Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
Eur J Radiol ; 171: 111297, 2024 Feb.
Article En | MEDLINE | ID: mdl-38237517

Hepatic diffuse conditions and focal liver lesions represent two of the most common scenarios to face in everyday radiological clinical practice. Thanks to the advances in technology, radiology has gained a central role in the management of patients with liver disease, especially due to its high sensitivity and specificity. Since the introduction of computed tomography (CT) and magnetic resonance imaging (MRI), radiology has been considered the non-invasive reference modality to assess and characterize liver pathologies. In recent years, clinical practice has moved forward to a quantitative approach to better evaluate and manage each patient with a more fitted approach. In this setting, radiomics has gained an important role in helping radiologists and clinicians characterize hepatic pathological entities, in managing patients, and in determining prognosis. Radiomics can extract a large amount of data from radiological images, which can be associated with different liver scenarios. Thanks to its wide applications in ultrasonography (US), CT, and MRI, different studies were focused on specific aspects related to liver diseases. Even if broadly applied, radiomics has some advantages and different pitfalls. This review aims to summarize the most important and robust studies published in the field of liver radiomics, underlying their main limitations and issues, and what they can add to the current and future clinical practice and literature.


Liver Neoplasms , Radiomics , Humans , Tomography, X-Ray Computed , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Radiography , Magnetic Resonance Imaging
2.
Eur Radiol ; 2024 Jan 04.
Article En | MEDLINE | ID: mdl-38177618

OBJECTIVES: The aims of this study are to develop and validate a clinical decision support system based on demographics, prostate-specific antigen (PSA), microRNA (miRNA), and MRI for the detection of prostate cancer (PCa) and clinical significant (cs) PCa, and to assess if this system performs better compared to MRI alone. METHODS: This retrospective, multicenter, observational study included 222 patients (mean age 66, range 46-75 years) who underwent prostate MRI, miRNA (let-7a-5p and miR-103a-3p) assessment, and biopsy. Monoparametric and multiparametric models including age, PSA, miRNA, and MRI outcome were trained on 65% of the data and then validated on the remaining 35% to predict both PCa (any Gleason grade [GG]) and csPCa (GG ≥ 2 vs GG = 1/negative). Accuracy, sensitivity, specificity, positive and negative predictive value (NPV), and area under the receiver operating characteristic curve were calculated. RESULTS: MRI outcome was the best predictor in the monoparametric model for both detection of PCa, with sensitivity of 90% (95%CI 73-98%) and NPV of 93% (95%CI 82-98%), and for csPCa identification, with sensitivity of 91% (95%CI 72-99%) and NPV of 95% (95%CI 84-99%). Sensitivity and NPV of PSA + miRNA for the detection of csPCa were not statistically different from the other models including MRI alone. CONCLUSION: MRI stand-alone yielded the best prediction models for both PCa and csPCa detection in biopsy-naïve patients. The use of miRNAs let-7a-5p and miR-103a-3p did not improve classification performances compared to MRI stand-alone results. CLINICAL RELEVANCE STATEMENT: The use of miRNA (let-7a-5p and miR-103a-3p), PSA, and MRI in a clinical decision support system (CDSS) does not improve MRI stand-alone performance in the detection of PCa and csPCa. KEY POINTS: • Clinical decision support systems including MRI improve the detection of both prostate cancer and clinically significant prostate cancer with respect to PSA test and/or microRNA. • The use of miRNAs let-7a-5p and miR-103a-3p did not significantly improve MRI stand-alone performance. • Results of this study were in line with previous works on MRI and microRNA.

3.
Cancers (Basel) ; 16(1)2024 Jan 01.
Article En | MEDLINE | ID: mdl-38201630

In the last years, several studies demonstrated that low-aggressive (Grade Group (GG) ≤ 2) and high-aggressive (GG ≥ 3) prostate cancers (PCas) have different prognoses and mortality. Therefore, the aim of this study was to develop and externally validate a radiomic model to noninvasively classify low-aggressive and high-aggressive PCas based on biparametric magnetic resonance imaging (bpMRI). To this end, 283 patients were retrospectively enrolled from four centers. Features were extracted from apparent diffusion coefficient (ADC) maps and T2-weighted (T2w) sequences. A cross-validation (CV) strategy was adopted to assess the robustness of several classifiers using two out of the four centers. Then, the best classifier was externally validated using the other two centers. An explanation for the final radiomics signature was provided through Shapley additive explanation (SHAP) values and partial dependence plots (PDP). The best combination was a naïve Bayes classifier trained with ten features that reached promising results, i.e., an area under the receiver operating characteristic (ROC) curve (AUC) of 0.75 and 0.73 in the construction and external validation set, respectively. The findings of our work suggest that our radiomics model could help distinguish between low- and high-aggressive PCa. This noninvasive approach, if further validated and integrated into a clinical decision support system able to automatically detect PCa, could help clinicians managing men with suspicion of PCa.

4.
Insights Imaging ; 15(1): 8, 2024 Jan 17.
Article En | MEDLINE | ID: mdl-38228979

PURPOSE: To propose a new quality scoring tool, METhodological RadiomICs Score (METRICS), to assess and improve research quality of radiomics studies. METHODS: We conducted an online modified Delphi study with a group of international experts. It was performed in three consecutive stages: Stage#1, item preparation; Stage#2, panel discussion among EuSoMII Auditing Group members to identify the items to be voted; and Stage#3, four rounds of the modified Delphi exercise by panelists to determine the items eligible for the METRICS and their weights. The consensus threshold was 75%. Based on the median ranks derived from expert panel opinion and their rank-sum based conversion to importance scores, the category and item weights were calculated. RESULT: In total, 59 panelists from 19 countries participated in selection and ranking of the items and categories. Final METRICS tool included 30 items within 9 categories. According to their weights, the categories were in descending order of importance: study design, imaging data, image processing and feature extraction, metrics and comparison, testing, feature processing, preparation for modeling, segmentation, and open science. A web application and a repository were developed to streamline the calculation of the METRICS score and to collect feedback from the radiomics community. CONCLUSION: In this work, we developed a scoring tool for assessing the methodological quality of the radiomics research, with a large international panel and a modified Delphi protocol. With its conditional format to cover methodological variations, it provides a well-constructed framework for the key methodological concepts to assess the quality of radiomic research papers. CRITICAL RELEVANCE STATEMENT: A quality assessment tool, METhodological RadiomICs Score (METRICS), is made available by a large group of international domain experts, with transparent methodology, aiming at evaluating and improving research quality in radiomics and machine learning. KEY POINTS: • A methodological scoring tool, METRICS, was developed for assessing the quality of radiomics research, with a large international expert panel and a modified Delphi protocol. • The proposed scoring tool presents expert opinion-based importance weights of categories and items with a transparent methodology for the first time. • METRICS accounts for varying use cases, from handcrafted radiomics to entirely deep learning-based pipelines. • A web application has been developed to help with the calculation of the METRICS score ( https://metricsscore.github.io/metrics/METRICS.html ) and a repository created to collect feedback from the radiomics community ( https://github.com/metricsscore/metrics ).

6.
World J Gastroenterol ; 29(36): 5180-5197, 2023 Sep 28.
Article En | MEDLINE | ID: mdl-37901445

The liver is one of the organs most commonly involved in metastatic disease, especially due to its unique vascularization. It's well known that liver metastases represent the most common hepatic malignant tumors. From a practical point of view, it's of utmost importance to evaluate the presence of liver metastases when staging oncologic patients, to select the best treatment possible, and finally to predict the overall prognosis. In the past few years, imaging techniques have gained a central role in identifying liver metastases, thanks to ultrasonography, contrast-enhanced computed tomography (CT), and magnetic resonance imaging (MRI). All these techniques, especially CT and MRI, can be considered the non-invasive reference standard techniques for the assessment of liver involvement by metastases. On the other hand, the liver can be affected by different focal lesions, sometimes benign, and sometimes malignant. On these bases, radiologists should face the differential diagnosis between benign and secondary lesions to correctly allocate patients to the best management. Considering the above-mentioned principles, it's extremely important to underline and refresh the broad spectrum of liver metastases features that can occur in everyday clinical practice. This review aims to summarize the most common imaging features of liver metastases, with a special focus on typical and atypical appearance, by using MRI.


Contrast Media , Liver Neoplasms , Humans , Gadolinium DTPA , Liver Neoplasms/pathology , Magnetic Resonance Imaging/methods , Liver/diagnostic imaging , Liver/pathology
7.
Diagnostics (Basel) ; 13(18)2023 Sep 21.
Article En | MEDLINE | ID: mdl-37761380

High-resolution intraoperative PET/CT specimen imaging, coupled with prostate-specific membrane antigen (PSMA) molecular targeting, holds great potential for the rapid ex vivo identification of disease localizations in high-risk prostate cancer patients undergoing surgery. However, the accurate analysis of radiotracer uptake would require time-consuming manual volumetric segmentation of 3D images. The aim of this study was to test the feasibility of using machine learning to perform automatic nodal segmentation of intraoperative 68Ga-PSMA-11 PET/CT specimen images. Six (n = 6) lymph-nodal specimens were imaged in the operating room after an e.v. injection of 2.1 MBq/kg of 68Ga-PSMA-11. A machine learning-based approach for automatic lymph-nodal segmentation was developed using only open-source Python libraries (Scikit-learn, SciPy, Scikit-image). The implementation of a k-means clustering algorithm (n = 3 clusters) allowed to identify lymph-nodal structures by leveraging differences in tissue density. Refinement of the segmentation masks was performed using morphological operations and 2D/3D-features filtering. Compared to manual segmentation (ITK-SNAP v4.0.1), the automatic segmentation model showed promising results in terms of weighted average precision (97-99%), recall (68-81%), Dice coefficient (80-88%) and Jaccard index (67-79%). Finally, the ML-based segmentation masks allowed to automatically compute semi-quantitative PET metrics (i.e., SUVmax), thus holding promise for facilitating the semi-quantitative analysis of PET/CT images in the operating room.

8.
Eur J Radiol Open ; 11: 100505, 2023 Dec.
Article En | MEDLINE | ID: mdl-37484979

Objectives: To develop a mutation-based radiomics signature to predict response to imatinib in Gastrointestinal Stromal Tumors (GISTs). Methods: Eighty-two patients with GIST were enrolled in this retrospective study, including 52 patients from one center that were used to develop the model, and 30 patients from a second center to validate it. Reference standard was the mutational status of tyrosine-protein kinase (KIT) and platelet-derived growth factor α (PDGFRA). Patients were dichotomized in imatinib sensitive (group 0 - mutation in KIT or PDGFRA, different from exon 18-D842V), and imatinib non-responsive (group 1 - PDGFRA exon 18-D842V mutation or absence of mutation in KIT/PDGFRA). Initially, 107 texture features were extracted from the tumor masks of baseline computed tomography scans. Different machine learning methods were then implemented to select the best combination of features for the development of the radiomics signature. Results: The best performance was obtained with the 5 features selected by the ANOVA model and the Bayes classifier, using a threshold of 0.36. With this setting the radiomics signature had an accuracy and precision for sensitive patients of 82 % (95 % CI:60-95) and 90 % (95 % CI:73-97), respectively. Conversely, a precision of 80 % (95 % CI:34-97) was obtained in non-responsive patients using a threshold of 0.9. Indeed, with the latter setting 4 patients out of 5 were correctly predicted as non-responders. Conclusions: The results are a first step towards using radiomics to improve the management of patients with GIST, especially when tumor tissue is unavailable for molecular analysis or when molecular profiling is inconclusive.

9.
Biomed Phys Eng Express ; 9(5)2023 07 17.
Article En | MEDLINE | ID: mdl-37413967

Radiomics-based systems could improve the management of oncological patients by supporting cancer diagnosis, treatment planning, and response assessment. However, one of the main limitations of these systems is the generalizability and reproducibility of results when they are applied to images acquired in different hospitals by different scanners. Normalization has been introduced to mitigate this issue, and two main approaches have been proposed: one rescales the image intensities (image normalization), the other the feature distributions for each center (feature normalization). The aim of this study is to evaluate how different image and feature normalization methods impact the robustness of 93 radiomics features acquired using a multicenter and multi-scanner abdominal Magnetic Resonance Imaging (MRI) dataset. To this scope, 88 rectal MRIs were retrospectively collected from 3 different institutions (4 scanners), and for each patient, six 3D regions of interest on the obturator muscle were considered. The methods applied were min-max, 1st-99th percentiles and 3-Sigma normalization, z-score standardization, mean centering, histogram normalization, Nyul-Udupa and ComBat harmonization. The Mann-Whitney U-test was applied to assess features repeatability between scanners, by comparing the feature values obtained for each normalization method, including the case in which no normalization was applied. Most image normalization methods allowed to reduce the overall variability in terms of intensity distributions, while worsening or showing unpredictable results in terms of feature robustness, except for thez-score, which provided a slight improvement by increasing the number of statistically similar features from 9/93 to 10/93. Conversely, feature normalization methods positively reduced the overall variability across the scanners, in particular, 3sigma,z_scoreandComBatthat increased the number of similar features (79/93). According to our results, it emerged that none of the image normalization methods was able to strongly increase the number of statistically similar features.


Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Humans , Reproducibility of Results , Retrospective Studies , Image Processing, Computer-Assisted/methods , Phantoms, Imaging
10.
World J Gastroenterol ; 29(19): 2888-2904, 2023 May 21.
Article En | MEDLINE | ID: mdl-37274803

The main therapeutic options for colorectal cancer are surgical resection and adjuvant chemotherapy in non-metastatic disease. However, the evaluation of the overall adjuvant chemotherapy benefit in patients with a high risk of recurrence is challenging. Radiological images can represent a source of data that can be analyzed by using automated computer-based techniques, working on numerical information coded within Digital Imaging and Communications in Medicine files: This image numerical analysis has been named "radiomics". Radiomics allows the extraction of quantitative features from radiological images, mainly invisible to the naked eye, that can be further analyzed by artificial intelligence algorithms. Radiomics is expanding in oncology to either understand tumor biology or for the development of imaging biomarkers for diagnosis, staging, and prognosis, prediction of treatment response and diseases monitoring and surveillance. Several efforts have been made to develop radiomics signatures for colorectal cancer patient using computed tomography (CT) images with different aims: The preoperative prediction of lymph node metastasis, detecting BRAF and RAS gene mutations. Moreover, the use of delta-radiomics allows the analysis of variations of the radiomics parameters extracted from CT scans performed at different timepoints. Most published studies concerning radiomics and magnetic resonance imaging (MRI) mainly focused on the response of advanced tumors that underwent neoadjuvant therapy. Nodes status is the main determinant of adjuvant chemotherapy. Therefore, several radiomics model based on MRI, especially on T2-weighted images and ADC maps, for the preoperative prediction of nodes metastasis in rectal cancer has been developed. Current studies mostly focused on the applications of radiomics in positron emission tomography/CT for the prediction of survival after curative surgical resection and assessment of response following neoadjuvant chemoradiotherapy. Since colorectal liver metastases develop in about 25% of patients with colorectal carcinoma, the main diagnostic tasks of radiomics should be the detection of synchronous and metachronous lesions. Radiomics could be an additional tool in clinical setting, especially in identifying patients with high-risk disease. Nevertheless, radiomics has numerous shortcomings that make daily use extremely difficult. Further studies are needed to assess performance of radiomics in stratifying patients with high-risk disease.


Artificial Intelligence , Rectal Neoplasms , Humans , Positron Emission Tomography Computed Tomography/methods , Rectal Neoplasms/pathology , Prognosis , Lymphatic Metastasis , Magnetic Resonance Imaging/methods , Retrospective Studies
11.
IEEE Open J Eng Med Biol ; 4: 67-76, 2023.
Article En | MEDLINE | ID: mdl-37283773

Goal: Artificial intelligence applied to medical image analysis has been extensively used to develop non-invasive diagnostic and prognostic signatures. However, these imaging biomarkers should be largely validated on multi-center datasets to prove their robustness before they can be introduced into clinical practice. The main challenge is represented by the great and unavoidable image variability which is usually addressed using different pre-processing techniques including spatial, intensity and feature normalization. The purpose of this study is to systematically summarize normalization methods and to evaluate their correlation with the radiomics model performances through meta-analyses. This review is carried out according to the PRISMA statement: 4777 papers were collected, but only 74 were included. Two meta-analyses were carried out according to two clinical aims: characterization and prediction of response. Findings of this review demonstrated that there are some commonly used normalization approaches, but not a commonly agreed pipeline that can allow to improve performance and to bridge the gap between bench and bedside.

12.
BJR Open ; 5(1): 20220055, 2023.
Article En | MEDLINE | ID: mdl-37035771

In recent years, researchers have explored new ways to obtain information from pathological tissues, also exploring non-invasive techniques, such as virtual biopsy (VB). VB can be defined as a test that provides promising outcomes compared to traditional biopsy by extracting quantitative information from radiological images not accessible through traditional visual inspection. Data are processed in such a way that they can be correlated with the patient's phenotypic expression, or with molecular patterns and mutations, creating a bridge between traditional radiology, pathology, genomics, and artificial intelligence (AI). Radiomics is the backbone of VB, since it allows the extraction and selection of features from radiological images, feeding them into AI models in order to derive lesions' pathological characteristics and molecular status. Presently, the output of VB provides only a gross approximation of the findings of tissue biopsy. However, in the future, with the improvement of imaging resolution and processing techniques, VB could partially substitute the classical surgical or percutaneous biopsy, with the advantage of being non-invasive, comprehensive, accounting for lesion heterogeneity, and low cost. In this review, we investigate the concept of VB in abdominal pathology, focusing on its pipeline development and potential benefits.

13.
Annu Int Conf IEEE Eng Med Biol Soc ; 2022: 5066-5069, 2022 07.
Article En | MEDLINE | ID: mdl-36086406

The aim of the study is to present and tune a fully automatic deep learning algorithm to segment colorectal cancers (CRC) on MR images, based on a U-Net structure. It is a multicenter study, including 3 different Italian institutions, that used 4 different MRI scanners. Two of them were used for training and tuning the systems, while the other two for the validation. The implemented algorithm consists of a pre-processing step to normalize and to highlight the tumoral area, followed by the CRC segmentation using different U-net structures. Automatic masks were compared with manual segmentations performed by three experienced radiologists, one at each center. The two best performing systems (called mdl2 and mdl3), obtained a median Dice Similarity Coefficient of 0.68(mdl2) - 0.69(mdl3), precision of 0.75(md/2) - 0.71(md/3), and recall of 0.69(mdl2) - 0.73(mdl3) on the validation set. Both systems reached high detection rates, 0.98 and 0.95, respectively, on the validation set. These encouraging results, if confirmed on larger dataset, might improve the management of patients with CRC, since it can be used as a fast and precise tool for further radiomics analyses. Clinical Relevance - To provide a reliable tool able to automatically segment CRC tumors that can be used as first step in future radiomics studies aimed at predicting response to chemotherapy and personalizing treatment.


Deep Learning , Rectal Neoplasms , Algorithms , Humans , Magnetic Resonance Imaging/methods , Rectal Neoplasms/diagnostic imaging
14.
Eur Radiol Exp ; 6(1): 19, 2022 05 03.
Article En | MEDLINE | ID: mdl-35501512

BACKGROUND: Pathological complete response after neoadjuvant chemoradiotherapy in locally advanced rectal cancer (LARC) is achieved in 15-30% of cases. Our aim was to implement and externally validate a magnetic resonance imaging (MRI)-based radiomics pipeline to predict response to treatment and to investigate the impact of manual and automatic segmentations on the radiomics models. METHODS: Ninety-five patients with stage II/III LARC who underwent multiparametric MRI before chemoradiotherapy and surgical treatment were enrolled from three institutions. Patients were classified as responders if tumour regression grade was 1 or 2 and nonresponders otherwise. Sixty-seven patients composed the construction dataset, while 28 the external validation. Tumour volumes were manually and automatically segmented using a U-net algorithm. Three approaches for feature selection were tested and combined with four machine learning classifiers. RESULTS: Using manual segmentation, the best result reached an accuracy of 68% on the validation set, with sensitivity 60%, specificity 77%, negative predictive value (NPV) 63%, and positive predictive value (PPV) 75%. The automatic segmentation achieved an accuracy of 75% on the validation set, with sensitivity 80%, specificity 69%, and both NPV and PPV 75%. Sensitivity and NPV on the validation set were significantly higher (p = 0.047) for the automatic versus manual segmentation. CONCLUSION: Our study showed that radiomics models can pave the way to help clinicians in the prediction of tumour response to chemoradiotherapy of LARC and to personalise per-patient treatment. The results from the external validation dataset are promising for further research into radiomics approaches using both manual and automatic segmentations.


Rectal Neoplasms , Rectum , Chemoradiotherapy , Humans , Magnetic Resonance Imaging/methods , Neoadjuvant Therapy/methods , Rectal Neoplasms/diagnostic imaging , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Rectum/pathology
15.
J Imaging ; 8(5)2022 May 11.
Article En | MEDLINE | ID: mdl-35621897

Magnetic resonance imaging (MRI) has a growing role in the clinical workup of prostate cancer. However, manual three-dimensional (3D) segmentation of the prostate is a laborious and time-consuming task. In this scenario, the use of automated algorithms for prostate segmentation allows us to bypass the huge workload of physicians. In this work, we propose a fully automated hybrid approach for prostate gland segmentation in MR images using an initial segmentation of prostate volumes using a custom-made 3D deep network (VNet-T2), followed by refinement using an Active Shape Model (ASM). While the deep network focuses on three-dimensional spatial coherence of the shape, the ASM relies on local image information and this joint effort allows for improved segmentation of the organ contours. Our method is developed and tested on a dataset composed of T2-weighted (T2w) MRI prostatic volumes of 60 male patients. In the test set, the proposed method shows excellent segmentation performance, achieving a mean dice score and Hausdorff distance of 0.851 and 7.55 mm, respectively. In the future, this algorithm could serve as an enabling technology for the development of computer-aided systems for prostate cancer characterization in MR imaging.

16.
Cancers (Basel) ; 14(1)2022 Jan 04.
Article En | MEDLINE | ID: mdl-35008405

The purpose of this paper is to develop and validate a delta-radiomics score to predict the response of individual colorectal cancer liver metastases (lmCRC) to first-line FOLFOX chemotherapy. Three hundred one lmCRC were manually segmented on both CT performed at baseline and after the first cycle of first-line FOLFOX, and 107 radiomics features were computed by subtracting textural features of CT at baseline from those at timepoint 1 (TP1). LmCRC were classified as nonresponders (R-) if they showed progression of disease (PD), according to RECIST1.1, before 8 months, and as responders (R+), otherwise. After feature selection, we developed a decision tree statistical model trained using all lmCRC coming from one hospital. The final output was a delta-radiomics signature subsequently validated on an external dataset. Sensitivity, specificity, positive (PPV), and negative (NPV) predictive values in correctly classifying individual lesions were assessed on both datasets. Per-lesion sensitivity, specificity, PPV, and NPV were 99%, 94%, 95%, 99%, 85%, 92%, 90%, and 87%, respectively, in the training and validation datasets. The delta-radiomics signature was able to reliably predict R- lmCRC, which were wrongly classified by lesion RECIST as R+ at TP1, (93%, averaging training and validation set, versus 67% of RECIST). The delta-radiomics signature developed in this study can reliably predict the response of individual lmCRC to oxaliplatin-based chemotherapy. Lesions forecasted as poor or nonresponders by the signature could be further investigated, potentially paving the way to lesion-specific therapies.

17.
Annu Int Conf IEEE Eng Med Biol Soc ; 2021: 3305-3308, 2021 11.
Article En | MEDLINE | ID: mdl-34891947

Colorectal cancer (CRC) has the second-highest tumor incidence and is a leading cause of death by cancer. Nearly 20% of patients with CRC will have metastases (mts) at the time of diagnosis, and more than 50% of patients with CRC develop metastases during their disease. Unfortunately, only 45% of patients after a chemotherapy will respond to treatment. The aim of this study is to develop and validate a machine learning algorithm to predict response of individual liver mts, using CT scans. Understanding which mts will respond or not will help clinicians in providing a more efficient per-lesion treatment based on patient specific response and not only following a standard treatment. A group of 92 patients was enrolled from two Italian institutions. CT scans were collected, and the portal venous phase was manually segmented by an expert radiologist. Then, 75 radiomics features were extracted both from 7x7 ROIs that moved across the image and from the whole 3D mts. Feature selection was performed using a genetic algorithm. Results are presented as a comparison of the two different approaches of features extraction and different classification algorithms. Accuracy (ACC), sensitivity (SE), specificity (SP), negative and positive predictive values (NPV and PPV) were evaluated for all lesions (per-lesion analysis) and patients (per-patient analysis) in the construction and validation sets. Best results were obtained in the per-lesion analysis from the 3D approach using a Support Vector Machine as classifier. We reached on the training set an ACC of 81%, while on test set, we obtained SE of 76%, SP of 67%, PPV of 69% and NPV of 75%. On the validation set a SE of 61%, SP of 60%, PPV of 57% and NPV of 64% were reached. The promising results obtained in the validation dataset should be extended to a larger cohort of patient to further validate our method.Clinical Relevance- to develop a radiomics signatures predicting single liver mts response to therapy. A personalized mts approach is important to avoid unnecessary toxicity offering more suitable treatments and a better quality of life to oncological patients.


Colonic Neoplasms , Liver Neoplasms , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/drug therapy , Quality of Life , Tomography, X-Ray Computed
18.
Annu Int Conf IEEE Eng Med Biol Soc ; 2021: 3370-3373, 2021 11.
Article En | MEDLINE | ID: mdl-34891962

Automatic segmentation of the prostate on Magnetic Resonance Imaging (MRI) is one of the topics on which research has focused in recent years as it is a fundamental first step in the building process of a Computer aided diagnosis (CAD) system for cancer detection. Unfortunately, MRI acquired in different centers with different scanners leads to images with different characteristics. In this work, we propose an automatic algorithm for prostate segmentation, based on a U-Net applying transfer learning method in a bi-center setting. First, T2w images with and without endorectal coil from 80 patients acquired at Center A were used as training set and internal validation set. Then, T2w images without endorectal coil from 20 patients acquired at Center B were used as external validation. The reference standard for this study was manual segmentation of the prostate gland performed by an expert operator. The results showed a Dice similarity coefficient >85% in both internal and external validation datasets.Clinical Relevance- This segmentation algorithm could be integrated into a CAD system to optimize computational effort in prostate cancer detection.


Deep Learning , Prostatic Neoplasms , Algorithms , Humans , Magnetic Resonance Imaging , Male , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging
19.
Annu Int Conf IEEE Eng Med Biol Soc ; 2021: 3374-3377, 2021 11.
Article En | MEDLINE | ID: mdl-34891963

In the last decades, MRI was proven a useful tool for the diagnosis and characterization of Prostate Cancer (PCa). In the literature, many studies focused on characterizing PCa aggressiveness, but a few have distinguished between low-aggressive (Gleason Grade Group (GG) <=2) and high-aggressive (GG>=3) PCas based on biparametric MRI (bpMRI). In this study, 108 PCas were collected from two different centers and were divided into training, testing, and validation set. From Apparent Diffusion Coefficient (ADC) map and T2-Weighted Image (T2WI), we extracted texture features, both 3D and 2D, and we implemented three different methods of Feature Selection (FS): Minimum Redundance Maximum Relevance (MRMR), Affinity Propagation (AP), and Genetic Algorithm (GA). From the resulting subsets of predictors, we trained Support Vector Machine (SVM), Decision Tree, and Ensemble Learning classifiers on the training set, and we evaluated their prediction ability on the testing set. Then, for each FS method, we chose the best classifier, based on both training and testing performances, and we further assessed their generalization capability on the validation set. Between the three best models, a Decision Tree was trained using only two features extracted from the ADC map and selected by MRMR, achieving, on the validation set, an Area Under the ROC (AUC) equal to 81%, with sensitivity and specificity of 77% and 93%, respectively.Clinical Relevance- Our best model demonstrated to be able to distinguish low-aggressive from high-aggressive PCas with high accuracy. Potentially, this approach could help clinician to noninvasively distinguish between PCas that might need active treatment and those that could potentially benefit from active surveillance, avoiding biopsy-related complications.


Magnetic Resonance Imaging , Prostatic Neoplasms , Biopsy , Humans , Machine Learning , Male , Prostatic Neoplasms/diagnostic imaging , Retrospective Studies
20.
Front Oncol ; 11: 718155, 2021.
Article En | MEDLINE | ID: mdl-34660282

In the last years, the widespread use of the prostate-specific antigen (PSA) blood examination to triage patients who will enter the diagnostic/therapeutic path for prostate cancer (PCa) has almost halved PCa-specific mortality. As a counterpart, millions of men with clinically insignificant cancer not destined to cause death are treated, with no beneficial impact on overall survival. Therefore, there is a compelling need to develop tools that can help in stratifying patients according to their risk, to support physicians in the selection of the most appropriate treatment option for each individual patient. The aim of this study was to develop and validate on multivendor data a fully automated computer-aided diagnosis (CAD) system to detect and characterize PCas according to their aggressiveness. We propose a CAD system based on artificial intelligence algorithms that a) registers all images coming from different MRI sequences, b) provides candidates suspicious to be tumor, and c) provides an aggressiveness score of each candidate based on the results of a support vector machine classifier fed with radiomics features. The dataset was composed of 131 patients (149 tumors) from two different institutions that were divided in a training set, a narrow validation set, and an external validation set. The algorithm reached an area under the receiver operating characteristic (ROC) curve in distinguishing between low and high aggressive tumors of 0.96 and 0.81 on the training and validation sets, respectively. Moreover, when the output of the classifier was divided into three classes of risk, i.e., indolent, indeterminate, and aggressive, our method did not classify any aggressive tumor as indolent, meaning that, according to our score, all aggressive tumors would undergo treatment or further investigations. Our CAD performance is superior to that of previous studies and overcomes some of their limitations, such as the need to perform manual segmentation of the tumor or the fact that analysis is limited to single-center datasets. The results of this study are promising and could pave the way to a prediction tool for personalized decision making in patients harboring PCa.

...