Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Nucl Med Mol Imaging ; 50(9): 2669-2682, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37017737

RESUMO

PURPOSE: Pittsburgh Compound-B (11C-PiB) and 18F-florbetapir are amyloid-ß (Aß) positron emission tomography (PET) radiotracers that have been used as endpoints in Alzheimer's disease (AD) clinical trials to evaluate the efficacy of anti-Aß monoclonal antibodies. However, comparing drug effects between and within trials may become complicated if different Aß radiotracers were used. To study the consequences of using different Aß radiotracers to measure Aß clearance, we performed a head-to-head comparison of 11C-PiB and 18F-florbetapir in a Phase 2/3 clinical trial of anti-Aß monoclonal antibodies. METHODS: Sixty-six mutation-positive participants enrolled in the gantenerumab and placebo arms of the first Dominantly Inherited Alzheimer Network Trials Unit clinical trial (DIAN-TU-001) underwent both 11C-PiB and 18F-florbetapir PET imaging at baseline and during at least one follow-up visit. For each PET scan, regional standardized uptake value ratios (SUVRs), regional Centiloids, a global cortical SUVR, and a global cortical Centiloid value were calculated. Longitudinal changes in SUVRs and Centiloids were estimated using linear mixed models. Differences in longitudinal change between PET radiotracers and between drug arms were estimated using paired and Welch two sample t-tests, respectively. Simulated clinical trials were conducted to evaluate the consequences of some research sites using 11C-PiB while other sites use 18F-florbetapir for Aß PET imaging. RESULTS: In the placebo arm, the absolute rate of longitudinal change measured by global cortical 11C-PiB SUVRs did not differ from that of global cortical 18F-florbetapir SUVRs. In the gantenerumab arm, global cortical 11C-PiB SUVRs decreased more rapidly than global cortical 18F-florbetapir SUVRs. Drug effects were statistically significant across both Aß radiotracers. In contrast, the rates of longitudinal change measured in global cortical Centiloids did not differ between Aß radiotracers in either the placebo or gantenerumab arms, and drug effects remained statistically significant. Regional analyses largely recapitulated these global cortical analyses. Across simulated clinical trials, type I error was higher in trials where both Aß radiotracers were used versus trials where only one Aß radiotracer was used. Power was lower in trials where 18F-florbetapir was primarily used versus trials where 11C-PiB was primarily used. CONCLUSION: Gantenerumab treatment induces longitudinal changes in Aß PET, and the absolute rates of these longitudinal changes differ significantly between Aß radiotracers. These differences were not seen in the placebo arm, suggesting that Aß-clearing treatments may pose unique challenges when attempting to compare longitudinal results across different Aß radiotracers. Our results suggest converting Aß PET SUVR measurements to Centiloids (both globally and regionally) can harmonize these differences without losing sensitivity to drug effects. Nonetheless, until consensus is achieved on how to harmonize drug effects across radiotracers, and since using multiple radiotracers in the same trial may increase type I error, multisite studies should consider potential variability due to different radiotracers when interpreting Aß PET biomarker data and, if feasible, use a single radiotracer for the best results. TRIAL REGISTRATION: ClinicalTrials.gov NCT01760005. Registered 31 December 2012. Retrospectively registered.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Compostos de Anilina , Etilenoglicóis , Encéfalo/metabolismo
2.
Eur J Nucl Med Mol Imaging ; 47(5): 1168-1182, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31807885

RESUMO

INTRODUCTION: Immunotherapy has improved outcomes for patients with non-small cell lung cancer (NSCLC), yet durable clinical benefit (DCB) is experienced in only a fraction of patients. Here, we test the hypothesis that radiomics features from baseline pretreatment 18F-FDG PET/CT scans can predict clinical outcomes of NSCLC patients treated with checkpoint blockade immunotherapy. METHODS: This study included 194 patients with histologically confirmed stage IIIB-IV NSCLC with pretreatment PET/CT images. Radiomics features were extracted from PET, CT, and PET+CT fusion images based on minimum Kullback-Leibler divergence (KLD) criteria. The radiomics features from 99 retrospective patients were used to train a multiparametric radiomics signature (mpRS) to predict DCB using an improved least absolute shrinkage and selection operator (LASSO) method, which was subsequently validated in both retrospective (N = 47) and prospective test cohorts (N = 48). Using these cohorts, the mpRS was also used to predict progression-free survival (PFS) and overall survival (OS) by training nomogram models using multivariable Cox regression analyses with additional clinical characteristics incorporated. RESULTS: The mpRS could predict patients who will receive DCB, with areas under receiver operating characteristic curves (AUCs) of 0.86 (95%CI 0.79-0.94), 0.83 (95%CI 0.71-0.94), and 0.81 (95%CI 0.68-0.92) in the training, retrospective test, and prospective test cohorts, respectively. In the same three cohorts, respectively, nomogram models achieved C-indices of 0.74 (95%CI 0.68-0.80), 0.74 (95%CI 0.66-0.82), and 0.77 (95%CI 0.69-0.84) to predict PFS and C-indices of 0.83 (95%CI 0.77-0.88), 0.83 (95%CI 0.71-0.94), and 0.80 (95%CI 0.69-0.91) to predict OS. CONCLUSION: PET/CT-based signature can be used prior to initiation of immunotherapy to identify NSCLC patients most likely to benefit from immunotherapy. As such, these data may be leveraged to improve more precise and individualized decision support in the treatment of patients with advanced NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/terapia , Fluordesoxiglucose F18 , Humanos , Imunoterapia , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/terapia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Estudos Prospectivos , Estudos Retrospectivos
3.
Alzheimers Dement (N Y) ; 9(3): e12415, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600216

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is characterized by the presence of both amyloid and tau pathology. In vivo diagnosis can be made with amyloid and tau positron emission tomography (PET) imaging. Emergent evidence supports that amyloid and tau accumulation are associated and that amyloid accumulation may precede that of tau. This report further investigates the relationship between amyloid and tau to assess whether elevated cortical tau can predict elevated amyloid in participants with early symptomatic AD. METHODS: Florbetapir F18 and flortaucipir F18 uptake were evaluated from baseline PET scans collected in three multi-center studies with cognitively impaired participants, including A05 (N = 306; NCT02016560), TB (N = 310; TRAILBLAZER-ALZ; NCT03367403), and TB2 (N = 1165; TRAILBLAZER-ALZ 2; NCT04437511). Images were assessed using visual and quantitative approaches to establish amyloid (A+) and tau (T+) positivity, as well as a combination method (tauVQ) to establish T+. Associations between global amyloid and tau were evaluated with positive and negative predictive values (PPV, NPV) and likelihood ratios (LR+, LR-). Predictive values within subgroups according to ethnicity, race, cognitive score, age, and sex were also evaluated. The relationship between regional tau (four target and two reference regions were tested) and global amyloid was investigated in A05 participant scans using receiver-operating characteristic (ROC) curves. RESULTS: PPV for amyloid positivity was ≥93% for all three trials using various A+ and T+ definitions, including visual, quantitative, and combination methods. Population characteristics did not have an impact on A+ predictability. Regional analyses (early tau (Eτ) volume of interest (VOI), temporal, parietal, frontal) revealed significant area under the ROC curve in Eτ VOI compared to frontal region, regardless of reference region and consistent among visual and quantitative A+ definitions (p < 0.001). DISCUSSION: These findings suggest that a positive tau PET scan is associated (≥93%) with amyloid positivity in individuals with early symptomatic AD, with the potential benefits of reducing clinical trial and health care expenses, radiation exposure, and participant time. Highlights: Positron emission tomography (PET) evaluates candidates for Alzheimer's disease (AD) research. A positive tau PET scan is associated (≥93%) with amyloid positivity.A positive amyloid PET is not necessarily associated with tau positivity.Tau PET could be the sole diagnostic tool to confirm candidates for AD trials.

4.
Cancer Biomark ; 33(4): 489-501, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35491768

RESUMO

BACKGROUND: Image-based biomarkers could have translational implications by characterizing tumor behavior of lung cancers diagnosed during lung cancer screening. In this study, peritumoral and intratumoral radiomics and volume doubling time (VDT) were used to identify high-risk subsets of lung patients diagnosed in lung cancer screening that are associated with poor survival outcomes. METHODS: Data and images were acquired from the National Lung Screening Trial. VDT was calculated between two consequent screening intervals approximately 1 year apart; peritumoral and intratumoral radiomics were extracted from the baseline screen. Overall survival (OS) was the main endpoint. Classification and Regression Tree analyses identified the most predictive covariates to classify patient outcomes. RESULTS: Decision tree analysis stratified patients into three risk-groups (low, intermediate, and high) based on VDT and one radiomic feature (compactness). High-risk patients had extremely poor survival outcomes (hazard ratio [HR] = 8.15; 25% 5-year OS) versus low-risk patients (HR = 1.00; 83.3% 5-year OS). Among early-stage lung cancers, high-risk patients had poor survival outcomes (HR = 9.07; 44.4% 5-year OS) versus the low-risk group (HR = 1.00; 90.9% 5-year OS). For VDT, the decision tree analysis identified a novel cut-point of 279 days and using this cut-point VDT alone discriminated between aggressive (HR = 4.18; 45% 5-year OS) versus indolent/low-risk cancers (HR = 1.00; 82.8% 5-year OS). CONCLUSION: We utilized peritumoral and intratumoral radiomic features and VDT to generate a model that identify a high-risk group of screen-detected lung cancers associated with poor survival outcomes. These vulnerable subset of screen-detected lung cancers may be candidates for more aggressive surveillance/follow-up and treatment, such as adjuvant therapy.


Assuntos
Neoplasias Pulmonares , Detecção Precoce de Câncer , Humanos , Pulmão/patologia , Neoplasias Pulmonares/diagnóstico por imagem , Fatores de Risco , Tomografia Computadorizada por Raios X/métodos
5.
Artigo em Inglês | MEDLINE | ID: mdl-33431509

RESUMO

Medical imaging is the standard-of-care for early detection, diagnosis, treatment planning, monitoring, and image-guided interventions of lung cancer patients. Most medical images are stored digitally in a standardized Digital Imaging and Communications in Medicine format that can be readily accessed and used for qualitative and quantitative analysis. Over the several last decades, medical images have been shown to contain complementary and interchangeable data orthogonal to other sources such as pathology, hematology, genomics, and/or proteomics. As such, "radiomics" has emerged as a field of research that involves the process of converting standard-of-care images into quantitative image-based data that can be merged with other data sources and subsequently analyzed using conventional biostatistics or artificial intelligence (AI) methods. As radiomic features capture biological and pathophysiological information, these quantitative radiomic features have shown to provide rapid and accurate noninvasive biomarkers for lung cancer risk prediction, diagnostics, prognosis, treatment response monitoring, and tumor biology. In this review, radiomics and emerging AI methods in lung cancer research are highlighted and discussed including advantages, challenges, and pitfalls.


Assuntos
Inteligência Artificial , Neoplasias Pulmonares/diagnóstico por imagem , Medicina de Precisão , Diagnóstico por Imagem/tendências , Humanos , Processamento de Imagem Assistida por Computador
6.
J Immunother Cancer ; 9(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34135101

RESUMO

BACKGROUND: Currently, only a fraction of patients with non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs) experience a durable clinical benefit (DCB). According to NCCN guidelines, Programmed death-ligand 1 (PD-L1) expression status determined by immunohistochemistry (IHC) of biopsies is the only clinically approved companion biomarker to trigger the use of ICI therapy. Based on prior work showing a relationship between quantitative imaging and gene expression, we hypothesize that quantitative imaging (radiomics) can provide an alternative surrogate for PD-L1 expression status in clinical decision support. METHODS: 18F-FDG-PET/CT images and clinical data were curated from 697 patients with NSCLC from three institutions and these were analyzed using a small-residual-convolutional-network (SResCNN) to develop a deeply learned score (DLS) to predict the PD-L1 expression status. This developed model was further used to predict DCB, progression-free survival (PFS), and overall survival (OS) in two retrospective and one prospective test cohorts of ICI-treated patients with advanced stage NSCLC. RESULTS: The PD-L1 DLS significantly discriminated between PD-L1 positive and negative patients (area under receiver operating characteristics curve ≥0.82 in the training, validation, and two external test cohorts). Importantly, the DLS was indistinguishable from IHC-derived PD-L1 status in predicting PFS and OS, suggesting the utility of DLS as a surrogate for IHC. A score generated by combining the DLS with clinical characteristics was able to accurately (C-indexes of 0.70-0.87) predict DCB, PFS, and OS in retrospective training, prospective testing and external validation cohorts. CONCLUSION: Hence, we propose DLS as a surrogate or substitute for IHC-determined PD-L1 measurement to guide individual pretherapy decisions pending in larger prospective trials.


Assuntos
Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Aprendizado Profundo/normas , Imunoterapia/métodos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
7.
JNCI Cancer Spectr ; 5(4)2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34409252

RESUMO

Background: Immunotherapy yields survival benefit for some advanced stage non-small cell lung cancer (NSCLC) patients. Because highly predictive biomarkers of immunotherapy response are an unmet clinical need, we used pretreatment radiomics and clinical data to train and validate a parsimonious model associated with survival outcomes among NSCLC patients treated with immunotherapy. Methods: Three cohorts of NSCLC patients treated with immunotherapy were analyzed: training (n = 180), validation 1 (n = 90), and validation 2 (n = 62). The most informative clinical and radiomic features were subjected to decision tree analysis, which stratified patients into risk groups of low, moderate, high, and very high risk of death after initiation of immunotherapy. All statistical tests were 2-sided. Results: The very high-risk group was associated with extremely poor overall survival (OS) in validation cohorts 1 (hazard ratio [HR] = 5.35, 95% confidence interval [CI] = 2.14 to 13.36; 1-year OS = 11.1%, 95% CI = 1.9% to 29.8%; 3-year OS = 0%) and 2 (HR = 13.81, 95% CI = 2.58 to 73.93; 1-year OS = 47.6%, 95% CI = 18.2% to 72.4%; 3-year OS = 0%) when compared with the low-risk group (HR = 1.00) in validation cohorts 1 (1-year OS = 85.0%, 95% CI = 60.4% to 94.9%; 3-year OS = 38.9%, 95% CI = 17.1% to 60.3%) and 2 (1-year OS = 80.2%, 95% CI = 40.3% to 94.8%; 3-year OS = 40.1%, 95% CI = 1.3% to 83.5%). The most informative radiomic feature, gray-level co-occurrence matrix (GLCM) inverse difference, was positively associated with hypoxia-related carbonic anhydrase 9 using gene-expression profiling and immunohistochemistry. Conclusion: Utilizing standard-of-care imaging and clinical data, we identified and validated a novel parsimonious model associated with survival outcomes among NSCLC patients treated with immunotherapy. Based on this model, clinicians can identify patients who are unlikely to respond to immunotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/terapia , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/terapia , Hipóxia Tumoral , Idoso , Antígenos de Neoplasias/genética , Anidrase Carbônica IX/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Estudos de Coortes , Intervalos de Confiança , Árvores de Decisões , Feminino , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Risco , Tomografia Computadorizada por Raios X , Resultado do Tratamento
8.
Radiol Artif Intell ; 2(1): e190063, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33937811

RESUMO

PURPOSE: To investigate the performance of pretreatment fluorine 18 (18F) fluorodeoxyglucose PET/CT radiomics in predicting severe immune-related adverse events (irSAEs) among patients with advanced non-small cell lung cancer (NSCLC) treated with immunotherapy, which is important in optimizing treatment plans and alleviating future complications with early interventions. MATERIALS AND METHODS: The retrospective arm of this study included 146 patients with histologically confirmed stage IIIB-IV NSCLC who were treated with immune checkpoint blockade between June 2011 and December 2017 and who were split into training (n = 97) and test (n = 49) cohorts. A prospective validation arm enrolled 48 patients before initiation of immunotherapy between January 2018 and June 2019 as an independent test cohort. Radiomics features extracted from baseline (preimmunotherapy treatment) PET, CT, and PET/CT fusion images were used to generate a radiomics score (RS) to quantify patient risk for developing irSAEs by an improved least absolute shrinkage and selection operator method. Weighted multivariable logistic regression analysis was then used to develop a nomogram model to predict irSAEs, which was assessed by its calibration, discrimination, and clinical usefulness. RESULTS: The radiomics nomogram, incorporating the RS, type of immune checkpoint blockade, and dosing schedule, was able to predict patients with and without irSAEs with area under the receiver operating characteristic curve of 0.92 (95% confidence interval [CI]: 0.86, 0.98), 0.92 (95% CI: 0.86, 0.99), and 0.88 (95% CI: 0.78, 0.97) in the training, test, and prospective validation cohorts, respectively. Decision curve analysis showed that the radiomics nomogram model had the highest overall net benefit. CONCLUSION: A high RS is a significant risk factor for development of irSAEs, demonstrating the value of PET/CT images in predicting irSAEs. By the identification, at baseline, of patients with NSCLC most likely to have irSAEs, treatment plans can be optimized before initiation of immunotherapy.Supplemental material is available for this article.© RSNA, 2020See also the commentary by Yousefi.

9.
Sci Rep ; 10(1): 10528, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601340

RESUMO

The National Lung Screening Trial (NLST) demonstrated that screening with low-dose computed tomography (LDCT) is associated with a 20% reduction in lung cancer mortality. One potential limitation of LDCT screening is overdiagnosis of slow growing and indolent cancers. In this study, peritumoral and intratumoral radiomics was used to identify a vulnerable subset of lung patients associated with poor survival outcomes. Incident lung cancer patients from the NLST were split into training and test cohorts and an external cohort of non-screen detected adenocarcinomas was used for further validation. After removing redundant and non-reproducible radiomics features, backward elimination analyses identified a single model which was subjected to Classification and Regression Tree to stratify patients into three risk-groups based on two radiomics features (NGTDM Busyness and Statistical Root Mean Square [RMS]). The final model was validated in the test cohort and the cohort of non-screen detected adenocarcinomas. Using a radio-genomics dataset, Statistical RMS was significantly associated with FOXF2 gene by both correlation and two-group analyses. Our rigorous approach generated a novel radiomics model that identified a vulnerable high-risk group of early stage patients associated with poor outcomes. These patients may require aggressive follow-up and/or adjuvant therapy to mitigate their poor outcomes.


Assuntos
Adenocarcinoma de Pulmão/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Pulmão/diagnóstico por imagem , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/mortalidade , Idoso , Detecção Precoce de Câncer , Feminino , Fatores de Transcrição Forkhead/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Masculino , Programas de Rastreamento , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida , Tomografia Computadorizada por Raios X
10.
Nat Commun ; 11(1): 5228, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33067442

RESUMO

Two major treatment strategies employed in non-small cell lung cancer, NSCLC, are tyrosine kinase inhibitors, TKIs, and immune checkpoint inhibitors, ICIs. The choice of strategy is based on heterogeneous biomarkers that can dynamically change during therapy. Thus, there is a compelling need to identify comprehensive biomarkers that can be used longitudinally to help guide therapy choice. Herein, we report a 18F-FDG-PET/CT-based deep learning model, which demonstrates high accuracy in EGFR mutation status prediction across patient cohorts from different institutions. A deep learning score (EGFR-DLS) was significantly and positively associated with longer progression free survival (PFS) in patients treated with EGFR-TKIs, while EGFR-DLS is significantly and negatively associated with higher durable clinical benefit, reduced hyperprogression, and longer PFS among patients treated with ICIs. Thus, the EGFR-DLS provides a non-invasive method for precise quantification of EGFR mutation status in NSCLC patients, which is promising to identify NSCLC patients sensitive to EGFR-TKI or ICI-treatments.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Aprendizado Profundo , Neoplasias Pulmonares/diagnóstico por imagem , Inibidores de Proteínas Quinases/administração & dosagem , Idoso , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Mutação , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Intervalo Livre de Progressão
11.
Med Phys ; 46(11): 5075-5085, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31494946

RESUMO

PURPOSE: Recent efforts have demonstrated that radiomic features extracted from the peritumoral region, the area surrounding the tumor parenchyma, have clinical utility in various cancer types. However, as like any radiomic features, peritumoral features could also be unstable and/or nonreproducible. Hence, the purpose of this study was to assess the stability and reproducibility of computed tomography (CT) radiomic features extracted from the peritumoral regions of lung lesions where stability was defined as the consistency of a feature by different segmentations, and reproducibility was defined as the consistency of a feature to different image acquisitions. METHODS: Stability was measured utilizing the "moist run" dataset and reproducibility was measured utilizing the Reference Image Database to Evaluate Therapy Response test-retest dataset. Peritumoral radiomic features were extracted from incremental distances of 3-12 mm outside the tumor segmentation. A total of 264 statistical, histogram, and texture radiomic features were assessed from the selected peritumoral region-of-interests (ROIs). All features (except wavelet texture features) were extracted using standardized algorithms defined by the Image Biomarker Standardisation Initiative. Stability and reproducibility of features were assessed using the concordance correlation coefficient. The clinical utility of stable and reproducible peritumoral features was tested in three previously published lung cancer datasets using overall survival as the endpoint. RESULTS: Features found to be stable and reproducible, regardless of the peritumoral distances, included statistical, histogram, and a subset of texture features suggesting that these features are less affected by changes (e.g., size or shape) of the peritumoral region due to different segmentations and image acquisitions. The stability and reproducibility of Laws and wavelet texture features were inconsistent across all peritumoral distances. The analyses also revealed that a subset of features were consistently stable irrespective of the initial parameters (e.g., seed point) for a given segmentation algorithm. No significant differences were found in stability for features that were extracted from ROIs bounded by a lung parenchyma mask versus ROIs that were not bounded by a lung parenchyma mask (i.e., peritumoral regions that extended outside of lung parenchyma). After testing the clinical utility of peritumoral features, stable and reproducible features were shown to be more likely to create repeatable models than unstable and nonreproducible features. CONCLUSIONS: This study identified a subset of stable and reproducible CT radiomic features extracted from the peritumoral region of lung lesions. The stable and reproducible features identified in this study could be applied to a feature selection pipeline for CT radiomic analyses. According to our findings, top performing features in survival models were more likely to be stable and reproducible hence, it may be best practice to utilize them to achieve repeatable studies and reduce the chance of overfitting.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Humanos
12.
Lung Cancer ; 129: 75-79, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30797495

RESUMO

OBJECTIVES: Immune-checkpoint blockades have exhibited durable responses and improved long-term survival in a subset of advanced non-small cell lung cancer (NSCLC) patients. However, highly predictive markers of positive and negative responses to immunotherapy are a significant unmet clinical need. The objective of this study was to identify clinical and computational image-based predictors of rapid disease progression phenotypes in NSCLC patients treated with immune-checkpoint blockades. MATERIALS AND METHODS: Using time-to-progression (TTP) and/or tumor growth rates, rapid disease progression phenotypes were developed including hyperprogressive disease. The pre-treatment baseline predictors that were used to identify these phenotypes included patient demographics, clinical data, driver mutations, hematology data, and computational image-based features (radiomics) that were extracted from pre-treatment computed tomography scans. Synthetic Minority Oversampling Technique (SMOTE) was used to subsample minority groups to eliminate classification bias. Patient-level probabilities were calculated from the final clinical-radiomic models to subgroup patients by progression-free survival (PFS). RESULTS: Among 228 NSCLC patients treated with single agent or double agent immunotherapy, we identified parsimonious clinical-radiomic models with modest to high ability to predict rapid disease progression phenotypes with area under the receiver-operator characteristics ranging from 0.804 to 0.865. Patients who had TTP < 2 months or hyperprogressive disease were classified with 73.41% and 82.28% accuracy after SMOTE subsampling, respectively. When the patient subgroups based on patient-level probabilities were analyzed for survival outcomes, patients with higher probability scores had significantly worse PFS. CONCLUSIONS: The models found in this study have potential important translational implications to identify highly vulnerable NSCLC patients treated with immunotherapy that experience rapid disease progression and poor survival outcomes.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Imunoterapia/métodos , Neoplasias Pulmonares/diagnóstico , Pulmão/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Idoso , Biomarcadores Farmacológicos , Carcinoma Pulmonar de Células não Pequenas/terapia , Biologia Computacional , Diagnóstico por Imagem , Progressão da Doença , Feminino , Humanos , Neoplasias Pulmonares/terapia , Masculino , Fenótipo , Prognóstico
13.
Oncotarget ; 8(56): 96013-96026, 2017 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-29221183

RESUMO

The goal of this study was to extract features from radial deviation and radial gradient maps which were derived from thoracic CT scans of patients diagnosed with lung adenocarcinoma and assess whether these features are associated with overall survival. We used two independent cohorts from different institutions for training (n= 61) and test (n= 47) and focused our analyses on features that were non-redundant and highly reproducible. To reduce the number of features and covariates into a single parsimonious model, a backward elimination approach was applied. Out of 48 features that were extracted, 31 were eliminated because they were not reproducible or were redundant. We considered 17 features for statistical analysis and identified a final model containing the two most highly informative features that were associated with lung cancer survival. One of the two features, radial deviation outside-border separation standard deviation, was replicated in a test cohort exhibiting a statistically significant association with lung cancer survival (multivariable hazard ratio = 0.40; 95% confidence interval 0.17-0.97). Additionally, we explored the biological underpinnings of these features and found radial gradient and radial deviation image features were significantly associated with semantic radiological features.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA