Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
PLoS Comput Biol ; 18(7): e1010254, 2022 07.
Article in English | MEDLINE | ID: mdl-35867773

ABSTRACT

Quantitative systems pharmacology (QSP) models and spatial agent-based models (ABM) are powerful and efficient approaches for the analysis of biological systems and for clinical applications. Although QSP models are becoming essential in discovering predictive biomarkers and developing combination therapies through in silico virtual trials, they are inadequate to capture the spatial heterogeneity and randomness that characterize complex biological systems, and specifically the tumor microenvironment. Here, we extend our recently developed spatial QSP (spQSP) model to analyze tumor growth dynamics and its response to immunotherapy at different spatio-temporal scales. In the model, the tumor spatial dynamics is governed by the ABM, coupled to the QSP model, which includes the following compartments: central (blood system), tumor, tumor-draining lymph node, and peripheral (the rest of the organs and tissues). A dynamic recruitment of T cells and myeloid-derived suppressor cells (MDSC) from the QSP central compartment has been implemented as a function of the spatial distribution of cancer cells. The proposed QSP-ABM coupling methodology enables the spQSP model to perform as a coarse-grained model at the whole-tumor scale and as an agent-based model at the regions of interest (ROIs) scale. Thus, we exploit the spQSP model potential to characterize tumor growth, identify T cell hotspots, and perform qualitative and quantitative descriptions of cell density profiles at the invasive front of the tumor. Additionally, we analyze the effects of immunotherapy at both whole-tumor and ROI scales under different tumor growth and immune response conditions. A digital pathology computational analysis of triple-negative breast cancer specimens is used as a guide for modeling the immuno-architecture of the invasive front.


Subject(s)
Neoplasms , Pharmacology , Combined Modality Therapy , Humans , Immunotherapy/methods , Models, Biological , Neoplasms/therapy , Network Pharmacology , Pharmacology/methods , Tumor Microenvironment
2.
Article in English | MEDLINE | ID: mdl-34708216

ABSTRACT

Response to cancer immunotherapies depends on the complex and dynamic interactions between T cell recognition and killing of cancer cells that are counteracted through immunosuppressive pathways in the tumor microenvironment. Therefore, while measurements such as tumor mutational burden provide biomarkers to select patients for immunotherapy, they neither universally predict patient response nor implicate the mechanisms that underlie immunotherapy resistance. Recent advances in single-cell RNA sequencing technology measure cellular heterogeneity within cells of an individual tumor but have yet to realize the promise of predictive oncology. In addition to data, mechanistic multiscale computational models are developed to predict treatment response. Incorporating single-cell data from tumors to parameterize these computational models provides deeper insights into prediction of clinical outcome in individual patients. Here, we integrate whole-exome sequencing and scRNA-seq data from Triple-Negative Breast Cancer patients to model neoantigen burden in tumor cells as input to a spatial Quantitative System Pharmacology model. The model comprises a four-compartmental Quantitative System Pharmacology sub-model to represent a whole patient and a spatial agent-based sub-model to represent tumor volumes at the cellular scale. We use the high-throughput single-cell data to model the role of antigen burden and heterogeneity relative to the tumor microenvironment composition on predicted immunotherapy response. We demonstrate how this integrated modeling and single-cell analysis framework can be used to relate neoantigen heterogeneity to immunotherapy treatment outcomes. Our results demonstrate feasibility of merging single-cell data to initialize cell states in multiscale computational models such as the spQSP for personalized prediction of clinical outcomes to immunotherapy.

3.
Cancers (Basel) ; 13(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34359653

ABSTRACT

Quantitative systems pharmacology (QSP) models have become increasingly common in fundamental mechanistic studies and drug discovery in both academic and industrial environments. With imaging techniques widely adopted and other spatial quantification of tumor such as spatial transcriptomics gaining traction, it is crucial that these data reflecting tumor spatial heterogeneity be utilized to inform the QSP models to enhance their predictive power. We developed a hybrid computational model platform, spQSP-IO, to extend QSP models of immuno-oncology with spatially resolved agent-based models (ABM), combining their powers to track whole patient-scale dynamics and recapitulate the emergent spatial heterogeneity in the tumor. Using a model of non-small-cell lung cancer developed based on this platform, we studied the role of the tumor microenvironment and cancer-immune cell interactions in tumor development and applied anti-PD-1 treatment to virtual patients and studied how the spatial distribution of cells changes during tumor growth in response to the immune checkpoint inhibition treatment. Using parameter sensitivity analysis and biomarker analysis, we are able to identify mechanisms and pretreatment measurements correlated with treatment efficacy. By incorporating spatial data that highlight both heterogeneity in tumors and variability among individual patients, spQSP-IO models can extend the QSP framework and further advance virtual clinical trials.

4.
J Theor Biol ; 484: 110006, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31539529

ABSTRACT

Quantitative predictions of FtsZ protein polymerization are essential for understanding the self-regulating mechanisms in biochemical systems. Due to structural complexity and parametric uncertainty, existing kinetic models remain incomplete and their predictions error-prone. To address such challenges, we perform probabilistic uncertainty quantification and global sensitivity analysis of the concentrations of various protein species predicted with a recent FtsZ protein polymerization model. Our results yield a ranked list of modeling shortcomings that can be improved in order to develop more accurate predictions and more realistic representations of key mechanisms of such biochemical systems and their response to changes in internal or external conditions. Our conclusions and improvement recommendations can be extended to other kinetics models.


Subject(s)
Bacterial Proteins , Cytoskeletal Proteins , Escherichia coli , Models, Biological , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Uncertainty
5.
Proc Natl Acad Sci U S A ; 115(19): 4933-4938, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29686085

ABSTRACT

High protein concentrations complicate modeling of polymer assembly kinetics by introducing structural complexity and a large variety of protein forms. We present a modeling approach that achieves orders of magnitude speed-up by replacing distributions of lengths and widths with their average counterparts and by introducing a hierarchical classification of species and reactions into sets. We have used this model to study FtsZ ring assembly in Escherichia coli The model's prediction of key features of the ring formation, such as time to reach the steady state, total concentration of FtsZ species in the ring, total concentration of monomers, and average dimensions of filaments and bundles, are all in agreement with the experimentally observed values. Besides validating our model against the in vivo observations, this study fills some knowledge gaps by proposing a specific structure of the ring, describing the influence of the total concentration in short and long kinetics processes, determining some characteristic mechanisms in polymer assembly regulation, and providing insights about the role of ZapA proteins, critical components for both positioning and stability of the ring.


Subject(s)
Bacterial Proteins/chemistry , Cytoskeletal Proteins/chemistry , Escherichia coli/chemistry , Models, Biological , Models, Chemical , Protein Multimerization , Bacterial Proteins/metabolism , Cytoskeletal Proteins/metabolism , Escherichia coli/metabolism
6.
Biophys J ; 111(1): 185-96, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27410746

ABSTRACT

Protein polymerization and bundling play a central role in cell physiology. Predictive modeling of these processes remains an open challenge, especially when the proteins involved become large and their concentrations high. We present an effective kinetics model of filament formation, bundling, and depolymerization after GTP hydrolysis, which involves a relatively small number of species and reactions, and remains robust over a wide range of concentrations and timescales. We apply this general model to study assembly of FtsZ protein, a basic element in the division process of prokaryotic cells such as Escherichia coli, Bacillus subtilis, or Caulobacter crescentus. This analysis demonstrates that our model outperforms its counterparts in terms of both accuracy and computational efficiency. Because our model comprises only 17 ordinary differential equations, its computational cost is orders-of-magnitude smaller than the current alternatives consisting of up to 1000 ordinary differential equations. It also provides, to our knowledge, a new insight into the characteristics and functioning of FtsZ proteins at high concentrations. The simplicity and versatility of our model render it a powerful computational tool, which can be used either as a standalone descriptor of other biopolymers' assembly or as a component in more complete kinetic models.


Subject(s)
Bacterial Proteins/chemistry , Cytoskeletal Proteins/chemistry , Models, Molecular , Protein Multimerization , Kinetics , Protein Structure, Quaternary , Thermodynamics
7.
La Paz; 1993. 107 p. ilus.
Thesis in Spanish | LIBOCS, LIBOSP | ID: biblio-1310506

ABSTRACT

El objetivo del presente trabajo, es relacionar los diferentes indicadores de estabilidad de voltaje, analizando sus caracteristicas y propiedades con el fin de proponer una metodologia confiable y robusta para la evaluacion de la proximidad al colapso de voltaje. Este procedimiento tendra la capacidad de medir con mucha exactitud: la cercania de un punto de operacion al punto de bifurcacion del sistema o colapso de voltaje. Proporcionar informacion identificando que region o regiones son las mas comprometidas con este fenomeno de manera que se pueda tomar una accion que evite este problema. La finalidad del trabajo es establecer un procedimiento de control centralizado de los diferentes estados de operacion de un sistema de potencia, que permita evaluar y cuantificar la estabilidad de voltaje y si es necesario, identificar los puntos neuralgicos de la red en los cuales deben tomarse medidas correctivas como por ejemplo, la desconexion de carga.

SELECTION OF CITATIONS
SEARCH DETAIL
...