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-38532525

RESUMO

Trastuzumab deruxtecan (T-DXd; DS-8201; ENHERTU®) is a human epithelial growth factor receptor 2 (HER2)-directed antibody drug conjugate (ADC) with demonstrated antitumor activity against a range of tumor types. Aiming to understand the relationship between antigen expression and downstream efficacy outcomes, T-DXd was administered in tumor-bearing mice carrying NCI-N87, Capan-1, JIMT-1, and MDA-MB-468 xenografts, characterized by varying HER2 levels. Plasma pharmacokinetics (PK) of total antibody, T-DXd, and released DXd and tumor concentrations of released DXd were evaluated, in addition to monitoring γΗ2AX and pRAD50 pharmacodynamic (PD) response. A positive relationship was observed between released DXd concentrations in tumor and HER2 expression, with NCI-N87 xenografts characterized by the highest exposures compared to the remaining cell lines. γΗ2AX and pRAD50 demonstrated a sustained increase over several days occurring with a time delay relative to tumoral-released DXd concentrations. In vitro investigations of cell-based DXd disposition facilitated the characterization of DXd kinetics across tumor cells. These outputs were incorporated into a mechanistic mathematical model, utilized to describe PK/PD trends. The model captured plasma PK across dosing arms as well as tumor PK in NCI-N87, Capan-1, and MDA-MB-468 models; tumor concentrations in JIMT-1 xenografts required additional parameter adjustments reflective of complex receptor dynamics. γΗ2AX longitudinal trends were well characterized via a unified PD model implemented across xenografts demonstrating the robustness of measured PD trends. This work supports the application of a mechanistic model as a quantitative tool, reliably projecting tumor payload concentrations upon T-DXd administration, as the first step towards preclinical-to-clinical translation.

2.
J Med Chem ; 66(13): 8782-8807, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37343272

RESUMO

Recent clinical reports have highlighted the need for wild-type (WT) and mutant dual inhibitors of c-MET kinase for the treatment of cancer. We report herein a novel chemical series of ATP competitive type-III inhibitors of WT and D1228V mutant c-MET. Using a combination of structure-based drug design and computational analyses, ligand 2 was optimized to a highly selective chemical series with nanomolar activities in biochemical and cellular settings. Representatives of the series demonstrate excellent pharmacokinetic profiles in rat in vivo studies with promising free-brain exposures, paving the way for the design of brain permeable drugs for the treatment of c-MET driven cancers.


Assuntos
Antineoplásicos , Neoplasias , Ratos , Animais , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met , Desenho de Fármacos , Trifosfato de Adenosina , Antineoplásicos/farmacologia
3.
AAPS J ; 25(4): 66, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37380821

RESUMO

Capturing human equivalent drug exposures preclinically is a key challenge in the translational process. Motivated by the need to recapitulate the pharmacokinetic (PK) profile of the clinical stage Mcl-1 inhibitor AZD5991 in mice, we describe the methodology used to develop a refined mathematical model relating clinically relevant concentration profiles to efficacy. Administration routes were explored to achieve target exposures matching the clinical exposure of AZD5991. Intravenous infusion using vascular access button (VAB) technology was found to best reproduce clinical target exposures of AZD5991 in mice. Exposure-efficacy relationships were evaluated, demonstrating that dissimilar PK profiles result in differences in target engagement and efficacy outcomes. Thus, these data underscore the importance of accurately ascribing key PK metrics in the translational process to enable clinically meaningful predictions of efficacy.


Assuntos
Compostos Macrocíclicos , Humanos , Animais , Camundongos , Modelos Animais de Doenças , Oncologia , Tecnologia
4.
PLoS One ; 18(5): e0285798, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37195991

RESUMO

The exposure of a dendritic nanoparticle and its conjugated active pharmaceutical ingredient (API) was determined in mouse, rat and dog, with the aim of investigating interspecies differences facilitating clinical translation. Plasma area under the curves (AUCs) were found to be dose proportional across species, while dose normalized concentration time course profiles in plasma, liver and spleen were superimposable in mouse, rat and dog. A physiologically based pharmacokinetic (PBPK) model, previously developed for mouse, was evaluated as a suitable framework to prospectively capture concentration dynamics in rat and dog. The PBPK model, parameterized either by considering species-specific physiology or using alternate scaling methods such as allometry, was shown to capture exposure profiles across species. A sensitivity analysis highlighted API systemic clearance as a key parameter influencing released API levels. The PBPK model was utilized to simulate human exposure profiles, which overlaid dose-normalized data from mouse, rat and dog. The consistency in measured interspecies exposures as well as the capability of the PBPK model to simulate observed dynamics support its use as a powerful translational tool.


Assuntos
Modelos Biológicos , Nanopartículas , Ratos , Camundongos , Humanos , Animais , Cães , Distribuição Tecidual , Área Sob a Curva , Fígado
5.
Xenobiotica ; 52(8): 770-785, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36314242

RESUMO

 The therapeutic concept of antibody drug conjugates (ADCs) is to selectively target tumour cells with small molecule cytotoxic drugs to maximise cell kill benefit and minimise healthy tissue toxicity.An ADC generally consists of an antibody that targets a protein on the surface of tumour cells chemically linked to a warhead small molecule cytotoxic drug.To deliver the warhead to the tumour cell, the antibody must bind to the target protein and in general be internalised into the cell. Following internalisation, the cytotoxic agent can be released in the endosomal or lysosomal compartment (via different mechanisms). Diffusion or transport out of the endosome or lysosome allows the cytotoxic drug to express its cell-killing pharmacology. Alternatively, some ADCs (e.g. EDB-ADCs) rely on extracellular cleavage releasing membrane permeable warheads.One potentially important aspect of the ADC mechanism is the 'bystander effect' whereby the cytotoxic drug released in the targeted cell can diffuse out of that cell and into other (non-target expressing) tumour cells to exert its cytotoxic effect. This is important as solid tumours tend to be heterogeneous and not all cells in a tumour will express the targeted protein.The combination of large and small molecule aspects in an ADC poses significant challenges to the disposition scientist in describing the ADME properties of the entire molecule.This article will review the ADC landscape and the ADME properties of successful ADCs, with the aim of outlining best practice and providing a perspective of how the field can further facilitate the discovery and development of these important therapeutic modalities.


Assuntos
Antineoplásicos , Imunoconjugados , Neoplasias , Humanos , Antineoplásicos/farmacocinética , Imunoconjugados/farmacocinética , Neoplasias/tratamento farmacológico
6.
Mol Pharm ; 19(1): 172-187, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34890209

RESUMO

A physiologically based pharmacokinetic model was developed to describe the tissue distribution kinetics of a dendritic nanoparticle and its conjugated active pharmaceutical ingredient (API) in plasma, liver, spleen, and tumors. Tumor growth data from MV-4-11 tumor-bearing mice were incorporated to investigate the exposure/efficacy relationship. The nanoparticle demonstrated improved antitumor activity compared to the conventional API formulation, owing to the extended released API concentrations at the site of action. Model simulations further enabled the identification of critical parameters that influence API exposure in tumors and downstream efficacy outcomes upon nanoparticle administration. The model was utilized to explore a range of dosing schedules and their effect on tumor growth kinetics, demonstrating the improved antitumor activity of nanoparticles with less frequent dosing compared to the same dose of naked APIs in conventional formulations.


Assuntos
Antineoplásicos/administração & dosagem , Dendrímeros/farmacocinética , Nanopartículas/metabolismo , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos SCID , Transplante de Neoplasias , Distribuição Tecidual , Resultado do Tratamento
7.
PLoS One ; 10(3): e0118977, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25786126

RESUMO

Antibody drug conjugates (ADCs) represent novel anti-cancer modalities engineered to specifically target and kill tumor cells expressing corresponding antigens. Due to their large size and their complex kinetics, these therapeutic agents often face heterogeneous distributions in tumors, leading to large untargeted regions that escape therapy. We present a modeling framework which includes the systemic distribution, vascular permeability, interstitial transport, as well as binding and payload release kinetics of ADC-therapeutic agents in mouse xenografts. We focused, in particular, on receptor dynamics such as endocytic trafficking mechanisms within cancer cells, to simulate their impact on tumor mass shrinkage upon ADC administration. Our model identified undesirable tumor properties that can impair ADC tissue homogeneity, further compromising ADC success, and explored ADC design optimization scenarios to counteract upon such unfavorable intrinsic tumor tissue attributes. We further demonstrated the profound impact of cytotoxic payload release mechanisms and the role of bystander killing effects on tumor shrinkage. This model platform affords a customizable simulation environment which can aid with experimental data interpretation and the design of ADC therapeutic treatments.


Assuntos
Desenho de Fármacos , Imunoconjugados/uso terapêutico , Modelos Biológicos , Neoplasias/tratamento farmacológico , Animais , Transformação Celular Neoplásica , Humanos , Imunoconjugados/metabolismo , Cinética , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Theor Biol ; 288: 44-56, 2011 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-21871462

RESUMO

We developed a multicellular model of the mammalian circadian clock characterized by a high degree of heterogeneity with respect to single cell periodicity and behavior (intrinsic and driven oscillators), neurotransmitter release (VIP, GABA and glutamate synthesis) and spatial organization (core and shell regions), mimicking structural patterns within the suprachiasmatic nucleus (SCN) associated with distinct circadian functions. We simulated the SCN core and shell separately utilizing experimentally derived connectivity schemes for the two subdivisions as observed within the rat SCN. The core was modeled via a small world network characterized by VIP and GABA co-localization, whereas the shell was simulated as a nearest neighbor network promoting local GABAergic connections. To study the function of the axonal plexus extending from the densely innervated ventrolateral region to distal areas across the dorsomedial SCN, directed long range links from the core to the shell were gradually introduced via a probability p(cs) that ranged from 0 to 1. A probability value of 0 excluded core-shell interactions, whereas p(cs)=1 achieved maximal connectivity between the two regions. Our model exhibited a threshold in the number of core-to-shell links required for sufficient cell-to-cell coordination to maintain periodicity and rhythmic behavior across the entire model network (including both shell and core populations) in constant darkness as well as 12:12h light-dark cycles. By contrast, constant light was shown to increase phase synchronization across the shell while core populations remained poorly synchronized, suggesting differential light response across the two SCN compartments. We further simulated increasing percentages of intrinsic oscillators and demonstrated a negative correlation between the number of intrinsic oscillators distributed across the SCN and the ability of the system to produce synchronized signals. Simulations that differed with respect to the placement of intrinsic oscillators supported the hypothesis that improved synchronization is achieved with networks characterized by localized intrinsic oscillators placed exclusively within the shell versus networks containing uniformly distributed intrinsic oscillators in both SCN compartments. This study has successfully reproduced a number of spatiotemporal and behavioral attributes of the SCN, providing a useful computational tool to correlate observed circadian phenotypes with distinct chemoarchitectural properties of spatially localized neural populations.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Modelos Biológicos , Núcleo Supraquiasmático/fisiologia , Animais , Escuridão , Neurotransmissores/fisiologia , Estimulação Luminosa/métodos , Transdução de Sinais/fisiologia , Núcleo Supraquiasmático/citologia , Biologia de Sistemas/métodos
9.
Biophys J ; 101(1): 12-20, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21723810

RESUMO

We developed a multicellular model characterized by a high degree of heterogeneity to investigate possible mechanisms that underlie circadian network synchronization and rhythmicity in the suprachiasmatic nucleus (SCN). We populated a two-dimensional grid with 400 model neurons coupled via γ-aminobutyric acid (GABA) and vasoactive intestinal polypeptide (VIP) neurotransmitters through a putative Ca(2+) mediated signaling cascade to investigate their roles in gene expression and electrical firing activity of cell populations. As observed experimentally, our model predicted that GABA would affect the amplitude of circadian oscillations but not synchrony among individual oscillators. Our model recapitulated experimental findings of decreased synchrony and average periods, loss of rhythmicity, and reduced circadian amplitudes as VIP signaling was eliminated. In addition, simulated increases of VIP reduced periodicity and synchrony. We therefore postulated a physiological range of VIP within which the system is able to produce sustained and synchronized oscillations. Our model recapitulated experimental findings of diminished amplitudes and periodicity with decreasing intracellular Ca(2+) concentrations, suggesting that such behavior could be due to simultaneous decrease of individual oscillation amplitudes and population synchrony. Simulated increases in Cl(-) levels resulted in increased Cl(-) influx into the cytosol, a decrease of inhibitory postsynaptic currents, and ultimately a shift of GABA-elicited responses from inhibitory to excitatory. The simultaneous reduction of IPSCs and increase in membrane resting potential produced GABA dose-dependent increases in firing rates across the population, as has been observed experimentally. By integrating circadian gene regulation and electrophysiology with intracellular and intercellular signaling, we were able to develop the first (to our knowledge) multicellular model that allows the effects of clock genes, electrical firing, Ca(2+), GABA, and VIP on circadian system behavior to be predicted.


Assuntos
Ritmo Circadiano/fisiologia , Espaço Extracelular/fisiologia , Modelos Biológicos , Rede Nervosa/fisiologia , Cálcio/metabolismo , Cloretos/metabolismo , Citosol/metabolismo , Espaço Intracelular/metabolismo , Neurônios/fisiologia , Transdução de Sinais , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Ácido gama-Aminobutírico/metabolismo
10.
PLoS Comput Biol ; 6(3): e1000706, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20300645

RESUMO

The suprachiasmatic nucleus (SCN) of the hypothalamus is a multicellular system that drives daily rhythms in mammalian behavior and physiology. Although the gene regulatory network that produces daily oscillations within individual neurons is well characterized, less is known about the electrophysiology of the SCN cells and how firing rate correlates with circadian gene expression. We developed a firing rate code model to incorporate known electrophysiological properties of SCN pacemaker cells, including circadian dependent changes in membrane voltage and ion conductances. Calcium dynamics were included in the model as the putative link between electrical firing and gene expression. Individual ion currents exhibited oscillatory patterns matching experimental data both in current levels and phase relationships. VIP and GABA neurotransmitters, which encode synaptic signals across the SCN, were found to play critical roles in daily oscillations of membrane excitability and gene expression. Blocking various mechanisms of intracellular calcium accumulation by simulated pharmacological agents (nimodipine, IP3- and ryanodine-blockers) reproduced experimentally observed trends in firing rate dynamics and core-clock gene transcription. The intracellular calcium concentration was shown to regulate diverse circadian processes such as firing frequency, gene expression and system periodicity. The model predicted a direct relationship between firing frequency and gene expression amplitudes, demonstrated the importance of intracellular pathways for single cell behavior and provided a novel multiscale framework which captured characteristics of the SCN at both the electrophysiological and gene regulatory levels.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Modelos Neurológicos , Rede Nervosa/fisiologia , Neurônios/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Simulação por Computador , Humanos
11.
J Biol Rhythms ; 24(3): 243-54, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19465701

RESUMO

The suprachiasmatic nucleus (SCN) of the hypothalamus is a multioscillator system that drives daily rhythms in mammalian behavior and physiology. Based on recent data implicating vasoactive intestinal polypeptide (VIP) as the key intercellular synchronizing agent, we developed a multicellular SCN model to investigate the effects of cellular heterogeneity and intercellular connectivity on circadian behavior. A 2-dimensional grid was populated with 400 model cells that were heterogeneous with respect to their uncoupled rhythmic behavior (intrinsic and damped pacemakers with a range of oscillation periods) and VIP release characteristics (VIP producers and nonproducers). We constructed small-world network architectures in which local connections between VIP producing cells and their 4 nearest neighbors were augmented with random connections, resulting in long-range coupling across the grid. With only 10% of the total possible connections, the small-world network model was able to produce similar phase synchronization indices as a mean-field model with VIP producing cells connected to all other cells. Partial removal of random connections decreased the synchrony among neurons, the amplitude of VIP and cAMP response element binding protein oscillations, the mean period of intrinsic periods across the population, and the percentage of oscillating cells. These results indicate that small-world connectivity provides the optimal compromise between the number of connections and control of circadian amplitude and synchrony. This model predicts that small decreases in long-range VIP connections in the SCN could have dramatic effects on period and amplitude of daily rhythms, features commonly described with aging.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Modelos Neurológicos , Rede Nervosa/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Mamíferos/metabolismo , Rede Nervosa/anatomia & histologia , Transdução de Sinais/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...