially broader ability to treat gefitinib-resistant NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Quinazolinas/farmacologia , Animais , Antineoplásicos/farmacologia , Butadienos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Receptores ErbB/biossíntese , Receptores ErbB/metabolismo , Gefitinibe , Células HeLa , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Camundongos , Mutação , Nitrilas/farmacologia , Fosforilação
18.
Mol Syst Biol ; 5: 239, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19156131

RESUMO

The ErbB signaling pathways, which regulate diverse physiological responses such as cell survival, proliferation and motility, have been subjected to extensive molecular analysis. Nonetheless, it remains poorly understood how different ligands induce different responses and how this is affected by oncogenic mutations. To quantify signal flow through ErbB-activated pathways we have constructed, trained and analyzed a mass action model of immediate-early signaling involving ErbB1-4 receptors (EGFR, HER2/Neu2, ErbB3 and ErbB4), and the MAPK and PI3K/Akt cascades. We find that parameter sensitivity is strongly dependent on the feature (e.g. ERK or Akt activation) or condition (e.g. EGF or heregulin stimulation) under examination and that this context dependence is informative with respect to mechanisms of signal propagation. Modeling predicts log-linear amplification so that significant ERK and Akt activation is observed at ligand concentrations far below the K(d) for receptor binding. However, MAPK and Akt modules isolated from the ErbB model continue to exhibit switch-like responses. Thus, key system-wide features of ErbB signaling arise from nonlinear interaction among signaling elements, the properties of which appear quite different in context and in isolation.


Assuntos
Proteínas Oncogênicas v-erbB/metabolismo , Transdução de Sinais , Cinética , Sistema de Sinalização das MAP Quinases , Sensibilidade e Especificidade
19.
Eur J Immunol ; 37(8): 2290-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17628859

RESUMO

We developed IgH-transgenic rabbits carrying a productive VDJ-Cmu Tg and found the rabbits were B cell-deficient, with a 50-100% reduction in serum IgM and IgG levels. The bone marrow of newborn Tg rabbits contained severely reduced levels of preB cells and almost no B cells. The few preB cells present in the bone marrow were large, cycling cells that expressed the VDJ-Cmu Tg, indicating that the block in B cell development likely occurred at or before the transition from large (early) preB to small (late) preB cells. By immunoprecipitation, the Tg mu-chain paired with VpreB and lambda5, suggesting that the B cell deficiency is not due to an inability to form a preB cell receptor. Despite the block in B cell development, a few B cells, expressing predominantly endogenous mu-chains, began the second stage of development in GALT. B cells were localized in and beneath the follicle-associated epithelium of GALT prior to B cell follicle formation, suggesting to us that B cell follicle formation is initiated near the follicle-associated epithelium, possibly through contact with intestinal microbiota. These IgH-Tg rabbits should provide a useful model for studies of B cell development both in bone marrow and in GALT.


Assuntos
Animais Geneticamente Modificados , Linfócitos B/imunologia , Genes de Cadeia Pesada de Imunoglobulina , Intestinos/imunologia , Coelhos/imunologia , Sequência de Aminoácidos , Animais , Linfócitos B/citologia , Diferenciação Celular/imunologia , Citometria de Fluxo , Imunofluorescência , Imunidade nas Mucosas , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Cadeias mu de Imunoglobulina/genética , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Éxons VDJ
20.
J Exp Med ; 201(1): 55-62, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15623575

RESUMO

Gut-associated lymphoid tissues (GALTs) interact with intestinal microflora to drive GALT development and diversify the primary antibody repertoire; however, the molecular mechanisms that link these events remain elusive. Alicia rabbits provide an excellent model to investigate the relationship between GALT, intestinal microflora, and modulation of the antibody repertoire. Most B cells in neonatal Alicia rabbits express V(H)n allotype immunoglobulin (Ig)M. Within weeks, the number of V(H)n B cells decreases, whereas V(H)a allotype B cells increase in number and become predominant. We hypothesized that the repertoire shift from V(H)n to V(H)a B cells results from interactions between GALT and intestinal microflora. To test this hypothesis, we surgically removed organized GALT from newborn Alicia pups and ligated the appendix to sequester it from intestinal microflora. Flow cytometry and nucleotide sequence analyses revealed that the V(H)n to V(H)a repertoire shift did not occur, demonstrating the requirement for interactions between GALT and intestinal microflora in the selective expansion of V(H)a B cells. By comparing amino acid sequences of V(H)n and V(H)a Ig, we identified a putative V(H) ligand binding site for a bacterial or endogenous B cell superantigen. We propose that interaction of such a superantigen with V(H)a B cells results in their selective expansion.


Assuntos
Linfócitos B/imunologia , Alótipos de Imunoglobulina/metabolismo , Switching de Imunoglobulina/genética , Imunoglobulina M/metabolismo , Região Variável de Imunoglobulina/genética , Tecido Linfoide/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Cálcio/metabolismo , Clonagem Molecular , DNA Complementar/genética , Citometria de Fluxo , Imunofluorescência , Genes de Imunoglobulinas/genética , Alótipos de Imunoglobulina/imunologia , Switching de Imunoglobulina/imunologia , Imunoglobulina M/imunologia , Região Variável de Imunoglobulina/biossíntese , Tecido Linfoide/microbiologia , Modelos Moleculares , Dados de Sequência Molecular , Coelhos , Análise de Sequência de DNA , Superantígenos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA