1
|
Qiao J, Liu B, Xin J, Shen S, Ma H, Pan S. Prediction of Prognosis and Response to Androgen Deprivation Therapy in Intermediate to High-Risk Prostate Cancer Using 18F-FDG PET/CT Radiomics. Acad Radiol 2024:S1076-6332(24)00420-3. [PMID: 39019687 DOI: 10.1016/j.acra.2024.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 07/19/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to predict intermediate to high-risk prostate cancer (PCa) prognosis based on 18-fluoro-2-deoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) radiomics. Additionally, subgroup analysis will be performed on the androgen deprivation therapy (ADT) group and the metastatic PCa group. MATERIALS AND METHODS In the retrospective analysis of 104 intermediate to high-risk PCa patients who underwent 18F-FDG PET/CT prior to treatment. The data set was divided into a training set (n = 72) and a testing set (n = 32). Two different PET/CT models were constructed using multivariate logistic regression with cross-validation: radiomics model A and an alternative ensemble learning-based model B. The superior model was then selected to develop a radiomics nomogram. Separate models were also developed for the ADT and metastatic PCa subgroups. RESULTS Model A, which integrates eight radiomics features showed excellent performance with an area under curve (AUC) of 0.844 in the training set and 0.804 in the testing set. The radiomics nomogram incorporating the radiomics score (radscore) from model A and the tumor-to-liver ratio (TLR) showed good prognostic accuracy in the testing set with an AUC of 0.827. In the subgroup analyses for endocrine therapy and metastatic cancer, the PET/CT radiomics model showed AUCs of 0.845 and 0.807 respectively, suggesting its potential effectiveness. CONCLUSION The study establishes the utility of the 18F-FDG PET/CT radiomics nomogram in predicting the prognosis of intermediate to high-risk PCa patients, indicating its potential for clinical application.
Collapse
Affiliation(s)
- Jianyi Qiao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bitian Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Xin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Siang Shen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Han Ma
- Department of Nuclear Medicine, People's Hospital of Liaoning Province, Shenyang, China
| | - Shen Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Wu X, Ruan Z, Ke Z, Lin F, Chen J, Xue Y, Lin B, Chen S, Chen D, Zheng Q, Xue X, Wei Y, Xu N. Magnetic resonance imaging-based radiomics nomogram for the evaluation of therapeutic responses to neoadjuvant chemohormonal therapy in high-risk non-metastatic prostate cancer. Cancer Med 2024; 13:e70001. [PMID: 39031016 PMCID: PMC11258568 DOI: 10.1002/cam4.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/30/2024] [Accepted: 06/30/2024] [Indexed: 07/22/2024] Open
Abstract
PURPOSE The aim of this study was to assess the potential application of a radiomics features-based nomogram for predicting therapeutic responses to neoadjuvant chemohormonal therapy (NCHT) in patients with high-risk non-metastatic prostate cancer (PCa). METHODS Clinicopathologic information was retrospectively collected from 162 patients with high-risk non-metastatic PCa receiving NCHT and radical prostatectomy at our center. The postoperative pathological findings were used as the gold standard for evaluating the efficacy of NCHT. The least absolute shrinkage and selection operator (LASSO) was conducted to develop radiomics signature. Multivariate logistic regression analyses were conducted to identify the predictors of a positive pathological response to NCHT, and a nomogram was constructed based on these predictors. RESULTS Sixty-three patients (38.89%) experienced positive pathological response to NCHT. Receiver operating characteristic analyses showed that the area under the curve (AUC) of periprostatic fat (PPF) radiomics signature was 0.835 (95% CI, 0.754-0.898), while the AUC of intratumoral radiomics signature was 0.822 (95% CI, 0.739-0.888). Multivariate logistic regression analysis revealed that PSA level, PPF radiomics signature and intratumoral radiomics signature were independent predictors of positive pathological response. A nomogram based on these three predictors was constructed. The AUC was 0.908 (95% CI, 0.839-0.954). The Hosmer-Lemeshow goodness-of-fit test showed that the nomogram was well calibrated. Decision curve analysis revealed the favorable clinical practicability of the nomogram. The nomogram was successfully validated in the validation cohort. Kaplan-Meier analyses showed that nomogram and positive pathological response were significantly related with survival of PCa. CONCLUSION The radiomics-clinical nomogram based on mpMRI radiomics features exhibited superior predictive ability for positive pathological response to NCHT in high-risk non-metastatic PCa.
Collapse
Affiliation(s)
- Xiao‐Hui Wu
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Zhong‐Tian Ruan
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Zhi‐Bin Ke
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Jia‐Yin Chen
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yu‐Ting Xue
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Bin Lin
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Shao‐Hao Chen
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Dong‐Ning Chen
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Qing‐Shui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Xue‐Yi Xue
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
3
|
Riaz IB, Harmon S, Chen Z, Naqvi SAA, Cheng L. Applications of Artificial Intelligence in Prostate Cancer Care: A Path to Enhanced Efficiency and Outcomes. Am Soc Clin Oncol Educ Book 2024; 44:e438516. [PMID: 38935882 DOI: 10.1200/edbk_438516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
The landscape of prostate cancer care has rapidly evolved. We have transitioned from the use of conventional imaging, radical surgeries, and single-agent androgen deprivation therapy to an era of advanced imaging, precision diagnostics, genomics, and targeted treatment options. Concurrently, the emergence of large language models (LLMs) has dramatically transformed the paradigm for artificial intelligence (AI). This convergence of advancements in prostate cancer management and AI provides a compelling rationale to comprehensively review the current state of AI applications in prostate cancer care. Here, we review the advancements in AI-driven applications across the continuum of the journey of a patient with prostate cancer from early interception to survivorship care. We subsequently discuss the role of AI in prostate cancer drug discovery, clinical trials, and clinical practice guidelines. In the localized disease setting, deep learning models demonstrated impressive performance in detecting and grading prostate cancer using imaging and pathology data. For biochemically recurrent diseases, machine learning approaches are being tested for improved risk stratification and treatment decisions. In advanced prostate cancer, deep learning can potentially improve prognostication and assist in clinical decision making. Furthermore, LLMs are poised to revolutionize information summarization and extraction, clinical trial design and operations, drug development, evidence synthesis, and clinical practice guidelines. Synergistic integration of multimodal data integration and human-AI integration are emerging as a key strategy to unlock the full potential of AI in prostate cancer care.
Collapse
Affiliation(s)
- Irbaz Bin Riaz
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic, Phoenix, AZ
- Department of AI and Informatics, Mayo Clinic, Rochester, MN
| | - Stephanie Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Zhijun Chen
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, Lifespan Health, and the Legorreta Cancer Center at Brown University, Providence, RI
| |
Collapse
|
4
|
Hu C, Qiao X, Huang R, Hu C, Bao J, Wang X. Development and Validation of a Multimodality Model Based on Whole-Slide Imaging and Biparametric MRI for Predicting Postoperative Biochemical Recurrence in Prostate Cancer. Radiol Imaging Cancer 2024; 6:e230143. [PMID: 38758079 PMCID: PMC11148661 DOI: 10.1148/rycan.230143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/16/2024] [Accepted: 03/25/2024] [Indexed: 05/18/2024]
Abstract
Purpose To develop and validate a machine learning multimodality model based on preoperative MRI, surgical whole-slide imaging (WSI), and clinical variables for predicting prostate cancer (PCa) biochemical recurrence (BCR) following radical prostatectomy (RP). Materials and Methods In this retrospective study (September 2015 to April 2021), 363 male patients with PCa who underwent RP were divided into training (n = 254; median age, 69 years [IQR, 64-74 years]) and testing (n = 109; median age, 70 years [IQR, 65-75 years]) sets at a ratio of 7:3. The primary end point was biochemical recurrence-free survival. The least absolute shrinkage and selection operator Cox algorithm was applied to select independent clinical variables and construct the clinical signature. The radiomics signature and pathomics signature were constructed using preoperative MRI and surgical WSI data, respectively. A multimodality model was constructed by combining the radiomics signature, pathomics signature, and clinical signature. Using Harrell concordance index (C index), the predictive performance of the multimodality model for BCR was assessed and compared with all single-modality models, including the radiomics signature, pathomics signature, and clinical signature. Results Both radiomics and pathomics signatures achieved good performance for BCR prediction (C index: 0.742 and 0.730, respectively) on the testing cohort. The multimodality model exhibited the best predictive performance, with a C index of 0.860 on the testing set, which was significantly higher than all single-modality models (all P ≤ .01). Conclusion The multimodality model effectively predicted BCR following RP in patients with PCa and may therefore provide an emerging and accurate tool to assist postoperative individualized treatment. Keywords: MR Imaging, Urinary, Pelvis, Comparative Studies Supplemental material is available for this article. © RSNA, 2024.
Collapse
Affiliation(s)
| | | | - Renpeng Huang
- From the Departments of Radiology (Chenhan Hu, X.Q., Chunhong Hu,
J.B., X.W.) and Pathology (R.H.), the First Affiliated Hospital of Soochow
University, 188 Shizi Road, Suzhou 215006, China
| | - Chunhong Hu
- From the Departments of Radiology (Chenhan Hu, X.Q., Chunhong Hu,
J.B., X.W.) and Pathology (R.H.), the First Affiliated Hospital of Soochow
University, 188 Shizi Road, Suzhou 215006, China
| | - Jie Bao
- From the Departments of Radiology (Chenhan Hu, X.Q., Chunhong Hu,
J.B., X.W.) and Pathology (R.H.), the First Affiliated Hospital of Soochow
University, 188 Shizi Road, Suzhou 215006, China
| | - Ximing Wang
- From the Departments of Radiology (Chenhan Hu, X.Q., Chunhong Hu,
J.B., X.W.) and Pathology (R.H.), the First Affiliated Hospital of Soochow
University, 188 Shizi Road, Suzhou 215006, China
| |
Collapse
|
5
|
Hiremath A, Corredor G, Li L, Leo P, Magi-Galluzzi C, Elliott R, Purysko A, Shiradkar R, Madabhushi A. An integrated radiology-pathology machine learning classifier for outcome prediction following radical prostatectomy: Preliminary findings. Heliyon 2024; 10:e29602. [PMID: 38665576 PMCID: PMC11044050 DOI: 10.1016/j.heliyon.2024.e29602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
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).
Collapse
Affiliation(s)
| | - Germán Corredor
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Lin Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Patrick Leo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | | - Robin Elliott
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrei Purysko
- Department of Radiology and Nuclear Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Rakesh Shiradkar
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| |
Collapse
|
6
|
Wang H, Wang K, Ma S, Gao G, Wang X. Investigation of radiomics models for predicting biochemical recurrence of advanced prostate cancer on pretreatment MR ADC maps based on automatic image segmentation. J Appl Clin Med Phys 2024; 25:e14244. [PMID: 38146796 PMCID: PMC11005965 DOI: 10.1002/acm2.14244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/16/2023] [Accepted: 12/03/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVES To develop radiomics models based on automatic segmentation of the pretreatment apparent diffusion coefficient (ADC) maps for predicting the biochemical recurrence (BCR) of advanced prostate cancer (PCa). METHODS A total of 100 cases with pathologically confirmed PCa were retrospectively included in this study. These cases were randomly divided into training (n = 70) and test (n = 30) datasets. Two predictive models were constructed based on the combination of age, prostate specific antigen (PSA) level, Gleason score, and clinical staging before therapy and the prostate area (Model_1) or PCa area (Model_2). Another two predictive models were constructed based on only prostate area (Model_3) or PCa area (Model_4). The area under the receiver operating characteristic curve (ROC AUC) and precision-recall (PR) curve analysis were used to analyze the models' performance. RESULTS Sixty-five patients without BCR (BCR-) and 35 patients with BCR (BCR+) were confirmed. The age, PSA, volume, diameter and ADC value of the prostate and PCa were not significantly different between the BCR- and BCR+ groups or between the training and test datasets (all p > 0.05). The AUCs were 0.637 (95% CI: 0.434-0.838), 0.841 (95% CI: 0.695-0.940), 0.840 (95% CI: 0.698-0.983), and 0.808 (95% CI: 0.627-0.988) for Model_1 to Model_4 in the test dataset without significant difference. The 95% bootstrap confidence intervals for the areas under the PR curve of the four models were not statistically different. CONCLUSION The radiomics models based on automatically segmented prostate and PCa areas on the pretreatment ADC maps developed in our study can be promising in predicting BCR of advanced PCa.
Collapse
Affiliation(s)
- Huihui Wang
- Department of RadiologyPeking University First HospitalBeijingChina
| | - Kexin Wang
- School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Shuai Ma
- Department of RadiologyPeking University First HospitalBeijingChina
| | - Ge Gao
- Department of RadiologyPeking University First HospitalBeijingChina
| | - Xiaoying Wang
- Department of RadiologyPeking University First HospitalBeijingChina
| |
Collapse
|
7
|
Baydoun A, Jia AY, Zaorsky NG, Kashani R, Rao S, Shoag JE, Vince RA, Bittencourt LK, Zuhour R, Price AT, Arsenault TH, Spratt DE. Artificial intelligence applications in prostate cancer. Prostate Cancer Prostatic Dis 2024; 27:37-45. [PMID: 37296271 DOI: 10.1038/s41391-023-00684-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/05/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
Artificial intelligence (AI) applications have enabled remarkable advancements in healthcare delivery. These AI tools are often aimed to improve accuracy and efficiency of histopathology assessment and diagnostic imaging interpretation, risk stratification (i.e., prognostication), and prediction of therapeutic benefit for personalized treatment recommendations. To date, multiple AI algorithms have been explored for prostate cancer to address automation of clinical workflow, integration of data from multiple domains in the decision-making process, and the generation of diagnostic, prognostic, and predictive biomarkers. While many studies remain within the pre-clinical space or lack validation, the last few years have witnessed the emergence of robust AI-based biomarkers validated on thousands of patients, and the prospective deployment of clinically-integrated workflows for automated radiation therapy design. To advance the field forward, multi-institutional and multi-disciplinary collaborations are needed in order to prospectively implement interoperable and accountable AI technology routinely in clinic.
Collapse
Affiliation(s)
- Atallah Baydoun
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Angela Y Jia
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rojano Kashani
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Santosh Rao
- Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jonathan E Shoag
- Department of Urology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Randy A Vince
- Department of Urology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Leonardo Kayat Bittencourt
- Department of Radiology, University Hospitals Cleveland Medical Center Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Raed Zuhour
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Alex T Price
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Theodore H Arsenault
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
8
|
Wang H, Wang K, Zhang Y, Chen Y, Zhang X, Wang X. Deep learning-based radiomics model from pretreatment ADC to predict biochemical recurrence in advanced prostate cancer. Front Oncol 2024; 14:1342104. [PMID: 38476369 PMCID: PMC10928490 DOI: 10.3389/fonc.2024.1342104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose To develop deep-learning radiomics model for predicting biochemical recurrence (BCR) of advanced prostate cancer (PCa) based on pretreatment apparent diffusion coefficient (ADC) maps. Methods Data were collected retrospectively from 131 patients diagnosed with advanced PCa, randomly divided into training (n = 93) and test (n = 38) datasets. Pre-treatment ADC images were segmented using a pre-trained artificial intelligence (AI) model to identify suspicious PCa areas. Three models were constructed, including a clinical model, a conventional radiomics model and a deep-radiomics model. The receiver operating characteristic (ROC), precision-recall (PR) curve and decision curve analysis (DCA) were used to assess predictive performance in test dataset. The net reclassification index (NRI) and integrated discrimination improvement (IDI) were employed to compare the performance enhancement of the deep-radiomics model in relation to the other two models. Results The deep-radiomics model exhibited a significantly higher area under the curve (AUC) of ROC than the other two (P = 0.033, 0.026), as well as PR curve (AUC difference 0.420, 0.432). The DCA curve demonstrated superior performance for the deep-radiomics model across all risk thresholds than the other two. Taking the clinical model as reference, the NRI and IDI was 0.508 and 0.679 for the deep-radiomics model with significant difference. Compared with the conventional radiomics model, the NRI and IDI was 0.149 and 0.164 for the deep-radiomics model without significant difference. Conclusion The deep-radiomics model exhibits promising potential in predicting BCR in advanced PCa, compared to both the clinical model and the conventional radiomics model.
Collapse
Affiliation(s)
- Huihui Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Kexin Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yaofeng Zhang
- Beijing Smart Tree Medical Technology Co. Ltd., Beijing, China
| | - Yuke Chen
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Xiaodong Zhang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Xiaoying Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| |
Collapse
|
9
|
Hu C, Qiao X, Hu C, Cao C, Wang X, Bao J. The practical clinical role of machine learning models with different algorithms in predicting prostate cancer local recurrence after radical prostatectomy. Cancer Imaging 2024; 24:23. [PMID: 38326860 PMCID: PMC10848341 DOI: 10.1186/s40644-024-00667-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/21/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND The detection of local recurrence for prostate cancer (PCa) patients following radical prostatectomy (RP) is challenging and can influence the treatment plan. Our aim was to construct and verify machine learning models with three different algorithms based on post-operative mpMRI for predicting local recurrence of PCa after RP and explore their potential clinical value compared with the Prostate Imaging for Recurrence Reporting (PI-RR) score of expert-level radiologists. METHODS A total of 176 patients were retrospectively enrolled and randomly divided into training (n = 123) and testing (n = 53) sets. The PI-RR assessments were performed by two expert-level radiologists with access to the operative histopathological and pre-surgical clinical results. The radiomics models to predict local recurrence were built by utilizing three different algorithms (i.e., support vector machine [SVM], linear discriminant analysis [LDA], and logistic regression-least absolute shrinkage and selection operator [LR-LASSO]). The combined model integrating radiomics features and PI-RR score was developed using the most effective classifier. The classification performances of the proposed models were assessed by receiver operating characteristic (ROC) curve analysis. RESULTS There were no significant differences between the training and testing sets concerning age, prostate-specific antigen (PSA), Gleason score, T-stage, seminal vesicle invasion (SVI), perineural invasion (PNI), and positive surgical margins (PSM). The radiomics model based on LR-LASSO exhibited superior performance than other radiomics models, with an AUC of 0.858 in the testing set; the PI-RR yielded an AUC of 0.833, and there was no significant difference between the best radiomics model and the PI-RR score. The combined model achieved the best predictive performance with an AUC of 0.924, and a significant difference was observed between the combined model and PI-RR score. CONCLUSIONS Our radiomics model is an effective tool to predict PCa local recurrence after RP. By integrating radiomics features with the PI-RR score, our combined model exhibited significantly better predictive performance of local recurrence than expert-level radiologists' PI-RR assessment.
Collapse
Affiliation(s)
- Chenhan Hu
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Xiaomeng Qiao
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Chunhong Hu
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Changhao Cao
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Ximing Wang
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China.
| | - Jie Bao
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China.
| |
Collapse
|
10
|
Taddese AA, Tilahun BC, Awoke T, Atnafu A, Mamuye A, Mengiste SA. Deep-learning models for image-based gynecological cancer diagnosis: a systematic review and meta- analysis. Front Oncol 2024; 13:1216326. [PMID: 38273847 PMCID: PMC10809847 DOI: 10.3389/fonc.2023.1216326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/13/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Gynecological cancers pose a significant threat to women worldwide, especially those in resource-limited settings. Human analysis of images remains the primary method of diagnosis, but it can be inconsistent and inaccurate. Deep learning (DL) can potentially enhance image-based diagnosis by providing objective and accurate results. This systematic review and meta-analysis aimed to summarize the recent advances of deep learning (DL) techniques for gynecological cancer diagnosis using various images and explore their future implications. Methods The study followed the PRISMA-2 guidelines, and the protocol was registered in PROSPERO. Five databases were searched for articles published from January 2018 to December 2022. Articles that focused on five types of gynecological cancer and used DL for diagnosis were selected. Two reviewers assessed the articles for eligibility and quality using the QUADAS-2 tool. Data was extracted from each study, and the performance of DL techniques for gynecological cancer classification was estimated by pooling and transforming sensitivity and specificity values using a random-effects model. Results The review included 48 studies, and the meta-analysis included 24 studies. The studies used different images and models to diagnose different gynecological cancers. The most popular models were ResNet, VGGNet, and UNet. DL algorithms showed more sensitivity but less specificity compared to machine learning (ML) methods. The AUC of the summary receiver operating characteristic plot was higher for DL algorithms than for ML methods. Of the 48 studies included, 41 were at low risk of bias. Conclusion This review highlights the potential of DL in improving the screening and diagnosis of gynecological cancer, particularly in resource-limited settings. However, the high heterogeneity and quality of the studies could affect the validity of the results. Further research is necessary to validate the findings of this study and to explore the potential of DL in improving gynecological cancer diagnosis.
Collapse
Affiliation(s)
- Asefa Adimasu Taddese
- Department of Health Informatics, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- eHealthlab Ethiopia Research Center, University of Gondar, Gondar, Ethiopia
| | - Binyam Chakilu Tilahun
- Department of Health Informatics, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- eHealthlab Ethiopia Research Center, University of Gondar, Gondar, Ethiopia
| | - Tadesse Awoke
- Department of Epidemiology and Biostatistics, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Asmamaw Atnafu
- eHealthlab Ethiopia Research Center, University of Gondar, Gondar, Ethiopia
- Department of Health Systems and Policy, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Adane Mamuye
- eHealthlab Ethiopia Research Center, University of Gondar, Gondar, Ethiopia
- School of Information Technology and Engineering, Addis Ababa University, Addis Ababa, Ethiopia
| | - Shegaw Anagaw Mengiste
- Department of Business, History and Social Sciences, University of Southeastern Norway, Vestfold, Vestfold, Norway
| |
Collapse
|
11
|
Hou Y, Jiang KW, Wang LL, Zhi R, Bao ML, Li Q, Zhang J, Qu JR, Zhu FP, Zhang YD. Biopsy-free AI-aided precision MRI assessment in prediction of prostate cancer biochemical recurrence. Br J Cancer 2023; 129:1625-1633. [PMID: 37758837 PMCID: PMC10646026 DOI: 10.1038/s41416-023-02441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND To investigate the predictive ability of high-throughput MRI with deep survival networks for biochemical recurrence (BCR) of prostate cancer (PCa) after prostatectomy. METHODS Clinical-MRI and histopathologic data of 579 (train/test, 463/116) PCa patients were retrospectively collected. The deep survival network (iBCR-Net) is based on stepwise processing operations, which first built an MRI radiomics signature (RadS) for BCR, and predicted the T3 stage and lymph node metastasis (LN+) of tumour using two predefined AI models. Subsequently, clinical, imaging and histopathological variables were integrated into iBCR-Net for BCR prediction. RESULTS RadS, derived from 2554 MRI features, was identified as an independent predictor of BCR. Two predefined AI models achieved an accuracy of 82.6% and 78.4% in staging T3 and LN+. The iBCR-Net, when expressed as a presurgical model by integrating RadS, AI-diagnosed T3 stage and PSA, can match a state-of-the-art histopathological model (C-index, 0.81 to 0.83 vs 0.79 to 0.81, p > 0.05); and has maximally 5.16-fold, 12.8-fold, and 2.09-fold (p < 0.05) benefit to conventional D'Amico score, the Cancer of the Prostate Risk Assessment (CAPRA) score and the CAPRA Postsurgical score. CONCLUSIONS AI-aided iBCR-Net using high-throughput MRI can predict PCa BCR accurately and thus may provide an alternative to the conventional method for PCa risk stratification.
Collapse
Affiliation(s)
- Ying Hou
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Ke-Wen Jiang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Li-Li Wang
- Department of Breast Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, 350014, Fuzhou, China
| | - Rui Zhi
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Mei-Ling Bao
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Qiao Li
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Jing Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Jin-Rong Qu
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450008, Zhengzhou, Henan, China
| | - Fei-Peng Zhu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Yu-Dong Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China.
| |
Collapse
|
12
|
Yilmaz EC, Harmon SA, Belue MJ, Merriman KM, Phelps TE, Lin Y, Garcia C, Hazen L, Patel KR, Merino MJ, Wood BJ, Choyke PL, Pinto PA, Citrin DE, Turkbey B. Evaluation of a Deep Learning-based Algorithm for Post-Radiotherapy Prostate Cancer Local Recurrence Detection Using Biparametric MRI. Eur J Radiol 2023; 168:111095. [PMID: 37717420 PMCID: PMC10615746 DOI: 10.1016/j.ejrad.2023.111095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVE To evaluate a biparametric MRI (bpMRI)-based artificial intelligence (AI) model for the detection of local prostate cancer (PCa) recurrence in patients with radiotherapy history. MATERIALS AND METHODS This study included post-radiotherapy patients undergoing multiparametric MRI and subsequent MRI/US fusion-guided and/or systematic biopsy. Histopathology results were used as ground truth. The recurrent cancer detection sensitivity of a bpMRI-based AI model, which was developed on a large dataset to primarily identify lesions in treatment-naïve patients, was compared to a prospective radiologist assessment using the Wald test. Subanalysis was conducted on patients stratified by the treatment modality (external beam radiation treatment [EBRT] and brachytherapy) and the prostate volume quartiles. RESULTS Of the 62 patients included (median age = 70 years; median PSA = 3.51 ng/ml; median prostate volume = 27.55 ml), 56 recurrent PCa foci were identified within 46 patients. The AI model detected 40 lesions in 35 patients. The AI model performance was lower than the prospective radiology interpretation (Rad) on a patient-(AI: 76.1% vs. Rad: 91.3%, p = 0.02) and lesion-level (AI: 71.4% vs. Rad: 87.5%, p = 0.01). The mean number of false positives per patient was 0.35 (range: 0-2). The AI model performance was higher in EBRT group both on patient-level (EBRT: 81.5% [22/27] vs. brachytherapy: 68.4% [13/19]) and lesion-level (EBRT: 79.4% [27/34] vs. brachytherapy: 59.1% [13/22]). In patients with gland volumes >34 ml (n = 25), detection sensitivities were 100% (11/11) and 94.1% (16/17) on patient- and lesion-level, respectively. CONCLUSION The reported bpMRI-based AI model detected the majority of locally recurrent prostate cancer after radiotherapy. Further testing including external validation of this model is warranted prior to clinical implementation.
Collapse
Affiliation(s)
- Enis C Yilmaz
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Stephanie A Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mason J Belue
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Katie M Merriman
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tim E Phelps
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yue Lin
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Charisse Garcia
- Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Lindsey Hazen
- Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Krishnan R Patel
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Maria J Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Bradford J Wood
- Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Peter L Choyke
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, MD, United States.
| |
Collapse
|
13
|
Zhang G, Zhang Z, Pei Y, Hu W, Xue Y, Ning R, Guo X, Sun Y, Zhang Q. Biological and clinical significance of radiomics features obtained from magnetic resonance imaging preceding pre-carbon ion radiotherapy in prostate cancer based on radiometabolomics. Front Endocrinol (Lausanne) 2023; 14:1272806. [PMID: 38027108 PMCID: PMC10644841 DOI: 10.3389/fendo.2023.1272806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction We aimed to investigate the feasibility of metabolomics to explain the underlying biological implications of radiomics features obtained from magnetic resonance imaging (MRI) preceding carbon ion radiotherapy (CIRT) in patients with prostate cancer and to further explore the clinical significance of radiomics features on the prognosis of patients, based on their biochemical recurrence (BCR) status. Methods Metabolomic results obtained using high-performance liquid chromatography coupled with tandem mass spectrometry of urine samples, combined with pre-RT radiomic features extracted from MRI images, were evaluated to investigate their biological significance. Receiver operating characteristic (ROC) curve analysis was subsequently conducted to examine the correlation between these biological implications and clinical BCR status. Statistical and metabolic pathway analyses were performed using MetaboAnalyst and R software. Results Correlation analysis revealed that methionine alteration extent was significantly related to four radiomic features (Contrast, Difference Variance, Small Dependence High Gray Level Emphasis, and Mean Absolute Deviation), which were significantly correlated with BCR status. The area under the curve (AUC) for BCR prediction of these four radiomic features ranged from 0.704 to 0.769, suggesting that the higher the value of these four radiomic features, the greater the decrease in methionine levels after CIRT and the lower the probability of BCR. Pre-CIRT MRI radiomic features were associated with CIRT-suppressed metabolites. Discussion These radiomic features can be used to predict the alteration in the amplitude of methionine after CIRT and the BCR status, which may contribute to the optimization of the CIRT strategy and deepen the understanding of PCa.
Collapse
Affiliation(s)
- Guangyuan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yulei Pei
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yushan Xue
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Renli Ning
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Xiaomao Guo
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yun Sun
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Qing Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| |
Collapse
|
14
|
Ye Y, Liu Z, Zhu J, Wu J, Sun K, Peng Y, Qiu J, Gong L. Development trends and knowledge framework in the application of magnetic resonance imaging in prostate cancer: a bibliometric analysis from 1984 to 2022. Quant Imaging Med Surg 2023; 13:6761-6777. [PMID: 37869318 PMCID: PMC10585509 DOI: 10.21037/qims-23-446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/07/2023] [Indexed: 10/24/2023]
Abstract
Background Prostate cancer (PCa) is the most common tumor of the male genitourinary system. With the development of imaging technology, the role of magnetic resonance imaging (MRI) in the management of PCa is increasing. The present study summarizes research on the application of MRI in the field of PCa using bibliometric analysis and predicts future research hotspots. Methods Articles regarding the application of MRI in PCa between January 1, 1984 and June 30, 2022 were selected from the Web of Science Core Collection (WoSCC) on November 6, 2022. Microsoft Excel 2016 and the Bibliometrix Biblioshiny R-package software were used for data analysis and bibliometric indicator extraction. CiteSpace (version 6.1.R3) was used to visualize literature feature clustering, including co-occurrence analysis of countries, institutions, authors, references, and burst keywords analysis. Results A total of 10,230 articles were included in the study. Turkbey was the most prolific author. The USA was the most productive country and had strong partnerships with other countries. The most productive institution was Memorial Sloan Kettering Cancer Center. Journal of Magnetic Resonance Imaging and Radiology were the most productive and highest impact factor (IF) journals in the field, respectively. Timeline views showed that "#1 multiparametric magnetic resonance imaging", "#4 pi-rads", and "#8 psma" were currently the latest research hotspots. Keywords burst analysis showed that "machine learning", "psa density", "multi parametric mri", "deep learning", and "artificial intelligence" were the most frequently used keywords in the past 3 years. Conclusions MRI has a wide range of applications in PCa. The USA is the leading country in this field, with a concentration of highly productive and high-level institutions. Meanwhile, it can be projected that "deep learning", "radiomics", and "artificial intelligence" will be research hotspots in the future.
Collapse
Affiliation(s)
- Yinquan Ye
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhixuan Liu
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianghua Zhu
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialong Wu
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ke Sun
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yun Peng
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jia Qiu
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lianggeng Gong
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Duenweg SR, Bobholz SA, Barrett MJ, Lowman AK, Winiarz A, Nath B, Stebbins M, Bukowy J, Iczkowski KA, Jacobsohn KM, Vincent-Sheldon S, LaViolette PS. T2-Weighted MRI Radiomic Features Predict Prostate Cancer Presence and Eventual Biochemical Recurrence. Cancers (Basel) 2023; 15:4437. [PMID: 37760407 PMCID: PMC10526331 DOI: 10.3390/cancers15184437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/23/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Prostate cancer (PCa) is the most diagnosed non-cutaneous cancer in men. Despite therapies such as radical prostatectomy, which is considered curative, distant metastases may form, resulting in biochemical recurrence (BCR). This study used radiomic features calculated from multi-parametric magnetic resonance imaging (MP-MRI) to evaluate their ability to predict BCR and PCa presence. Data from a total of 279 patients, of which 46 experienced BCR, undergoing MP-MRI prior to surgery were assessed for this study. After surgery, the prostate was sectioned using patient-specific 3D-printed slicing jigs modeled using the T2-weighted imaging (T2WI). Sectioned tissue was stained, digitized, and annotated by a GU-fellowship trained pathologist for cancer presence. Digitized slides and annotations were co-registered to the T2WI and radiomic features were calculated across the whole prostate and cancerous lesions. A tree regression model was fitted to assess the ability of radiomic features to predict BCR, and a tree classification model was fitted with the same radiomic features to classify regions of cancer. We found that 10 radiomic features predicted eventual BCR with an AUC of 0.97 and classified cancer at an accuracy of 89.9%. This study showcases the application of a radiomic feature-based tool to screen for the presence of prostate cancer and assess patient prognosis, as determined by biochemical recurrence.
Collapse
Affiliation(s)
- Savannah R. Duenweg
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA; (S.R.D.); (M.S.)
| | - Samuel A. Bobholz
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Michael J. Barrett
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Allison K. Lowman
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Aleksandra Winiarz
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA; (S.R.D.); (M.S.)
| | - Biprojit Nath
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA; (S.R.D.); (M.S.)
| | - Margaret Stebbins
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA; (S.R.D.); (M.S.)
| | - John Bukowy
- Department of Electrical Engineering and Computer Science, Milwaukee School of Engineering, 1025 N Broadway, Milwaukee, WI 53202, USA
| | - Kenneth A. Iczkowski
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA;
| | - Kenneth M. Jacobsohn
- Department of Urology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Stephanie Vincent-Sheldon
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Peter S. LaViolette
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA; (S.R.D.); (M.S.)
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| |
Collapse
|
16
|
Midya A, Hiremath A, Huber J, Sankar Viswanathan V, Omil-Lima D, Mahran A, Bittencourt LK, Harsha Tirumani S, Ponsky L, Shiradkar R, Madabhushi A. Delta radiomic patterns on serial bi-parametric MRI are associated with pathologic upgrading in prostate cancer patients on active surveillance: preliminary findings. Front Oncol 2023; 13:1166047. [PMID: 37731630 PMCID: PMC10508842 DOI: 10.3389/fonc.2023.1166047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/24/2023] [Indexed: 09/22/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Abhishek Midya
- Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
| | | | - Jacob Huber
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | | | | | - Amr Mahran
- Department of Urology, Assiut University, Asyut, Egypt
| | - Leonardo K. Bittencourt
- Department of Radiology, University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Sree Harsha Tirumani
- Department of Radiology, University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Lee Ponsky
- Department of Urology, University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Rakesh Shiradkar
- Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
- Atlanta Veterans Administration Medical Center, Atlanta, GA, United States
| |
Collapse
|
17
|
Jha AK, Mithun S, Sherkhane UB, Dwivedi P, Puts S, Osong B, Traverso A, Purandare N, Wee L, Rangarajan V, Dekker A. Emerging role of quantitative imaging (radiomics) and artificial intelligence in precision oncology. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:569-582. [PMID: 37720353 PMCID: PMC10501896 DOI: 10.37349/etat.2023.00153] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/20/2023] [Indexed: 09/19/2023] Open
Abstract
Cancer is a fatal disease and the second most cause of death worldwide. Treatment of cancer is a complex process and requires a multi-modality-based approach. Cancer detection and treatment starts with screening/diagnosis and continues till the patient is alive. Screening/diagnosis of the disease is the beginning of cancer management and continued with the staging of the disease, planning and delivery of treatment, treatment monitoring, and ongoing monitoring and follow-up. Imaging plays an important role in all stages of cancer management. Conventional oncology practice considers that all patients are similar in a disease type, whereas biomarkers subgroup the patients in a disease type which leads to the development of precision oncology. The utilization of the radiomic process has facilitated the advancement of diverse imaging biomarkers that find application in precision oncology. The role of imaging biomarkers and artificial intelligence (AI) in oncology has been investigated by many researchers in the past. The existing literature is suggestive of the increasing role of imaging biomarkers and AI in oncology. However, the stability of radiomic features has also been questioned. The radiomic community has recognized that the instability of radiomic features poses a danger to the global generalization of radiomic-based prediction models. In order to establish radiomic-based imaging biomarkers in oncology, the robustness of radiomic features needs to be established on a priority basis. This is because radiomic models developed in one institution frequently perform poorly in other institutions, most likely due to radiomic feature instability. To generalize radiomic-based prediction models in oncology, a number of initiatives, including Quantitative Imaging Network (QIN), Quantitative Imaging Biomarkers Alliance (QIBA), and Image Biomarker Standardisation Initiative (IBSI), have been launched to stabilize the radiomic features.
Collapse
Affiliation(s)
- Ashish Kumar Jha
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Sneha Mithun
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Umeshkumar B. Sherkhane
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
| | - Pooj Dwivedi
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
- Department of Nuclear Medicine, Advance Center for Treatment, Research, Education in Cancer, Kharghar, Navi-Mumbai 410210, Maharashtra, India
| | - Senders Puts
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Biche Osong
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| |
Collapse
|
18
|
Sánchez Iglesias Á, Morillo Macías V, Picó Peris A, Fuster-Matanzo A, Nogué Infante A, Muelas Soria R, Bellvís Bataller F, Domingo Pomar M, Casillas Meléndez C, Yébana Huertas R, Ferrer Albiach C. Prostate Region-Wise Imaging Biomarker Profiles for Risk Stratification and Biochemical Recurrence Prediction. Cancers (Basel) 2023; 15:4163. [PMID: 37627191 PMCID: PMC10453281 DOI: 10.3390/cancers15164163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Identifying prostate cancer (PCa) patients with a worse prognosis and a higher risk of biochemical recurrence (BCR) is essential to guide treatment choices. Here, we aimed to identify possible imaging biomarker (perfusion/diffusion + radiomic features) profiles extracted from MRIs that were able to discriminate patients according to their risk or the occurrence of BCR 10 years after diagnosis, as well as to evaluate their predictive value with or without clinical data. METHODS Patients with localized PCa receiving neoadjuvant androgen deprivation therapy and radiotherapy were retrospectively evaluated. Imaging features were extracted from MRIs for each prostate region or for the whole gland. Univariate and multivariate analyses were conducted. RESULTS 128 patients (mean [range] age, 71 [50-83] years) were included. Prostate region-wise imaging biomarker profiles mainly composed of radiomic features allowed discriminating risk groups and patients experiencing BCR. Heterogeneity-related radiomic features were increased in patients with worse prognosis and with BCR. Overall, imaging biomarkers profiles retained good predictive ability (AUC values superior to 0.725 in most cases), which generally improved when clinical data were included (particularly evident for the prediction of the BCR, with AUC values ranging from 0.841 to 0.877 for combined models and sensitivity values above 0.960) and when models were built per prostate region vs. the whole gland. CONCLUSIONS Prostate region-aware imaging profiles enable identification of patients with worse prognosis and with a higher risk of BCR, retaining higher predictive values when combined with clinical variables.
Collapse
Affiliation(s)
- Ángel Sánchez Iglesias
- Radiation Oncology Department, Hospital Provincial de Castellón, 12002 Castellón, Spain; (Á.S.I.); (V.M.M.); (R.M.S.)
| | - Virginia Morillo Macías
- Radiation Oncology Department, Hospital Provincial de Castellón, 12002 Castellón, Spain; (Á.S.I.); (V.M.M.); (R.M.S.)
| | - Alfonso Picó Peris
- Quantitative Imaging Biomarkers in Medicine (Quibim), 46021 Valencia, Spain; (A.P.P.); (A.F.-M.); (A.N.I.); (F.B.B.); (M.D.P.); (R.Y.H.)
| | - Almudena Fuster-Matanzo
- Quantitative Imaging Biomarkers in Medicine (Quibim), 46021 Valencia, Spain; (A.P.P.); (A.F.-M.); (A.N.I.); (F.B.B.); (M.D.P.); (R.Y.H.)
| | - Anna Nogué Infante
- Quantitative Imaging Biomarkers in Medicine (Quibim), 46021 Valencia, Spain; (A.P.P.); (A.F.-M.); (A.N.I.); (F.B.B.); (M.D.P.); (R.Y.H.)
| | - Rodrigo Muelas Soria
- Radiation Oncology Department, Hospital Provincial de Castellón, 12002 Castellón, Spain; (Á.S.I.); (V.M.M.); (R.M.S.)
| | - Fuensanta Bellvís Bataller
- Quantitative Imaging Biomarkers in Medicine (Quibim), 46021 Valencia, Spain; (A.P.P.); (A.F.-M.); (A.N.I.); (F.B.B.); (M.D.P.); (R.Y.H.)
| | - Marcos Domingo Pomar
- Quantitative Imaging Biomarkers in Medicine (Quibim), 46021 Valencia, Spain; (A.P.P.); (A.F.-M.); (A.N.I.); (F.B.B.); (M.D.P.); (R.Y.H.)
| | | | - Raúl Yébana Huertas
- Quantitative Imaging Biomarkers in Medicine (Quibim), 46021 Valencia, Spain; (A.P.P.); (A.F.-M.); (A.N.I.); (F.B.B.); (M.D.P.); (R.Y.H.)
| | - Carlos Ferrer Albiach
- Radiation Oncology Department, Hospital Provincial de Castellón, 12002 Castellón, Spain; (Á.S.I.); (V.M.M.); (R.M.S.)
| |
Collapse
|
19
|
Zhu X, Shao L, Liu Z, Liu Z, He J, Liu J, Ping H, Lu J. MRI-derived radiomics models for diagnosis, aggressiveness, and prognosis evaluation in prostate cancer. J Zhejiang Univ Sci B 2023; 24:663-681. [PMID: 37551554 PMCID: PMC10423970 DOI: 10.1631/jzus.b2200619] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/11/2023] [Indexed: 08/09/2023]
Abstract
Prostate cancer (PCa) is a pernicious tumor with high heterogeneity, which creates a conundrum for making a precise diagnosis and choosing an optimal treatment approach. Multiparametric magnetic resonance imaging (mp-MRI) with anatomical and functional sequences has evolved as a routine and significant paradigm for the detection and characterization of PCa. Moreover, using radiomics to extract quantitative data has emerged as a promising field due to the rapid growth of artificial intelligence (AI) and image data processing. Radiomics acquires novel imaging biomarkers by extracting imaging signatures and establishes models for precise evaluation. Radiomics models provide a reliable and noninvasive alternative to aid in precision medicine, demonstrating advantages over traditional models based on clinicopathological parameters. The purpose of this review is to provide an overview of related studies of radiomics in PCa, specifically around the development and validation of radiomics models using MRI-derived image features. The current landscape of the literature, focusing mainly on PCa detection, aggressiveness, and prognosis evaluation, is reviewed and summarized. Rather than studies that exclusively focus on image biomarker identification and method optimization, models with high potential for universal clinical implementation are identified. Furthermore, we delve deeper into the critical concerns that can be addressed by different models and the obstacles that may arise in a clinical scenario. This review will encourage researchers to design models based on actual clinical needs, as well as assist urologists in gaining a better understanding of the promising results yielded by radiomics.
Collapse
Affiliation(s)
- Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Lizhi Shao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100080, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Jide He
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Jiangang Liu
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing 100191, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
20
|
Kim H, Kang SW, Kim JH, Nagar H, Sabuncu M, Margolis DJA, Kim CK. The role of AI in prostate MRI quality and interpretation: Opportunities and challenges. Eur J Radiol 2023; 165:110887. [PMID: 37245342 DOI: 10.1016/j.ejrad.2023.110887] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/06/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
Prostate MRI plays an important role in imaging the prostate gland and surrounding tissues, particularly in the diagnosis and management of prostate cancer. With the widespread adoption of multiparametric magnetic resonance imaging in recent years, the concerns surrounding the variability of imaging quality have garnered increased attention. Several factors contribute to the inconsistency of image quality, such as acquisition parameters, scanner differences and interobserver variabilities. While efforts have been made to standardize image acquisition and interpretation via the development of systems, such as PI-RADS and PI-QUAL, the scoring systems still depend on the subjective experience and acumen of humans. Artificial intelligence (AI) has been increasingly used in many applications, including medical imaging, due to its ability to automate tasks and lower human error rates. These advantages have the potential to standardize the tasks of image interpretation and quality control of prostate MRI. Despite its potential, thorough validation is required before the implementation of AI in clinical practice. In this article, we explore the opportunities and challenges of AI, with a focus on the interpretation and quality of prostate MRI.
Collapse
Affiliation(s)
- Heejong Kim
- Department of Radiology, Weill Cornell Medical College, 525 E 68th St Box 141, New York, NY 10021, United States
| | - Shin Won Kang
- Research Institute for Future Medicine, Samsung Medical Center, Republic of Korea
| | - Jae-Hun Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Himanshu Nagar
- Department of Radiation Oncology, Weill Cornell Medical College, 525 E 68th St, New York, NY 10021, United States
| | - Mert Sabuncu
- Department of Radiology, Weill Cornell Medical College, 525 E 68th St Box 141, New York, NY 10021, United States
| | - Daniel J A Margolis
- Department of Radiology, Weill Cornell Medical College, 525 E 68th St Box 141, New York, NY 10021, United States.
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| |
Collapse
|
21
|
Spohn SKB, Schmidt-Hegemann NS, Ruf J, Mix M, Benndorf M, Bamberg F, Makowski MR, Kirste S, Rühle A, Nouvel J, Sprave T, Vogel MME, Galitsnaya P, Gschwend JE, Gratzke C, Stief C, Löck S, Zwanenburg A, Trapp C, Bernhardt D, Nekolla SG, Li M, Belka C, Combs SE, Eiber M, Unterrainer L, Unterrainer M, Bartenstein P, Grosu AL, Zamboglou C, Peeken JC. Development of PSMA-PET-guided CT-based radiomic signature to predict biochemical recurrence after salvage radiotherapy. Eur J Nucl Med Mol Imaging 2023; 50:2537-2547. [PMID: 36929180 PMCID: PMC10250433 DOI: 10.1007/s00259-023-06195-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE To develop a CT-based radiomic signature to predict biochemical recurrence (BCR) in prostate cancer patients after sRT guided by positron-emission tomography targeting prostate-specific membrane antigen (PSMA-PET). MATERIAL AND METHODS Consecutive patients, who underwent 68Ga-PSMA11-PET/CT-guided sRT from three high-volume centers in Germany, were included in this retrospective multicenter study. Patients had PET-positive local recurrences and were treated with intensity-modulated sRT. Radiomic features were extracted from volumes of interests on CT guided by focal PSMA-PET uptakes. After preprocessing, clinical, radiomics, and combined clinical-radiomic models were developed combining different feature reduction techniques and Cox proportional hazard models within a nested cross validation approach. RESULTS Among 99 patients, median interval until BCR was the radiomic models outperformed clinical models and combined clinical-radiomic models for prediction of BCR with a C-index of 0.71 compared to 0.53 and 0.63 in the test sets, respectively. In contrast to the other models, the radiomic model achieved significantly improved patient stratification in Kaplan-Meier analysis. The radiomic and clinical-radiomic model achieved a significantly better time-dependent net reclassification improvement index (0.392 and 0.762, respectively) compared to the clinical model. Decision curve analysis demonstrated a clinical net benefit for both models. Mean intensity was the most predictive radiomic feature. CONCLUSION This is the first study to develop a PSMA-PET-guided CT-based radiomic model to predict BCR after sRT. The radiomic models outperformed clinical models and might contribute to guide personalized treatment decisions.
Collapse
Affiliation(s)
- Simon K B Spohn
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany.
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany.
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | | | - Juri Ruf
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Michael Mix
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Matthias Benndorf
- Department of Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fabian Bamberg
- Department of Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcus R Makowski
- Department of Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Simon Kirste
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
| | - Jerome Nouvel
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
| | - Tanja Sprave
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
| | - Marco M E Vogel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Polina Galitsnaya
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Jürgen E Gschwend
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christian Gratzke
- Department of Urology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Alex Zwanenburg
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK) Partner Site Dresden, Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Christian Trapp
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Denise Bernhardt
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stephan G Nekolla
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Radiation Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lena Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, Heidelberg, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Oncology Center, European University of Cyprus, Limassol, Cyprus
| | - Jan C Peeken
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Radiation Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
22
|
Pierre K, Gupta M, Raviprasad A, Sadat Razavi SM, Patel A, Peters K, Hochhegger B, Mancuso A, Forghani R. Medical imaging and multimodal artificial intelligence models for streamlining and enhancing cancer care: opportunities and challenges. Expert Rev Anticancer Ther 2023; 23:1265-1279. [PMID: 38032181 DOI: 10.1080/14737140.2023.2286001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION Artificial intelligence (AI) has the potential to transform oncologic care. There have been significant developments in AI applications in medical imaging and increasing interest in multimodal models. These are likely to enable improved oncologic care through more precise diagnosis, increasingly in a more personalized and less invasive manner. In this review, we provide an overview of the current state and challenges that clinicians, administrative personnel and policy makers need to be aware of and mitigate for the technology to reach its full potential. AREAS COVERED The article provides a brief targeted overview of AI, a high-level review of the current state and future potential AI applications in diagnostic radiology and to a lesser extent digital pathology, focusing on oncologic applications. This is followed by a discussion of emerging approaches, including multimodal models. The article concludes with a discussion of technical, regulatory challenges and infrastructure needs for AI to realize its full potential. EXPERT OPINION There is a large volume of promising research, and steadily increasing commercially available tools using AI. For the most advanced and promising precision diagnostic applications of AI to be used clinically, robust and comprehensive quality monitoring systems and informatics platforms will likely be required.
Collapse
Affiliation(s)
- Kevin Pierre
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Manas Gupta
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
| | - Abheek Raviprasad
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Seyedeh Mehrsa Sadat Razavi
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Anjali Patel
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Keith Peters
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Bruno Hochhegger
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Anthony Mancuso
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Reza Forghani
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
- Division of Medical Physics, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Neurology, Division of Movement Disorders, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
23
|
Lee HW, Kim E, Na I, Kim CK, Seo SI, Park H. Novel Multiparametric Magnetic Resonance Imaging-Based Deep Learning and Clinical Parameter Integration for the Prediction of Long-Term Biochemical Recurrence-Free Survival in Prostate Cancer after Radical Prostatectomy. Cancers (Basel) 2023; 15:3416. [PMID: 37444526 DOI: 10.3390/cancers15133416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Radical prostatectomy (RP) is the main treatment of prostate cancer (PCa). Biochemical recurrence (BCR) following RP remains the first sign of aggressive disease; hence, better assessment of potential long-term post-RP BCR-free survival is crucial. Our study aimed to evaluate a combined clinical-deep learning (DL) model using multiparametric magnetic resonance imaging (mpMRI) for predicting long-term post-RP BCR-free survival in PCa. A total of 437 patients with PCa who underwent mpMRI followed by RP between 2008 and 2009 were enrolled; radiomics features were extracted from T2-weighted imaging, apparent diffusion coefficient maps, and contrast-enhanced sequences by manually delineating the index tumors. Deep features from the same set of imaging were extracted using a deep neural network based on pretrained EfficentNet-B0. Here, we present a clinical model (six clinical variables), radiomics model, DL model (DLM-Deep feature), combined clinical-radiomics model (CRM-Multi), and combined clinical-DL model (CDLM-Deep feature) that were built using Cox models regularized with the least absolute shrinkage and selection operator. We compared their prognostic performances using stratified fivefold cross-validation. In a median follow-up of 61 months, 110/437 patients experienced BCR. CDLM-Deep feature achieved the best performance (hazard ratio [HR] = 7.72), followed by DLM-Deep feature (HR = 4.37) or RM-Multi (HR = 2.67). CRM-Multi performed moderately. Our results confirm the superior performance of our mpMRI-derived DL algorithm over conventional radiomics.
Collapse
Affiliation(s)
- Hye Won Lee
- Samsung Medical Center, Department of Urology, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Eunjin Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Inye Na
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Seong Il Seo
- Samsung Medical Center, Department of Urology, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon 16419, Republic of Korea
| |
Collapse
|
24
|
Qi X, Wang K, Feng B, Sun X, Yang J, Hu Z, Zhang M, Lv C, Jin L, Zhou L, Wang Z, Yao J. Comparison of machine learning models based on multi-parametric magnetic resonance imaging and ultrasound videos for the prediction of prostate cancer. Front Oncol 2023; 13:1157949. [PMID: 37260984 PMCID: PMC10227569 DOI: 10.3389/fonc.2023.1157949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Objective To establish machine learning (ML) prediction models for prostate cancer (PCa) using transrectal ultrasound videos and multi-parametric magnetic resonance imaging (mpMRI) and compare their diagnostic performance. Materials and methods We systematically collated the data of 383 patients, including 187 with PCa and 196 with benign lesions. Of them, 307 patients (150 with PCa and 157 with benign lesions) were randomly selected to train and validate the ML models, 76 patients were used as test set. B-Ultrasound videos (BUS), mpMRI T2 sequence (T2), and ADC sequence (ADC) were obtained from all patients. We extracted 851 features of each patient in the BUS, T2, and ADC groups and used a t-test, the Mann-Whitney U test, and LASSO regression to screen the features. Support vector machine (SVM), random forest (RF), adaptive boosting (ADB), and gradient boosting machine (GBM) models were used to establish radiomics models. In addition, we fused the features screened via LASSO regression from three groups as new features and rebuilt ML models. The performance of the ML models in diagnosing PCa in the BUS, T2, ADC, and fusion groups was compared using the area under the ROC curve (AUC), sensitivity, specificity, and accuracy. Results In the test cohort, the AUC of each model in the ADC group was higher than that of in the.BUS and T2 groups. Among the models, the RF model had the best diagnostic performance, with an AUC of 0.85, sensitivity of 0.78 (0.61-0.89), specificity of 0.84 (0.69-0.94), and accuracy of 0.83 (0.66-0.93). The SVM model in both the BUS and T2 groups performed best. Based on the features screened in the BUS, T2, and ADC groups fused to construct the models, the SVM model was found to perform best, with an AUC of 0.87, sensitivity of 0.73 (0.56-0.86), specificity of 0.79 (0.63-0.90), and accuracy of 0.77 (0.59-0.89). The difference in the results was statistically significant (p<0.05). Conclusion The ML prediction models had a good diagnostic ability for PCa. Among them, the SVM model in the fusion group showed the best performance in diagnosing PCa. These prediction models can help radiologists make better diagnoses.
Collapse
Affiliation(s)
- Xiaoyang Qi
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Kai Wang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Bojian Feng
- Department of Ultrasound, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, Zhejiang, China
| | - Xingbo Sun
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Jie Yang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Zhengbiao Hu
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Maoliang Zhang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Cheng Lv
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Liyuan Jin
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Lingyan Zhou
- Department of Ultrasound, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, Zhejiang, China
| | - Zhengping Wang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Jincao Yao
- Department of Ultrasound, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Stanzione A, Ponsiglione A, Alessandrino F, Brembilla G, Imbriaco M. Beyond diagnosis: is there a role for radiomics in prostate cancer management? Eur Radiol Exp 2023; 7:13. [PMID: 36907973 PMCID: PMC10008761 DOI: 10.1186/s41747-023-00321-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/05/2023] [Indexed: 03/13/2023] Open
Abstract
The role of imaging in pretreatment staging and management of prostate cancer (PCa) is constantly evolving. In the last decade, there has been an ever-growing interest in radiomics as an image analysis approach able to extract objective quantitative features that are missed by human eye. However, most of PCa radiomics studies have been focused on cancer detection and characterisation. With this narrative review we aimed to provide a synopsis of the recently proposed potential applications of radiomics for PCa with a management-based approach, focusing on primary treatments with curative intent and active surveillance as well as highlighting on recurrent disease after primary treatment. Current evidence is encouraging, with radiomics and artificial intelligence appearing as feasible tools to aid physicians in planning PCa management. However, the lack of external independent datasets for validation and prospectively designed studies casts a shadow on the reliability and generalisability of radiomics models, delaying their translation into clinical practice.Key points• Artificial intelligence solutions have been proposed to streamline prostate cancer radiotherapy planning.• Radiomics models could improve risk assessment for radical prostatectomy patient selection.• Delta-radiomics appears promising for the management of patients under active surveillance.• Radiomics might outperform current nomograms for prostate cancer recurrence risk assessment.• Reproducibility of results, methodological and ethical issues must still be faced before clinical implementation.
Collapse
Affiliation(s)
- Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.
| | | | - Giorgio Brembilla
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Imbriaco
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
26
|
Chilaca-Rosas MF, Garcia-Lezama M, Moreno-Jimenez S, Roldan-Valadez E. Diagnostic Performance of Selected MRI-Derived Radiomics Able to Discriminate Progression-Free and Overall Survival in Patients with Midline Glioma and the H3F3AK27M Mutation. Diagnostics (Basel) 2023; 13:849. [PMID: 36899993 PMCID: PMC10001394 DOI: 10.3390/diagnostics13050849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Radiomics refers to a recent area of knowledge that studies features extracted from different imaging techniques and subsequently transformed into high-dimensional data that can be associated with biological events. Diffuse midline gliomas (DMG) are one of the most devastating types of cancer, with a median survival of approximately 11 months after diagnosis and 4-5 months after radiological and clinical progression. METHODS A retrospective study. From a database of 91 patients with DMG, only 12 had the H3.3K27M mutation and brain MRI DICOM files available. Radiomic features were extracted from MRI T1 and T2 sequences using LIFEx software. Statistical analysis included normal distribution tests and the Mann-Whitney U test, ROC analysis, and calculation of cut-off values. RESULTS A total of 5760 radiomic values were included in the analyses. AUROC demonstrated 13 radiomics with statistical significance for progression-free survival (PFS) and overall survival (OS). Diagnostic performance tests showed nine radiomics with specificity for PFS above 90% and one with a sensitivity of 97.2%. For OS, 3 out of 4 radiomics demonstrated between 80 and 90% sensitivity. CONCLUSIONS Several radiomic features demonstrated statistical significance and have the potential to further aid DMG diagnostic assessment non-invasively. The most significant radiomics were first- and second-order features with GLCM texture profile, GLZLM_GLNU, and NGLDM_Contrast.
Collapse
Affiliation(s)
- Maria-Fatima Chilaca-Rosas
- Radiotherapy Department, Hospital de Oncología, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Melissa Garcia-Lezama
- Directorate of Research, Hospital General de Mexico “Dr Eduardo Liceaga”, Mexico City 06720, Mexico
| | - Sergio Moreno-Jimenez
- Directorate of Surgery, Instituto Nacional de Neurología y Neurocirugia, “Manuel Velasco Suarez”, Mexico City 14269, Mexico
| | - Ernesto Roldan-Valadez
- Directorate of Research, Hospital General de Mexico “Dr Eduardo Liceaga”, Mexico City 06720, Mexico
- Department of Radiology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| |
Collapse
|
27
|
Canellas R, Kohli MD, Westphalen AC. The Evidence for Using Artificial Intelligence to Enhance Prostate Cancer MR Imaging. Curr Oncol Rep 2023; 25:243-250. [PMID: 36749494 DOI: 10.1007/s11912-023-01371-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current status of artificial intelligence applied to prostate cancer MR imaging. RECENT FINDINGS Artificial intelligence has been applied to prostate cancer MR imaging to improve its diagnostic accuracy and reproducibility of interpretation. Multiple models have been tested for gland segmentation and volume calculation, automated lesion detection, localization, and characterization, as well as prediction of tumor aggressiveness and tumor recurrence. Studies show, for example, that very robust automated gland segmentation and volume calculations can be achieved and that lesions can be detected and accurately characterized. Although results are promising, we should view these with caution. Most studies included a small sample of patients from a single institution and most models did not undergo proper external validation. More research is needed with larger and well-design studies for the development of reliable artificial intelligence tools.
Collapse
Affiliation(s)
- Rodrigo Canellas
- Department of Radiology, University of Washington, 1959 NE Pacific St., 2nd Floor, Seattle, WA, 98195, USA
| | - Marc D Kohli
- Clinical Informatics, Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94143, USA.,Imaging Informatics, UCSF Health, 500 Parnassus Ave, 3rd Floor, San Francisco, CA, 94143, USA
| | - Antonio C Westphalen
- Department of Radiology, University of Washington, 1959 NE Pacific St., 2nd Floor, Seattle, WA, 98195, USA. .,Department of Urology, University of Washington, 1959 NE Pacific St., 2nd Floor, Seattle, WA, 98195, USA. .,Department Radiation Oncology, University of Washington, 1959 NE Pacific St., 2nd Floor, Seattle, WA, 98195, USA.
| |
Collapse
|
28
|
Bologna M, Tenconi C, Corino VDA, Annunziata G, Orlandi E, Calareso G, Pignoli E, Valdagni R, Mainardi LT, Rancati T. Repeatability and reproducibility of MRI-radiomic features: A phantom experiment on a 1.5 T scanner. Med Phys 2023; 50:750-762. [PMID: 36310346 DOI: 10.1002/mp.16054] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/11/2022] Open
Abstract
PURPOSE Aim of this study is to assess the repeatability of radiomic features on magnetic resonance images (MRI) and their stability to variations in time of repetition (TR), time of echo (TE), slice thickness (ST), and pixel spacing (PS) using vegetable phantoms. METHODS The organic phantom was realized using two cucumbers placed inside a cylindrical container, and the analysis was performed using T1-weighted (T1w), T2-weighted (T2w), and diffusion-weighted images. One dataset was used to test the repeatability of the radiomic features, whereas other four datasets were used to test the sensitivity of the different MRI sequences to image acquisition parameters (TR, TE, ST, and PS). Four regions of interest (ROIs) were segmented: two for the central part of each cucumber and two for the external parts. Radiomic features were extracted from each ROI using Pyradiomics. To assess the effect of preprocessing on the reduction of variability, features were extracted both before and after the preprocessing. The coefficient of variation (CV) and intra-class correlation coefficient (ICC) were used to evaluate variability. RESULTS The use of intensity standardization increased the stability for the first-order statistics features. Shape and size features were always stable for all the analyses. Textural features were particularly sensitive to changes in ST and PS, although some increase in stability could be obtained by voxel size resampling. When images underwent image preprocessing, the number of stable features (ICC > 0.75 and mean absolute CV < 0.3) was 33 for apparent diffusion coefficient (ADC), 52 for T1w, and 73 for T2w. CONCLUSIONS The most critical source of variability is related to changes in voxel size (either caused by changes in ST or PS). Preprocessing increases features stability to both test-retest and variation of the image acquisition parameters for all the types of analyzed MRI (T1w, T2w, and ADC), except for ST.
Collapse
Affiliation(s)
- Marco Bologna
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Chiara Tenconi
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy.,Department of Medical Physics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina D A Corino
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Gaetano Annunziata
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ester Orlandi
- Radiation Oncology 2, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppina Calareso
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Emanuele Pignoli
- Department of Medical Physics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Riccardo Valdagni
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy.,Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.,Department of Radiation Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca T Mainardi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Tiziana Rancati
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
29
|
He K, Zhang Y, Li S, Yuan G, Liang P, Zhang Q, Xie Q, Xiao P, Li H, Meng X, Li Z. Incremental prognostic value of ADC histogram analysis in patients with high-risk prostate cancer receiving adjuvant hormonal therapy after radical prostatectomy. Front Oncol 2023; 13:1076400. [PMID: 36761966 PMCID: PMC9907778 DOI: 10.3389/fonc.2023.1076400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Purpose To investigate the incremental prognostic value of preoperative apparent diffusion coefficient (ADC) histogram analysis in patients with high-risk prostate cancer (PCa) who received adjuvant hormonal therapy (AHT) after radical prostatectomy (RP). Methods Sixty-two PCa patients in line with the criteria were enrolled in this study. The 10th, 50th, and 90th percentiles of ADC (ADC10, ADC50, ADC90), the mean value of ADC (ADCmean), kurtosis, and skewness were obtained from the whole-lesion ADC histogram. The Kaplan-Meier method and Cox regression analysis were used to analyze the relationship between biochemical recurrence-free survival (BCR-fs) and ADC parameters and other clinicopathological factors. Prognostic models were constructed with and without ADC parameters. Results The median follow-up time was 53.4 months (range, 41.1-79.3 months). BCR was found in 19 (30.6%) patients. Kaplan-Meier curves showed that lower ADCmean, ADC10, ADC50, and ADC90 and higher kurtosis could predict poorer BCR-fs (all p<0.05). After adjusting for clinical parameters, ADC50 and kurtosis remained independent prognostic factors for BCR-fs (HR: 0.172, 95% CI: 0.055-0.541, p=0.003; HR: 7.058, 95% CI: 2.288-21.773, p=0.001, respectively). By adding ADC parameters to the clinical model, the C index and diagnostic accuracy for the 24- and 36-month BCR-fs were improved. Conclusion ADC histogram analysis has incremental prognostic value in patients with high-risk PCa who received AHT after RP. Combining ADC50, kurtosis and clinical parameters can improve the accuracy of BCR-fs prediction.
Collapse
Affiliation(s)
- Kangwen He
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shichao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanjie Yuan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Liang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qingguo Xie
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Xiao
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Heng Li, ; Xiaoyan Meng,
| | - Xiaoyan Meng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Heng Li, ; Xiaoyan Meng,
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Ching JCF, Lam S, Lam CCH, Lui AOY, Kwong JCK, Lo AYH, Chan JWH, Cai J, Leung WS, Lee SWY. Integrating CT-based radiomic model with clinical features improves long-term prognostication in high-risk prostate cancer. Front Oncol 2023; 13:1060687. [PMID: 37205204 PMCID: PMC10186349 DOI: 10.3389/fonc.2023.1060687] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Objective High-risk prostate cancer (PCa) is often treated by prostate-only radiotherapy (PORT) owing to its favourable toxicity profile compared to whole-pelvic radiotherapy. Unfortunately, more than 50% patients still developed disease progression following PORT. Conventional clinical factors may be unable to identify at-risk subgroups in the era of precision medicine. In this study, we aimed to investigate the prognostic value of pre-treatment planning computed tomography (pCT)-based radiomic features and clinical attributes to predict 5-year progression-free survival (PFS) in high-risk PCa patients following PORT. Materials and methods A total of 176 biopsy-confirmed PCa patients who were treated at the Hong Kong Princess Margaret Hospital were retrospectively screened for eligibility. Clinical data and pCT of one hundred eligible high-risk PCa patients were analysed. Radiomic features were extracted from the gross-tumour-volume (GTV) with and without applying Laplacian-of-Gaussian (LoG) filter. The entire patient cohort was temporally stratified into a training and an independent validation cohort in a ratio of 3:1. Radiomics (R), clinical (C) and radiomic-clinical (RC) combined models were developed by Ridge regression through 5-fold cross-validation with 100 iterations on the training cohort. A model score was calculated for each model based on the included features. Model classification performance on 5-year PFS was evaluated in the independent validation cohort by average area-under-curve (AUC) of receiver-operating-characteristics (ROC) curve and precision-recall curve (PRC). Delong's test was used for model comparison. Results The RC combined model which contains 6 predictive features (tumour flatness, root-mean-square on fine LoG-filtered image, prostate-specific antigen serum concentration, Gleason score, Roach score and GTV volume) was the best-performing model (AUC = 0.797, 95%CI = 0.768-0.826), which significantly outperformed the R-model (AUC = 0.795, 95%CI = 0.774-0.816) and C-model (AUC = 0.625, 95%CI = 0.585-0.665) in the independent validation cohort. Besides, only the RC model score significantly classified patients in both cohorts into progression and progression-free groups regarding their 5-year PFS (p< 0.05). Conclusion Combining pCT-based radiomic and clinical attributes provided superior prognostication value regarding 5-year PFS in high-risk PCa patients following PORT. A large multi-centre study will potentially aid clinicians in implementing personalised treatment for this vulnerable subgroup in the future.
Collapse
Affiliation(s)
- Jerry C. F. Ching
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Saikit Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Research Institute for Smart Aging, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Cody C. H. Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Angie O. Y. Lui
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Joanne C. K. Kwong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Anson Y. H. Lo
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jason W. H. Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - W. S. Leung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- *Correspondence: Shara W. Y. Lee, ; W. S. Leung,
| | - Shara W. Y. Lee
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- *Correspondence: Shara W. Y. Lee, ; W. S. Leung,
| |
Collapse
|
31
|
Sminia P, Guipaud O, Viktorsson K, Ahire V, Baatout S, Boterberg T, Cizkova J, Dostál M, Fernandez-Palomo C, Filipova A, François A, Geiger M, Hunter A, Jassim H, Edin NFJ, Jordan K, Koniarová I, Selvaraj VK, Meade AD, Milliat F, Montoro A, Politis C, Savu D, Sémont A, Tichy A, Válek V, Vogin G. Clinical Radiobiology for Radiation Oncology. RADIOBIOLOGY TEXTBOOK 2023:237-309. [DOI: 10.1007/978-3-031-18810-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
AbstractThis chapter is focused on radiobiological aspects at the molecular, cellular, and tissue level which are relevant for the clinical use of ionizing radiation (IR) in cancer therapy. For radiation oncology, it is critical to find a balance, i.e., the therapeutic window, between the probability of tumor control and the probability of side effects caused by radiation injury to the healthy tissues and organs. An overview is given about modern precision radiotherapy (RT) techniques, which allow optimal sparing of healthy tissues. Biological factors determining the width of the therapeutic window are explained. The role of the six typical radiobiological phenomena determining the response of both malignant and normal tissues in the clinic, the 6R’s, which are Reoxygenation, Redistribution, Repopulation, Repair, Radiosensitivity, and Reactivation of the immune system, is discussed. Information is provided on tumor characteristics, for example, tumor type, growth kinetics, hypoxia, aberrant molecular signaling pathways, cancer stem cells and their impact on the response to RT. The role of the tumor microenvironment and microbiota is described and the effects of radiation on the immune system including the abscopal effect phenomenon are outlined. A summary is given on tumor diagnosis, response prediction via biomarkers, genetics, and radiomics, and ways to selectively enhance the RT response in tumors. Furthermore, we describe acute and late normal tissue reactions following exposure to radiation: cellular aspects, tissue kinetics, latency periods, permanent or transient injury, and histopathology. Details are also given on the differential effect on tumor and late responding healthy tissues following fractionated and low dose rate irradiation as well as the effect of whole-body exposure.
Collapse
|
32
|
Krithika alias AnbuDevi M, Suganthi K. Review of Semantic Segmentation of Medical Images Using Modified Architectures of UNET. Diagnostics (Basel) 2022; 12:diagnostics12123064. [PMID: 36553071 PMCID: PMC9777361 DOI: 10.3390/diagnostics12123064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
In biomedical image analysis, information about the location and appearance of tumors and lesions is indispensable to aid doctors in treating and identifying the severity of diseases. Therefore, it is essential to segment the tumors and lesions. MRI, CT, PET, ultrasound, and X-ray are the different imaging systems to obtain this information. The well-known semantic segmentation technique is used in medical image analysis to identify and label regions of images. The semantic segmentation aims to divide the images into regions with comparable characteristics, including intensity, homogeneity, and texture. UNET is the deep learning network that segments the critical features. However, UNETs basic architecture cannot accurately segment complex MRI images. This review introduces the modified and improved models of UNET suitable for increasing segmentation accuracy.
Collapse
|
33
|
Chiti G, Grazzini G, Flammia F, Matteuzzi B, Tortoli P, Bettarini S, Pasqualini E, Granata V, Busoni S, Messserini L, Pradella S, Massi D, Miele V. Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): a radiomic model to predict tumor grade. Radiol Med 2022; 127:928-938. [DOI: 10.1007/s11547-022-01529-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
|
34
|
Dang S, Guo Y, Han D, Ma G, Yu N, Yang Q, Duan X, Duan H, Ren J. MRI-based radiomics analysis in differentiating solid non-small-cell from small-cell lung carcinoma: a pilot study. Clin Radiol 2022; 77:e749-e757. [PMID: 35817610 DOI: 10.1016/j.crad.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/29/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
AIM To investigate the ability of a T2-weighted (W) magnetic resonance imaging (MRI)-based radiomics signature to differentiate solid non-small-cell lung carcinoma (NSCLC) from small-cell lung carcinoma (SCLC). MATERIALS AND METHODS The present retrospective study enrolled 152 eligible patients (NSCLC = 125, SCLC = 27). All patients underwent MRI using a 3 T scanner and radiomics features were extracted from T2W MRI. The least absolute shrinkage and selection operator (LASSO) logistic regression model was used to identify the optimal radiomics features for the construction of a radiomics model to differentiate solid NSCLC from SCLC. Threefold cross validation repeated 10 times was used for model training and evaluation. The conventional MRI morphology features of the lesions were also evaluated. The performance of the conventional MRI morphological features, and the radiomics signature model and nomogram model (combining radiomics signature with conventional MRI morphological features) was evaluated using receiver operating characteristic (ROC) curve analysis. RESULTS Five optimal features were chosen to build a radiomics signature. There was no significant difference in age, gender, and the largest diameter. The radiomics signature and conventional MRI morphological features (only pleural indentation and lymph node enlargement) were independent predictive factors for differentiating solid NSCLC from SCLC. The area under the ROC curves (AUCs) for MRI morphological features, and the radiomics model, and nomogram model was 0.69, 0.85, and 0.90 (ROC), respectively. CONCLUSIONS The T2W MRI-based radiomics signature is a potential non-invasive approach for distinguishing solid NSCLC from SCLC.
Collapse
Affiliation(s)
- S Dang
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - Y Guo
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - D Han
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - G Ma
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - N Yu
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China; Shaanxi University of Chinese Medicine, Xianyang, China
| | - Q Yang
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - X Duan
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - H Duan
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China; Shaanxi University of Chinese Medicine, Xianyang, China.
| | - J Ren
- GE Healthcare China, Daxing District, Beijing, China
| |
Collapse
|
35
|
Ismail M, Prasanna P, Bera K, Statsevych V, Hill V, Singh G, Partovi S, Beig N, McGarry S, Laviolette P, Ahluwalia M, Madabhushi A, Tiwari P. Radiomic Deformation and Textural Heterogeneity (R-DepTH) Descriptor to Characterize Tumor Field Effect: Application to Survival Prediction in Glioblastoma. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:1764-1777. [PMID: 35108202 PMCID: PMC9575333 DOI: 10.1109/tmi.2022.3148780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The concept of tumor field effect implies that cancer is a systemic disease with its impact way beyond the visible tumor confines. For instance, in Glioblastoma (GBM), an aggressive brain tumor, the increase in intracranial pressure due to tumor burden often leads to brain herniation and poor outcomes. Our work is based on the rationale that highly aggressive tumors tend to grow uncontrollably, leading to pronounced biomechanical tissue deformations in the normal parenchyma, which when combined with local morphological differences in the tumor confines on MRI scans, will comprehensively capture tumor field effect. Specifically, we present an integrated MRI-based descriptor, radiomic-Deformation and Textural Heterogeneity (r-DepTH). This descriptor comprises measurements of the subtle perturbations in tissue deformations throughout the surrounding normal parenchyma due to mass effect. This involves non-rigidly aligning the patients' MRI scans to a healthy atlas via diffeomorphic registration. The resulting inverse mapping is used to obtain the deformation field magnitudes in the normal parenchyma. These measurements are then combined with a 3D texture descriptor, Co-occurrence of Local Anisotropic Gradient Orientations (COLLAGE), which captures the morphological heterogeneity and infiltration within the tumor confines, on MRI scans. In this work, we extensively evaluated r-DepTH for survival risk-stratification on a total of 207 GBM cases from 3 different cohorts (Cohort 1 ( n1 = 53 ), Cohort 2 ( n2 = 75 ), and Cohort 3 ( n3 = 79 )), where each of these three cohorts was used as a training set for our model separately, and the other two cohorts were used for testing, independently, for each training experiment. When employing Cohort 1 for training, r-DepTH yielded Concordance indices (C-indices) of 0.7 and 0.65, hazard ratios (HR) and Confidence Intervals (CI) of 10 (6 - 19) and 5 (3 - 8) on Cohorts 2 and 3, respectively. Similarly, training on Cohort 2 yielded C-indices of 0.6 and 0.7, HR and CI of 1 (0.7 - 2) and 3 (2 - 5) on Cohorts 1 and 3, respectively. Finally, training on Cohort 3 yielded C-indices of 0.75 and 0.63, HR and CI of 24 (10 - 57) and 12 (6 - 21) on Cohorts 1 and 2, respectively. Our results show that r-DepTH descriptor may serve as a comprehensive and a robust MRI-based prognostic marker of disease aggressiveness and survival in solid tumors.
Collapse
|
36
|
Jha AK, Mithun S, Purandare NC, Kumar R, Rangarajan V, Wee L, Dekker A. Radiomics: a quantitative imaging biomarker in precision oncology. Nucl Med Commun 2022; 43:483-493. [PMID: 35131965 DOI: 10.1097/mnm.0000000000001543] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cancer treatment is heading towards precision medicine driven by genetic and biochemical markers. Various genetic and biochemical markers are utilized to render personalized treatment in cancer. In the last decade, noninvasive imaging biomarkers have also been developed to assist personalized decision support systems in oncology. The imaging biomarkers i.e., radiomics is being researched to develop specific digital phenotype of tumor in cancer. Radiomics is a process to extract high throughput data from medical images by using advanced mathematical and statistical algorithms. The radiomics process involves various steps i.e., image generation, segmentation of region of interest (e.g. a tumor), image preprocessing, radiomic feature extraction, feature analysis and selection and finally prediction model development. Radiomics process explores the heterogeneity, irregularity and size parameters of the tumor to calculate thousands of advanced features. Our study investigates the role of radiomics in precision oncology. Radiomics research has witnessed a rapid growth in the last decade with several studies published that show the potential of radiomics in diagnosis and treatment outcome prediction in oncology. Several radiomics based prediction models have been developed and reported in the literature to predict various prediction endpoints i.e., overall survival, progression-free survival and recurrence in various cancer i.e., brain tumor, head and neck cancer, lung cancer and several other cancer types. Radiomics based digital phenotypes have shown promising results in diagnosis and treatment outcome prediction in oncology. In the coming years, radiomics is going to play a significant role in precision oncology.
Collapse
Affiliation(s)
- Ashish Kumar Jha
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Sneha Mithun
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Nilendu C Purandare
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Science, New Delhi, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
| |
Collapse
|
37
|
Shen Z, Wu H, Chen Z, Hu J, Pan J, Kong J, Lin T. The Global Research of Artificial Intelligence on Prostate Cancer: A 22-Year Bibliometric Analysis. Front Oncol 2022; 12:843735. [PMID: 35299747 PMCID: PMC8921533 DOI: 10.3389/fonc.2022.843735] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/28/2022] [Indexed: 01/03/2023] Open
Abstract
Background With the rapid development of technology, artificial intelligence (AI) has been widely used in the diagnosis and prognosis prediction of a variety of diseases, including prostate cancer. Facts have proved that AI has broad prospects in the accurate diagnosis and treatment of prostate cancer. Objective This study mainly summarizes the research on the application of artificial intelligence in the field of prostate cancer through bibliometric analysis and explores possible future research hotspots. Methods The articles and reviews regarding application of AI in prostate cancer between 1999 and 2020 were selected from Web of Science Core Collection on August 23, 2021. Microsoft Excel 2019 and GraphPad Prism 8 were applied to analyze the targeted variables. VOSviewer (version 1.6.16), Citespace (version 5.8.R2), and a widely used online bibliometric platform were used to conduct co-authorship, co-citation, and co-occurrence analysis of countries, institutions, authors, references, and keywords in this field. Results A total of 2,749 articles were selected in this study. AI-related research on prostate cancer increased exponentially in recent years, of which the USA was the most productive country with 1,342 publications, and had close cooperation with many countries. The most productive institution and researcher were the Henry Ford Health System and Tewari. However, the cooperation among most institutions or researchers was not close even if the high research outputs. The result of keyword analysis could divide all studies into three clusters: “Diagnosis and Prediction AI-related study”, “Non-surgery AI-related study”, and “Surgery AI-related study”. Meanwhile, the current research hotspots were “deep learning” and “multiparametric MRI”. Conclusions Artificial intelligence has broad application prospects in prostate cancer, and a growing number of scholars are devoted to AI-related research on prostate cancer. Meanwhile, the cooperation among various countries and institutions needs to be strengthened in the future. It can be projected that noninvasive diagnosis and accurate minimally invasive treatment through deep learning technology will still be the research focus in the next few years.
Collapse
Affiliation(s)
- Zefeng Shen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haiyang Wu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Zeshi Chen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jintao Hu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiexin Pan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| |
Collapse
|
38
|
Cho HH, Kim CK, Park H. Overview of radiomics in prostate imaging and future directions. Br J Radiol 2022; 95:20210539. [PMID: 34797688 PMCID: PMC8978251 DOI: 10.1259/bjr.20210539] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advancements in imaging technology and analysis methods have led to an analytic framework known as radiomics. This framework extracts comprehensive high-dimensional features from imaging data and performs data mining to build analytical models for improved decision-support. Its features include many categories spanning texture and shape; thus, it can provide abundant information for precision medicine. Many studies of prostate radiomics have shown promising results in the assessment of pathological features, prediction of treatment response, and stratification of risk groups. Herein, we aimed to provide a general overview of radiomics procedures, discuss technical issues, explain various clinical applications, and suggest future research directions, especially for prostate imaging.
Collapse
Affiliation(s)
- Hwan-Ho Cho
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea.,School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
39
|
Zhang L, Jiang D, Chen C, Yang X, Lei H, Kang Z, Huang H, Pang J. Development and validation of a multiparametric MRI-based radiomics signature for distinguishing between indolent and aggressive prostate cancer. Br J Radiol 2022; 95:20210191. [PMID: 34289319 PMCID: PMC8978240 DOI: 10.1259/bjr.20210191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To develop and validate a non-invasive MRI-based radiomics signature for distinguishing between indolent and aggressive prostate cancer (PCa) prior to therapy. METHODS In all, 139 qualified and pathology-confirmed PCa patients were divided into a training set (n = 93) and a validation set (n = 46). A total of 1576 radiomics features were extracted from the T2WI (n = 788) and diffusion-weighted imaging (n = 788) for each patient. The Select K Best and the least absolute shrinkage and selection operator regression algorithm were used to construct a radiomics signature in the training set. The predictive performance of the radiomics signature was assessed in the training set and then validated in the validation set by receiver operating characteristic curve analysis. We computed the calibration curve and the decision curve to evaluate the calibration and clinical usefulness of the signature. RESULTS Nine radiomics features were identified to form the radiomics signature. The radiomics score (Rad-score) was significantly different between indolent and aggressive PCa (p < 0.001). The radiomics signature exhibited favorable discrimination between the indolent and aggressive PCa groups in the training set (AUC: 0.853, 95% CI: 0.766 to 0.941) and validation set (AUC: 0.901, 95% CI: 0.793 to 1.000). The decision curve analysis showed that a greater net benefit would be obtained when the threshold probability ranged from 20 to 90%. CONCLUSION The multiparametric MRI-based radiomics signature can potentially serve as a non-invasive tool for distinguishing between indolent and aggressive PCa prior to therapy. ADVANCES IN KNOWLEDGE The multiparametric MRI-based radiomics signature has the potential to non-invasively distinguish between the indolent and aggressive PCa, which might aid clinicians in making personalized therapeutic decisions.
Collapse
Affiliation(s)
| | - Donggen Jiang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center,The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chujie Chen
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center,The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiangwei Yang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center,The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hanqi Lei
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center,The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhuang Kang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hai Huang
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center,The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
40
|
Bera K, Braman N, Gupta A, Velcheti V, Madabhushi A. Predicting cancer outcomes with radiomics and artificial intelligence in radiology. Nat Rev Clin Oncol 2022; 19:132-146. [PMID: 34663898 PMCID: PMC9034765 DOI: 10.1038/s41571-021-00560-7] [Citation(s) in RCA: 282] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2021] [Indexed: 12/14/2022]
Abstract
The successful use of artificial intelligence (AI) for diagnostic purposes has prompted the application of AI-based cancer imaging analysis to address other, more complex, clinical needs. In this Perspective, we discuss the next generation of challenges in clinical decision-making that AI tools can solve using radiology images, such as prognostication of outcome across multiple cancers, prediction of response to various treatment modalities, discrimination of benign treatment confounders from true progression, identification of unusual response patterns and prediction of the mutational and molecular profile of tumours. We describe the evolution of and opportunities for AI in oncology imaging, focusing on hand-crafted radiomic approaches and deep learning-derived representations, with examples of their application for decision support. We also address the challenges faced on the path to clinical adoption, including data curation and annotation, interpretability, and regulatory and reimbursement issues. We hope to demystify AI in radiology for clinicians by helping them to understand its limitations and challenges, as well as the opportunities it provides as a decision-support tool in cancer management.
Collapse
Affiliation(s)
- Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Nathaniel Braman
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Tempus Labs, Chicago, IL, USA
| | - Amit Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Vamsidhar Velcheti
- Department of Hematology and Oncology, NYU Langone Health, New York, NY, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
- Louis Stokes Cleveland Veterans Medical Center, Cleveland, OH, USA.
| |
Collapse
|
41
|
Huang G, Cui Y, Wang P, Ren J, Wang L, Ma Y, Jia Y, Ma X, Zhao L. Multi-Parametric Magnetic Resonance Imaging-Based Radiomics Analysis of Cervical Cancer for Preoperative Prediction of Lymphovascular Space Invasion. Front Oncol 2022; 11:663370. [PMID: 35096556 PMCID: PMC8790703 DOI: 10.3389/fonc.2021.663370] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 12/17/2021] [Indexed: 01/03/2023] Open
Abstract
Background Detection of lymphovascular space invasion (LVSI) in early cervical cancer (CC) is challenging. To date, no standard clinical markers or screening tests have been used to detect LVSI preoperatively. Therefore, non-invasive risk stratification tools are highly desirable. Objective To train and validate a multi-parametric magnetic resonance imaging (mpMRI)-based radiomics model to detect LVSI in patients with CC and investigate its potential as a complementary tool to enhance the efficiency of risk assessment strategies. Materials and Methods The model was developed from the tumor volume of interest (VOI) of 125 patients with CC. A total of 1037 radiomics features obtained from conventional magnetic resonance imaging (MRI), including a small field-of-view (sFOV) high-resolution (HR)-T2-weighted MRI (T2WI), apparent diffusion coefficient (ADC), T2WI, fat-suppressed (FS)-T2WI, as well as axial and sagittal contrast-enhanced T1-weighted MRI (T1c). We conducted a radiomics-based characterization of each tumor region using pretreatment image data. Feature selection was performed using the least absolute shrinkage and selection operator method on the training set. The predictive performance was compared with single variates (clinical data and single-layer radiomics signatures) analyzed using a receiver operating characteristic (ROC) curve. Three-fold cross-validation performed 20 times was used to evaluate the accuracy of the trained classifiers and the stability of the selected features. The models were validated by using a validation set. Results Feature selection extracted the six most important features (3 from sFOV HR-T2WI, 1 T2WI, 1 FS-T2WI, and 1 T1c) for model construction. The mpMRI-combined radiomics model (area under the curve [AUC]: 0.940) reached a significantly higher performance (better than the clinical parameters [AUC: 0.730]), including any single-layer model using sFOV HR-T2WI (AUC: 0.840), T2WI (AUC: 0.770), FS-T2WI (AUC: 0.710), ADC maps (AUC: 0.650), sagittal, and axial T1c values (AUC: 0.710, 0.680) in the validation set. Conclusion Biomarkers using multi-parametric radiomics features derived from preoperative MR images could predict LVSI in patients with CC.
Collapse
Affiliation(s)
- Gang Huang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | - Yaqiong Cui
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China.,The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Wang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | | | - Lili Wang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | - Yaqiong Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | - Yingmei Jia
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | - Xiaomei Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | - Lianping Zhao
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
42
|
Li H, Lee CH, Chia D, Lin Z, Huang W, Tan CH. Machine Learning in Prostate MRI for Prostate Cancer: Current Status and Future Opportunities. Diagnostics (Basel) 2022; 12:diagnostics12020289. [PMID: 35204380 PMCID: PMC8870978 DOI: 10.3390/diagnostics12020289] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/31/2021] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Advances in our understanding of the role of magnetic resonance imaging (MRI) for the detection of prostate cancer have enabled its integration into clinical routines in the past two decades. The Prostate Imaging Reporting and Data System (PI-RADS) is an established imaging-based scoring system that scores the probability of clinically significant prostate cancer on MRI to guide management. Image fusion technology allows one to combine the superior soft tissue contrast resolution of MRI, with real-time anatomical depiction using ultrasound or computed tomography. This allows the accurate mapping of prostate cancer for targeted biopsy and treatment. Machine learning provides vast opportunities for automated organ and lesion depiction that could increase the reproducibility of PI-RADS categorisation, and improve co-registration across imaging modalities to enhance diagnostic and treatment methods that can then be individualised based on clinical risk of malignancy. In this article, we provide a comprehensive and contemporary review of advancements, and share insights into new opportunities in this field.
Collapse
Affiliation(s)
- Huanye Li
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore; (H.L.); (Z.L.)
| | - Chau Hung Lee
- Department of Diagnostic Radiology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
| | - David Chia
- Department of Radiation Oncology, National University Cancer Institute (NUH), Singapore 119074, Singapore;
| | - Zhiping Lin
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore; (H.L.); (Z.L.)
| | - Weimin Huang
- Institute for Infocomm Research, A*Star, Singapore 138632, Singapore;
| | - Cher Heng Tan
- Department of Diagnostic Radiology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798, Singapore
- Correspondence:
| |
Collapse
|
43
|
Ferro M, de Cobelli O, Musi G, del Giudice F, Carrieri G, Busetto GM, Falagario UG, Sciarra A, Maggi M, Crocetto F, Barone B, Caputo VF, Marchioni M, Lucarelli G, Imbimbo C, Mistretta FA, Luzzago S, Vartolomei MD, Cormio L, Autorino R, Tătaru OS. Radiomics in prostate cancer: an up-to-date review. Ther Adv Urol 2022; 14:17562872221109020. [PMID: 35814914 PMCID: PMC9260602 DOI: 10.1177/17562872221109020] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most common worldwide diagnosed malignancy in male population. The diagnosis, the identification of aggressive disease, and the post-treatment follow-up needs a more comprehensive and holistic approach. Radiomics is the extraction and interpretation of images phenotypes in a quantitative manner. Radiomics may give an advantage through advancements in imaging modalities and through the potential power of artificial intelligence techniques by translating those features into clinical outcome prediction. This article gives an overview on the current evidence of methodology and reviews the available literature on radiomics in PCa patients, highlighting its potential for personalized treatment and future applications.
Collapse
Affiliation(s)
- Matteo Ferro
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy, via Ripamonti 435 Milano, Italy
| | - Ottavio de Cobelli
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Gennaro Musi
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Francesco del Giudice
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Carrieri
- Department of Urology and Organ Transplantation, University of Foggia, Foggia, Italy
| | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, Foggia, Italy
| | | | - Alessandro Sciarra
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Martina Maggi
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Biagio Barone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Vincenzo Francesco Caputo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio, University of Chieti, Chieti, Italy; Urology Unit, ‘SS. Annunziata’ Hospital, Chieti, Italy
- Department of Urology, ASL Abruzzo 2, Chieti, Italy
| | - Giuseppe Lucarelli
- Department of Emergency and Organ Transplantation, Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Francesco Alessandro Mistretta
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Stefano Luzzago
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Mihai Dorin Vartolomei
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mures, Târgu Mures, Romania
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Luigi Cormio
- Urology and Renal Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Urology Unit, Bonomo Teaching Hospital, Foggia, Italy
| | | | - Octavian Sabin Tătaru
- Institution Organizing University Doctoral Studies, I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mures, Târgu Mures, Romania
| |
Collapse
|
44
|
Differentiating Central Lung Tumors from Atelectasis with Contrast-Enhanced CT-Based Radiomics Features. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5522452. [PMID: 34820455 PMCID: PMC8608546 DOI: 10.1155/2021/5522452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/20/2021] [Indexed: 01/29/2023]
Abstract
Objectives To evaluate the utility of radiomics features in differentiating central lung cancers and atelectasis on contrast-enhanced computed tomography (CT) images. This study is retrospective. Materials and Methods In this study, 36 patients with central pulmonary cancer and atelectasis between July 2013 and June 2018 were identified. A total of 1,653 2D and 2,327 3D radiomics features were extracted from segmented lung cancers and atelectasis on contrast-enhanced CT. The refined features were investigated for usefulness in classifying lung cancer and atelectasis according to the information gain, and 10 models were trained based on these features. The classification model is trained and tested at the region level and pixel level, respectively. Results Among all the extracted features, 334 2D features and 1,507 3D features had an information gain (IG) greater than 0.1. The highest accuracy (AC) of the region classifiers was 0.9375. The best Dice score, Hausdorff distance, and voxel AC were 0.2076, 45.28, and 0.8675, respectively. Conclusions Radiomics features derived from contrast-enhanced CT images can differentiate lung cancers and atelectasis at the regional and voxel levels.
Collapse
|
45
|
Ghezzo S, Bezzi C, Presotto L, Mapelli P, Bettinardi V, Savi A, Neri I, Preza E, Samanes Gajate AM, De Cobelli F, Scifo P, Picchio M. State of the art of radiomic analysis in the clinical management of prostate cancer: A systematic review. Crit Rev Oncol Hematol 2021; 169:103544. [PMID: 34801699 DOI: 10.1016/j.critrevonc.2021.103544] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 02/04/2023] Open
Abstract
We present the current clinical applications of radiomics in the context of prostate cancer (PCa) management. Several online databases for original articles using a combination of the following keywords: "(radiomic or radiomics) AND (prostate cancer or prostate tumour or prostate tumor or prostate neoplasia)" have been searched. The selected papers have been pooled as focus on (i) PCa detection, (ii) assessing the clinical significance of PCa, (iii) biochemical recurrence prediction, (iv) radiation-therapy outcome prediction and treatment efficacy monitoring, (v) metastases detection, (vi) metastases prediction, (vii) prediction of extra-prostatic extension. Seventy-six studies were included for qualitative analyses. Classifiers powered with radiomic features were able to discriminate between healthy tissue and PCa and between low- and high-risk PCa. However, before radiomics can be proposed for clinical use its methods have to be standardized, and these first encouraging results need to be robustly replicated in large and independent cohorts.
Collapse
Affiliation(s)
| | | | - Luca Presotto
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Mapelli
- Vita-Salute San Raffaele University, Milan, Italy; Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentino Bettinardi
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Annarita Savi
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Neri
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Erik Preza
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesco De Cobelli
- Vita-Salute San Raffaele University, Milan, Italy; Radiology Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Scifo
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Picchio
- Vita-Salute San Raffaele University, Milan, Italy; Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
46
|
Mendes B, Domingues I, Silva A, Santos J. Prostate Cancer Aggressiveness Prediction Using CT Images. Life (Basel) 2021; 11:life11111164. [PMID: 34833040 PMCID: PMC8618689 DOI: 10.3390/life11111164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Prostate Cancer (PCa) is mostly asymptomatic at an early stage and often painless requiring active surveillance screening. Transrectal Ultrasound Guided Biopsy (TRUS) is the principal method to diagnose PCa following a histological examination by observing cell pattern irregularities and assigning the Gleason Score (GS) according to the recommended guidelines. This procedure presents sampling errors and, being invasive may cause complications to the patients. External Beam Radiotherapy Treatment (EBRT) is presented as curative option for localised and locally advanced disease, as a palliative option for metastatic low-volume disease or after prostatectomy for prostate bed and pelvic nodes salvage. In the EBRT worflow a Computed Tomography (CT) scan is performed as the basis for dose calculations and volume delineations. In this work, we evaluated the use of data-characterization algorithms (radiomics) from CT images for PCa aggressiveness assessment. The fundamental motivation relies on the wide availability of CT images and the need to provide tools to assess EBRT effectiveness. We used Pyradiomics and Local Image Features Extraction (LIFEx) to extract features and search for a radiomic signature within CT images. Finnaly, when applying Principal Component Analysis (PCA) to the features, we were able to show promising results.
Collapse
Affiliation(s)
- Bruno Mendes
- Centro de Investigação do Instituto Português de Oncologia do Porto (CI-IPOP), Grupo de Física Médica, Radiobiologia e Protecção Radiológica, 4200-072 Porto, Portugal; (I.D.); (J.S.)
- Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
- Correspondence:
| | - Inês Domingues
- Centro de Investigação do Instituto Português de Oncologia do Porto (CI-IPOP), Grupo de Física Médica, Radiobiologia e Protecção Radiológica, 4200-072 Porto, Portugal; (I.D.); (J.S.)
- Instituto Superior de Engenharia de Coimbra (ISEC), 3030-199 Coimbra, Portugal
| | - Augusto Silva
- IEETA, Universidade de Aveiro (UA), 3810-193 Aveiro, Portugal;
| | - João Santos
- Centro de Investigação do Instituto Português de Oncologia do Porto (CI-IPOP), Grupo de Física Médica, Radiobiologia e Protecção Radiológica, 4200-072 Porto, Portugal; (I.D.); (J.S.)
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), 4050-313 Porto, Portugal
| |
Collapse
|
47
|
Machine Learning: Applications and Advanced Progresses of Radiomics in Endocrine Neoplasms. JOURNAL OF ONCOLOGY 2021; 2021:8615450. [PMID: 34671399 PMCID: PMC8523238 DOI: 10.1155/2021/8615450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Endocrine neoplasms remain a great threat to human health. It is extremely important to make a clear diagnosis and timely treatment of endocrine tumors. Machine learning includes radiomics, which has long been utilized in clinical cancer research. Radiomics refers to the extraction of valuable information by analyzing a large amount of standard data with high-throughput medical images mainly including computed tomography, positron emission tomography, magnetic resonance imaging, and ultrasound. With the quantitative imaging analysis and model building, radiomics can reflect specific underlying characteristics of a disease that otherwise could not be evaluated visually. More and more promising results of radiomics in oncological practice have been seen in recent years. Radiomics may have the potential to supplement traditional imaging analysis and assist in providing precision medicine for patients. Radiomics had developed rapidly in endocrine neoplasms practice in the past decade. In this review, we would introduce the general workflow of radiomics and summarize the applications and developments of radiomics in endocrine neoplasms in recent years. The limitations of current radiomic research studies and future development directions would also be discussed.
Collapse
|
48
|
Gu L, Liu Y, Guo X, Tian Y, Ye H, Zhou S, Gao F. Computed tomography-based radiomic analysis for prediction of treatment response to salvage chemoradiotherapy for locoregional lymph node recurrence after curative esophagectomy. J Appl Clin Med Phys 2021; 22:71-79. [PMID: 34614265 PMCID: PMC8598151 DOI: 10.1002/acm2.13434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/15/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Objective To investigate the capability of computed tomography (CT) radiomic features to predict the therapeutic response and local control of the locoregional recurrence lymph node (LN) after curative esophagectomy by chemoradiotherapy. Methods This retrospective study included 129 LN from 77 patients (training cohort: 102 LN from 59 patients; validation cohort: 27 LN from 18 patients) with postoperative esophageal squamous cell carcinoma (ESCC). The region of the tumor was contoured in pretreatment contrast‐enhanced CT images. The least absolute shrinkage and selection operator with logistic regression was used to identify radiomic predictors in the training cohort. Model performance was evaluated using the area under the receiver operating characteristic curves (AUC). The Kaplan–Meier method was used to determine the local recurrence time of cancer. Results The radiomic model suggested seven features that could be used to predict treatment response. The AUCs in training and validated cohorts were 0.777 (95% CI: 0.667–0.878) and 0.765 (95% CI: 0.556–0.975), respectively. A significant difference in the radiomic scores (Rad‐scores) between response and nonresponse was observed in the two cohorts (p < 0.001, 0.034, respectively). Two features were identified for classifying whether there will be relapse in 2 years. AUC was 0.857 (95% CI: 0.780–0.935) in the training cohort. The local control time of the high Rad‐score group was higher than the low group in both cohorts (p < 0.001 and 0.025, respectively). As inferred from the Cox regression analysis, the low Rad‐score was a high‐risk factor for local recurrence within 2 years. Conclusions The radiomic approach can be used as a potential imaging biomarker to predict treatment response and local control of recurrence LN in ESCC patients.
Collapse
Affiliation(s)
- Liang Gu
- Department of Radiation Oncology, Taixing People's Hospital, Tai Xing, Jiangsu Province, China.,Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Su Zhou, Jiangsu Province, China
| | - Yangchen Liu
- Department of Radiation Oncology, Taixing People's Hospital, Tai Xing, Jiangsu Province, China
| | - Xinwei Guo
- Department of Radiation Oncology, Taixing People's Hospital, Tai Xing, Jiangsu Province, China
| | - Ye Tian
- Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Su Zhou, Jiangsu Province, China
| | - Hongxun Ye
- Department of Radiation Oncology, Taixing People's Hospital, Tai Xing, Jiangsu Province, China
| | - Shaobin Zhou
- Department of Radiation Oncology, Taixing People's Hospital, Tai Xing, Jiangsu Province, China
| | - Fei Gao
- Department of Radiation Oncology, Taixing People's Hospital, Tai Xing, Jiangsu Province, China
| |
Collapse
|
49
|
Hiremath A, Shiradkar R, Fu P, Mahran A, Rastinehad AR, Tewari A, Tirumani SH, Purysko A, Ponsky L, Madabhushi A. An integrated nomogram combining deep learning, Prostate Imaging-Reporting and Data System (PI-RADS) scoring, and clinical variables for identification of clinically significant prostate cancer on biparametric MRI: a retrospective multicentre study. LANCET DIGITAL HEALTH 2021; 3:e445-e454. [PMID: 34167765 PMCID: PMC8261599 DOI: 10.1016/s2589-7500(21)00082-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/20/2021] [Accepted: 04/27/2021] [Indexed: 12/23/2022]
Abstract
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.
Collapse
Affiliation(s)
- Amogh Hiremath
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Rakesh Shiradkar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Amr Mahran
- Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Ashutosh Tewari
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sree Harsha Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrei Purysko
- Department of Radiology and Nuclear Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Lee Ponsky
- Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA.
| |
Collapse
|
50
|
Sakai K. [2. Radiomics of MRI]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2021; 77:866-875. [PMID: 34421076 DOI: 10.6009/jjrt.2021_jsrt_77.8.866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Koji Sakai
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| |
Collapse
|