Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Lancet Oncol ; 25(7): 879-887, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38876123

RESUMEN

BACKGROUND: Artificial intelligence (AI) systems can potentially aid the diagnostic pathway of prostate cancer by alleviating the increasing workload, preventing overdiagnosis, and reducing the dependence on experienced radiologists. We aimed to investigate the performance of AI systems at detecting clinically significant prostate cancer on MRI in comparison with radiologists using the Prostate Imaging-Reporting and Data System version 2.1 (PI-RADS 2.1) and the standard of care in multidisciplinary routine practice at scale. METHODS: In this international, paired, non-inferiority, confirmatory study, we trained and externally validated an AI system (developed within an international consortium) for detecting Gleason grade group 2 or greater cancers using a retrospective cohort of 10 207 MRI examinations from 9129 patients. Of these examinations, 9207 cases from three centres (11 sites) based in the Netherlands were used for training and tuning, and 1000 cases from four centres (12 sites) based in the Netherlands and Norway were used for testing. In parallel, we facilitated a multireader, multicase observer study with 62 radiologists (45 centres in 20 countries; median 7 [IQR 5-10] years of experience in reading prostate MRI) using PI-RADS (2.1) on 400 paired MRI examinations from the testing cohort. Primary endpoints were the sensitivity, specificity, and the area under the receiver operating characteristic curve (AUROC) of the AI system in comparison with that of all readers using PI-RADS (2.1) and in comparison with that of the historical radiology readings made during multidisciplinary routine practice (ie, the standard of care with the aid of patient history and peer consultation). Histopathology and at least 3 years (median 5 [IQR 4-6] years) of follow-up were used to establish the reference standard. The statistical analysis plan was prespecified with a primary hypothesis of non-inferiority (considering a margin of 0·05) and a secondary hypothesis of superiority towards the AI system, if non-inferiority was confirmed. This study was registered at ClinicalTrials.gov, NCT05489341. FINDINGS: Of the 10 207 examinations included from Jan 1, 2012, through Dec 31, 2021, 2440 cases had histologically confirmed Gleason grade group 2 or greater prostate cancer. In the subset of 400 testing cases in which the AI system was compared with the radiologists participating in the reader study, the AI system showed a statistically superior and non-inferior AUROC of 0·91 (95% CI 0·87-0·94; p<0·0001), in comparison to the pool of 62 radiologists with an AUROC of 0·86 (0·83-0·89), with a lower boundary of the two-sided 95% Wald CI for the difference in AUROC of 0·02. At the mean PI-RADS 3 or greater operating point of all readers, the AI system detected 6·8% more cases with Gleason grade group 2 or greater cancers at the same specificity (57·7%, 95% CI 51·6-63·3), or 50·4% fewer false-positive results and 20·0% fewer cases with Gleason grade group 1 cancers at the same sensitivity (89·4%, 95% CI 85·3-92·9). In all 1000 testing cases where the AI system was compared with the radiology readings made during multidisciplinary practice, non-inferiority was not confirmed, as the AI system showed lower specificity (68·9% [95% CI 65·3-72·4] vs 69·0% [65·5-72·5]) at the same sensitivity (96·1%, 94·0-98·2) as the PI-RADS 3 or greater operating point. The lower boundary of the two-sided 95% Wald CI for the difference in specificity (-0·04) was greater than the non-inferiority margin (-0·05) and a p value below the significance threshold was reached (p<0·001). INTERPRETATION: An AI system was superior to radiologists using PI-RADS (2.1), on average, at detecting clinically significant prostate cancer and comparable to the standard of care. Such a system shows the potential to be a supportive tool within a primary diagnostic setting, with several associated benefits for patients and radiologists. Prospective validation is needed to test clinical applicability of this system. FUNDING: Health~Holland and EU Horizon 2020.


Asunto(s)
Inteligencia Artificial , Imagen por Resonancia Magnética , Neoplasias de la Próstata , Radiólogos , Humanos , Masculino , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Anciano , Estudios Retrospectivos , Persona de Mediana Edad , Clasificación del Tumor , Países Bajos , Curva ROC
2.
J Magn Reson Imaging ; 59(5): 1800-1806, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37572098

RESUMEN

BACKGROUND: Single center MRI radiomics models are sensitive to data heterogeneity, limiting the diagnostic capabilities of current prostate cancer (PCa) radiomics models. PURPOSE: To study the impact of image resampling on the diagnostic performance of radiomics in a multicenter prostate MRI setting. STUDY TYPE: Retrospective. POPULATION: Nine hundred thirty patients (nine centers, two vendors) with 737 eligible PCa lesions, randomly split into training (70%, N = 500), validation (10%, N = 89), and a held-out test set (20%, N = 148). FIELD STRENGTH/SEQUENCE: 1.5T and 3T scanners/T2-weighted imaging (T2W), diffusion-weighted imaging (DWI), and apparent diffusion coefficient maps. ASSESSMENT: A total of 48 normalized radiomics datasets were created using various resampling methods, including different target resolutions (T2W: 0.35, 0.5, and 0.8 mm; DWI: 1.37, 2, and 2.5 mm), dimensionalities (2D/3D) and interpolation techniques (nearest neighbor, linear, Bspline and Blackman windowed-sinc). Each of the datasets was used to train a radiomics model to detect clinically relevant PCa (International Society of Urological Pathology grade ≥ 2). Baseline models were constructed using 2D and 3D datasets without image resampling. The resampling configurations with highest validation performance were evaluated in the test dataset and compared to the baseline models. STATISTICAL TESTS: Area under the curve (AUC), DeLong test. The significance level used was 0.05. RESULTS: The best 2D resampling model (T2W: Bspline and 0.5 mm resolution, DWI: nearest neighbor and 2 mm resolution) significantly outperformed the 2D baseline (AUC: 0.77 vs. 0.64). The best 3D resampling model (T2W: linear and 0.8 mm resolution, DWI: nearest neighbor and 2.5 mm resolution) significantly outperformed the 3D baseline (AUC: 0.79 vs. 0.67). DATA CONCLUSION: Image resampling has a significant effect on the performance of multicenter radiomics artificial intelligence in prostate MRI. The recommended 2D resampling configuration is isotropic resampling with T2W at 0.5 mm (Bspline interpolation) and DWI at 2 mm (nearest neighbor interpolation). For the 3D radiomics, this work recommends isotropic resampling with T2W at 0.8 mm (linear interpolation) and DWI at 2.5 mm (nearest neighbor interpolation). EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 2.


Asunto(s)
Próstata , Neoplasias de la Próstata , Masculino , Humanos , Próstata/diagnóstico por imagen , Próstata/patología , Estudios Retrospectivos , Inteligencia Artificial , Radiómica , Imagen por Resonancia Magnética/métodos , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología
3.
Eur Radiol ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937295

RESUMEN

OBJECTIVE: To review the components of past and present active surveillance (AS) protocols, provide an overview of the current studies employing artificial intelligence (AI) in AS of prostate cancer, discuss the current challenges of AI in AS, and offer recommendations for future research. METHODS: Research studies on the topic of MRI-based AI were reviewed to summarize current possibilities and diagnostic accuracies for AI methods in the context of AS. Established guidelines were used to identify possibilities for future refinement using AI. RESULTS: Preliminary results show the role of AI in a range of diagnostic tasks in AS populations, including the localization, follow-up, and prognostication of prostate cancer. Current evidence is insufficient to support a shift to AI-based AS, with studies being limited by small dataset sizes, heterogeneous inclusion and outcome definitions, or lacking appropriate benchmarks. CONCLUSION: The AI-based integration of prostate MRI is a direction that promises substantial benefits for AS in the future, but evidence is currently insufficient to support implementation. Studies with standardized inclusion criteria and standardized progression definitions are needed to support this. The increasing inclusion of patients in AS protocols and the incorporation of MRI as a scheduled examination in AS protocols may help to alleviate these challenges in future studies. CLINICAL RELEVANCE STATEMENT: This manuscript provides an overview of available evidence for the integration of prostate MRI and AI in active surveillance, addressing its potential for clinical optimizations in the context of established guidelines, while highlighting the main challenges for implementation. KEY POINTS: Active surveillance is currently based on diagnostic tests such as PSA, biopsy, and imaging. Prostate MRI and AI demonstrate promising diagnostic accuracy across a variety of tasks, including the localization, follow-up and risk estimation in active surveillance cohorts. A transition to AI-based active surveillance is not currently realistic; larger studies using standardized inclusion criteria and outcomes are necessary to improve and validate existing evidence.

4.
Eur Radiol ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724765

RESUMEN

OBJECTIVE: Deep learning (DL) MRI reconstruction enables fast scan acquisition with good visual quality, but the diagnostic impact is often not assessed because of large reader study requirements. This study used existing diagnostic DL to assess the diagnostic quality of reconstructed images. MATERIALS AND METHODS: A retrospective multisite study of 1535 patients assessed biparametric prostate MRI between 2016 and 2020. Likely clinically significant prostate cancer (csPCa) lesions (PI-RADS ≥ 4) were delineated by expert radiologists. T2-weighted scans were retrospectively undersampled, simulating accelerated protocols. DL reconstruction (DLRecon) and diagnostic DL detection (DLDetect) were developed. The effect on the partial area under (pAUC), the Free-Response Operating Characteristic (FROC) curve, and the structural similarity (SSIM) were compared as metrics for diagnostic and visual quality, respectively. DLDetect was validated with a reader concordance analysis. Statistical analysis included Wilcoxon, permutation, and Cohen's kappa tests for visual quality, diagnostic performance, and reader concordance. RESULTS: DLRecon improved visual quality at 4- and 8-fold (R4, R8) subsampling rates, with SSIM (range: -1 to 1) improved to 0.78 ± 0.02 (p < 0.001) and 0.67 ± 0.03 (p < 0.001) from 0.68 ± 0.03 and 0.51 ± 0.03, respectively. However, diagnostic performance at R4 showed a pAUC FROC of 1.33 (CI 1.28-1.39) for DL and 1.29 (CI 1.23-1.35) for naive reconstructions, both significantly lower than fully sampled pAUC of 1.58 (DL: p = 0.024, naïve: p = 0.02). Similar trends were noted for R8. CONCLUSION: DL reconstruction produces visually appealing images but may reduce diagnostic accuracy. Incorporating diagnostic AI into the assessment framework offers a clinically relevant metric essential for adopting reconstruction models into clinical practice. CLINICAL RELEVANCE STATEMENT: In clinical settings, caution is warranted when using DL reconstruction for MRI scans. While it recovered visual quality, it failed to match the prostate cancer detection rates observed in scans not subjected to acceleration and DL reconstruction.

5.
Radiology ; 308(3): e230275, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37724961

RESUMEN

Background A priori identification of patients at risk of artificial intelligence (AI) failure in diagnosing cancer would contribute to the safer clinical integration of diagnostic algorithms. Purpose To evaluate AI prediction variability as an uncertainty quantification (UQ) metric for identifying cases at risk of AI failure in diagnosing cancer at MRI and CT across different cancer types, data sets, and algorithms. Materials and Methods Multicenter data sets and publicly available AI algorithms from three previous studies that evaluated detection of pancreatic cancer on contrast-enhanced CT images, detection of prostate cancer on MRI scans, and prediction of pulmonary nodule malignancy on low-dose CT images were analyzed retrospectively. Each task's algorithm was extended to generate an uncertainty score based on ensemble prediction variability. AI accuracy percentage and partial area under the receiver operating characteristic curve (pAUC) were compared between certain and uncertain patient groups in a range of percentile thresholds (10%-90%) for the uncertainty score using permutation tests for statistical significance. The pulmonary nodule malignancy prediction algorithm was compared with 11 clinical readers for the certain group (CG) and uncertain group (UG). Results In total, 18 022 images were used for training and 838 images were used for testing. AI diagnostic accuracy was higher for the cases in the CG across all tasks (P < .001). At an 80% threshold of certain predictions, accuracy in the CG was 21%-29% higher than in the UG and 4%-6% higher than in the overall test data sets. The lesion-level pAUC in the CG was 0.25-0.39 higher than in the UG and 0.05-0.08 higher than in the overall test data sets (P < .001). For pulmonary nodule malignancy prediction, accuracy of AI was on par with clinicians for cases in the CG (AI results vs clinician results, 80% [95% CI: 76, 85] vs 78% [95% CI: 70, 87]; P = .07) but worse for cases in the UG (AI results vs clinician results, 50% [95% CI: 37, 64] vs 68% [95% CI: 60, 76]; P < .001). Conclusion An AI-prediction UQ metric consistently identified reduced performance of AI in cancer diagnosis. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Babyn in this issue.


Asunto(s)
Neoplasias Pulmonares , Trastornos Mentales , Masculino , Humanos , Inteligencia Artificial , Estudios Retrospectivos , Imagen por Resonancia Magnética , Neoplasias Pulmonares/diagnóstico por imagen , Tomografía Computarizada por Rayos X
6.
Eur Radiol ; 33(1): 64-76, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35900376

RESUMEN

OBJECTIVES: To evaluate the effect of a deep learning-based computer-aided diagnosis (DL-CAD) system on experienced and less-experienced radiologists in reading prostate mpMRI. METHODS: In this retrospective, multi-reader multi-case study, a consecutive set of 184 patients examined between 01/2018 and 08/2019 were enrolled. Ground truth was combined targeted and 12-core systematic transrectal ultrasound-guided biopsy. Four radiologists, two experienced and two less-experienced, evaluated each case twice, once without (DL-CAD-) and once assisted by DL-CAD (DL-CAD+). ROC analysis, sensitivities, specificities, PPV and NPV were calculated to compare the diagnostic accuracy for the diagnosis of prostate cancer (PCa) between the two groups (DL-CAD- vs. DL-CAD+). Spearman's correlation coefficients were evaluated to assess the relationship between PI-RADS category and Gleason score (GS). Also, the median reading times were compared for the two reading groups. RESULTS: In total, 172 patients were included in the final analysis. With DL-CAD assistance, the overall AUC of the less-experienced radiologists increased significantly from 0.66 to 0.80 (p = 0.001; cutoff ISUP GG ≥ 1) and from 0.68 to 0.80 (p = 0.002; cutoff ISUP GG ≥ 2). Experienced radiologists showed an AUC increase from 0.81 to 0.86 (p = 0.146; cutoff ISUP GG ≥ 1) and from 0.81 to 0.84 (p = 0.433; cutoff ISUP GG ≥ 2). Furthermore, the correlation between PI-RADS category and GS improved significantly in the DL-CAD + group (0.45 vs. 0.57; p = 0.03), while the median reading time was reduced from 157 to 150 s (p = 0.023). CONCLUSIONS: DL-CAD assistance increased the mean detection performance, with the most significant benefit for the less-experienced radiologist; with the help of DL-CAD less-experienced radiologists reached performances comparable to that of experienced radiologists. KEY POINTS: • DL-CAD used as a concurrent reading aid helps radiologists to distinguish between benign and cancerous lesions in prostate MRI. • With the help of DL-CAD, less-experienced radiologists may achieve detection performances comparable to that of experienced radiologists. • DL-CAD assistance increases the correlation between PI-RADS category and cancer grade.


Asunto(s)
Aprendizaje Profundo , Imágenes de Resonancia Magnética Multiparamétrica , Neoplasias de la Próstata , Masculino , Humanos , Próstata/diagnóstico por imagen , Próstata/patología , Imagen por Resonancia Magnética , Estudios Retrospectivos , Neoplasias de la Próstata/patología , Clasificación del Tumor , Biopsia Guiada por Imagen , Radiólogos , Computadores
7.
Eur Radiol ; 32(4): 2224-2234, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34786615

RESUMEN

OBJECTIVES: To assess Prostate Imaging Reporting and Data System (PI-RADS)-trained deep learning (DL) algorithm performance and to investigate the effect of data size and prior knowledge on the detection of clinically significant prostate cancer (csPCa) in biopsy-naïve men with a suspicion of PCa. METHODS: Multi-institution data included 2734 consecutive biopsy-naïve men with elevated PSA levels (≥ 3 ng/mL) that underwent multi-parametric MRI (mpMRI). mpMRI exams were prospectively reported using PI-RADS v2 by expert radiologists. A DL framework was designed and trained on center 1 data (n = 1952) to predict PI-RADS ≥ 4 (n = 1092) lesions from bi-parametric MRI (bpMRI). Experiments included varying the number of cases and the use of automatic zonal segmentation as a DL prior. Independent center 2 cases (n = 296) that included pathology outcome (systematic and MRI targeted biopsy) were used to compute performance for radiologists and DL. The performance of detecting PI-RADS 4-5 and Gleason > 6 lesions was assessed on 782 unseen cases (486 center 1, 296 center 2) using free-response ROC (FROC) and ROC analysis. RESULTS: The DL sensitivity for detecting PI-RADS ≥ 4 lesions was 87% (193/223, 95% CI: 82-91) at an average of 1 false positive (FP) per patient, and an AUC of 0.88 (95% CI: 0.84-0.91). The DL sensitivity for the detection of Gleason > 6 lesions was 85% (79/93, 95% CI: 77-83) @ 1 FP compared to 91% (85/93, 95% CI: 84-96) @ 0.3 FP for a consensus panel of expert radiologists. Data size and prior zonal knowledge significantly affected performance (4%, [Formula: see text]). CONCLUSION: PI-RADS-trained DL can accurately detect and localize Gleason > 6 lesions. DL could reach expert performance using substantially more than 2000 training cases, and DL zonal segmentation. KEY POINTS: • AI for prostate MRI analysis depends strongly on data size and prior zonal knowledge. • AI needs substantially more than 2000 training cases to achieve expert performance.


Asunto(s)
Aprendizaje Profundo , Imágenes de Resonancia Magnética Multiparamétrica , Neoplasias de la Próstata , Humanos , Biopsia Guiada por Imagen , Imagen por Resonancia Magnética/métodos , Masculino , Próstata/patología , Neoplasias de la Próstata/patología , Estudios Retrospectivos
8.
Eur Radiol ; 32(5): 3236-3247, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34913991

RESUMEN

OBJECTIVES: Multiparametric MRI has high diagnostic accuracy for detecting prostate cancer, but non-invasive prediction of tumor grade remains challenging. Characterizing tumor perfusion by exploiting the fractal nature of vascular anatomy might elucidate the aggressive potential of a tumor. This study introduces the concept of fractal analysis for characterizing prostate cancer perfusion and reports about its usefulness for non-invasive prediction of tumor grade. METHODS: We retrospectively analyzed the openly available PROSTATEx dataset with 112 cancer foci in 99 patients. In all patients, histological grading groups specified by the International Society of Urological Pathology (ISUP) were obtained from in-bore MRI-guided biopsy. Fractal analysis of dynamic contrast-enhanced perfusion MRI sequences was performed, yielding fractal dimension (FD) as quantitative descriptor. Two-class and multiclass diagnostic accuracy was analyzed using area under the curve (AUC) receiver operating characteristic analysis, and optimal FD cutoffs were established. Additionally, we compared fractal analysis to conventional apparent diffusion coefficient (ADC) measurements. RESULTS: Fractal analysis of perfusion allowed accurate differentiation of non-significant (group 1) and clinically significant (groups 2-5) cancer with a sensitivity of 91% (confidence interval [CI]: 83-96%) and a specificity of 86% (CI: 73-94%). FD correlated linearly with ISUP groups (r2 = 0.874, p < 0.001). Significant groupwise differences were obtained between low, intermediate, and high ISUP group 1-4 (p ≤ 0.001) but not group 5 tumors. Fractal analysis of perfusion was significantly more reliable than ADC in predicting non-significant and clinically significant cancer (AUCFD = 0.97 versus AUCADC = 0.77, p < 0.001). CONCLUSION: Fractal analysis of perfusion MRI accurately predicts prostate cancer grading in low-, intermediate-, and high-, but not highest-grade, tumors. KEY POINTS: • In 112 prostate carcinomas, fractal analysis of MR perfusion imaging accurately differentiated low-, intermediate-, and high-grade cancer (ISUP grade groups 1-4). • Fractal analysis detected clinically significant prostate cancer with a sensitivity of 91% (83-96%) and a specificity of 86% (73-94%). • Fractal dimension of perfusion at the tumor margin may provide an imaging biomarker to predict prostate cancer grading.


Asunto(s)
Próstata , Neoplasias de la Próstata , Fractales , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Clasificación del Tumor , Perfusión , Próstata/patología , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Estudios Retrospectivos
9.
Eur Radiol ; 32(4): 2372-2383, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34921618

RESUMEN

OBJECTIVES: Multiparametric MRI with Prostate Imaging Reporting and Data System (PI-RADS) assessment is sensitive but not specific for detecting clinically significant prostate cancer. This study validates the diagnostic accuracy of the recently suggested fractal dimension (FD) of perfusion for detecting clinically significant cancer. MATERIALS AND METHODS: Routine clinical MR imaging data, acquired at 3 T without an endorectal coil including dynamic contrast-enhanced sequences, of 72 prostate cancer foci in 64 patients were analyzed. In-bore MRI-guided biopsy with International Society of Urological Pathology (ISUP) grading served as reference standard. Previously established FD cutoffs for predicting tumor grade were compared to measurements of the apparent diffusion coefficient (25th percentile, ADC25) and PI-RADS assessment with and without inclusion of the FD as separate criterion. RESULTS: Fractal analysis allowed prediction of ISUP grade groups 1 to 4 but not 5, with high agreement to the reference standard (κFD = 0.88 [CI: 0.79-0.98]). Integrating fractal analysis into PI-RADS allowed a strong improvement in specificity and overall accuracy while maintaining high sensitivity for significant cancer detection (ISUP > 1; PI-RADS alone: sensitivity = 96%, specificity = 20%, area under the receiver operating curve [AUC] = 0.65; versus PI-RADS with fractal analysis: sensitivity = 95%, specificity = 88%, AUC = 0.92, p < 0.001). ADC25 only differentiated low-grade group 1 from pooled higher-grade groups 2-5 (κADC = 0.36 [CI: 0.12-0.59]). Importantly, fractal analysis was significantly more reliable than ADC25 in predicting non-significant and clinically significant cancer (AUCFD = 0.96 versus AUCADC = 0.75, p < 0.001). Diagnostic accuracy was not significantly affected by zone location. CONCLUSIONS: Fractal analysis is accurate in noninvasively predicting tumor grades in prostate cancer and adds independent information when implemented into PI-RADS assessment. This opens the opportunity to individually adjust biopsy priority and method in individual patients. KEY POINTS: • Fractal analysis of perfusion is accurate in noninvasively predicting tumor grades in prostate cancer using dynamic contrast-enhanced sequences (κFD = 0.88). • Including the fractal dimension into PI-RADS as a separate criterion improved specificity (from 20 to 88%) and overall accuracy (AUC from 0.86 to 0.96) while maintaining high sensitivity (96% versus 95%) for predicting clinically significant cancer. • Fractal analysis was significantly more reliable than ADC25 in predicting clinically significant cancer (AUCFD = 0.96 versus AUCADC = 0.75).


Asunto(s)
Próstata , Neoplasias de la Próstata , Fractales , Humanos , Biopsia Guiada por Imagen/métodos , Imagen por Resonancia Magnética/métodos , Masculino , Próstata/patología , Neoplasias de la Próstata/patología , Estudios Retrospectivos
10.
Eur Radiol ; 32(9): 6526-6535, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35420303

RESUMEN

OBJECTIVES: To determine the value of a deep learning masked (DLM) auto-fixed volume of interest (VOI) segmentation method as an alternative to manual segmentation for radiomics-based diagnosis of clinically significant (CS) prostate cancer (PCa) on biparametric magnetic resonance imaging (bpMRI). MATERIALS AND METHODS: This study included a retrospective multi-center dataset of 524 PCa lesions (of which 204 are CS PCa) on bpMRI. All lesions were both semi-automatically segmented with a DLM auto-fixed VOI method (averaging < 10 s per lesion) and manually segmented by an expert uroradiologist (averaging 5 min per lesion). The DLM auto-fixed VOI method uses a spherical VOI (with its center at the location of the lowest apparent diffusion coefficient of the prostate lesion as indicated with a single mouse click) from which non-prostate voxels are removed using a deep learning-based prostate segmentation algorithm. Thirteen different DLM auto-fixed VOI diameters (ranging from 6 to 30 mm) were explored. Extracted radiomics data were split into training and test sets (4:1 ratio). Performance was assessed with receiver operating characteristic (ROC) analysis. RESULTS: In the test set, the area under the ROC curve (AUCs) of the DLM auto-fixed VOI method with a VOI diameter of 18 mm (0.76 [95% CI: 0.66-0.85]) was significantly higher (p = 0.0198) than that of the manual segmentation method (0.62 [95% CI: 0.52-0.73]). CONCLUSIONS: A DLM auto-fixed VOI segmentation can provide a potentially more accurate radiomics diagnosis of CS PCa than expert manual segmentation while also reducing expert time investment by more than 97%. KEY POINTS: • Compared to traditional expert-based segmentation, a deep learning mask (DLM) auto-fixed VOI placement is more accurate at detecting CS PCa. • Compared to traditional expert-based segmentation, a DLM auto-fixed VOI placement is faster and can result in a 97% time reduction. • Applying deep learning to an auto-fixed VOI radiomics approach can be valuable.


Asunto(s)
Aprendizaje Profundo , Neoplasias de la Próstata , Imagen de Difusión por Resonancia Magnética/métodos , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Próstata/diagnóstico por imagen , Próstata/patología , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Estudios Retrospectivos
11.
Radiology ; 298(1): E18-E28, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32729810

RESUMEN

Background The coronavirus disease 2019 (COVID-19) pandemic has spread across the globe with alarming speed, morbidity, and mortality. Immediate triage of patients with chest infections suspected to be caused by COVID-19 using chest CT may be of assistance when results from definitive viral testing are delayed. Purpose To develop and validate an artificial intelligence (AI) system to score the likelihood and extent of pulmonary COVID-19 on chest CT scans using the COVID-19 Reporting and Data System (CO-RADS) and CT severity scoring systems. Materials and Methods The CO-RADS AI system consists of three deep-learning algorithms that automatically segment the five pulmonary lobes, assign a CO-RADS score for the suspicion of COVID-19, and assign a CT severity score for the degree of parenchymal involvement per lobe. This study retrospectively included patients who underwent a nonenhanced chest CT examination because of clinical suspicion of COVID-19 at two medical centers. The system was trained, validated, and tested with data from one of the centers. Data from the second center served as an external test set. Diagnostic performance and agreement with scores assigned by eight independent observers were measured using receiver operating characteristic analysis, linearly weighted κ values, and classification accuracy. Results A total of 105 patients (mean age, 62 years ± 16 [standard deviation]; 61 men) and 262 patients (mean age, 64 years ± 16; 154 men) were evaluated in the internal and external test sets, respectively. The system discriminated between patients with COVID-19 and those without COVID-19, with areas under the receiver operating characteristic curve of 0.95 (95% CI: 0.91, 0.98) and 0.88 (95% CI: 0.84, 0.93), for the internal and external test sets, respectively. Agreement with the eight human observers was moderate to substantial, with mean linearly weighted κ values of 0.60 ± 0.01 for CO-RADS scores and 0.54 ± 0.01 for CT severity scores. Conclusion With high diagnostic performance, the CO-RADS AI system correctly identified patients with COVID-19 using chest CT scans and assigned standardized CO-RADS and CT severity scores that demonstrated good agreement with findings from eight independent observers and generalized well to external data. © RSNA, 2020 Supplemental material is available for this article.


Asunto(s)
Inteligencia Artificial , COVID-19/diagnóstico por imagen , Índice de Severidad de la Enfermedad , Tórax/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Anciano , Sistemas de Datos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos de Investigación , Estudios Retrospectivos
12.
Eur Radiol ; 31(12): 9567-9578, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33991226

RESUMEN

Artificial intelligence developments are essential to the successful deployment of community-wide, MRI-driven prostate cancer diagnosis. AI systems should ensure that the main benefits of biopsy avoidance are delivered while maintaining consistent high specificities, at a range of disease prevalences. Since all current artificial intelligence / computer-aided detection systems for prostate cancer detection are experimental, multiple developmental efforts are still needed to bring the vision to fruition. Initial work needs to focus on developing systems as diagnostic supporting aids so their results can be integrated into the radiologists' workflow including gland and target outlining tasks for fusion biopsies. Developing AI systems as clinical decision-making tools will require greater efforts. The latter encompass larger multicentric, multivendor datasets where the different needs of patients stratified by diagnostic settings, disease prevalence, patient preference, and clinical setting are considered. AI-based, robust, standard operating procedures will increase the confidence of patients and payers, thus enabling the wider adoption of the MRI-directed approach for prostate cancer diagnosis. KEY POINTS: • AI systems need to ensure that the benefits of biopsy avoidance are delivered with consistent high specificities, at a range of disease prevalence. • Initial work has focused on developing systems as diagnostic supporting aids for outlining tasks, so they can be integrated into the radiologists' workflow to support MRI-directed biopsies. • Decision support tools require a larger body of work including multicentric, multivendor studies where the clinical needs, disease prevalence, patient preferences, and clinical setting are additionally defined.


Asunto(s)
Inteligencia Artificial , Neoplasias de la Próstata , Humanos , Biopsia Guiada por Imagen , Imagen por Resonancia Magnética , Masculino , Neoplasias de la Próstata/diagnóstico por imagen
13.
Eur Radiol ; 30(3): 1313-1324, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31776744

RESUMEN

OBJECTIVES: To create a radiomics approach based on multiparametric magnetic resonance imaging (mpMRI) features extracted from an auto-fixed volume of interest (VOI) that quantifies the phenotype of clinically significant (CS) peripheral zone (PZ) prostate cancer (PCa). METHODS: This study included 206 patients with 262 prospectively called mpMRI prostate imaging reporting and data system 3-5 PZ lesions. Gleason scores > 6 were defined as CS PCa. Features were extracted with an auto-fixed 12-mm spherical VOI placed around a pin point in each lesion. The value of dynamic contrast-enhanced imaging(DCE), multivariate feature selection and extreme gradient boosting (XGB) vs. univariate feature selection and random forest (RF), expert-based feature pre-selection, and the addition of image filters was investigated using the training (171 lesions) and test (91 lesions) datasets. RESULTS: The best model with features from T2-weighted (T2-w) + diffusion-weighted imaging (DWI) + DCE had an area under the curve (AUC) of 0.870 (95% CI 0.980-0.754). Removal of DCE features decreased AUC to 0.816 (95% CI 0.920-0.710), although not significantly (p = 0.119). Multivariate and XGB outperformed univariate and RF (p = 0.028). Expert-based feature pre-selection and image filters had no significant contribution. CONCLUSIONS: The phenotype of CS PZ PCa lesions can be quantified using a radiomics approach based on features extracted from T2-w + DWI using an auto-fixed VOI. Although DCE features improve diagnostic performance, this is not statistically significant. Multivariate feature selection and XGB should be preferred over univariate feature selection and RF. The developed model may be a valuable addition to traditional visual assessment in diagnosing CS PZ PCa. KEY POINTS: • T2-weighted and diffusion-weighted imaging features are essential components of a radiomics model for clinically significant prostate cancer; addition of dynamic contrast-enhanced imaging does not significantly improve diagnostic performance. • Multivariate feature selection and extreme gradient outperform univariate feature selection and random forest. • The developed radiomics model that extracts multiparametric MRI features with an auto-fixed volume of interest may be a valuable addition to visual assessment in diagnosing clinically significant prostate cancer.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Imágenes de Resonancia Magnética Multiparamétrica/métodos , Neoplasias de la Próstata/diagnóstico por imagen , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Medios de Contraste , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Neoplasias de la Próstata/patología
15.
World J Urol ; 35(12): 1849-1855, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28871396

RESUMEN

PURPOSE: To compare clinically significant prostate cancer (csPCa) detection rates between magnetic resonance imaging (MRI)-transrectal ultrasound (TRUS) fusion-guided prostate biopsy (FGB) and direct in-bore MRI-guided biopsy (MRGB). METHODS: We performed a comparison of csPCa detection rates between FGB and MRGB. Included patients had (1) at least one prior negative TRUS biopsy; (2) a Prostate Imaging Reporting and Data System (PI-RADS) 4 or 5 lesion and (3) a lesion size of ≥8 mm measured in at least one direction. We considered a Gleason score ≥7 being csPCa. Descriptive statistics with 95% confidence intervals (CI) were used to determine any differences. RESULTS: We included 51 patients with FGB (59 PI-RADS 4 and 41% PI-RADS 5) and 227 patients with MRGB (34 PI-RADS 4 and 66% PI-RADS 5). Included patients had a median age of 69 years (IQR, 65-72) and a median PSA level of 11.0 ng/ml (IQR, 7.4-15.1) and a median age of 67 years (IQR, 61-70), the median PSA 12.8 ng/ml (IQR, 9.1-19.0) within the FGB and the MRGB group, respectively. Detection rates of csPCA did not differ significantly between FGB and MRGB, 49 vs. 61%, respectively. CONCLUSION: We did not detect significant differences between FGB and MRGB in the detection of csPCa. The differences in detection ratios between both biopsy techniques are narrow with an increasing lesion size. This study warrants further studies to optimize selection of best biopsy modality.


Asunto(s)
Imagen por Resonancia Magnética Intervencional/métodos , Imagen por Resonancia Magnética/métodos , Próstata , Neoplasias de la Próstata/patología , Ultrasonografía Intervencional/métodos , Anciano , Humanos , Biopsia Guiada por Imagen/métodos , Masculino , Clasificación del Tumor , Próstata/diagnóstico por imagen , Próstata/patología , Reproducibilidad de los Resultados , Estudios Retrospectivos
16.
Radiology ; 278(1): 135-45, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26192734

RESUMEN

PURPOSE: To determine the best features to discriminate prostate cancer from benign disease and its relationship to benign disease class and cancer grade. MATERIALS AND METHODS: The institutional review board approved this study and waived the need for informed consent. A retrospective cohort of 70 patients (age range, 48-70 years; median, 62 years), all of whom were scheduled to undergo radical prostatectomy and underwent preoperative 3-T multiparametric magnetic resonance (MR) imaging, including T2-weighted, diffusion-weighted, and dynamic contrast material-enhanced imaging, were included. The digitized prostatectomy slides were annotated for cancer and noncancerous disease and coregistered to MR imaging with an interactive deformable coregistration scheme. Computer-identified features for each of the noncancerous disease categories (eg, benign prostatic hyperplasia [BPH], prostatic intraepithelial neoplasia [PIN], inflammation, and atrophy) and prostate cancer were extracted. Feature selection was performed to identify the features with the highest discriminatory power. The performance of these five features was evaluated by using the area under the receiver operating characteristic curve (AUC). RESULTS: High-b-value diffusion-weighted images were more discriminative in distinguishing BPH from prostate cancer than apparent diffusion coefficient, which was most suitable for distinguishing PIN from prostate cancer. The focal appearance of lesions on dynamic contrast-enhanced images may help discriminate atrophy and inflammation from cancer. Which imaging features are discriminative for different benign lesions is influenced by cancer grade. The apparent diffusion coefficient appeared to be the most discriminative feature in identifying high-grade cancer. Classification results showed increased performance by taking into account specific benign types (AUC = 0.70) compared with grouping all noncancerous findings together (AUC = 0.62). CONCLUSION: The best features with which to discriminate prostate cancer from noncancerous benign disease depend on the type of benign disease and cancer grade. Use of the best features may result in better diagnostic performance.


Asunto(s)
Adenocarcinoma/diagnóstico , Imagen por Resonancia Magnética/métodos , Neoplasias de la Próstata/diagnóstico , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Anciano , Diagnóstico Diferencial , Humanos , Masculino , Persona de Mediana Edad , Prostatectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Estudios Retrospectivos
17.
Eur Radiol ; 25(11): 3187-99, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26060063

RESUMEN

OBJECTIVES: To investigate the added value of computer-aided diagnosis (CAD) on the diagnostic accuracy of PIRADS reporting and the assessment of cancer aggressiveness. METHODS: Multi-parametric MRI and histopathological outcome of MR-guided biopsies of a consecutive set of 130 patients were included. All cases were prospectively PIRADS reported and the reported lesions underwent CAD analysis. Logistic regression combined the CAD prediction and radiologist PIRADS score into a combination score. Receiver-operating characteristic (ROC) analysis and Spearman's correlation coefficient were used to assess the diagnostic accuracy and correlation to cancer grade. Evaluation was performed for discriminating benign lesions from cancer and for discriminating indolent from aggressive lesions. RESULTS: In total 141 lesions (107 patients) were included for final analysis. The area-under-the-ROC-curve of the combination score was higher than for the PIRADS score of the radiologist (benign vs. cancer, 0.88 vs. 0.81, p = 0.013 and indolent vs. aggressive, 0.88 vs. 0.78, p < 0.01). The combination score correlated significantly stronger with cancer grade (0.69, p = 0.0014) than the individual CAD system or radiologist (0.54 and 0.58). CONCLUSIONS: Combining CAD prediction and PIRADS into a combination score has the potential to improve diagnostic accuracy. Furthermore, such a combination score has a strong correlation with cancer grade. KEY POINTS: • Computer-aided diagnosis helps radiologists discriminate benign findings from cancer in prostate MRI. • Combining PIRADS and computer-aided diagnosis improves differentiation between indolent and aggressive cancer. • Adding computer-aided diagnosis to PIRADS increases the correlation coefficient with respect to cancer grade.


Asunto(s)
Diagnóstico por Computador/estadística & datos numéricos , Imagen por Resonancia Magnética/estadística & datos numéricos , Neoplasias de la Próstata/patología , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Biopsia con Aguja/métodos , Estudios de Cohortes , Medios de Contraste , Imagen de Difusión por Resonancia Magnética/estadística & datos numéricos , Humanos , Imagen por Resonancia Magnética Intervencional/métodos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Valor Predictivo de las Pruebas , Estudios Prospectivos , Neoplasias de la Próstata/clasificación , Curva ROC , Sensibilidad y Especificidad , Resultado del Tratamiento
18.
Acta Radiol ; 56(4): 500-11, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24819231

RESUMEN

BACKGROUND: The use of multiparametric magnetic resonance imaging (mpMRI) to detect and localize prostate cancer has increased in recent years. In 2010, the European Society of Urogenital Radiology (ESUR) published guidelines for mpMRI and introduced the Prostate Imaging Reporting and Data System (PI-RADS) for scoring the different parameters. PURPOSE: To evaluate the reliability and diagnostic performance of endorectal 1.5-T mpMRI using the PI-RADS to localize the index tumor of prostate cancer in patients undergoing prostatectomy. MATERIAL AND METHODS: This institutional review board IRB-approved, retrospective study included 63 patients (mean age, 60.7 years, median PSA, 8.0). Three observers read mpMRI parameters (T2W, DWI, and DCE) using the PI-RADS, which were compared with the results from whole-mount histopathology that analyzed 27 regions of interest. Inter-observer agreement was calculated as well as sensitivity, specificity, positive predictive value (PPV), and negative predicted value (NPV) by dichotomizing the PI-RADS criteria scores ≥3. A receiver-operating curve (ROC) analysis was performed for the different MR parameters and overall score. RESULTS: Inter-observer agreement on the overall score was 0.41. The overall score in the peripheral zone achieved sensitivities of 0.41, 0.60, and 0.55 with an NPV of 0.80, 0.84, and 0.83, and in the transitional zone, sensitivities of 0.26, 0.15, and 0.19 with an NPV of 0.92, 0.91, and 0.92 for Observers 1, 2, and 3, respectively. The ROC analysis showed a significantly increased area under the curve (AUC) for the overall score when compared to T2W alone for two of the three observers. CONCLUSION: 1.5 T mpMRI using the PI-RADS to localize the index tumor achieved moderate reliability and diagnostic performance.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Prostatectomía/métodos , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/cirugía , Sistemas de Información Radiológica , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Masculino , Persona de Mediana Edad , Próstata/patología , Próstata/cirugía , Curva ROC , Reproducibilidad de los Resultados , Estudios Retrospectivos , Sensibilidad y Especificidad
19.
Eur Radiol ; 24(10): 2597-605, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25033819

RESUMEN

OBJECTIVES: To correlate pharmacokinetic parameters of 3-T dynamic contrast-enhanced (DCE-)MRI with histopathologic microvascular and lymphatic parameters in organ-confined prostate cancer. METHODS: In 18 patients with unilateral peripheral zone (pT2a) tumours who underwent DCE-MRI prior to radical prostatectomy (RP), the following pharmacokinetic parameters were assessed: permeability surface area volume transfer constant (K (trans)), extravascular extracellular volume (Ve) and rate constant (K ep). In the RP sections blood and lymph vessels were visualised immunohistochemically and automatically examined and analysed. Parameters assessed included microvessel density (MVD), area (MVA) and perimeter (MVP) as well as lymph vessel density (LVD), area (LVA) and perimeter (LVP). RESULTS: A negative correlation was found between age and K (trans) and K ep for tumour (r = -0.60, p = 0.009; r = -0.67, p = 0.002) and normal (r = -0.54, p = 0.021; r = -0.46, p = 0.055) tissue. No correlation existed between absolute values of microvascular parameters from histopathology and DCE-MRI. In contrast, the ratio between tumour and normal tissue (correcting for individual microvascularity variations) significantly correlated between K ep and MVD (r = 0.61, p = 0.007) and MVP (r = 0.54, p = 0.022). The lymphovascular parameters showed only a correlation between LVA and K ep (r = -0.66, p = 0.003). CONCLUSIONS: Significant correlations between DCE-MRI and histopathologic parameters were found when correcting for interpatient variations in microvascularity. KEY POINTS: • Normal prostate tissue shows strong heterogeneity in microvascularity. • Peripheral zone prostate cancer shows increased and less heterogeneous microvascularity. • Normal and tumour tissue shows considerable variation in microvascularity between patients. • DCE-MRI should take into account the interprostatic heterogeneity of microvasculature between patients.


Asunto(s)
Medios de Contraste , Detección Precoz del Cáncer/métodos , Imagen por Resonancia Magnética/métodos , Microvasos/patología , Próstata/patología , Neoplasias de la Próstata/diagnóstico , Anciano , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Próstata/irrigación sanguínea , Prostatectomía , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/cirugía , Reproducibilidad de los Resultados , Estudios Retrospectivos
20.
Cancers (Basel) ; 16(3)2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38339328

RESUMEN

CT perfusion (CTP) analysis is difficult to implement in clinical practice. Therefore, we investigated a novel semi-automated CTP AI biomarker and applied it to identify vascular phenotypes of pancreatic ductal adenocarcinoma (PDAC) and evaluate their association with overall survival (OS). METHODS: From January 2018 to November 2022, 107 PDAC patients were prospectively included, who needed to undergo CTP and a diagnostic contrast-enhanced CT (CECT). We developed a semi-automated CTP AI biomarker, through a process that involved deformable image registration, a deep learning segmentation model of tumor and pancreas parenchyma volume, and a trilinear non-parametric CTP curve model to extract the enhancement slope and peak enhancement in segmented tumors and pancreas. The biomarker was validated in terms of its use to predict vascular phenotypes and their association with OS. A receiver operating characteristic (ROC) analysis with five-fold cross-validation was performed. OS was assessed with Kaplan-Meier curves. Differences between phenotypes were tested using the Mann-Whitney U test. RESULTS: The final analysis included 92 patients, in whom 20 tumors (21%) were visually isovascular. The AI biomarker effectively discriminated tumor types, and isovascular tumors showed higher enhancement slopes (2.9 Hounsfield unit HU/s vs. 2.0 HU/s, p < 0.001) and peak enhancement (70 HU vs. 47 HU, p < 0.001); the AUC was 0.86. The AI biomarker's vascular phenotype significantly differed in OS (p < 0.01). CONCLUSIONS: The AI biomarker offers a promising tool for robust CTP analysis. In PDAC, it can distinguish vascular phenotypes with significant OS prognostication.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA