Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Heliyon ; 10(8): e29602, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38665576

RESUMO

Objectives: To evaluate the added benefit of integrating features from pre-treatment MRI (radiomics) and digitized post-surgical pathology slides (pathomics) in prostate cancer (PCa) patients for prognosticating outcomes post radical-prostatectomy (RP) including a) rising prostate specific antigen (PSA), and b) extraprostatic-extension (EPE). Methods: Multi-institutional data (N = 58) of PCa patients who underwent pre-treatment 3-T MRI prior to RP were included in this retrospective study. Radiomic and pathomic features were extracted from PCa regions on MRI and RP specimens delineated by expert clinicians. On training set (D1, N = 44), Cox Proportional-Hazards models MR, MP and MRaP were trained using radiomics, pathomics, and their combination, respectively, to prognosticate rising PSA (PSA > 0.03 ng/mL). Top features from MRaP were used to train a model to predict EPE on D1 and test on external dataset (D2, N = 14). C-index, Kalplan-Meier curves were used for survival analysis, and area under ROC (AUC) was used for EPE. MRaP was compared with the existing post-treatment risk-calculator, CAPRA (MC). Results: Patients had median follow-up of 34 months. MRaP (c-index = 0.685 ± 0.05) significantly outperformed MR (c-index = 0.646 ± 0.05), MP (c-index = 0.631 ± 0.06) and MC (c-index = 0.601 ± 0.071) (p < 0.0001). Cross-validated Kaplan-Meier curves showed significant separation among risk groups for rising PSA for MRaP (p < 0.005, Hazard Ratio (HR) = 11.36) as compared to MR (p = 0.64, HR = 1.33), MP (p = 0.19, HR = 2.82) and MC (p = 0.10, HR = 3.05). Integrated radio-pathomic model MRaP (AUC = 0.80) outperformed MR (AUC = 0.57) and MP (AUC = 0.76) in predicting EPE on external-data (D2). Conclusions: Results from this preliminary study suggest that a combination of radiomic and pathomic features can better predict post-surgical outcomes (rising PSA and EPE) compared to either of them individually as well as extant prognostic nomogram (CAPRA).

2.
Int J Med Inform ; 182: 105303, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38088002

RESUMO

BACKGROUND: Studies about racial disparities in healthcare are increasing in quantity; however, they are subject to vast differences in definition, classification, and utilization of race/ethnicity data. Improved standardization of this information can strengthen conclusions drawn from studies using such data. The objective of this study is to examine how data related to race/ethnicity are recorded in research through examining articles on race/ethnicity health disparities and examine problems and solutions in data reporting that may impact overall data quality. METHODS: In this systematic review, Business Source Complete, Embase.com, IEEE Xplore, PubMed, Scopus and Web of Science Core Collection were searched for relevant articles published from 2000 to 2020. Search terms related to the concepts of electronic medical records, race/ethnicity, and data entry related to race/ethnicity were used. Exclusion criteria included articles not in the English language and those describing pediatric populations. Data were extracted from published articles. This review was organized and reported in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 statement for systematic reviews. FINDINGS: In this systematic review, 109 full text articles were reviewed. Weaknesses and possible solutions have been discussed in current literature, with the predominant problem and solution as follows: the electronic medical record (EMR) is vulnerable to inaccuracies and incompleteness in the methods that research staff collect this data; however, improved standardization of the collection and use of race data in patient care may help alleviate these inaccuracies. INTERPRETATION: Conclusions drawn from large datasets concerning peoples of certain race/ethnic groups should be made cautiously, and a careful review of the methodology of each publication should be considered prior to implementation in patient care.


Assuntos
Registros Eletrônicos de Saúde , Projetos de Pesquisa , Criança , Humanos , Etnicidade , Confiabilidade dos Dados , Disparidades em Assistência à Saúde
3.
Med Phys ; 51(4): 2549-2562, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37742344

RESUMO

BACKGROUND: Accurate delineations of regions of interest (ROIs) on multi-parametric magnetic resonance imaging (mpMRI) are crucial for development of automated, machine learning-based prostate cancer (PCa) detection and segmentation models. However, manual ROI delineations are labor-intensive and susceptible to inter-reader variability. Histopathology images from radical prostatectomy (RP) represent the "gold standard" in terms of the delineation of disease extents, for example, PCa, prostatitis, and benign prostatic hyperplasia (BPH). Co-registering digitized histopathology images onto pre-operative mpMRI enables automated mapping of the ground truth disease extents onto mpMRI, thus enabling the development of machine learning tools for PCa detection and risk stratification. Still, MRI-histopathology co-registration is challenging due to various artifacts and large deformation between in vivo MRI and ex vivo whole-mount histopathology images (WMHs). Furthermore, the artifacts on WMHs, such as tissue loss, may introduce unrealistic deformation during co-registration. PURPOSE: This study presents a new registration pipeline, MSERgSDM, a multi-scale feature-based registration (MSERg) with a statistical deformation (SDM) constraint, which aims to improve accuracy of MRI-histopathology co-registration. METHODS: In this study, we collected 85 pairs of MRI and WMHs from 48 patients across three cohorts. Cohort 1 (D1), comprised of a unique set of 3D printed mold data from six patients, facilitated the generation of ground truth deformations between ex vivo WMHs and in vivo MRI. The other two clinically acquired cohorts (D2 and D3) included 42 patients. Affine and nonrigid registrations were employed to minimize the deformation between ex vivo WMH and ex vivo T2-weighted MRI (T2WI) in D1. Subsequently, ground truth deformation between in vivo T2WI and ex vivo WMH was approximated as the deformation between in vivo T2WI and ex vivo T2WI. In D2 and D3, the prostate anatomical annotations, for example, tumor and urethra, were made by a pathologist and a radiologist in collaboration. These annotations included ROI boundary contours and landmark points. Before applying the registration, manual corrections were made for flipping and rotation of WMHs. MSERgSDM comprises two main components: (1) multi-scale representation construction, and (2) SDM construction. For the SDM construction, we collected N = 200 reasonable deformation fields generated using MSERg, verified through visual inspection. Three additional methods, including intensity-based registration, ProsRegNet, and MSERg, were also employed for comparison against MSERgSDM. RESULTS: Our results suggest that MSERgSDM performed comparably to the ground truth (p > 0.05). Additionally, MSERgSDM (ROI Dice ratio = 0.61, landmark distance = 3.26 mm) exhibited significant improvement over MSERg (ROI Dice ratio = 0.59, landmark distance = 3.69 mm) and ProsRegNet (ROI Dice ratio = 0.56, landmark distance = 4.00 mm) in local alignment. CONCLUSIONS: This study presents a novel registration method, MSERgSDM, for mapping ex vivo WMH onto in vivo prostate MRI. Our preliminary results demonstrate that MSERgSDM can serve as a valuable tool to map ground truth disease annotations from histopathology images onto MRI, thereby assisting in the development of machine learning models for PCa detection on MRI.


Assuntos
Próstata , Neoplasias da Próstata , Masculino , Humanos , Próstata/diagnóstico por imagem , Próstata/cirurgia , Próstata/patologia , Imageamento por Ressonância Magnética/métodos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/cirurgia , Prostatectomia , Pelve
4.
Front Oncol ; 13: 1166047, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37731630

RESUMO

Objective: The aim of this study was to quantify radiomic changes in prostate cancer (PCa) progression on serial MRI among patients on active surveillance (AS) and evaluate their association with pathologic progression on biopsy. Methods: This retrospective study comprised N = 121 biopsy-proven PCa patients on AS at a single institution, of whom N = 50 at baseline conformed to the inclusion criteria. ISUP Gleason Grade Groups (GGG) were obtained from 12-core TRUS-guided systematic biopsies at baseline and follow-up. A biopsy upgrade (AS+) was defined as an increase in GGG (or in number of positive cores) and no upgrade (AS-) was defined when GGG remained the same during a median period of 18 months. Of N = 50 patients at baseline, N = 30 had MRI scans available at follow-up (median interval = 18 months) and were included for delta radiomic analysis. A total of 252 radiomic features were extracted from the PCa region of interest identified by board-certified radiologists on 3T bi-parametric MRI [T2-weighted (T2W) and apparent diffusion coefficient (ADC)]. Delta radiomic features were computed as the difference of radiomic feature between baseline and follow-up scans. The association of AS+ with age, prostate-specific antigen (PSA), Prostate Imaging Reporting and Data System (PIRADS v2.1) score, and tumor size was evaluated at baseline and follow-up. Various prediction models were built using random forest (RF) classifier within a threefold cross-validation framework leveraging baseline radiomics (Cbr), baseline radiomics + baseline clinical (Cbrbcl), delta radiomics (CΔr), delta radiomics + baseline clinical (CΔrbcl), and delta radiomics + delta clinical (CΔrΔcl). Results: An AUC of 0.64 ± 0.09 was obtained for Cbr, which increased to 0.70 ± 0.18 with the integration of clinical variables (Cbrbcl). CΔr yielded an AUC of 0.74 ± 0.15. Integrating delta radiomics with baseline clinical variables yielded an AUC of 0.77 ± 0.23. CΔrΔclresulted in the best AUC of 0.84 ± 0.20 (p < 0.05) among all combinations. Conclusion: Our preliminary findings suggest that delta radiomics were more strongly associated with upgrade events compared to PIRADS and other clinical variables. Delta radiomics on serial MRI in combination with changes in clinical variables (PSA and tumor volume) between baseline and follow-up showed the strongest association with biopsy upgrade in PCa patients on AS. Further independent multi-site validation of these preliminary findings is warranted.

5.
Eur J Radiol Open ; 10: 100496, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37396490

RESUMO

Background: around one third of clinically significant prostate cancer (CsPCa) foci are reported to be MRI non-visible (MRI─). Objective: To quantify the differences between MR visible (MRI+) and MRI─ CsPCa using intra- and peri-lesional radiomic features on bi-parametric MRI (bpMRI). Methods: This retrospective and multi-institutional study comprised 164 patients with pre-biopsy 3T prostate multi-parametric MRI from 2014 to 2017. The MRI─ CsPCa referred to lesions with PI-RADS v2 score < 3 but ISUP grade group > 1. Three experienced radiologists were involved in annotating lesions and PI-RADS assignment. The validation set (Dv) comprised 52 patients from a single institution, the remaining 112 patients were used for training (Dt). 200 radiomic features were extracted from intra-lesional and peri-lesional regions on bpMRI.Logistic regression with least absolute shrinkage and selection operator (LASSO) and 10-fold cross-validation was applied on Dt to identify radiomic features associated with MRI─ and MRI+ CsPCa to generate corresponding risk scores RMRI─ and RMRI+. RbpMRI was further generated by integrating RMRI─ and RMRI+. Statistical significance was determined using the Wilcoxon signed-rank test. Results: Both intra-lesional and peri-lesional bpMRI Haralick and CoLlAGe radiomic features were significantly associated with MRI─ CsPCa (p < 0.05). Intra-lesional ADC Haralick and CoLlAGe radiomic features were significantly different among MRI─ and MRI+ CsPCa (p < 0.05). RbpMRI yielded the highest AUC of 0.82 (95 % CI 0.72-0.91) compared to AUCs of RMRI+ 0.76 (95 % CI 0.63-0.89), and PI-RADS 0.58 (95 % CI 0.50-0.72) on Dv. RbpMRI correctly reclassified 10 out of 14 MRI─ CsPCa on Dv. Conclusion: Our preliminary results demonstrated that both intra-lesional and peri-lesional bpMRI radiomic features were significantly associated with MRI─ CsPCa. These features could assist in CsPCa identification on bpMRI.

6.
J Am Coll Radiol ; 20(9): 842-851, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37506964

RESUMO

Despite the expert-level performance of artificial intelligence (AI) models for various medical imaging tasks, real-world performance failures with disparate outputs for various subgroups limit the usefulness of AI in improving patients' lives. Many definitions of fairness have been proposed, with discussions of various tensions that arise in the choice of an appropriate metric to use to evaluate bias; for example, should one aim for individual or group fairness? One central observation is that AI models apply "shortcut learning" whereby spurious features (such as chest tubes and portable radiographic markers on intensive care unit chest radiography) on medical images are used for prediction instead of identifying true pathology. Moreover, AI has been shown to have a remarkable ability to detect protected attributes of age, sex, and race, while the same models demonstrate bias against historically underserved subgroups of age, sex, and race in disease diagnosis. Therefore, an AI model may take shortcut predictions from these correlations and subsequently generate an outcome that is biased toward certain subgroups even when protected attributes are not explicitly used as inputs into the model. As a result, these subgroups became nonprivileged subgroups. In this review, the authors discuss the various types of bias from shortcut learning that may occur at different phases of AI model development, including data bias, modeling bias, and inference bias. The authors thereafter summarize various tool kits that can be used to evaluate and mitigate bias and note that these have largely been applied to nonmedical domains and require more evaluation for medical AI. The authors then summarize current techniques for mitigating bias from preprocessing (data-centric solutions) and during model development (computational solutions) and postprocessing (recalibration of learning). Ongoing legal changes where the use of a biased model will be penalized highlight the necessity of understanding, detecting, and mitigating biases from shortcut learning and will require diverse research teams looking at the whole AI pipeline.


Assuntos
Inteligência Artificial , Radiologia , Humanos , Radiografia , Causalidade , Viés
7.
PLoS Comput Biol ; 19(1): e1010778, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36602952

RESUMO

Medical imaging is a great asset for modern medicine, since it allows physicians to spatially interrogate a disease site, resulting in precise intervention for diagnosis and treatment, and to observe particular aspect of patients' conditions that otherwise would not be noticeable. Computational analysis of medical images, moreover, can allow the discovery of disease patterns and correlations among cohorts of patients with the same disease, thus suggesting common causes or providing useful information for better therapies and cures. Machine learning and deep learning applied to medical images, in particular, have produced new, unprecedented results that can pave the way to advanced frontiers of medical discoveries. While computational analysis of medical images has become easier, however, the possibility to make mistakes or generate inflated or misleading results has become easier, too, hindering reproducibility and deployment. In this article, we provide ten quick tips to perform computational analysis of medical images avoiding common mistakes and pitfalls that we noticed in multiple studies in the past. We believe our ten guidelines, if taken into practice, can help the computational-medical imaging community to perform better scientific research that eventually can have a positive impact on the lives of patients worldwide.


Assuntos
Aprendizado de Máquina , Humanos , Reprodutibilidade dos Testes
8.
Front Oncol ; 12: 841801, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35669420

RESUMO

Objective: To derive and evaluate the association of prostate shape distension descriptors from T2-weighted MRI (T2WI) with prostate cancer (PCa) biochemical recurrence (BCR) post-radical prostatectomy (RP) independently and in conjunction with texture radiomics of PCa. Methods: This retrospective study comprised 133 PCa patients from two institutions who underwent 3T-MRI prior to RP and were followed up with PSA measurements for ≥3 years. A 3D shape atlas-based approach was adopted to derive prostate shape distension descriptors from T2WI, and these descriptors were used to train a random forest classifier (CS ) to predict BCR. Texture radiomics was derived within PCa regions of interest from T2WI and ADC maps, and another machine learning classifier (CR ) was trained for BCR. An integrated classifier CS + R was then trained using predictions from CS and CR . These models were trained on D1 (N = 71, 27 BCR+) and evaluated on independent hold-out set D2 (N = 62, 12 BCR+). CS + R was compared against pre-RP, post-RP clinical variables, and extant nomograms for BCR-free survival (bFS) at 3 years. Results: CS + R resulted in a higher AUC (0.75) compared to CR (0.70, p = 0.04) and CS (0.69, p = 0.01) on D2 in predicting BCR. On univariable analysis, CS + R achieved a higher hazard ratio (2.89, 95% CI 0.35-12.81, p < 0.01) compared to other pre-RP clinical variables for bFS. CS + R , pathologic Gleason grade, extraprostatic extension, and positive surgical margins were associated with bFS (p < 0.05). CS + R resulted in a higher C-index (0.76 ± 0.06) compared to CAPRA (0.69 ± 0.09, p < 0.01) and Decipher risk (0.59 ± 0.06, p < 0.01); however, it was comparable to post-RP CAPRA-S (0.75 ± 0.02, p = 0.07). Conclusions: Radiomic shape descriptors quantifying prostate surface distension complement texture radiomics of prostate cancer on MRI and result in an improved association with biochemical recurrence post-radical prostatectomy.

9.
Chin J Cancer Res ; 33(5): 563-573, 2021 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-34815630

RESUMO

In the last decade, the focus of computational pathology research community has shifted from replicating the pathological examination for diagnosis done by pathologists to unlocking and discovering "sub-visual" prognostic image cues from the histopathological image. While we are getting more knowledge and experience in digital pathology, the emerging goal is to integrate other-omics or modalities that will contribute for building a better prognostic assay. In this paper, we provide a brief review of representative works that focus on integrating pathomics with radiomics and genomics for cancer prognosis. It includes: correlation of pathomics and genomics; fusion of pathomics and genomics; fusion of pathomics and radiomics. We also present challenges, potential opportunities, and avenues for future work.

10.
Lancet Digit Health ; 3(7): e445-e454, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34167765

RESUMO

BACKGROUND: Biparametric MRI (comprising T2-weighted MRI and apparent diffusion coefficient maps) is increasingly being used to characterise prostate cancer. Although previous studies have combined Prostate Imaging-Reporting & Data System (PI-RADS)-based MRI findings with routinely available clinical variables and with deep learning-based imaging predictors, respectively, for prostate cancer risk stratification, none have combined all three. We aimed to construct an integrated nomogram (referred to as ClaD) combining deep learning-based imaging predictions, PI-RADS scoring, and clinical variables to identify clinically significant prostate cancer on biparametric MRI. METHODS: In this retrospective multicentre study, we included patients with prostate cancer, with histopathology or biopsy reports and a screening or diagnostic MRI scan in the axial view, from four cohorts in the USA (from University Hospitals Cleveland Medical Center, Icahn School of Medicine at Mount Sinai, Cleveland Clinic, and Long Island Jewish Medical Center) and from the PROSTATEx Challenge dataset in the Netherlands. We constructed an integrated nomogram combining deep learning, PI-RADS score, and clinical variables (prostate-specific antigen, prostate volume, and lesion volume) using multivariable logistic regression to identify clinically significant prostate cancer on biparametric MRI. We used data from the first three cohorts to train the nomogram and data from the remaining two cohorts for independent validation. We compared the performance of our ClaD integrated nomogram with that of integrated nomograms combining clinical variables with either the deep learning-based imaging predictor (referred to as DIN) or PI-RADS score (referred to as PIN) using area under the receiver operating characteristic curves (AUCs). We also compared the ability of the nomograms to predict biochemical recurrence on a subset of patients who had undergone radical prostatectomy. We report cross-validation AUCs as means for the training set and used AUCs with 95% CIs to assess the performance on the test set. The difference in AUCs between the models were tested for statistical significance using DeLong's test. We used log-rank tests and Kaplan-Meier curves to analyse survival. FINDINGS: We investigated 592 patients (823 lesions) with prostate cancer who underwent 3T multiparametric MRI at five hospitals in the USA between Jan 8, 2009, and June 3, 2017. The training data set consisted of 368 patients from three sites (the PROSTATEx Challenge cohort [n=204], University Hospitals Cleveland Medical Center [n=126], and Icahn School of Medicine at Mount Sinai [n=38]), and the independent validation data set consisted of 224 patients from two sites (Cleveland Clinic [n=151] and Long Island Jewish Medical Center [n=73]). The ClaD clinical nomogram yielded an AUC of 0·81 (95% CI 0·76-0·85) for identification of clinically significant prostate cancer in the validation data set, significantly improving performance over the DIN (0·74 [95% CI 0·69-0·80], p=0·0005) and PIN (0·76 [0·71-0·81], p<0·0001) nomograms. In the subset of patients who had undergone radical prostatectomy (n=81), the ClaD clinical nomogram resulted in a significant separation in Kaplan-Meier survival curves between patients with and without biochemical recurrence (HR 5·92 [2·34-15·00], p=0·044), whereas the DIN (1·22 [0·54-2·79], p=0·65) and PIN nomograms did not (1·30 [0·62-2·71], p=0·51). INTERPRETATION: Risk stratification of patients with prostate cancer using the integrated ClaD nomogram could help to identify patients with prostate cancer who are at low risk, very low risk, and favourable intermediate risk, who might be candidates for active surveillance, and could also help to identify patients with lethal prostate cancer who might benefit from adjuvant therapy. FUNDING: National Cancer Institute of the US National Institutes of Health, National Institute for Biomedical Imaging and Bioengineering, National Center for Research Resources, US Department of Veterans Affairs Biomedical Laboratory Research and Development Service, US Department of Defense, US National Institute of Diabetes and Digestive and Kidney Diseases, The Ohio Third Frontier Technology Validation Fund, Case Western Reserve University, Dana Foundation, and Clinical and Translational Science Collaborative.


Assuntos
Aprendizado Profundo , Imageamento por Ressonância Magnética Multiparamétrica , Nomogramas , Neoplasias da Próstata/diagnóstico por imagem , Sistemas de Dados , Humanos , Masculino , Projetos de Pesquisa , Estudos Retrospectivos
12.
NPJ Precis Oncol ; 5(1): 35, 2021 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-33941830

RESUMO

Existing tools for post-radical prostatectomy (RP) prostate cancer biochemical recurrence (BCR) prognosis rely on human pathologist-derived parameters such as tumor grade, with the resulting inter-reviewer variability. Genomic companion diagnostic tests such as Decipher tend to be tissue destructive, expensive, and not routinely available in most centers. We present a tissue non-destructive method for automated BCR prognosis, termed "Histotyping", that employs computational image analysis of morphologic patterns of prostate tissue from a single, routinely acquired hematoxylin and eosin slide. Patients from two institutions (n = 214) were used to train Histotyping for identifying high-risk patients based on six features of glandular morphology extracted from RP specimens. Histotyping was validated for post-RP BCR prognosis on a separate set of n = 675 patients from five institutions and compared against Decipher on n = 167 patients. Histotyping was prognostic of BCR in the validation set (p < 0.001, univariable hazard ratio [HR] = 2.83, 95% confidence interval [CI]: 2.03-3.93, concordance index [c-index] = 0.68, median years-to-BCR: 1.7). Histotyping was also prognostic in clinically stratified subsets, such as patients with Gleason grade group 3 (HR = 4.09) and negative surgical margins (HR = 3.26). Histotyping was prognostic independent of grade group, margin status, pathological stage, and preoperative prostate-specific antigen (PSA) (multivariable p < 0.001, HR = 2.09, 95% CI: 1.40-3.10, n = 648). The combination of Histotyping, grade group, and preoperative PSA outperformed Decipher (c-index = 0.75 vs. 0.70, n = 167). These results suggest that a prognostic classifier for prostate cancer based on digital images could serve as an alternative or complement to molecular-based companion diagnostic tests.

13.
Eur Radiol ; 31(3): 1336-1346, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32876839

RESUMO

OBJECTIVES: To explore the associations between T1 and T2 magnetic resonance fingerprinting (MRF) measurements and corresponding tissue compartment ratios (TCRs) on whole mount histopathology of prostate cancer (PCa) and prostatitis. MATERIALS AND METHODS: A retrospective, IRB-approved, HIPAA-compliant cohort consisting of 14 PCa patients who underwent 3 T multiparametric MRI along with T1 and T2 MRF maps prior to radical prostatectomy was used. Correspondences between whole mount specimens and MRI and MRF were manually established. Prostatitis, PCa, and normal peripheral zone (PZ) regions of interest (ROIs) on pathology were segmented for TCRs of epithelium, lumen, and stroma using two U-net deep learning models. Corresponding ROIs were mapped to T2-weighted MRI (T2w), apparent diffusion coefficient (ADC), and T1 and T2 MRF maps. Their correlations with TCRs were computed using Pearson's correlation coefficient (R). Statistically significant differences in means were assessed using one-way ANOVA. RESULTS: Statistically significant differences (p < 0.01) in means of TCRs and T1 and T2 MRF were observed between PCa, prostatitis, and normal PZ. A negative correlation was observed between T1 and T2 MRF and epithelium (R = - 0.38, - 0.44, p < 0.05) of PCa. T1 MRF was correlated in opposite directions with stroma of PCa and prostatitis (R = 0.35, - 0.44, p < 0.05). T2 MRF was positively correlated with lumen of PCa and prostatitis (R = 0.57, 0.46, p < 0.01). Mean T2 MRF showed significant differences (p < 0.01) between PCa and prostatitis across both transition zone (TZ) and PZ, while mean T1 MRF was significant (p = 0.02) in TZ. CONCLUSION: Significant associations between MRF (T1 in the TZ and T2 in the PZ) and tissue compartments on corresponding histopathology were observed. KEY POINTS: • Mean T2 MRF measurements and ADC within cancerous regions of interest dropped with increasing ISUP prognostic groups (IPG). • Mean T1 and T2 MRF measurements were significantly different (p < 0.001) across IPGs, prostatitis, and normal peripheral zone (NPZ). • T2 MRF showed stronger correlations in the peripheral zone, while T1 MRF showed stronger correlations in the transition zone with histopathology for prostate cancer.


Assuntos
Aprendizado Profundo , Neoplasias da Próstata , Prostatite , Imagem de Difusão por Ressonância Magnética , Epitélio , Humanos , Imageamento por Ressonância Magnética , Masculino , Neoplasias da Próstata/diagnóstico por imagem , Prostatite/diagnóstico por imagem , Estudos Retrospectivos
14.
Eur Radiol ; 31(1): 379-391, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32700021

RESUMO

OBJECTIVES: To evaluate short-term test-retest repeatability of a deep learning architecture (U-Net) in slice- and lesion-level detection and segmentation of clinically significant prostate cancer (csPCa: Gleason grade group > 1) using diffusion-weighted imaging fitted with monoexponential function, ADCm. METHODS: One hundred twelve patients with prostate cancer (PCa) underwent 2 prostate MRI examinations on the same day. PCa areas were annotated using whole mount prostatectomy sections. Two U-Net-based convolutional neural networks were trained on three different ADCm b value settings for (a) slice- and (b) lesion-level detection and (c) segmentation of csPCa. Short-term test-retest repeatability was estimated using intra-class correlation coefficient (ICC(3,1)), proportionate agreement, and dice similarity coefficient (DSC). A 3-fold cross-validation was performed on training set (N = 78 patients) and evaluated for performance and repeatability on testing data (N = 34 patients). RESULTS: For the three ADCm b value settings, repeatability of mean ADCm of csPCa lesions was ICC(3,1) = 0.86-0.98. Two CNNs with U-Net-based architecture demonstrated ICC(3,1) in the range of 0.80-0.83, agreement of 66-72%, and DSC of 0.68-0.72 for slice- and lesion-level detection and segmentation of csPCa. Bland-Altman plots suggest that there is no systematic bias in agreement between inter-scan ground truth segmentation repeatability and segmentation repeatability of the networks. CONCLUSIONS: For the three ADCm b value settings, two CNNs with U-Net-based architecture were repeatable for the problem of detection of csPCa at the slice-level. The network repeatability in segmenting csPCa lesions is affected by inter-scan variability and ground truth segmentation repeatability and may thus improve with better inter-scan reproducibility. KEY POINTS: • For the three ADCm b value settings, two CNNs with U-Net-based architecture were repeatable for the problem of detection of csPCa at the slice-level. • The network repeatability in segmenting csPCa lesions is affected by inter-scan variability and ground truth segmentation repeatability and may thus improve with better inter-scan reproducibility.


Assuntos
Aprendizado Profundo , Neoplasias da Próstata , Imagem de Difusão por Ressonância Magnética , Humanos , Masculino , Redes Neurais de Computação , Neoplasias da Próstata/diagnóstico por imagem , Reprodutibilidade dos Testes
16.
EBioMedicine ; 63: 103163, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33321450

RESUMO

BACKGROUND: We developed and validated an integrated radiomic-clinicopathologic nomogram (RadClip) for post-surgical biochemical recurrence free survival (bRFS) and adverse pathology (AP) prediction in men with prostate cancer (PCa). RadClip was further compared against extant prognostics tools like CAPRA and Decipher. METHODS: A retrospective study of 198 patients with PCa from four institutions who underwent pre-operative 3 Tesla MRI followed by radical prostatectomy, between 2009 and 2017 with a median 35-month follow-up was performed. Radiomic features were extracted from prostate cancer regions on bi-parametric magnetic resonance imaging (bpMRI). Cox Proportional-Hazards (CPH) model warped with minimum redundancy maximum relevance (MRMR) feature selection was employed to select bpMRI radiomic features for bRFS prediction in the training set (D1, N = 71). In addition, a bpMRI radiomic risk score (RadS) and associated nomogram, RadClip, were constructed in D1 and then compared against the Decipher, pre-operative (CAPRA), and post-operative (CAPRA-S) nomograms for bRFS and AP prediction in the testing set (D2, N = 127). FINDINGS: "RadClip yielded a higher C-index (0.77, 95% CI 0.65-0.88) compared to CAPRA (0.68, 95% CI 0.57-0.8) and Decipher (0.51, 95% CI 0.33-0.69) and was found to be comparable to CAPRA-S (0.75, 95% CI 0.65-0.85). RadClip resulted in a higher AUC (0.71, 95% CI 0.62-0.81) for predicting AP compared to Decipher (0.66, 95% CI 0.56-0.77) and CAPRA (0.69, 95% CI 0.59-0.79)." INTERPRETATION: RadClip was more prognostic of bRFS and AP compared to Decipher and CAPRA. It could help pre-operatively identify PCa patients at low risk of biochemical recurrence and AP and who therefore might defer additional therapy. FUNDING: The National Institutes of Health, the U.S. Department of Veterans Affairs, and the Department of Defense.


Assuntos
Diagnóstico por Imagem , Assistência Perioperatória , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/mortalidade , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais , Tomada de Decisão Clínica , Diagnóstico por Imagem/métodos , Gerenciamento Clínico , Humanos , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Nomogramas , Seleção de Pacientes , Prognóstico , Prostatectomia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Estudos Retrospectivos , Fluxo de Trabalho
17.
Cancers (Basel) ; 12(8)2020 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-32781640

RESUMO

Background: Prostate cancer (PCa) influences its surrounding habitat, which tends to manifest as different phenotypic appearances on magnetic resonance imaging (MRI). This region surrounding the PCa lesion, or the peri-tumoral region, may encode useful information that can complement intra-tumoral information to enable better risk stratification. Purpose: To evaluate the role of peri-tumoral radiomic features on bi-parametric MRI (T2-weighted and Diffusion-weighted) to distinguish PCa risk categories as defined by D'Amico Risk Classification System. Materials and Methods: We studied a retrospective, HIPAA-compliant, 4-institution cohort of 231 PCa patients (n = 301 lesions) who underwent 3T multi-parametric MRI prior to biopsy. PCa regions of interest (ROIs) were delineated on MRI by experienced radiologists following which peri-tumoral ROIs were defined. Radiomic features were extracted within the intra- and peri-tumoral ROIs. Radiomic features differentiating low-risk from: (1) high-risk (L-vs.-H), and (2) (intermediate- and high-risk (L-vs.-I + H)) lesions were identified. Using a multi-institutional training cohort of 151 lesions (D1, N = 116 patients), machine learning classifiers were trained using peri- and intra-tumoral features individually and in combination. The remaining 150 lesions (D2, N = 115 patients) were used for independent hold-out validation and were evaluated using Receiver Operating Characteristic (ROC) analysis and compared with PI-RADS v2 scores. Results: Validation on D2 using peri-tumoral radiomics alone resulted in areas under the ROC curve (AUCs) of 0.84 and 0.73 for the L-vs.-H and L-vs.-I + H classifications, respectively. The best combination of intra- and peri-tumoral features resulted in AUCs of 0.87 and 0.75 for the L-vs.-H and L-vs.-I + H classifications, respectively. This combination improved the risk stratification results by 3-6% compared to intra-tumoral features alone. Our radiomics-based model resulted in a 53% accuracy in differentiating L-vs.-H compared to PI-RADS v2 (48%), on the validation set. Conclusion: Our findings suggest that peri-tumoral radiomic features derived from prostate bi-parametric MRI add independent predictive value to intra-tumoral radiomic features for PCa risk assessment.

18.
Magn Reson Med ; 83(6): 2293-2309, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31703155

RESUMO

PURPOSE: To evaluate repeatability of prostate DWI-derived radiomics and machine learning methods for prostate cancer (PCa) characterization. METHODS: A total of 112 patients with diagnosed PCa underwent 2 prostate MRI examinations (Scan1 and Scan2) performed on the same day. DWI was performed using 12 b-values (0-2000 s/mm2 ), post-processed using kurtosis function, and PCa areas were annotated using whole mount prostatectomy sections. A total of 1694 radiomic features including Sobel, Kirch, Gradient, Zernike Moments, Gabor, Haralick, CoLIAGe, Haar wavelet coefficients, 3D analogue to Laws features, 2D contours, and corner detectors were calculated. Radiomics and 4 feature pruning methods (area under the receiver operator characteristic curve, maximum relevance minimum redundancy, Spearman's ρ, Wilcoxon rank-sum) were evaluated in terms of Scan1-Scan2 repeatability using intraclass correlation coefficient (ICC)(3,1). Classification performance for clinically significant and insignificant PCa with Gleason grade groups 1 versus >1 was evaluated by area under the receiver operator characteristic curve in unseen random 30% data split. RESULTS: The ICC(3,1) values for conventional radiomics and feature pruning methods were in the range of 0.28-0.90. The machine learning classifications varied between Scan1 and Scan2 with % of same class labels between Scan1 and Scan2 in the range of 61-81%. Surface-to-volume ratio and corner detector-based features were among the most represented features with high repeatability, ICC(3,1) >0.75, consistently high ranking using all 4 feature pruning methods, and classification performance with area under the receiver operator characteristic curve >0.70. CONCLUSION: Surface-to-volume ratio and corner detectors for prostate DWI led to good classification of unseen data and performed similarly in Scan1 and Scan2 in contrast to multiple conventional radiomic features.


Assuntos
Neoplasias da Próstata , Humanos , Aprendizado de Máquina , Masculino , Gradação de Tumores , Prostatectomia , Neoplasias da Próstata/diagnóstico por imagem
19.
J Magn Reson Imaging ; 48(6): 1626-1636, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29734484

RESUMO

BACKGROUND: Radiomics or computer-extracted texture features derived from MRI have been shown to help quantitatively characterize prostate cancer (PCa). Radiomics have not been explored depth in the context of predicting biochemical recurrence (BCR) of PCa. PURPOSE: To identify a set of radiomic features derived from pretreatment biparametric MRI (bpMRI) that may be predictive of PCa BCR. STUDY TYPE: Retrospective. SUBJECTS: In all, 120 PCa patients from two institutions, I1 and I2 , partitioned into training set D1 (N = 70) from I1 and independent validation set D2 (N = 50) from I2 . All patients were followed for ≥3 years. SEQUENCE: 3T, T2 -weighted (T2 WI) and apparent diffusion coefficient (ADC) maps derived from diffusion-weighted sequences. ASSESSMENT: PCa regions of interest (ROIs) on T2 WI were annotated by two experienced radiologists. Radiomic features from bpMRI (T2 WI and ADC maps) were extracted from the ROIs. A machine-learning classifier (CBCR ) was trained with the best discriminating set of radiomic features to predict BCR (pBCR ). STATISTICAL TESTS: Wilcoxon rank-sum tests with P < 0.05 were considered statistically significant. Differences in BCR-free survival at 3 years using pBCR was assessed using the Kaplan-Meier method and compared with Gleason Score (GS), PSA, and PIRADS-v2. RESULTS: Distribution statistics of co-occurrence of local anisotropic gradient orientation (CoLlAGe) and Haralick features from T2 WI and ADC were associated with BCR (P < 0.05) on D1 . CBCR predictions resulted in a mean AUC = 0.84 on D1 and AUC = 0.73 on D2 . A significant difference in BCR-free survival between the predicted classes (BCR + and BCR-) was observed (P = 0.02) on D2 compared to those obtained from GS (P = 0.8), PSA (P = 0.93) and PIRADS-v2 (P = 0.23). DATA CONCLUSION: Radiomic features from pretreatment bpMRI can be predictive of PCa BCR after therapy and may help identify men who would benefit from adjuvant therapy. LEVEL OF EVIDENCE: 4 Technical Efficacy: Stage 5 J. Magn. Reson. Imaging 2018;48:1626-1636.


Assuntos
Diagnóstico por Computador , Imageamento por Ressonância Magnética , Neoplasias da Próstata/diagnóstico por imagem , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Imagem de Difusão por Ressonância Magnética , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Processamento de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Recidiva Local de Neoplasia , Reconhecimento Automatizado de Padrão , Curva ROC , Radiometria , Estudos Retrospectivos
20.
J Magn Reson Imaging ; 2018 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-29469937

RESUMO

BACKGROUND: Radiomic analysis is defined as computationally extracting features from radiographic images for quantitatively characterizing disease patterns. There has been recent interest in examining the use of MRI for identifying prostate cancer (PCa) aggressiveness in patients on active surveillance (AS). PURPOSE: To evaluate the performance of MRI-based radiomic features in identifying the presence or absence of clinically significant PCa in AS patients. STUDY TYPE: Retrospective. SUBJECTS MODEL: MRI/TRUS (transperineal grid ultrasound) fusion-guided biopsy was performed for 56 PCa patients on AS who had undergone prebiopsy. FIELD STRENGTH/SEQUENCE: 3T, T2 -weighted (T2 w) and diffusion-weighted (DW) MRI. ASSESSMENT: A pathologist histopathologically defined the presence of clinically significant disease. A radiologist manually delineated lesions on T2 w-MRs. Then three radiologists assessed MRIs using PIRADS v2.0 guidelines. Tumors were categorized into four groups: MRI-negative-biopsy-negative (Group 1, N = 15), MRI-positive-biopsy-positive (Group 2, N = 16), MRI-negative-biopsy-positive (Group 3, N = 10), and MRI-positive-biopsy-negative (Group 4, N = 15). In all, 308 radiomic features (First-order statistics, Gabor, Laws Energy, and Haralick) were extracted from within the annotated lesions on T2 w images and apparent diffusion coefficient (ADC) maps. The top 10 features associated with clinically significant tumors were identified using minimum-redundancy-maximum-relevance and used to construct three machine-learning models that were independently evaluated for their ability to identify the presence and absence of clinically significant disease. STATISTICAL TESTS: Wilcoxon rank-sum tests with P < 0.05 considered statistically significant. RESULTS: Seven T2 w-based (First-order Statistics, Haralick, Laws, and Gabor) and three ADC-based radiomic features (Laws, Gradient and Sobel) exhibited statistically significant differences (P < 0.001) between malignant and normal regions in the training groups. The three constructed models yielded overall accuracy improvement of 33, 60, 80% and 30, 40, 60% for patients in testing groups, when compared to PIRADS v2.0 alone. DATA CONCLUSION: Radiomic features could help in identifying the presence and absence of clinically significant disease in AS patients when PIRADS v2.0 assessment on MRI contradicted pathology findings of MRI-TRUS prostate biopsies. LEVEL OF EVIDENCE: 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2018.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA