Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Annu Rev Biomed Eng ; 26(1): 529-560, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38594947

RESUMO

Despite the remarkable advances in cancer diagnosis, treatment, and management over the past decade, malignant tumors remain a major public health problem. Further progress in combating cancer may be enabled by personalizing the delivery of therapies according to the predicted response for each individual patient. The design of personalized therapies requires the integration of patient-specific information with an appropriate mathematical model of tumor response. A fundamental barrier to realizing this paradigm is the current lack of a rigorous yet practical mathematical theory of tumor initiation, development, invasion, and response to therapy. We begin this review with an overview of different approaches to modeling tumor growth and treatment, including mechanistic as well as data-driven models based on big data and artificial intelligence. We then present illustrative examples of mathematical models manifesting their utility and discuss the limitations of stand-alone mechanistic and data-driven models. We then discuss the potential of mechanistic models for not only predicting but also optimizing response to therapy on a patient-specific basis. We describe current efforts and future possibilities to integrate mechanistic and data-driven models. We conclude by proposing five fundamental challenges that must be addressed to fully realize personalized care for cancer patients driven by computational models.


Assuntos
Inteligência Artificial , Big Data , Neoplasias , Medicina de Precisão , Humanos , Neoplasias/terapia , Medicina de Precisão/métodos , Simulação por Computador , Modelos Biológicos , Modelagem Computacional Específica para o Paciente
2.
Cancer Res Commun ; 4(3): 617-633, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38426815

RESUMO

Active surveillance (AS) is a suitable management option for newly diagnosed prostate cancer, which usually presents low to intermediate clinical risk. Patients enrolled in AS have their tumor monitored via longitudinal multiparametric MRI (mpMRI), PSA tests, and biopsies. Hence, treatment is prescribed when these tests identify progression to higher-risk prostate cancer. However, current AS protocols rely on detecting tumor progression through direct observation according to population-based monitoring strategies. This approach limits the design of patient-specific AS plans and may delay the detection of tumor progression. Here, we present a pilot study to address these issues by leveraging personalized computational predictions of prostate cancer growth. Our forecasts are obtained with a spatiotemporal biomechanistic model informed by patient-specific longitudinal mpMRI data (T2-weighted MRI and apparent diffusion coefficient maps from diffusion-weighted MRI). Our results show that our technology can represent and forecast the global tumor burden for individual patients, achieving concordance correlation coefficients from 0.93 to 0.99 across our cohort (n = 7). In addition, we identify a model-based biomarker of higher-risk prostate cancer: the mean proliferation activity of the tumor (P = 0.041). Using logistic regression, we construct a prostate cancer risk classifier based on this biomarker that achieves an area under the ROC curve of 0.83. We further show that coupling our tumor forecasts with this prostate cancer risk classifier enables the early identification of prostate cancer progression to higher-risk disease by more than 1 year. Thus, we posit that our predictive technology constitutes a promising clinical decision-making tool to design personalized AS plans for patients with prostate cancer. SIGNIFICANCE: Personalization of a biomechanistic model of prostate cancer with mpMRI data enables the prediction of tumor progression, thereby showing promise to guide clinical decision-making during AS for each individual patient.


Assuntos
Neoplasias da Próstata , Conduta Expectante , Masculino , Humanos , Projetos Piloto , Neoplasias da Próstata/diagnóstico por imagem , Próstata/diagnóstico por imagem , Antígeno Prostático Específico
3.
Cancer Biol Ther ; 25(1): 2321769, 2024 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-38411436

RESUMO

Tumor heterogeneity contributes significantly to chemoresistance, a leading cause of treatment failure. To better personalize therapies, it is essential to develop tools capable of identifying and predicting intra- and inter-tumor heterogeneities. Biology-inspired mathematical models are capable of attacking this problem, but tumor heterogeneity is often overlooked in in-vivo modeling studies, while phenotypic considerations capturing spatial dynamics are not typically included in in-vitro modeling studies. We present a data assimilation-prediction pipeline with a two-phenotype model that includes a spatiotemporal component to characterize and predict the evolution of in-vitro breast cancer cells and their heterogeneous response to chemotherapy. Our model assumes that the cells can be divided into two subpopulations: surviving cells unaffected by the treatment, and irreversibly damaged cells undergoing treatment-induced death. MCF7 breast cancer cells were previously cultivated in wells for up to 1000 hours, treated with various concentrations of doxorubicin and imaged with time-resolved microscopy to record spatiotemporally-resolved cell count data. Images were used to generate cell density maps. Treatment response predictions were initialized by a training set and updated by weekly measurements. Our mathematical model successfully calibrated the spatiotemporal cell growth dynamics, achieving median [range] concordance correlation coefficients of > .99 [.88, >.99] and .73 [.58, .85] across the whole well and individual pixels, respectively. Our proposed data assimilation-prediction approach achieved values of .97 [.44, >.99] and .69 [.35, .79] for the whole well and individual pixels, respectively. Thus, our model can capture and predict the spatiotemporal dynamics of MCF7 cells treated with doxorubicin in an in-vitro setting.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/farmacologia , Ciclo Celular , Proliferação de Células , Células MCF-7
4.
iScience ; 27(1): 108589, 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38169893

RESUMO

The heterogeneity inherent in cancer means that even a successful clinical trial merely results in a therapeutic regimen that achieves, on average, a positive result only in a subset of patients. The only way to optimize an intervention for an individual patient is to reframe their treatment as their own, personalized trial. Toward this goal, we formulate a computational framework for performing personalized trials that rely on four mathematical techniques. First, mathematical models that can be calibrated with patient-specific data to make accurate predictions of response. Second, digital twins built on these models capable of simulating the effects of interventions. Third, optimal control theory applied to the digital twins to optimize outcomes. Fourth, data assimilation to continually update and refine predictions in response to therapeutic interventions. In this perspective, we describe each of these techniques, quantify their "state of readiness", and identify use cases for personalized clinical trials.

5.
Front Artif Intell ; 6: 1222612, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37886348

RESUMO

We develop a methodology to create data-driven predictive digital twins for optimal risk-aware clinical decision-making. We illustrate the methodology as an enabler for an anticipatory personalized treatment that accounts for uncertainties in the underlying tumor biology in high-grade gliomas, where heterogeneity in the response to standard-of-care (SOC) radiotherapy contributes to sub-optimal patient outcomes. The digital twin is initialized through prior distributions derived from population-level clinical data in the literature for a mechanistic model's parameters. Then the digital twin is personalized using Bayesian model calibration for assimilating patient-specific magnetic resonance imaging data. The calibrated digital twin is used to propose optimal radiotherapy treatment regimens by solving a multi-objective risk-based optimization under uncertainty problem. The solution leads to a suite of patient-specific optimal radiotherapy treatment regimens exhibiting varying levels of trade-off between the two competing clinical objectives: (i) maximizing tumor control (characterized by minimizing the risk of tumor volume growth) and (ii) minimizing the toxicity from radiotherapy. The proposed digital twin framework is illustrated by generating an in silico cohort of 100 patients with high-grade glioma growth and response properties typically observed in the literature. For the same total radiation dose as the SOC, the personalized treatment regimens lead to median increase in tumor time to progression of around six days. Alternatively, for the same level of tumor control as the SOC, the digital twin provides optimal treatment options that lead to a median reduction in radiation dose by 16.7% (10 Gy) compared to SOC total dose of 60 Gy. The range of optimal solutions also provide options with increased doses for patients with aggressive cancer, where SOC does not lead to sufficient tumor control.

6.
ArXiv ; 2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37693182

RESUMO

Despite the remarkable advances in cancer diagnosis, treatment, and management that have occurred over the past decade, malignant tumors remain a major public health problem. Further progress in combating cancer may be enabled by personalizing the delivery of therapies according to the predicted response for each individual patient. The design of personalized therapies requires patient-specific information integrated into an appropriate mathematical model of tumor response. A fundamental barrier to realizing this paradigm is the current lack of a rigorous, yet practical, mathematical theory of tumor initiation, development, invasion, and response to therapy. In this review, we begin by providing an overview of different approaches to modeling tumor growth and treatment, including mechanistic as well as data-driven models based on ``big data" and artificial intelligence. Next, we present illustrative examples of mathematical models manifesting their utility and discussing the limitations of stand-alone mechanistic and data-driven models. We further discuss the potential of mechanistic models for not only predicting, but also optimizing response to therapy on a patient-specific basis. We then discuss current efforts and future possibilities to integrate mechanistic and data-driven models. We conclude by proposing five fundamental challenges that must be addressed to fully realize personalized care for cancer patients driven by computational models.

7.
Eng Comput ; : 1-25, 2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37362241

RESUMO

The rapid spread of the numerous outbreaks of the coronavirus disease 2019 (COVID-19) pandemic has fueled interest in mathematical models designed to understand and predict infectious disease spread, with the ultimate goal of contributing to the decision making of public health authorities. Here, we propose a computational pipeline that dynamically parameterizes a modified SEIRD (susceptible-exposed-infected-recovered-deceased) model using standard daily series of COVID-19 cases and deaths, along with isolated estimates of population-level seroprevalence. We test our pipeline in five heavily impacted states of the US (New York, California, Florida, Illinois, and Texas) between March and August 2020, considering two scenarios with different calibration time horizons to assess the update in model performance as new epidemiologic data become available. Our results show a median normalized root mean squared error (NRMSE) of 2.38% and 4.28% in calibrating cumulative cases and deaths in the first scenario, and 2.41% and 2.30% when new data are assimilated in the second scenario, respectively. Then, 2-week (4-week) forecasts of the calibrated model resulted in median NRMSE of cumulative cases and deaths of 5.85% and 4.68% (8.60% and 17.94%) in the first scenario, and 1.86% and 1.93% (2.21% and 1.45%) in the second. Additionally, we show that our method provides significantly more accurate predictions of cases and deaths than a constant parameterization in the second scenario (p < 0.05). Thus, we posit that our methodology is a promising approach to analyze the dynamics of infectious disease outbreaks, and that our forecasts could contribute to designing effective pandemic-arresting public health policies. Supplementary Information: The online version contains supplementary material available at 10.1007/s00366-023-01816-9.

8.
iScience ; 25(11): 105430, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36388979

RESUMO

The detection of prostate cancer recurrence after external beam radiotherapy relies on the measurement of a sustained rise of serum prostate-specific antigen (PSA). However, this biochemical relapse may take years to occur, thereby delaying the delivery of a secondary treatment to patients with recurring tumors. To address this issue, we propose to use patient-specific forecasts of PSA dynamics to predict biochemical relapse earlier. Our forecasts are based on a mechanistic model of prostate cancer response to external beam radiotherapy, which is fit to patient-specific PSA data collected during standard posttreatment monitoring. Our results show a remarkable performance of our model in recapitulating the observed changes in PSA and yielding short-term predictions over approximately 1 year (cohort median root mean squared error of 0.10-0.47 ng/mL and 0.13 to 1.39 ng/mL, respectively). Additionally, we identify 3 model-based biomarkers that enable accurate identification of biochemical relapse (area under the receiver operating characteristic curve > 0.80) significantly earlier than standard practice (p < 0.01).

9.
Front Mol Biosci ; 9: 972146, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36172049

RESUMO

The development of chemoresistance remains a significant cause of treatment failure in breast cancer. We posit that a mathematical understanding of chemoresistance could assist in developing successful treatment strategies. Towards that end, we have developed a model that describes the cytotoxic effects of the standard chemotherapeutic drug doxorubicin on the MCF-7 breast cancer cell line. We assume that treatment with doxorubicin induces a compartmentalization of the breast cancer cell population into surviving cells, which continue proliferating after treatment, and irreversibly damaged cells, which gradually transition from proliferating to treatment-induced death. The model is fit to experimental data including variations in drug concentration, inter-treatment interval, and number of doses. Our model recapitulates tumor cell dynamics in all these scenarios (as quantified by the concordance correlation coefficient, CCC > 0.95). In particular, superior tumor control is observed with higher doxorubicin concentrations, shorter inter-treatment intervals, and a higher number of doses (p < 0.05). Longer inter-treatment intervals require adapting the model parameterization after each doxorubicin dose, suggesting the promotion of chemoresistance. Additionally, we propose promising empirical formulas to describe the variation of model parameters as functions of doxorubicin concentration (CCC > 0.78). Thus, we conclude that our mathematical model could deepen our understanding of the cytotoxic effects of doxorubicin and could be used to explore practical drug regimens achieving optimal tumor control.

10.
J Biomech Eng ; 144(12)2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35771166

RESUMO

The computer simulation of organ-scale biomechanistic models of cancer personalized via routinely collected clinical and imaging data enables to obtain patient-specific predictions of tumor growth and treatment response over the anatomy of the patient's affected organ. These patient-specific computational forecasts have been regarded as a promising approach to personalize the clinical management of cancer and derive optimal treatment plans for individual patients, which constitute timely and critical needs in clinical oncology. However, the computer simulation of the underlying spatiotemporal models can entail a prohibitive computational cost, which constitutes a barrier to the successful development of clinically-actionable computational technologies for personalized tumor forecasting. To address this issue, here we propose to utilize dynamic-mode decomposition (DMD) to construct a low-dimensional representation of cancer models and accelerate their simulation. DMD is an unsupervised machine learning method based on the singular value decomposition that has proven useful in many applications as both a predictive and a diagnostic tool. We show that DMD may be applied to Fisher-Kolmogorov models, which constitute an established formulation to represent untreated solid tumor growth that can further accommodate other relevant cancer phenomena (e.g., therapeutic effects, mechanical deformation). Our results show that a DMD implementation of this model over a clinically relevant parameter space can yield promising predictions, with short to medium-term errors remaining under 1% and long-term errors remaining under 20%, despite very short training periods. In particular, we have found that, for moderate to high tumor cell diffusivity and low to moderate tumor cell proliferation rate, DMD reconstructions provide accurate, bounded-error reconstructions for all tested training periods. Additionally, we also show that the three-dimensional DMD reconstruction of the tumor field can be leveraged to accurately reconstruct the displacement fields of the tumor-induced deformation of the host tissue. Thus, we posit the proposed data-driven approach has the potential to greatly reduce the computational overhead of personalized simulations of cancer models, thereby facilitating tumor forecasting, parameter identification, uncertainty quantification, and treatment optimization.


Assuntos
Neoplasias , Simulação por Computador , Humanos
11.
Biophys Rev (Melville) ; 3(2): 021304, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35602761

RESUMO

Digital twins employ mathematical and computational models to virtually represent a physical object (e.g., planes and human organs), predict the behavior of the object, and enable decision-making to optimize the future behavior of the object. While digital twins have been widely used in engineering for decades, their applications to oncology are only just emerging. Due to advances in experimental techniques quantitatively characterizing cancer, as well as advances in the mathematical and computational sciences, the notion of building and applying digital twins to understand tumor dynamics and personalize the care of cancer patients has been increasingly appreciated. In this review, we present the opportunities and challenges of applying digital twins in clinical oncology, with a particular focus on integrating medical imaging with mechanism-based, tissue-scale mathematical modeling. Specifically, we first introduce the general digital twin framework and then illustrate existing applications of image-guided digital twins in healthcare. Next, we detail both the imaging and modeling techniques that provide practical opportunities to build patient-specific digital twins for oncology. We then describe the current challenges and limitations in developing image-guided, mechanism-based digital twins for oncology along with potential solutions. We conclude by outlining five fundamental questions that can serve as a roadmap when designing and building a practical digital twin for oncology and attempt to provide answers for a specific application to brain cancer. We hope that this contribution provides motivation for the imaging science, oncology, and computational communities to develop practical digital twin technologies to improve the care of patients battling cancer.

12.
IEEE Trans Biomed Eng ; 69(11): 3334-3344, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35439121

RESUMO

OBJECTIVE: This study establishes a fluid dynamics model personalized with patient-specific imaging data to optimize neoadjuvant therapy (i.e., doxorubicin) protocols for breast cancers. METHODS: Ten patients recruited at the University of Chicago were included in this study. Quantitative dynamic contrast-enhanced and diffusion weighted magnetic resonance imaging data are leveraged to estimate patient-specific hemodynamic properties, which are then used to constrain the mechanism-based drug delivery model. Then, computer simulations of this model yield the subsequent drug distribution throughout the breast. By systematically varying the dosing schedule, we identify an optimized regimen for each patient using the maximum safe therapeutic duration (MSTD), which is a metric balancing treatment efficacy and toxicity. RESULTS: With an individually optimized dose (range = 12.11-15.11 mg/m2 per injection), a 3-week regimen consisting of a uniform daily injection significantly outperforms all other scheduling strategies (P < 0.001). In particular, the optimal protocol is predicted to significantly outperform the standard protocol (P < 0.001), improving the MSTD by an average factor of 9.93 (range = 6.63 to 14.17). CONCLUSION: A clinical-mathematical framework was developed by integrating quantitative MRI data, advanced image processing, and computational fluid dynamics to predict the efficacy and toxicity of neoadjuvant therapy protocols, thus enabling the rational identification of an optimal therapeutic regimen on a patient-specific basis. SIGNIFICANCE: Our clinical-computational approach has the potential to enable optimization of therapeutic regimens on a patient-specific basis and provide guidance for prospective clinical trials aimed at refining neoadjuvant therapy protocols for breast cancers.


Assuntos
Neoplasias da Mama , Terapia Neoadjuvante , Humanos , Feminino , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Hidrodinâmica , Estudos Prospectivos , Doxorrubicina/uso terapêutico , Resultado do Tratamento
13.
Cancers (Basel) ; 13(12)2021 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-34208448

RESUMO

Tumor-associated vasculature is responsible for the delivery of nutrients, removal of waste, and allowing growth beyond 2-3 mm3. Additionally, the vascular network, which is changing in both space and time, fundamentally influences tumor response to both systemic and radiation therapy. Thus, a robust understanding of vascular dynamics is necessary to accurately predict tumor growth, as well as establish optimal treatment protocols to achieve optimal tumor control. Such a goal requires the intimate integration of both theory and experiment. Quantitative and time-resolved imaging methods have emerged as technologies able to visualize and characterize tumor vascular properties before and during therapy at the tissue and cell scale. Parallel to, but separate from those developments, mathematical modeling techniques have been developed to enable in silico investigations into theoretical tumor and vascular dynamics. In particular, recent efforts have sought to integrate both theory and experiment to enable data-driven mathematical modeling. Such mathematical models are calibrated by data obtained from individual tumor-vascular systems to predict future vascular growth, delivery of systemic agents, and response to radiotherapy. In this review, we discuss experimental techniques for visualizing and quantifying vascular dynamics including magnetic resonance imaging, microfluidic devices, and confocal microscopy. We then focus on the integration of these experimental measures with biologically based mathematical models to generate testable predictions.

15.
Appl Math Lett ; 111: 106617, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32834475

RESUMO

We present an early version of a Susceptible-Exposed-Infected-Recovered-Deceased (SEIRD) mathematical model based on partial differential equations coupled with a heterogeneous diffusion model. The model describes the spatio-temporal spread of the COVID-19 pandemic, and aims to capture dynamics also based on human habits and geographical features. To test the model, we compare the outputs generated by a finite-element solver with measured data over the Italian region of Lombardy, which has been heavily impacted by this crisis between February and April 2020. Our results show a strong qualitative agreement between the simulated forecast of the spatio-temporal COVID-19 spread in Lombardy and epidemiological data collected at the municipality level. Additional simulations exploring alternative scenarios for the relaxation of lockdown restrictions suggest that reopening strategies should account for local population densities and the specific dynamics of the contagion. Thus, we argue that data-driven simulations of our model could ultimately inform health authorities to design effective pandemic-arresting measures and anticipate the geographical allocation of crucial medical resources.

16.
iScience ; 23(12): 101807, 2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33299976

RESUMO

We provide an overview on the use of biological assays to calibrate and initialize mechanism-based models of cancer phenomena. Although artificial intelligence methods currently dominate the landscape in computational oncology, mathematical models that seek to explicitly incorporate biological mechanisms into their formalism are of increasing interest. These models can guide experimental design and provide insights into the underlying mechanisms of cancer progression. Historically, these models have included a myriad of parameters that have been difficult to quantify in biologically relevant systems, limiting their practical insights. Recently, however, there has been much interest calibrating biologically based models with the quantitative measurements available from (for example) RNA sequencing, time-resolved microscopy, and in vivo imaging. In this contribution, we summarize how a variety of experimental methods quantify tumor characteristics from the molecular to tissue scales and describe how such data can be directly integrated with mechanism-based models to improve predictions of tumor growth and treatment response.

17.
Sci Adv ; 6(36)2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32917585

RESUMO

Quantum communication is rapidly gaining popularity due to its high security and technological maturity. However, most implementations are limited to just two communicating parties (users). Quantum communication networks aim to connect a multitude of users. Here, we present a fully connected quantum communication network on a city-wide scale without active switching or trusted nodes. We demonstrate simultaneous and secure connections between all 28 pairings of eight users. Our novel network topology is easily scalable to many users, allows traffic management features, and minimizes the infrastructure as well as the user hardware needed.

18.
Comput Mech ; 66(5): 1131-1152, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32836602

RESUMO

The outbreak of COVID-19 in 2020 has led to a surge in interest in the research of the mathematical modeling of epidemics. Many of the introduced models are so-called compartmental models, in which the total quantities characterizing a certain system may be decomposed into two (or more) species that are distributed into two (or more) homogeneous units called compartments. We propose herein a formulation of compartmental models based on partial differential equations (PDEs) based on concepts familiar to continuum mechanics, interpreting such models in terms of fundamental equations of balance and compatibility, joined by a constitutive relation. We believe that such an interpretation may be useful to aid understanding and interdisciplinary collaboration. We then proceed to focus on a compartmental PDE model of COVID-19 within the newly-introduced framework, beginning with a detailed derivation and explanation. We then analyze the model mathematically, presenting several results concerning its stability and sensitivity to different parameters. We conclude with a series of numerical simulations to support our findings.

19.
IEEE Trans Med Imaging ; 39(9): 2760-2771, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32086203

RESUMO

The overall goal of this study is to employ quantitative magnetic resonance imaging (MRI) data to constrain a patient-specific, computational fluid dynamics (CFD) model of blood flow and interstitial transport in breast cancer. We develop image processing methodologies to generate tumor-related vasculature-interstitium geometry and realistic material properties, using dynamic contrast enhanced MRI (DCE-MRI) and diffusion weighted MRI (DW-MRI) data. These data are used to constrain CFD simulations for determining the tumor-associated blood supply and interstitial transport characteristics unique to each patient. We then perform a proof-of-principle statistical comparison between these hemodynamic characteristics in 11 malignant and 5 benign lesions from 12 patients. Significant differences between groups (i.e., malignant versus benign) were observed for the median of tumor-associated interstitial flow velocity ( P = 0.028 ), and the ranges of tumor-associated blood pressure (P = 0.016) and vascular extraction rate (P = 0.040). The implication is that malignant lesions tend to have larger magnitude of interstitial flow velocity, and higher heterogeneity in blood pressure and vascular extraction rate. Multivariable logistic models based on combinations of these hemodynamic data achieved excellent differentiation between malignant and benign lesions with an area under the receiver operator characteristic curve of 1.0, sensitivity of 1.0, and specificity of 1.0. This image-based model system is a fundamentally new way to map flow and pressure fields related to breast tumors using only non-invasive, clinically available imaging data and established laws of fluid mechanics. Furthermore, the results provide preliminary evidence for this methodology's utility for the quantitative characterization of breast cancer.


Assuntos
Neoplasias da Mama , Mama/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Meios de Contraste , Imagem de Difusão por Ressonância Magnética , Feminino , Hemodinâmica , Humanos , Hidrodinâmica , Imageamento por Ressonância Magnética , Curva ROC , Estudos Retrospectivos , Sensibilidade e Especificidade
20.
J R Soc Interface ; 16(157): 20190195, 2019 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-31409240

RESUMO

External beam radiation therapy is a widespread treatment for prostate cancer. The ensuing patient follow-up is based on the evolution of the prostate-specific antigen (PSA). Serum levels of PSA decay due to the radiation-induced death of tumour cells and cancer recurrence usually manifest as a rising PSA. The current definition of biochemical relapse requires that PSA reaches nadir and starts increasing, which delays the use of further treatments. Also, these methods do not account for the post-radiation tumour dynamics that may contain early information on cancer recurrence. Here, we develop three mechanistic models of post-radiation PSA evolution. Our models render superior fits of PSA data in a patient cohort and provide a biological justification for the most common empirical formulation of PSA dynamics. We also found three model-based prognostic variables: the proliferation rate of the survival fraction, the ratio of radiation-induced cell death rate to the survival proliferation rate, and the time to PSA nadir since treatment termination. We argue that these markers may enable the early identification of biochemical relapse, which would permit physicians to subsequently adapt patient monitoring to optimize the detection and treatment of cancer recurrence.


Assuntos
Modelos Biológicos , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/radioterapia , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Antígeno Prostático Específico/metabolismo , Taxa de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA