Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Cancer ; 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39023303

RESUMEN

The purpose of this study was to determine if dual-energy CT (DECT) vital iodine tumor burden (ViTB), a direct assessment of tumor vascularity, allows reliable response assessment in patients with GIST compared to established CT criteria such as RECIST1.1 and modified Choi (mChoi). From 03/2014 to 12/2019, 138 patients (64 years [32-94 years]) with biopsy proven GIST were entered in this prospective, multi-center trial. All patients were treated with tyrosine kinase inhibitors (TKI) and underwent pre-treatment and follow-up DECT examinations for a minimum of 24 months. Response assessment was performed according to RECIST1.1, mChoi, vascular tumor burden (VTB) and DECT ViTB. A change in therapy management could be because of imaging (RECIST1.1 or mChoi) and/or clinical progression. The DECT ViTB criteria had the highest discrimination ability for progression-free survival (PFS) of all criteria in both first line and second line and thereafter treatment, and was significantly superior to RECIST1.1 and mChoi (p < .034). Both, the mChoi and DECT ViTB criteria demonstrated a significantly early median time-to-progression (both delta 2.5 months; both p < .036). Multivariable analysis revealed 6 variables associated with shorter overall survival: secondary mutation (HR = 4.62), polymetastatic disease (HR = 3.02), metastatic second line and thereafter treatment (HR = 2.33), shorter PFS determined by the DECT ViTB criteria (HR = 1.72), multiple organ metastases (HR = 1.51) and lower age (HR = 1.04). DECT ViTB is a reliable response criteria and provides additional value for assessing TKI treatment in GIST patients. A significant superior response discrimination ability for median PFS was observed, including non-responders at first follow-up and patients developing resistance while on therapy.

2.
Sci Rep ; 14(1): 3758, 2024 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355768

RESUMEN

Stereotactic ablative radiotherapy (SABR) is a highly effective treatment for patients with early-stage lung cancer who are inoperable. However, SABR causes benign radiation-induced lung injury (RILI) which appears as lesion growth on follow-up CT scans. This triggers the standard definition of progressive disease, yet cancer recurrence is not usually present, and distinguishing RILI from recurrence when a lesion appears to grow in size is critical but challenging. In this study, we developed a tool to do this using scans with apparent lesion growth after SABR from 68 patients. We performed bootstrapped experiments using radiomics and explored the use of multiple regions of interest (ROIs). The best model had an area under the receiver operating characteristic curve of 0.66 and used a sphere with a diameter equal to the lesion's longest axial measurement as the ROI. We also investigated the effect of using inter-feature and volume correlation filters and found that the former was detrimental to performance and that the latter had no effect. We also found that the radiomics features ranked as highly important by the model were significantly correlated with outcomes. These findings represent a key step in developing a tool that can help determine who would benefit from follow-up invasive interventions when a SABR-treated lesion increases in size, which could help provide better treatment for patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Lesión Pulmonar , Neoplasias Pulmonares , Traumatismos por Radiación , Radiocirugia , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/patología , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/etiología , Criterios de Evaluación de Respuesta en Tumores Sólidos , Radiómica , Recurrencia Local de Neoplasia/patología , Traumatismos por Radiación/etiología , Tomografía Computarizada por Rayos X , Radiocirugia/efectos adversos
3.
Med Phys ; 51(5): 3510-3520, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38100260

RESUMEN

BACKGROUND: Patients with oropharyngeal cancer (OPC) treated with chemoradiation can experience weight loss and tumor shrinkage, altering the prescribed treatment. Treatment replanning ensures patients do not receive excessive doses to normal tissue. However, it is a time- and resource-intensive process, as it takes 1 to 2 weeks to acquire a new treatment plan, and during this time, overtreatment of normal tissues could lead to increased toxicities. Currently, there are limited prognostic factors to determine which patients will require a replan. There remains an unmet need for predictive models to assist in identifying patients who could benefit from the knowledge of a replan prior to treatment. PURPOSE: We aimed to develop and evaluate a CT-based radiomic model, integrating clinical and dosimetric information, to predict the need for a replan prior to treatment. METHODS: A dataset of patients (n = 315) with OPC treated with chemoradiation was used for this study. The dataset was split into independent training (n = 220) and testing (n = 95) datasets. Tumor volumes and organs at risk (OARs) were contoured on planning CT images. PyRadiomics was used to compute radiomic image features (n = 1218) on the original and filtered images from each of the primary tumor, nodal volumes, and ipsilateral and contralateral parotid glands. Nine clinical features and nine dose features extracted from the OARs were collected and those significantly (p < 0.05) associated with the need for a replan in the training dataset were used in a baseline model. Random forest feature selection was applied to select the optimal radiomic features to predict replanning. Logistic regression, Naïve Bayes, support vector machine, and random forest classifiers were built using the non-correlated selected radiomic, clinical, and dose features on the training dataset and performance was assessed in the testing dataset. The area under the curve (AUC) was used to assess the prognostic value. RESULTS: A total of 78 patients (25%) required a replan. Smoking status, nodal stage, base of tongue subsite, and larynx mean dose were found to be significantly associated with the need for a replan in the training dataset and incorporated into the baseline model, as well as into the combined models. Five predictive radiomic features were selected (one nodal volume, one primary tumor, two ipsilateral and one contralateral parotid gland). The baseline model comprised of clinical and dose features alone achieved an AUC of 0.66 [95% CI: 0.51-0.79] in the testing dataset. The random forest classifier was the top-performing radiomics model and achieved an AUC of 0.82 [0.75-0.89] in the training dataset and an AUC of 0.78 [0.68-0.87] in the testing dataset, which significantly outperformed the baseline model (p = 0.023, testing dataset). CONCLUSIONS: This is the first study to use radiomics from the primary tumor, nodal volumes, and parotid glands for the prediction of replanning for patients with OPC. Radiomic features augmented clinical and dose features for predicting the need for a replan in our testing dataset. Once validated, this model has the potential to assist physicians in identifying patients that may benefit from a replan, allowing for better resource allocation and reduced toxicities.


Asunto(s)
Neoplasias Orofaríngeas , Radiometría , Tomografía Computarizada por Rayos X , Neoplasias Orofaríngeas/diagnóstico por imagen , Neoplasias Orofaríngeas/radioterapia , Neoplasias Orofaríngeas/terapia , Humanos , Dosificación Radioterapéutica , Órganos en Riesgo/efectos de la radiación , Planificación de la Radioterapia Asistida por Computador/métodos , Quimioradioterapia , Masculino , Femenino , Persona de Mediana Edad , Carga Tumoral/efectos de la radiación , Anciano , Radiómica
4.
Radiother Oncol ; 178: 109434, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36464179

RESUMEN

BACKGROUND AND PURPOSE: Radiomics is a high-throughput approach that allows for quantitative analysis of imaging data for prognostic applications. Medical images are used in oropharyngeal cancer (OPC) diagnosis and treatment planning and these images may contain prognostic information allowing for treatment personalization. However, the lack of validated models has been a barrier to the translation of radiomic research to the clinic. We hypothesize that a previously developed radiomics model for risk stratification in OPC can be validated in a local dataset. MATERIALS AND METHODS: The radiomics signature predicting overall survival incorporates features derived from the primary gross tumor volume of OPC patients treated with radiation +/- chemotherapy at a single institution (n = 343). Model fit, calibration, discrimination, and utility were evaluated. The signature was compared with a clinical model using overall stage and a model incorporating both radiomics and clinical data. A model detecting dental artifacts on computed tomography images was also validated. RESULTS: The radiomics signature had a Concordance index (C-index) of 0.66 comparable to the clinical model's C-index of 0.65. The combined model significantly outperformed (C-index of 0.69, p = 0.024) the clinical model, suggesting that radiomics provides added value. The dental artifact model demonstrated strong ability in detecting dental artifacts with an area under the curve of 0.87. CONCLUSION: This work demonstrates model performance comparable to previous validation work and provides a framework for future independent and multi-center validation efforts. With sufficient validation, radiomic models have the potential to improve traditional systems of risk stratification, treatment personalization and patient outcomes.


Asunto(s)
Neoplasias Orofaríngeas , Tomografía Computarizada por Rayos X , Humanos , Pronóstico , Neoplasias Orofaríngeas/diagnóstico por imagen , Neoplasias Orofaríngeas/terapia , Estudios Retrospectivos
5.
J Med Imaging (Bellingham) ; 9(6): 066001, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36388142

RESUMEN

Purpose: We developed a model integrating multimodal quantitative imaging features from tumor and nontumor regions, qualitative features, and clinical data to improve the risk stratification of patients with resectable non-small cell lung cancer (NSCLC). Approach: We retrospectively analyzed 135 patients [mean age, 69 years (43 to 87, range); 100 male patients and 35 female patients] with NSCLC who underwent upfront surgical resection between 2008 and 2012. The tumor and peritumoral regions on both preoperative CT and FDG PET-CT and the vertebral bodies L3 to L5 on FDG PET were segmented to assess the tumor and bone marrow uptake, respectively. Radiomic features were extracted and combined with clinical and CT qualitative features. A random survival forest model was developed using the top-performing features to predict the time to recurrence/progression in the training cohort ( n = 101 ), validated in the testing cohort ( n = 34 ) using the concordance, and compared with a stage-only model. Patients were stratified into high- and low-risks of recurrence/progression using Kaplan-Meier analysis. Results: The model, consisting of stage, three wavelet texture features, and three wavelet first-order features, achieved a concordance of 0.78 and 0.76 in the training and testing cohorts, respectively, significantly outperforming the baseline stage-only model results of 0.67 ( p < 0.005 ) and 0.60 ( p = 0.008 ), respectively. Patients at high- and low-risks of recurrence/progression were significantly stratified in both the training ( p < 0.005 ) and the testing ( p = 0.03 ) cohorts. Conclusions: Our radiomic model, consisting of stage and tumor, peritumoral, and bone marrow features from CT and FDG PET-CT significantly stratified patients into low- and high-risk of recurrence/progression.

6.
PLoS One ; 17(11): e0278135, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36441690

RESUMEN

OBJECTIVES: In those undergoing treatment for head and neck cancer (HNC), sarcopenia is a strong prognostic factor for outcomes and mortality. This review identified working definitions and methods used to objectively assess sarcopenia in HNC. METHOD: The scoping review was performed in accordance with Arksey and O'Malley's five-stage methodology and the Joanna Briggs Institute guidelines. INFORMATION SOURCES: Eligible studies were identified using MEDLINE, Embase, Scopus, Cochrane Library, and CINAHL databases. STUDY SELECTION: Inclusion criteria represented studies of adult HNC patients in which sarcopenia was listed as an outcome, full-text articles written in English, and empirical research studies with a quantitative design. DATA EXTRACTION: Eligible studies were assessed using a proprietary data extraction form. General information, article details and characteristics, and details related to the concept of the scoping review were extracted in an iterative process. RESULTS: Seventy-six studies published internationally from 2016 to 2021 on sarcopenia in HNC were included. The majority were retrospective (n = 56; 74%) and the prevalence of sarcopenia ranged from 3.8% to 78.7%. Approximately two-thirds of studies used computed tomography (CT) to assess sarcopenia. Skeletal muscle index (SMI) at the third lumbar vertebra (L3) (n = 53; 70%) was the most prevalent metric used to identify sarcopenia, followed by SMI at the third cervical vertebra (C3) (n = 4; 5%). CONCLUSIONS: Currently, the most effective strategy to assess sarcopenia in HNC depends on several factors, including access to resources, patient and treatment characteristics, and the prognostic significance of outcomes used to represent sarcopenia. Skeletal muscle mass (SMM) measured at C3 may represent a practical, precise, and cost-effective biomarker for the detection of sarcopenia. However, combining SMM measurements at C3 with other sarcopenic parameters-including muscle strength and physical performance-may provide a more accurate risk profile for sarcopenia assessment and allow for a greater understanding of this condition in HNC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Sarcopenia , Adulto , Humanos , Sarcopenia/diagnóstico , Sarcopenia/epidemiología , Sarcopenia/etiología , Estudios Retrospectivos , Neoplasias de Cabeza y Cuello/complicaciones , Músculo Esquelético , Fuerza Muscular
7.
PLoS One ; 17(1): e0262639, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35061813

RESUMEN

One important metric of a radiologist's visibility and influence is their ability to participate in discussion within their community. The goal of our study was to compare the participation level of men and women in scientific discussions at the annual meeting of the Radiological Society of North America (RSNA). Eleven volunteers collected participation data by gender in 59 sessions (286 presentations) at the 2018 RSNA meeting. Data was analyzed using a combination of Chi-squared, paired Wilcoxon signed-rank and T-test. Of all RSNA professional attendees at the RSNA, 68% were men and 32% were women. Of the 2869 presentations listed in the program, 65% were presented by men and 35% were presented by women. Of the 286 presentations in our sample, 177 (61.8%) were presented by men and 109 (38.1%) were presented by women. Of these 286 presentations, 81 (63%) were moderated by men and 47 (37%) were moderated by women. From the audience, 190 male attendees participated in 134 question-and-answer (Q&A) sessions following presentations and 58 female attendees participated in 52 Q&A sessions (P<0.001). Female attendees who did participate in Q&A sessions talked for a significantly shorter period of time (mean 7.14 ± 17.7 seconds, median 0) compared to male attendees (28.7 ± 29.6 seconds, median 16; P<0.001). Overall, our findings demonstrate that women participated less than men in the Q&A sessions at RSNA 2018, and talked for a shorter period of time. The fact that women were outnumbered among their male peers may explain the difference in behavior by gender.


Asunto(s)
Congresos como Asunto/estadística & datos numéricos , Radiólogos/estadística & datos numéricos , Sexismo/estadística & datos numéricos , Movilidad Laboral , Femenino , Humanos , Masculino , Radiología/estadística & datos numéricos , Factores Sexuales
8.
Neuro Oncol ; 24(6): 986-994, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34850171

RESUMEN

BACKGROUND: The risk profile for posterior fossa ependymoma (EP) depends on surgical and molecular status [Group A (PFA) versus Group B (PFB)]. While subtotal tumor resection is known to confer worse prognosis, MRI-based EP risk-profiling is unexplored. We aimed to apply machine learning strategies to link MRI-based biomarkers of high-risk EP and also to distinguish PFA from PFB. METHODS: We extracted 1800 quantitative features from presurgical T2-weighted (T2-MRI) and gadolinium-enhanced T1-weighted (T1-MRI) imaging of 157 EP patients. We implemented nested cross-validation to identify features for risk score calculations and apply a Cox model for survival analysis. We conducted additional feature selection for PFA versus PFB and examined performance across three candidate classifiers. RESULTS: For all EP patients with GTR, we identified four T2-MRI-based features and stratified patients into high- and low-risk groups, with 5-year overall survival rates of 62% and 100%, respectively (P < .0001). Among presumed PFA patients with GTR, four T1-MRI and five T2-MRI features predicted divergence of high- and low-risk groups, with 5-year overall survival rates of 62.7% and 96.7%, respectively (P = .002). T1-MRI-based features showed the best performance distinguishing PFA from PFB with an AUC of 0.86. CONCLUSIONS: We present machine learning strategies to identify MRI phenotypes that distinguish PFA from PFB, as well as high- and low-risk PFA. We also describe quantitative image predictors of aggressive EP tumors that might assist risk-profiling after surgery. Future studies could examine translating radiomics as an adjunct to EP risk assessment when considering therapy strategies or trial candidacy.


Asunto(s)
Ependimoma , Ependimoma/diagnóstico por imagen , Ependimoma/genética , Ependimoma/patología , Humanos , Aprendizaje Automático , Imagen por Resonancia Magnética , Pronóstico , Estudios Retrospectivos
10.
Neurosurgery ; 89(3): 509-517, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34131749

RESUMEN

BACKGROUND: Clinicoradiologic differentiation between benign and malignant peripheral nerve sheath tumors (PNSTs) has important management implications. OBJECTIVE: To develop and evaluate machine-learning approaches to differentiate benign from malignant PNSTs. METHODS: We identified PNSTs treated at 3 institutions and extracted high-dimensional radiomics features from gadolinium-enhanced, T1-weighted magnetic resonance imaging (MRI) sequences. Training and test sets were selected randomly in a 70:30 ratio. A total of 900 image features were automatically extracted using the PyRadiomics package from Quantitative Imaging Feature Pipeline. Clinical data including age, sex, neurogenetic syndrome presence, spontaneous pain, and motor deficit were also incorporated. Features were selected using sparse regression analysis and retained features were further refined by gradient boost modeling to optimize the area under the curve (AUC) for diagnosis. We evaluated the performance of radiomics-based classifiers with and without clinical features and compared performance against human readers. RESULTS: A total of 95 malignant and 171 benign PNSTs were included. The final classifier model included 21 imaging and clinical features. Sensitivity, specificity, and AUC of 0.676, 0.882, and 0.845, respectively, were achieved on the test set. Using imaging and clinical features, human experts collectively achieved sensitivity, specificity, and AUC of 0.786, 0.431, and 0.624, respectively. The AUC of the classifier was statistically better than expert humans (P = .002). Expert humans were not statistically better than the no-information rate, whereas the classifier was (P = .001). CONCLUSION: Radiomics-based machine learning using routine MRI sequences and clinical features can aid in evaluation of PNSTs. Further improvement may be achieved by incorporating additional imaging sequences and clinical variables into future models.


Asunto(s)
Neoplasias de la Vaina del Nervio , Neurofibrosarcoma , Humanos , Aprendizaje Automático , Imagen por Resonancia Magnética , Neoplasias de la Vaina del Nervio/diagnóstico por imagen , Estudios Retrospectivos
11.
Neurooncol Adv ; 3(1): vdab042, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33977272

RESUMEN

BACKGROUND: Diffuse intrinsic pontine gliomas (DIPGs) are lethal pediatric brain tumors. Presently, MRI is the mainstay of disease diagnosis and surveillance. We identify clinically significant computational features from MRI and create a prognostic machine learning model. METHODS: We isolated tumor volumes of T1-post-contrast (T1) and T2-weighted (T2) MRIs from 177 treatment-naïve DIPG patients from an international cohort for model training and testing. The Quantitative Image Feature Pipeline and PyRadiomics was used for feature extraction. Ten-fold cross-validation of least absolute shrinkage and selection operator Cox regression selected optimal features to predict overall survival in the training dataset and tested in the independent testing dataset. We analyzed model performance using clinical variables (age at diagnosis and sex) only, radiomics only, and radiomics plus clinical variables. RESULTS: All selected features were intensity and texture-based on the wavelet-filtered images (3 T1 gray-level co-occurrence matrix (GLCM) texture features, T2 GLCM texture feature, and T2 first-order mean). This multivariable Cox model demonstrated a concordance of 0.68 (95% CI: 0.61-0.74) in the training dataset, significantly outperforming the clinical-only model (C = 0.57 [95% CI: 0.49-0.64]). Adding clinical features to radiomics slightly improved performance (C = 0.70 [95% CI: 0.64-0.77]). The combined radiomics and clinical model was validated in the independent testing dataset (C = 0.59 [95% CI: 0.51-0.67], Noether's test P = .02). CONCLUSIONS: In this international study, we demonstrate the use of radiomic signatures to create a machine learning model for DIPG prognostication. Standardized, quantitative approaches that objectively measure DIPG changes, including computational MRI evaluation, could offer new approaches to assessing tumor phenotype and serve a future role for optimizing clinical trial eligibility and tumor surveillance.

12.
Can Assoc Radiol J ; 72(1): 73-85, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32735452

RESUMEN

Artificial intelligence (AI)-based models have become a growing area of interest in predictive medicine and have the potential to aid physician decision-making to improve patient outcomes. Imaging and radiomics play an increasingly important role in these models. This review summarizes recent developments in the field of radiomics for AI in head and neck cancer. Prediction models for oncologic outcomes, treatment toxicity, and pathological findings have all been created. Exploratory studies are promising; however, validation studies that demonstrate consistency, reproducibility, and prognostic impact remain uncommon. Prospective clinical trials with standardized procedures are required for clinical translation.


Asunto(s)
Inteligencia Artificial , Diagnóstico por Imagen/métodos , Neoplasias de Cabeza y Cuello/diagnóstico por imagen , Interpretación de Imagen Asistida por Computador/métodos , Humanos , Pronóstico
13.
Can Assoc Radiol J ; 72(1): 86-97, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32735493

RESUMEN

Lung cancer remains the most common cause of cancer death worldwide. Recent advances in lung cancer screening, radiotherapy, surgical techniques, and systemic therapy have led to increasing complexity in diagnosis, treatment decision-making, and assessment of recurrence. Artificial intelligence (AI)-based prediction models are being developed to address these issues and may have a future role in screening, diagnosis, treatment selection, and decision-making around salvage therapy. Imaging plays an essential role in all components of lung cancer management and has the potential to play a key role in AI applications. Artificial intelligence has demonstrated value in prognostic biomarker discovery in lung cancer diagnosis, treatment, and response assessment, putting it at the forefront of the next phase of personalized medicine. However, although exploratory studies demonstrate potential utility, there is a need for rigorous validation and standardization before AI can be utilized in clinical decision-making. In this review, we will provide a summary of the current literature implementing AI for outcome prediction in lung cancer. We will describe the anticipated impact of AI on the management of patients with lung cancer and discuss the challenges of clinical implementation of these techniques.


Asunto(s)
Inteligencia Artificial , Toma de Decisiones Clínicas/métodos , Diagnóstico por Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Neoplasias Pulmonares/diagnóstico por imagen , Humanos , Pulmón/diagnóstico por imagen
14.
Radiat Oncol ; 15(1): 261, 2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33168055

RESUMEN

BACKGROUND: Transoral surgery (TOS), particularly transoral robotic surgery (TORS) has become the preferred modality in the United States for the treatment of early stage oropharyngeal cancer, largely due to assumptions of fewer toxicities and improved quality of life compared to primary radiotherapy (RT). However, these assumptions are based on retrospective analysis, a subset of which utilize primary RT groups not limited to T1-2 stage tumors for which transoral robotic surgery is FDA approved. Thus, there is potential for underestimating survival and overestimating toxicity, including treatment related mortality, in primary RT. METHODS: Consecutive cases of early T-stage (T1-T2) oropharyngeal cancer presenting to the London Health Sciences Centre between 2014 and 2018 treated with RT or chemoradiation (CRT) were reviewed. Patient demographics, treatment details, survival outcomes and toxicity were collected. Toxicities were retrospectively graded using the Common Terminology Criteria for Adverse Events criteria. RESULTS: A total of 198 patients were identified, of which 82% were male and 73% were HPV-positive. Sixty-eight percent of patients experienced a grade 2 toxicity, 48% a grade 3 and 4% a grade 4. The most frequent toxicities were dysphagia, neutropenia and ototoxicity. The rates of gastrostomy tube dependence at 1 and 2 years were 2.5% and 1% respectively. There were no grade 5 (fatal) toxicities. HPV-positive patients experienced improved 5-year overall survival (86% vs 64%, p = 0.0026). CONCLUSIONS: Primary RT or CRT provides outstanding survival for early T-stage disease, with low rates of severe toxicity and feeding tube dependence. This study provides a reference for comparison for patients treated with primary transoral surgery.


Asunto(s)
Neoplasias Orofaríngeas/radioterapia , Radioterapia de Intensidad Modulada/métodos , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Anciano , Quimioradioterapia Adyuvante , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Orofaríngeas/mortalidad , Neoplasias Orofaríngeas/patología , Radioterapia de Intensidad Modulada/efectos adversos , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Atención Terciaria de Salud
15.
Tomography ; 6(2): 111-117, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32548287

RESUMEN

Several institutions have developed image feature extraction software to compute quantitative descriptors of medical images for radiomics analyses. With radiomics increasingly proposed for use in research and clinical contexts, new techniques are necessary for standardizing and replicating radiomics findings across software implementations. We have developed a software toolkit for the creation of 3D digital reference objects with customizable size, shape, intensity, texture, and margin sharpness values. Using user-supplied input parameters, these objects are defined mathematically as continuous functions, discretized, and then saved as DICOM objects. Here, we present the definition of these objects, parameterized derivations of a subset of their radiomics values, computer code for object generation, example use cases, and a user-downloadable sample collection used for the examples cited in this paper.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Radiometría , Programas Informáticos , Radiometría/normas , Estándares de Referencia
16.
Med Phys ; 47(5): e185-e202, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32418336

RESUMEN

Radiomics is an emerging area in quantitative image analysis that aims to relate large-scale extracted imaging information to clinical and biological endpoints. The development of quantitative imaging methods along with machine learning has enabled the opportunity to move data science research towards translation for more personalized cancer treatments. Accumulating evidence has indeed demonstrated that noninvasive advanced imaging analytics, that is, radiomics, can reveal key components of tumor phenotype for multiple three-dimensional lesions at multiple time points over and beyond the course of treatment. These developments in the use of CT, PET, US, and MR imaging could augment patient stratification and prognostication buttressing emerging targeted therapeutic approaches. In recent years, deep learning architectures have demonstrated their tremendous potential for image segmentation, reconstruction, recognition, and classification. Many powerful open-source and commercial platforms are currently available to embark in new research areas of radiomics. Quantitative imaging research, however, is complex and key statistical principles should be followed to realize its full potential. The field of radiomics, in particular, requires a renewed focus on optimal study design/reporting practices and standardization of image acquisition, feature calculation, and rigorous statistical analysis for the field to move forward. In this article, the role of machine and deep learning as a major computational vehicle for advanced model building of radiomics-based signatures or classifiers, and diverse clinical applications, working principles, research opportunities, and available computational platforms for radiomics will be reviewed with examples drawn primarily from oncology. We also address issues related to common applications in medical physics, such as standardization, feature extraction, model building, and validation.


Asunto(s)
Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador/métodos , Humanos , Neoplasias/diagnóstico por imagen
17.
J Med Imaging (Bellingham) ; 7(4): 042803, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32206688

RESUMEN

Quantitative image features that can be computed from medical images are proving to be valuable biomarkers of underlying cancer biology that can be used for assessing treatment response and predicting clinical outcomes. However, validation and eventual clinical implementation of these tools is challenging due to the absence of shared software algorithms, architectures, and the tools required for computing, comparing, evaluating, and disseminating predictive models. Similarly, researchers need to have programming expertise in order to complete these tasks. The quantitative image feature pipeline (QIFP) is an open-source, web-based, graphical user interface (GUI) of configurable quantitative image-processing pipelines for both planar (two-dimensional) and volumetric (three-dimensional) medical images. This allows researchers and clinicians a GUI-driven approach to process and analyze images, without having to write any software code. The QIFP allows users to upload a repository of linked imaging, segmentation, and clinical data or access publicly available datasets (e.g., The Cancer Imaging Archive) through direct links. Researchers have access to a library of file conversion, segmentation, quantitative image feature extraction, and machine learning algorithms. An interface is also provided to allow users to upload their own algorithms in Docker containers. The QIFP gives researchers the tools and infrastructure for the assessment and development of new imaging biomarkers and the ability to use them for single and multicenter clinical and virtual clinical trials.

18.
Radiology ; 293(2): 451-459, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31526257

RESUMEN

Background Primary tumor maximum standardized uptake value is a prognostic marker for non-small cell lung cancer. In the setting of malignancy, bone marrow activity from fluorine 18-fluorodeoxyglucose (FDG) PET may be informative for clinical risk stratification. Purpose To determine whether integrating FDG PET radiomic features of the primary tumor, tumor penumbra, and bone marrow identifies lung cancer disease-free survival more accurately than clinical features alone. Materials and Methods Patients were retrospectively analyzed from two distinct cohorts collected between 2008 and 2016. Each tumor, its surrounding penumbra, and bone marrow from the L3-L5 vertebral bodies was contoured on pretreatment FDG PET/CT images. There were 156 bone marrow and 512 tumor and penumbra radiomic features computed from the PET series. Randomized sparse Cox regression by least absolute shrinkage and selection operator identified features that predicted disease-free survival in the training cohort. Cox proportional hazards models were built and locked in the training cohort, then evaluated in an independent cohort for temporal validation. Results There were 227 patients analyzed; 136 for training (mean age, 69 years ± 9 [standard deviation]; 101 men) and 91 for temporal validation (mean age, 72 years ± 10; 91 men). The top clinical model included stage; adding tumor region features alone improved outcome prediction (log likelihood, -158 vs -152; P = .007). Adding bone marrow features continued to improve performance (log likelihood, -158 vs -145; P = .001). The top model integrated stage, two bone marrow texture features, one tumor with penumbra texture feature, and two penumbra texture features (concordance, 0.78; 95% confidence interval: 0.70, 0.85; P < .001). This fully integrated model was a predictor of poor outcome in the independent cohort (concordance, 0.72; 95% confidence interval: 0.64, 0.80; P < .001) and a binary score stratified patients into high and low risk of poor outcome (P < .001). Conclusion A model that includes pretreatment fluorine 18-fluorodeoxyglucose PET texture features from the primary tumor, tumor penumbra, and bone marrow predicts disease-free survival of patients with non-small cell lung cancer more accurately than clinical features alone. © RSNA, 2019 Online supplemental material is available for this article.


Asunto(s)
Médula Ósea/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Neoplasias Pulmonares/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones , Anciano , Médula Ósea/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Fluorodesoxiglucosa F18 , Humanos , Neoplasias Pulmonares/patología , Masculino , Valor Predictivo de las Pruebas , Pronóstico , Radiofármacos , Estudios Retrospectivos , Medición de Riesgo
19.
Tomography ; 5(1): 145-153, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30854452

RESUMEN

We identified computational imaging features on 18F-fluorodeoxyglucose positron emission tomography (PET) that predict recurrence/progression in non-small cell lung cancer (NSCLC). We retrospectively identified 291 patients with NSCLC from 2 prospectively acquired cohorts (training, n = 145; validation, n = 146). We contoured the metabolic tumor volume (MTV) on all pretreatment PET images and added a 3-dimensional penumbra region that extended outward 1 cm from the tumor surface. We generated 512 radiomics features, selected 435 features based on robustness to contour variations, and then applied randomized sparse regression (LASSO) to identify features that predicted time to recurrence in the training cohort. We built Cox proportional hazards models in the training cohort and independently evaluated the models in the validation cohort. Two features including stage and a MTV plus penumbra texture feature were selected by LASSO. Both features were significant univariate predictors, with stage being the best predictor (hazard ratio [HR] = 2.15 [95% confidence interval (CI): 1.56-2.95], P < .001). However, adding the MTV plus penumbra texture feature to stage significantly improved prediction (P = .006). This multivariate model was a significant predictor of time to recurrence in the training cohort (concordance = 0.74 [95% CI: 0.66-0.81], P < .001) that was validated in a separate validation cohort (concordance = 0.74 [95% CI: 0.67-0.81], P < .001). A combined radiomics and clinical model improved NSCLC recurrence prediction. FDG PET radiomic features may be useful biomarkers for lung cancer prognosis and add clinical utility for risk stratification.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Neoplasias Pulmonares/diagnóstico por imagen , Recurrencia Local de Neoplasia/diagnóstico por imagen , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Fluorodesoxiglucosa F18 , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Variaciones Dependientes del Observador , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Radiofármacos , Reproducibilidad de los Resultados , Estudios Retrospectivos , Medición de Riesgo/métodos
20.
Int J Radiat Oncol Biol Phys ; 95(5): 1545-1546, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27479728
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...