Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1358019, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38515743

RESUMEN

Bladder cancer is an increasingly prevalent global disease that continues to cause morbidity and mortality despite recent advances in treatment. Immune checkpoint inhibitors (ICI) and fibroblast growth factor receptor (FGFR)-targeted therapeutics have had modest success in bladder cancer when used as monotherapy. Emerging data suggests that the combination of these two therapies could lead to improved clinical outcomes, but the optimal strategy for combining these agents remains uncertain. Mathematical models, specifically agent-based models (ABMs), have shown recent successes in uncovering the multiscale dynamics that shape the trajectory of cancer. They have enabled the optimization of treatment methods and the identification of novel therapeutic strategies. To assess the combined effects of anti-PD-1 and anti-FGFR3 small molecule inhibitors (SMI) on tumor growth and the immune response, we built an ABM that captures key facets of tumor heterogeneity and CD8+ T cell phenotypes, their spatial interactions, and their response to therapeutic pressures. Our model quantifies how tumor antigenicity and FGFR3 activating mutations impact disease trajectory and response to anti-PD-1 antibodies and anti-FGFR3 SMI. We find that even a small population of weakly antigenic tumor cells bearing an FGFR3 mutation can render the tumor resistant to combination therapy. However, highly antigenic tumors can overcome therapeutic resistance mediated by FGFR3 mutation. The optimal therapy depends on the strength of the FGFR3 signaling pathway. Under certain conditions, ICI alone is optimal; in others, ICI followed by anti-FGFR3 therapy is best. These results indicate the need to quantify FGFR3 signaling and the fitness advantage conferred on bladder cancer cells harboring this mutation. This ABM approach may enable rationally designed treatment plans to improve clinical outcomes.


Asunto(s)
Transducción de Señal , Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Terapia Combinada , Mutación , Línea Celular Tumoral , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo
2.
Bull Math Biol ; 86(1): 11, 2023 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-38159216

RESUMEN

Across a broad range of disciplines, agent-based models (ABMs) are increasingly utilized for replicating, predicting, and understanding complex systems and their emergent behavior. In the biological and biomedical sciences, researchers employ ABMs to elucidate complex cellular and molecular interactions across multiple scales under varying conditions. Data generated at these multiple scales, however, presents a computational challenge for robust analysis with ABMs. Indeed, calibrating ABMs remains an open topic of research due to their own high-dimensional parameter spaces. In response to these challenges, we extend and validate our novel methodology, Surrogate Modeling for Reconstructing Parameter Surfaces (SMoRe ParS), arriving at a computationally efficient framework for connecting high dimensional ABM parameter spaces with multidimensional data. Specifically, we modify SMoRe ParS to initially confine high dimensional ABM parameter spaces using unidimensional data, namely, single time-course information of in vitro cancer cell growth assays. Subsequently, we broaden the scope of our approach to encompass more complex ABMs and constrain parameter spaces using multidimensional data. We explore this extension with in vitro cancer cell inhibition assays involving the chemotherapeutic agent oxaliplatin. For each scenario, we validate and evaluate the effectiveness of our approach by comparing how well ABM simulations match the experimental data when using SMoRe ParS-inferred parameters versus parameters inferred by a commonly used direct method. In so doing, we show that our approach of using an explicitly formulated surrogate model as an interlocutor between the ABM and the experimental data effectively calibrates the ABM parameter space to multidimensional data. Our method thus provides a robust and scalable strategy for leveraging multidimensional data to inform multiscale ABMs and explore the uncertainty in their parameters.


Asunto(s)
Conceptos Matemáticos , Modelos Biológicos , Incertidumbre , Fagocitosis
3.
Sci Rep ; 13(1): 22541, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110479

RESUMEN

Immunotherapy has dramatically transformed the cancer treatment landscape largely due to the efficacy of immune checkpoint inhibitors (ICIs). Although ICIs have shown promising results for many patients, the low response rates in many cancers highlight the ongoing challenges in cancer treatment. Cytotoxic T lymphocytes (CTLs) execute their cell-killing function via two distinct mechanisms: a fast-acting, perforin-mediated process and a slower, Fas ligand (FasL)-driven pathway. Evidence also suggests that the preferred killing mechanism of CTLs depends on the antigenicity of tumor cells. To determine the critical factors affecting responses to ICIs, we construct an ordinary differential equation model describing in vivo tumor-immune dynamics in the presence of active or blocked PD-1/PD-L1 immune checkpoint. Specifically, we identify important aspects of the tumor-immune landscape that affect tumor size and composition in the short and long term. We also generate a virtual cohort of mice with diverse tumor and immune attributes to simulate the outcomes of immune checkpoint blockade in a heterogeneous population. By identifying key tumor and immune characteristics associated with tumor elimination, dormancy, and escape, we predict which fraction of a population potentially responds well to ICIs and ways to enhance therapeutic outcomes with combination therapy.


Asunto(s)
Neoplasias , Linfocitos T Citotóxicos , Humanos , Animales , Ratones , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Perforina , Modelos Teóricos
4.
bioRxiv ; 2023 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-37745323

RESUMEN

Cells are fundamental units of life, constantly interacting and evolving as dynamical systems. While recent spatial multi-omics can quantitate individual cells' characteristics and regulatory programs, forecasting their evolution ultimately requires mathematical modeling. We develop a conceptual framework-a cell behavior hypothesis grammar-that uses natural language statements (cell rules) to create mathematical models. This allows us to systematically integrate biological knowledge and multi-omics data to make them computable. We can then perform virtual "thought experiments" that challenge and extend our understanding of multicellular systems, and ultimately generate new testable hypotheses. In this paper, we motivate and describe the grammar, provide a reference implementation, and demonstrate its potential through a series of examples in tumor biology and immunotherapy. Altogether, this approach provides a bridge between biological, clinical, and systems biology researchers for mathematical modeling of biological systems at scale, allowing the community to extrapolate from single-cell characterization to emergent multicellular behavior.

5.
Commun Biol ; 4(1): 983, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408236

RESUMEN

During progression from carcinoma in situ to an invasive tumor, the immune system is engaged in complex sets of interactions with various tumor cells. Tumor cell plasticity alters disease trajectories via epithelial-to-mesenchymal transition (EMT). Several of the same pathways that regulate EMT are involved in tumor-immune interactions, yet little is known about the mechanisms and consequences of crosstalk between these regulatory processes. Here we introduce a multiscale evolutionary model to describe tumor-immune-EMT interactions and their impact on epithelial cancer progression from in situ to invasive disease. Through simulation of patient cohorts in silico, the model predicts that a controllable region maximizes invasion-free survival. This controllable region depends on properties of the mesenchymal tumor cell phenotype: its growth rate and its immune-evasiveness. In light of the model predictions, we analyze EMT-inflammation-associated data from The Cancer Genome Atlas, and find that association with EMT worsens invasion-free survival probabilities. This result supports the predictions of the model, and leads to the identification of genes that influence outcomes in bladder and uterine cancer, including FGF pathway members. These results suggest new means to delay disease progression, and demonstrate the importance of studying cancer-immune interactions in light of EMT.


Asunto(s)
Carcinoma/genética , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Sistema Inmunológico , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...