Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-37848637

RESUMO

Clinical studies have found there still exists a lack of gene therapy dose-toxicity and dose-efficacy data that causes gene therapy dose selection to remain elusive. Model informed drug development (MIDD) has become a standard tool implemented throughout the discovery, development, and approval of pharmaceutical therapies, and has the potential to inform dose-toxicity and dose-efficacy relationships to support gene therapy dose selection. Despite this potential, MIDD approaches for gene therapy remain immature and require standardization to be useful for gene therapy clinical programs. With the goal to advance MIDD approaches for gene therapy, in this review we first provide an overview of gene therapy types and how they differ from a bioanalytical, formulation, route of administration, and regulatory standpoint. With this biological and regulatory background, we propose how MIDD can be advanced for AAV-based gene therapies by utilizing physiological based pharmacokinetic modeling and quantitative systems pharmacology to holistically inform AAV and target protein dynamics following dosing. We discuss how this proposed model, allowing for in-depth exploration of AAV pharmacology, could be the key the field needs to treat these unmet disease populations.

2.
CPT Pharmacometrics Syst Pharmacol ; 12(11): 1726-1737, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36710368

RESUMO

Bispecific antibodies exhibit proven clinical benefit, and many bispecifics are currently in clinical development for oncology. Cytokine release syndrome (CRS) is a common clinical adverse effect observed following CD3-based bispecific dosing. However, the pathophysiology of CRS is not fully understood, and no computational model mechanistically describing clinical cytokine dynamics following bispecific dosing in solid tumors exists. Here, a quantitative systems pharmacology (QSP) model describing peripheral clinical cytokine dynamics following bispecific dosing in solid tumors is presented. Using tebentafusp as a case study, a CD3-bispecific approved for uveal melanoma, the model successfully captures the dynamics of five cytokines. The QSP model was shown to predict observed phenomena, such as cytokine maximum concentration suppression using step-up dosing regimens and the importance of on-target off-tumor binding toward CRS and toxicity. Furthermore, the QSP model provides rationale for these biological phenomena based on dynamics of immune cell activation and desensitization in tumors and healthy tissues. Overall, the QSP model structure presented here serves as a basis to infer cytokine dynamics for other CD3-based bispecifics or tumor types by altering model parameters to capture the scenario of interest, supporting applications including dose selection, candidate nomination, and disease area selection.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Humanos , Citocinas , Neoplasias/tratamento farmacológico
3.
Clin Transl Sci ; 14(1): 395-404, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33073529

RESUMO

Antibody drug conjugates (ADCs) provide targeted delivery of cytotoxic agents directly inside tumor cells. However, many ADCs targeting solid tumors have exhibited limited clinical efficacy, in part, due to insufficient penetration within tumors. To better understand the relationship between ADC tumor penetration and efficacy, previously applied Krogh cylinder models that explore tumor growth dynamics following ADC administration in preclinical species were expanded to a clinical framework by integrating clinical pharmacokinetics, tumor penetration, and tumor growth inhibition. The objective of this framework is to link ADC tumor penetration and distribution to clinical efficacy. The model was validated by comparing virtual patient population simulations to observed overall response rates from trastuzumab-DM1 treated patients with metastatic breast cancer. To capture clinical outcomes, we expanded upon previous Krogh cylinder models to include the additional mechanism of heterogeneous tumor growth inhibition spatially across the tumor. This expansion mechanistically captures clinical response rates by describing heterogeneous ADC binding and tumor cell killing; high binding and tumor cell death close to capillaries vs. low binding, and high tumor cell proliferation far from capillaries. Sensitivity analyses suggest that clinical efficacy could be optimized through dose fractionation, and that clinical efficacy is primarily dependent on the ADC-target affinity, payload potency, and tumor growth rate. This work offers a mechanistic basis to predict and optimize ADC clinical efficacy for solid tumors, allowing dosing strategy optimization to improve patient outcomes.


Assuntos
Ado-Trastuzumab Emtansina/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Imunoconjugados/farmacocinética , Modelos Biológicos , Ado-Trastuzumab Emtansina/administração & dosagem , Antineoplásicos/administração & dosagem , Área Sob a Curva , Mama/patologia , Neoplasias da Mama/patologia , Feminino , Humanos , Imunoconjugados/administração & dosagem , Distribuição Tecidual , Carga Tumoral
4.
Clin Pharmacol Ther ; 109(3): 605-618, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32686076

RESUMO

Drug development in oncology commonly exploits the tools of molecular biology to gain therapeutic benefit through reprograming of cellular responses. In immuno-oncology (IO) the aim is to direct the patient's own immune system to fight cancer. After remarkable successes of antibodies targeting PD1/PD-L1 and CTLA4 receptors in targeted patient populations, the focus of further development has shifted toward combination therapies. However, the current drug-development approach of exploiting a vast number of possible combination targets and dosing regimens has proven to be challenging and is arguably inefficient. In particular, the unprecedented number of clinical trials testing different combinations may no longer be sustainable by the population of available patients. Further development in IO requires a step change in selection and validation of candidate therapies to decrease development attrition rate and limit the number of clinical trials. Quantitative systems pharmacology (QSP) proposes to tackle this challenge through mechanistic modeling and simulation. Compounds' pharmacokinetics, target binding, and mechanisms of action as well as existing knowledge on the underlying tumor and immune system biology are described by quantitative, dynamic models aiming to predict clinical results for novel combinations. Here, we review the current QSP approaches, the legacy of mathematical models available to quantitative clinical pharmacologists describing interaction between tumor and immune system, and the recent development of IO QSP platform models. We argue that QSP and virtual patients can be integrated as a new tool in existing IO drug development approaches to increase the efficiency and effectiveness of the search for novel combination therapies.


Assuntos
Alergia e Imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Desenvolvimento de Medicamentos , Inibidores de Checkpoint Imunológico/uso terapêutico , Oncologia , Simulação de Dinâmica Molecular , Neoplasias/tratamento farmacológico , Biologia de Sistemas , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Simulação por Computador , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/farmacocinética , Modelos Imunológicos , Terapia de Alvo Molecular , Neoplasias/imunologia , Neoplasias/metabolismo , Microambiente Tumoral
5.
Sci Rep ; 10(1): 11001, 2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32601287

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
NPJ Syst Biol Appl ; 4: 1, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29263797

RESUMO

The ability to control vascular endothelial growth factor (VEGF) signaling offers promising therapeutic potential for vascular diseases and cancer. Despite this promise, VEGF-targeted therapies are not clinically effective for many pathologies, such as breast cancer. VEGFR1 has recently emerged as a predictive biomarker for anti-VEGF efficacy, implying a functional VEGFR1 role beyond its classically defined decoy receptor status. Here we introduce a computational approach that accurately predicts cellular responses elicited via VEGFR1 signaling. Aligned with our model prediction, we show empirically that VEGFR1 promotes macrophage migration through PLCγ and PI3K pathways and promotes macrophage proliferation through a PLCγ pathway. These results provide new insight into the basic function of VEGFR1 signaling while offering a computational platform to quantify signaling of any receptor.

7.
Sci Rep ; 7(1): 16439, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29180757

RESUMO

Nearly all studies of angiogenesis have focused on uni-family ligand-receptor binding, e.g., VEGFs bind to VEGF receptors, PDGFs bind to PDGF receptors, etc. The discovery of VEGF-PDGFRs binding challenges this paradigm and calls for investigation of other ligand-receptor binding possibilities. We utilized surface plasmon resonance to identify and measure PDGF-to-VEGFR binding rates, establishing cut-offs for binding and non-binding interactions. We quantified the kinetics of the recent VEGF-A:PDGFRß interaction for the first time with KD = 340 pM. We discovered new PDGF:VEGFR2 interactions with PDGF-AA:R2 KD = 530 nM, PDGF-AB:R2 KD = 110 pM, PDGF-BB:R2 KD = 40 nM, and PDGF-CC:R2 KD = 70 pM. We computationally predict that cross-family PDGF binding could contribute up to 96% of VEGFR2 ligation in healthy conditions and in cancer. Together the identification, quantification, and simulation of these novel cross-family interactions posits new mechanisms for understanding anti-angiogenic drug resistance and presents an expanded role of growth factor signaling with significance in health and disease.


Assuntos
Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Células Endoteliais/metabolismo , Humanos , Cinética , Ligantes , Modelos Biológicos , Neoplasias/metabolismo , Ligação Proteica , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Reprodutibilidade dos Testes , Ressonância de Plasmônio de Superfície
8.
Integr Biol (Camb) ; 9(5): 464-484, 2017 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-28436498

RESUMO

Recently, intracellular receptor signaling has been identified as a key component mediating cell responses for various receptor tyrosine kinases (RTKs). However, the extent each endocytic compartment (endocytic vesicle, early endosome, recycling endosome, late endosome, lysosome and nucleus) contributes to receptor signaling has not been quantified. Furthermore, our understanding of endocytosis and receptor signaling is complicated by cell- or receptor-specific endocytosis mechanisms. Therefore, towards understanding the differential endocytic compartment signaling roles, and identifying how to achieve signal transduction control for RTKs, we delineate how endocytosis regulates RTK signaling. We achieve this via a meta-analysis across eight RTKs, integrating computational modeling with experimentally derived cell (compartment volume, trafficking kinetics and pH) and ligand-receptor (ligand/receptor concentration and interaction kinetics) physiology. Our simulations predict the abundance of signaling from eight RTKs, identifying the following hierarchy in RTK signaling: PDGFRß > IGFR1 > EGFR > PDGFRα > VEGFR1 > VEGFR2 > Tie2 > FGFR1. We find that endocytic vesicles are the primary cell signaling compartment; over 43% of total receptor signaling occurs within the endocytic vesicle compartment for these eight RTKs. Mechanistically, we found that high RTK signaling within endocytic vesicles may be attributed to their low volume (5.3 × 10-19 L) which facilitates an enriched ligand concentration (3.2 µM per ligand molecule within the endocytic vesicle). Under the analyzed physiological conditions, we identified extracellular ligand concentration as the most sensitive parameter to change; hence the most significant one to modify when regulating absolute compartment signaling. We also found that the late endosome and nucleus compartments are important contributors to receptor signaling, where 26% and 18%, respectively, of average receptor signaling occurs across the eight RTKs. Conversely, we found very low membrane-based receptor signaling, exhibiting <1% of the total receptor signaling for these eight RTKs. Moreover, we found that nuclear translocation, mechanistically, requires late endosomal transport; when we blocked receptor trafficking from late endosomes to the nucleus we found a 57% reduction in nuclear translocation. In summary, our research has elucidated the significance of endocytic vesicles, late endosomes and the nucleus in RTK signal propagation.


Assuntos
Modelos Biológicos , Receptores Proteína Tirosina Quinases/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Compartimento Celular , Núcleo Celular/enzimologia , Endocitose , Endossomos/enzimologia , Humanos , Cinética , Ligantes , Fosforilação , Transdução de Sinais , Vesículas Transportadoras/enzimologia
9.
Methods Mol Biol ; 1570: 117-138, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28238133

RESUMO

Nanosensor-based detection of biomarkers can improve medical diagnosis; however, a critical factor in nanosensor development is deciding which biomarker to target, as most diseases present several biomarkers. Biomarker-targeting decisions can be informed via an understanding of biomarker expression. Currently, immunohistochemistry (IHC) is the accepted standard for profiling biomarker expression. While IHC provides a relative mapping of biomarker expression, it does not provide cell-by-cell readouts of biomarker expression or absolute biomarker quantification. Flow cytometry overcomes both these IHC challenges by offering biomarker expression on a cell-by-cell basis, and when combined with calibration standards, providing quantitation of biomarker concentrations: this is known as qFlow cytometry. Here, we outline the key components for applying qFlow cytometry to detect biomarkers within the angiogenic vascular endothelial growth factor receptor family. The key aspects of the qFlow cytometry methodology include: antibody specificity testing, immunofluorescent cell labeling, saturation analysis, fluorescent microsphere calibration, and quantitative analysis of both ensemble and cell-by-cell data. Together, these methods enable high-throughput quantification of biomarker expression.


Assuntos
Biomarcadores , Técnicas Biossensoriais/métodos , Citometria de Fluxo/métodos , Ensaios de Triagem em Larga Escala , Receptores de Superfície Celular/metabolismo , Descoberta de Drogas/métodos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Software , Estatística como Assunto/métodos
10.
PLoS One ; 10(4): e0124575, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25897758

RESUMO

Development of many conditions and disorders, such as atherosclerosis and stroke, are dependent upon hemodynamic forces. To accurately predict and prevent these conditions and disorders hemodynamic forces must be properly mapped. Here we compare a shear-rate dependent fluid (SDF) constitutive model, based on the works by Yasuda et al in 1981, against a Newtonian model of blood. We verify our stabilized finite element numerical method with the benchmark lid-driven cavity flow problem. Numerical simulations show that the Newtonian model gives similar velocity profiles in the 2-dimensional cavity given different height and width dimensions, given the same Reynolds number. Conversely, the SDF model gave dissimilar velocity profiles, differing from the Newtonian velocity profiles by up to 25% in velocity magnitudes. This difference can affect estimation in platelet distribution within blood vessels or magnetic nanoparticle delivery. Wall shear stress (WSS) is an important quantity involved in vascular remodeling through integrin and adhesion molecule mechanotransduction. The SDF model gave a 7.3-fold greater WSS than the Newtonian model at the top of the 3-dimensional cavity. The SDF model gave a 37.7-fold greater WSS than the Newtonian model at artery walls located immediately after bifurcations in the idealized femoral artery tree. The pressure drop across arteries reveals arterial sections highly resistive to flow which correlates with stenosis formation. Numerical simulations give the pressure drop across the idealized femoral artery tree with the SDF model which is approximately 2.3-fold higher than with the Newtonian model. In atherosclerotic lesion models, the SDF model gives over 1 Pa higher WSS than the Newtonian model, a difference correlated with over twice as many adherent monocytes to endothelial cells from the Newtonian model compared to the SDF model.


Assuntos
Aterosclerose/fisiopatologia , Artéria Femoral/fisiopatologia , Mecanotransdução Celular , Modelos Cardiovasculares , Aterosclerose/metabolismo , Aterosclerose/patologia , Velocidade do Fluxo Sanguíneo , Plaquetas/metabolismo , Plaquetas/patologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Simulação por Computador , Sistemas de Liberação de Medicamentos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Expressão Gênica , Humanos , Integrinas/genética , Integrinas/metabolismo , Nanopartículas de Magnetita/química , Monócitos/metabolismo , Monócitos/patologia , Fluxo Pulsátil , Estresse Mecânico
11.
PLoS One ; 9(5): e97271, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24827582

RESUMO

Cell population heterogeneity can affect cellular response and is a major factor in drug resistance. However, there are few techniques available to represent and explore how heterogeneity is linked to population response. Recent high-throughput genomic, proteomic, and cellomic approaches offer opportunities for profiling heterogeneity on several scales. We have recently examined heterogeneity in vascular endothelial growth factor receptor (VEGFR) membrane localization in endothelial cells. We and others processed the heterogeneous data through ensemble averaging and integrated the data into computational models of anti-angiogenic drug effects in breast cancer. Here we show that additional modeling insight can be gained when cellular heterogeneity is considered. We present comprehensive statistical and computational methods for analyzing cellomic data sets and integrating them into deterministic models. We present a novel method for optimizing the fit of statistical distributions to heterogeneous data sets to preserve important data and exclude outliers. We compare methods of representing heterogeneous data and show methodology can affect model predictions up to 3.9-fold. We find that VEGF levels, a target for tuning angiogenesis, are more sensitive to VEGFR1 cell surface levels than VEGFR2; updating VEGFR1 levels in the tumor model gave a 64% change in free VEGF levels in the blood compartment, whereas updating VEGFR2 levels gave a 17% change. Furthermore, we find that subpopulations of tumor cells and tumor endothelial cells (tEC) expressing high levels of VEGFR (>35,000 VEGFR/cell) negate anti-VEGF treatments. We show that lowering the VEGFR membrane insertion rate for these subpopulations recovers the anti-angiogenic effect of anti-VEGF treatment, revealing new treatment targets for specific tumor cell subpopulations. This novel method of characterizing heterogeneous distributions shows for the first time how different representations of the same data set lead to different predictions of drug efficacy.


Assuntos
Membrana Celular/metabolismo , Receptores de Superfície Celular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Simulação por Computador , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...