Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Mol Divers ; 27(4): 1675-1687, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36219381

RESUMEN

Optimizing the pharmacokinetics (PK) of a drug candidate to support oral dosing is a key challenge in drug development. PK parameters are usually estimated from the concentration-time profile following intravenous administration; however, traditional methods are time-consuming and expensive. In recent years, quantitative structure-pharmacokinetic relationship (QSPKR), an in silico tool that aims to develop a mathematical relationship between the structure of a molecule and its PK properties, has emerged as a useful alternative to experimental testing. Due to the complex nature of the various processes involved in dictating the fate of a drug, the development of adequate QSPKR models that can be used in real-world pre-screening situations has proved challenging. Given the crucial role played by a molecule's ionization state in determining its PK properties, this work aims to build predictive QSPKR models for PK parameters in humans using an ionization state-based strategy. We divide a high-quality dataset into clusters based on ionization state at physiological pH and build global and ion subset-based 'local' models for three major PK parameters: plasma clearance (CL), steady-state volume of distribution (VDss), and half-life (t1/2). We use a robust methodology developed in our lab entitled 'EigenValue ANalySis' that accounts for the stereospecificity in drug disposition and use the support vector machine algorithm for model building. Our findings suggest that categorizing compounds in accordance with ionization state does not result in improved QSPKR models. The narrow ranges in the endpoints along with redundancies in the data adversely affect the ion subset-based QSPKR models. We suggest alternative approaches such as elimination route-based models that account for drug-transporter interactions for CL and chemotype-specific QSPKR for VDss.


Asunto(s)
Algoritmos , Relación Estructura-Actividad Cuantitativa , Humanos , Preparaciones Farmacéuticas , Modelos Biológicos
2.
J Pharmacokinet Pharmacodyn ; 48(5): 743-762, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34146191

RESUMEN

In the past, our lab proposed a two-pore PBPK model for different-size protein therapeutics using de novo derived parameters and the model was validated using plasma PK data of different-size antibody fragments digitized from the literature (Li Z, Shah DK, J Pharmacokinet Pharmacodynam 46(3):305-318, 2009). To further validate the model using tissue distribution data, whole-body biodistribution study of 6 different-size proteins in mice were conducted. Studied molecules covered a wide MW range (13-150 kDa). Plasma PK and tissue distribution profiles is 9 tissues were measured, including heart, lung, liver, spleen, kidney, skin, muscle, small intestine, large intestine. Tumor exposure of different-size proteins were also evaluated. The PBPK model was validated by comparing percentage predictive errors (%PE) between observed and model predicted results for each type of molecule in each tissue. Model validation showed that the two-pore PBPK model was able to predict plasma, tissues and tumor PK of all studied molecules relatively well. This model could serve as a platform for developing a generic PBPK model for protein therapeutics in the future.


Asunto(s)
Distribución Tisular/fisiología , Trastuzumab/farmacocinética , Animales , Anticuerpos Monoclonales/farmacocinética , Línea Celular Tumoral , Humanos , Ratones , Ratones Desnudos , Modelos Biológicos , Neoplasias/metabolismo
3.
J Pharmacokinet Pharmacodyn ; 46(3): 305-318, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31028591

RESUMEN

Two-pore PBPK models have been used for characterizing the PK of protein therapeutics since 1990s. However, widespread utilization of these models is hampered by the lack of a priori parameter values, which are typically estimated using the observed data. To overcome this hurdle, here we have presented the development of a two-pore PBPK model using de novo derived parameters. The PBPK model was validated using plasma PK data for different size proteins in mice. Using the "two pore theory" we were able to establish the relationship between protein size and key model parameters, such as: permeability-surface area product (PS), vascular reflection coefficient (σ), peclet number (Pe), and glomerular sieving coefficient (θ). The model accounted for size dependent changes in tissue extravasation and glomerular filtration. The model was able to a priori predict the PK of 8 different proteins: IgG (150 kDa), scFv-Fc (105 kDa), F(ab)2 (100 kDa, minibody (80 kDa), scFv2 (55 kDa), Fab (50 kDa), diabody (50 kDa), scFv (27 kDa), and nanobody (13 kDa). In addition, the model was able to provide unprecedented quantitative insight into the relative contribution of convective and diffusive pathway towards trans-capillary mass transportation of different size proteins. The two-pore PBPK model was also able to predict systemic clearance (CL) versus Molecular Weight relationship for different size proteins reasonably well. As such, the PBPK model proposed here represents a bottom-up systems PK model for protein therapeutics, which can serve as a generalized platform for the development of truly translational PBPK model for protein therapeutics.


Asunto(s)
Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/farmacocinética , Proteínas/farmacocinética , Distribución Tisular/fisiología , Animales , Humanos , Cinética , Ratones , Modelos Biológicos
4.
Pharm Res ; 34(10): 2131-2141, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28681164

RESUMEN

PURPOSE: To establish a continuous relationship between the size of various antibody fragments and their systemic clearance (CL) in mice. METHODS: Two different orthogonal approaches have been used to establish the relationship. First approach uses CL values estimated by non-compartmental analysis (NCA) to establish a correlation with protein size. The second approach simultaneously characterizes the PK data for all the proteins using a 2-compartment model to establish a relationship between protein size and pharmacokinetic (PK) parameters. RESULTS: Simple mathematical functions (e.g. sigmoidal, power law) were able to characterize the CL vs. protein size relationship generated using the investigated proteins. The relationship established in mouse was used to predict rat, rabbit, monkey, and human relationships using allometric scaling. The predicted relationships were found to capture the available spares data from each species reasonably well. CONCLUSIONS: The CL vs. protein size relationship is important for establishing a robust quantitative structure-PK relationship (QSPKR) for protein therapeutics. The relationship presented here can help in a priori predicting plasma exposure of therapeutic proteins, and together with our previously established relationship between plasma and tissue concentrations of proteins, it can predict the tissue exposure of non-binding proteins simply based on molecular weight/radius and dose.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Fragmentos de Inmunoglobulinas/farmacología , Modelos Biológicos , Animales , Anticuerpos Monoclonales/química , Haplorrinos , Humanos , Fragmentos de Inmunoglobulinas/química , Cinética , Ratones , Estructura Molecular , Peso Molecular , Conejos , Ratas , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
5.
AAPS J ; 25(3): 48, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37118220

RESUMEN

Motivated by a series of work demonstrating the effect of molecular charge on antibody pharmacokinetics (PK), physiological-based pharmacokinetic (PBPK) models are emerging that relate in silico calculated charge or in vitro measures of polyspecificity to antibody PK parameters. However, only plasma data has been used for model development in these studies, leading to unvalidated assumptions. Here, we present an extended platform PBPK model for antibodies that incorporate charge-dependent endothelial cell pinocytosis rate and nonspecific off-target binding in the interstitial space and on circulating blood cells, to simultaneously characterize whole-body disposition of three antibody charge variants. Predictive potential of various charge metrics was also explored, and the difference between positive charge patches and negative charge patches (i.e., PPC-PNC) was used as the charge parameter to establish quantitative relationships with nonspecific binding affinities and endothelial cell uptake rate. Whole-body disposition of these charge variants was captured well by the model, with less than 2-fold predictive error in area under the curve of most plasma and tissue PK data. The model also predicted that with greater positive charge, nonspecific binding was more substantial, and pinocytosis rate increased especially in brain, heart, kidney, liver, lung, and spleen, but remained unchanged in adipose, bone, muscle, and skin. The presented PBPK model contributes to our understanding of the mechanisms governing the disposition of charged antibodies and can be used as a platform to guide charge engineering based on desired plasma and tissue exposures.


Asunto(s)
Anticuerpos Monoclonales , Hígado , Hígado/metabolismo , Modelos Biológicos
6.
MAbs ; 8(1): 113-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26496429

RESUMEN

Biodistribution coefficients (BC) allow estimation of the tissue concentrations of proteins based on the plasma pharmacokinetics. We have previously established the BC values for monoclonal antibodies. Here, this concept is extended by development of a relationship between protein size and BC values. The relationship was built by deriving the BC values for various antibody fragments of known molecular weight from published biodistribution studies. We found that there exists a simple exponential relationship between molecular weight and BC values that allows the prediction of tissue distribution of proteins based on molecular weight alone. The relationship was validated by a priori predicting BC values of 4 antibody fragments that were not used in building the relationship. The relationship was also used to derive BC50 values for all the tissues, which is the molecular weight increase that would result in 50% reduction in tissue uptake of a protein. The BC50 values for most tissues were found to be ~35 kDa. An ability to estimate tissue distribution of antibody fragments based on the BC vs. molecular size relationship established here may allow better understanding of the biologics concentrations in tissues responsible for efficacy or toxicity. This relationship can also be applied for rational development of new biotherapeutic modalities with optimal biodistribution properties to target (or avoid) specific tissues.


Asunto(s)
Fragmentos de Inmunoglobulinas/farmacología , Modelos Biológicos , Animales , Humanos , Peso Molecular , Distribución Tisular
7.
J Cheminform ; 7: 6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25767566

RESUMEN

BACKGROUND: Volume of distribution is an important pharmacokinetic property that indicates the extent of a drug's distribution in the body tissues. This paper addresses the problem of how to estimate the apparent volume of distribution at steady state (Vss) of chemical compounds in the human body using decision tree-based regression methods from the area of data mining (or machine learning). Hence, the pros and cons of several different types of decision tree-based regression methods have been discussed. The regression methods predict Vss using, as predictive features, both the compounds' molecular descriptors and the compounds' tissue:plasma partition coefficients (Kt:p) - often used in physiologically-based pharmacokinetics. Therefore, this work has assessed whether the data mining-based prediction of Vss can be made more accurate by using as input not only the compounds' molecular descriptors but also (a subset of) their predicted Kt:p values. RESULTS: Comparison of the models that used only molecular descriptors, in particular, the Bagging decision tree (mean fold error of 2.33), with those employing predicted Kt:p values in addition to the molecular descriptors, such as the Bagging decision tree using adipose Kt:p (mean fold error of 2.29), indicated that the use of predicted Kt:p values as descriptors may be beneficial for accurate prediction of Vss using decision trees if prior feature selection is applied. CONCLUSIONS: Decision tree based models presented in this work have an accuracy that is reasonable and similar to the accuracy of reported Vss inter-species extrapolations in the literature. The estimation of Vss for new compounds in drug discovery will benefit from methods that are able to integrate large and varied sources of data and flexible non-linear data mining methods such as decision trees, which can produce interpretable models. Graphical AbstractDecision trees for the prediction of tissue partition coefficient and volume of distribution of drugs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA