Next Article in Journal
Predicting Malignancy of Breast Imaging Findings Using Quantitative Analysis of Contrast-Enhanced Mammography (CEM)
Next Article in Special Issue
Automated Error Labeling in Radiation Oncology via Statistical Natural Language Processing
Previous Article in Journal
Hydatid Disease: A Radiological Pictorial Review of a Great Neoplasms Mimicker
Previous Article in Special Issue
Deep Hybrid Learning Prediction of Patient-Specific Quality Assurance in Radiotherapy: Implementation in Clinical Routine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Use of MRI-Derived Radiomic Models in Prostate Cancer Risk Stratification: A Critical Review of Contemporary Literature

1
Department of Radiation Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 987521 Nebraska Medical Center, Omaha, NE 68198-7521, USA
2
Department of Urology, University of California, Orange, CA 92868, USA
*
Author to whom correspondence should be addressed.
Diagnostics 2023, 13(6), 1128; https://doi.org/10.3390/diagnostics13061128
Submission received: 12 December 2022 / Revised: 7 March 2023 / Accepted: 10 March 2023 / Published: 16 March 2023
(This article belongs to the Special Issue Artificial Intelligence and Radiation Oncology)

Abstract

:
The development of precise medical imaging has facilitated the establishment of radiomics, a computer-based method of quantitatively analyzing subvisual imaging characteristics. The present review summarizes the current literature on the use of diagnostic magnetic resonance imaging (MRI)-derived radiomics in prostate cancer (PCa) risk stratification. A stepwise literature search of publications from 2017 to 2022 was performed. Of 218 articles on MRI-derived prostate radiomics, 33 (15.1%) generated models for PCa risk stratification. Prediction of Gleason score (GS), adverse pathology, postsurgical recurrence, and postradiation failure were the primary endpoints in 15 (45.5%), 11 (33.3%), 4 (12.1%), and 3 (9.1%) studies. In predicting GS and adverse pathology, radiomic models differentiated well, with receiver operator characteristic area under the curve (ROC-AUC) values of 0.50–0.92 and 0.60–0.92, respectively. For studies predicting post-treatment recurrence or failure, ROC-AUC for radiomic models ranged from 0.73 to 0.99 in postsurgical and radiation cohorts. Finally, of the 33 studies, 7 (21.2%) included external validation. Overall, most investigations showed good to excellent prediction of GS and adverse pathology with MRI-derived radiomic features. Direct prediction of treatment outcomes, however, is an ongoing investigation. As these studies mature and reach potential for clinical integration, concerted effort to validate these radiomic models must be undertaken.

1. Introduction

In recent years, the advancement of precise medical imaging has facilitated the establishment of radiomics, a computer-based method of extracting and quantitatively analyzing subvisual imaging characteristics [1]. Radiomic features (i.e., qualities of intensity, texture, shape, or wavelet) can be extracted from a variety of medical images (computed tomography (CT), magnetic resonance imaging (MRI), or positron emission tomography (PET) images) using advanced mathematical algorithms, aggregated into predictive models, and applied to enhance personalized therapy [2,3]. The radiomics pipeline includes (1) image acquisition and preprocessing, (2) high-throughput feature extraction, and (3) data integration and data analysis. This process ultimately results in a predictive or prognostic model based on extracted radiomic features and can be applied to any clinical endpoint. Although the methodology behind radiomics is rapidly evolving, several contemporary studies have highlighted the enormous potential of radiomics in a variety of diseases, including, but not limited to, cancers of the gastrointestinal tract [4,5], lung [6], brain [7], and genitourinary tract [8,9].
In prostate cancer (PCa) diagnosis, preoperative MRI is standard of care and most commonly used for treatment planning and prediction of adverse pathology [10,11]. In this regard, the literature to date highlights the use of MRI-derived radiomics as an extension of this purpose. Radiomic models are most often reported to be utilized in the prediction of high-risk pathology, high Gleason score (GS), and, more recently, treatment failure following surgery or radiation [1]. As PCa is highly heterogeneous, the identification of imaging-based biomarkers predictive of clinical outcomes would enable disease-tailored treatment planning and prediction of therapy response independent of tissue biopsy and molecular analysis. If applied to identify men at high risk for recurrence, for example, these models could enhance discussion on treatment strategy, tolerable risks and benefits to the patient, and the need or lack thereof for individual therapies. In this regard, the aim of the present review is to summarize the current literature on the use of diagnostic MRI-derived radiomics in PCa risk stratification via prediction of GS, adverse pathology, and postsurgical recurrence or postradiation failure.

2. Methods

A stepwise literature search of publications from 2017 to 2022 was performed. A search of Medical Literature Analysis and Retrieval System Online (MEDLINE) databases was completed utilizing the following keywords and combination(s) thereof: [radiomics] with/without [prostate cancer] or [prostate], interchanged with [mpMRI] and/or [MRI]. This yielded 218 articles. A hand-search was performed for articles assessing the use of diagnostic MRIs (defined as scans obtained prior to any treatment) in PCa risk stratification (defined with endpoints of Gleason score (GS), postsurgical high-risk pathology (i.e., extraprostatic extension (EPE), positive surgical margin (PSM), and/or lymph node invasion (LNI)), postsurgical recurrence, and/or postradiation therapy failure). If a study included other endpoints outside of the aforementioned, the study was still included in this review but data from other endpoints were not reported. Non-English publications, review articles, editorials, and commentaries were excluded, but the reference list of each was searched to ensure inclusion of all relevant studies.
Utilizing the following stepwise methodology, studies were reviewed by the study team for inclusion and exclusion criteria defined a priori. First, the titles and abstracts were screened such that nonrelevant studies were excluded. Second, full manuscripts were reviewed for their study populations and/or outcome measures. Two authors (LH and MB) independently agreed on the selection of eligible studies and achieved consensus of included studies. Data on the number of subjects, outcome measures, image series used, feature selection, region of interest, and model validation were systematically extracted from each article and summarized in Table 1, Table 2, Table 3 and Table 4. To ensure standardization, the International Society of Urological Pathology (ISUP) guidelines on GS [12], National Comprehensive Center Network (NCCN) and American Urological Association (AUA) guidelines for risk group stratification [11,13], and Prostate Imaging Reporting and Data System version 2 (PI-RADS v2) were utilized [14].

3. Results

3.1. Study Selection

Two-hundred and eighteen publications were initially identified and screened through a literature search of the MEDLINE journals via a PubMed interface. Of these, 52 review articles, 1 letter to the editor, and 1 clinical trial protocol were excluded, leaving 164 records for title and abstract review. After title and abstract review, 112 records were excluded for clinical endpoints outside of risk stratification for Gleason grade, adverse pathology, and post-treatment recurrence or failure, leaving 52 for full-paper review. Nineteen additional records were excluded during full-paper review, as they did not utilize diagnostic MRIs at the time of PCa diagnosis (n = 4), predicted biopsy Gleason grade (n = 7), predicted presence of overt radiographic features for increased staging (n = 5), or utilized registries without clinical endpoints (n = 3). After all inclusion and exclusion criteria were satisfied, 33 articles remained and were reviewed.

3.2. Description of Studies

Of the 33 included articles, all studies were published between 2017 and 2022, with the majority (n = 26, 78.7%) published in 2020 or later. Overall, in image acquisition and preprocessing, T2 weighted images (T2WI) were universally used, with diffusion-weighted images (DWI) and apparent diffusion coefficient (ADC) sequences in the second majority. Prediction of GS and adverse pathology (i.e., PSM, EPE, and/or LNI) postradical prostatectomy (RP) were primary endpoints in 15 (45.5%) and 11 (33.3%) studies, respectively. Four (12.1%) and three (9.1%) investigations highlighted the use of MRI-derived radiomics in predicting postsurgical recurrence and postradiation failure, respectively. Finally, of the 33 studies included, 7 (21.2%) reported external validation of their radiomic models.

3.2.1. Prediction of Gleason Grade

While many studies have been conducted to utilize MRI-derived radiomic models to discriminate between clinically significant (GS ≥ 3 + 3) [11,13] and insignificant (GS < 3 + 3) PCa lesions, these studies are out of the scope of the current review. Rather, we focused on stratification of clinically significant PCa by GS risk groups [15,16,17]. Table 1 summarizes the 15 studies utilizing MRI-derived radiomic models to predict pathologic GS [15,16,17,18,19,20,21,22,23,24,25,26,27,28,29], with one study predicting GS upgrading between biopsy and surgery [23].
To this effect, several of these studies compared the utility of radiomic features extracted from different imaging sequences (T2WI, DWI, ADC, etc.) [21,22,25,29]. Gong et al. found DWI to outperform T2WI image sequences in GS risk stratification, while two other groups combined radiomic features from T2WI and ADC image sequences into their final model [19,21,25].
In delineating regions of interest, eight studies [15,17,18,19,22,24,25,27] utilized only PI-RADS lesions, while the remaining seven utilized the full prostate [16,20,21,23,26,28,29]. Feature extraction and selection was most commonly done via minimum redundancy maximum relevance (mRMR) [21,28,29] or random forest (RF) [15,16,24], and the receiver operator characteristic–area under the curve (ROC-AUC) of the final models ranged from 0.50 to 0.92. Castillo and colleagues [27] were the only group to perform a multi-institutional external validation of their model for the prediction of GS ≤ 6 versus GS ≥ 7, yielding an ROC-AUC of 0.75.

3.2.2. Prediction of Adverse Pathology

Table 2 summarizes the 11 studies utilizing MRI-derived radiomic models to predict EPE, PSM, or LNI [25,30,31,32,33,34,35,36,37,38,39]. Seven [25,31,32,33,38,39], six [25,34,35,36,37,39], and one [38] study(ies) reported on EPE, LNI, and PSM, respectively. Uniquely, Damascelli et al. utilized both EPE and LNI [25] as their primary endpoint, while He and colleagues utilized both PSM and EPE [38]. Furthermore, Li et al. constructed a radiomic model to predict “adverse pathology”, inclusive of any EPE, SVI, and/or LNI [39]. All studies predicting adverse pathology utilized final pathology following RP as the gold standard comparator.
Of the studies utilizing radiomics to predict EPE, the prostate capsule [30], PI-RADS lesions [31,38], the index lesion [25,33], and prostate [39] were delineated as regions of interest. The ROC-AUC on training sets ranged from 0.674 to 0.92, while the ROC-AUC on testing sets ranged from 0.598 to 0.92. For studies predicting LNI, the prostate [34,37,39], PI-RADS lesions [35,37], and index lesions [25,36] were used as the regions of interest. The ROC-AUC on training sets ranged from 0.82 to 1.0, and the ROC-AUC for testing sets ranged from 0.73 to 0.94. Of note, out of the 11 studies predicting adverse pathology, external validation was pursued in three (27.3%) [32,33,34], of which yielded highly heterogeneous results with ROC-AUC values between 0.598 and 0.80.
In predicting adverse pathology, two studies integrated clinical characteristics with radiomic features to construct a combined model [35,38]. First, a study by Bourbonne and colleagues generated a combined clinical and radiomic model to predict LNI [35]. This model included the six most important radiomic features combined with clinical parameters of tumor size, t-stage, Gleason score, pre- and postoperative prostate-specific antigen (PSA), margin status, age at surgery, and the University of California San Francisco Cancer of the Prostate Risk Assessment Score (UCSF-CAPRA) score. Training of this integrated model yielded an ROC-AUC of 1.0, and model testing in their internal cohort resulted in an ROC-AUC of 0.87. Similarly, a study by He et al. integrated clinical parameters with radiomic models predicting ECE and PSM. Inclusion of these variables increased the ROC-AUC for ECE prediction from 0.625 to 0.728 and increased the ROC-AUC for PSM prediction from 0.733 to 0.766 [38]. Neither of these studies provided correlational analysis between the selected radiomic features and clinical characteristics.

3.2.3. Prediction of Postsurgical Biochemical Recurrence

To date, there have been only a handful of investigations utilizing radiomic features to predict biochemical recurrence (BCR) following RP. Table 3 illustrates the four studies [39,40,41,42] utilizing preoperative MRI-derived radiomics to predict post-RP BCR, defined as two consecutive serum PSA levels greater than or equal to 0.2 ng/mL.
Bourbonne et al. was the first to train and validate an MRI-derived radiomic model against post-RP BCR and BCR-free survival [40]. After feature extraction from T2WI and ADC maps of 107 patients, independent factors correlating with BCR were identified via Cox regression analysis. The final radiomic model had a high negative predictive value of 96% and could be reliably used to identify patients at a very low risk of BCR.
Similar results for BCR prediction at various timepoints were obtained by three other studies, which all included external cohorts in the construction of their radiomic models [39,41,42]. Furthermore, two of these three studies included head-to-head comparisons between their radiomic model and commonly used clinical nomograms for prediction of BCR. Yan et al. reported ROC-AUCs ranging from 0.84 to 0.88 in predicting three-year BCR [41], while Shiradkar et al. reported their radiomic model [42] to be significantly and independently correlated with three-year BCR in cox proportional hazards regression modeling (HR: 2.91, 95% CI: 1.45–11.51, p = 0.02). In addition, both studies compared their radiomic models to the UCSF-CAPRA and CAPRA-S scores. Compared to these clinical nomograms, the radiomic model proposed by Yan et al. [41] maintained significantly improved concordance with three-year BCR (p < 0.05). While ROC-AUCs for these clinical models ranged from 0.535 to 0.689 across the internal and external datasets, their radiomic signature yielded ROC-AUCs ranging from 0.685 to 0.877. A statistical comparison on this improvement was not reported. These results were similarly echoed by Li and colleagues in 2021 [39], who reported on a combined radiomic-clinicopathologic model (RadClip) integrating radiomic features and clinical characteristics utilized in the UCSF-CAPRA score. In multivariate analysis, RadClip was independently associated with BCR (HR: 7.01, 95% CI: +1.21–40.68, p < 0.05). Diagnostic performance via ROC-AUC, sensitivity, and specificity were not reported.

3.2.4. Prediction of Postradiation Biochemical Failure

Finally, Table 4 summarizes three studies [43,44,45] utilizing pretreatment MRI-derived radiomic models to predict postradiation therapy biochemical failure (BF), defined as a PSA nadir > 2.0 ng/mL. Each of the three studies defined the region of interest differently: one study included the lesion, prostate, peripheral zone, and transitional zone [43], another utilized the prostate and 2 mm of margin [44], and the last contoured only the prostate [45]. In similar regard, each of the three studies predicting postradiation BF approached feature selection and extraction differently. Gnep and colleagues selected five radiomic features most significantly correlated with BF: tumor volume, T2W difference variance mean, T2W contrast mean, maximal tumor area, and ADC median [43]. Combined, these features yielded a radiomic model with a C-index of 0.90 ± 0.09. In contrast, Fernandes et al. developed a radiomic model with T2WI images, predicting BF with an ROC-AUC of 0.63 [44]. Finally, Zhong and colleagues generated a radiomic model with an ROC-AUC of 0.99 in training and an ROC-AUC of 0.73 in validation [45]. Of note, none of these studies included external validation, integration of clinical characteristics with the radiomic models, or head-to-head comparisons with clinical nomograms.

4. Discussion

With the rise of big data, the use of radiomics in personalized medicine is anticipated. More specifically, PCa radiomics is a continuously evolving research field with high potential to offer noninvasive, personalized biomarkers for risk stratification. Expectedly, the number of research articles on MRI-derived prostate radiomics has exponentially increased since 2017, accounting for 218 original articles in the last five years. In our qualitative review of these articles, however, it is clear that only a minority concentrate on the use of MRI-derived radiomics in PCa risk stratification. Rather, most of these investigations have concentrated on its use as a screening or diagnostic tool, for instance, in the correlation of radiomic models with PI-RADS lesions [46], in confirming prostate biopsy findings [47], or as a PCa screening tool [48]. While these are valuable explorations, there is little room for clinical integration of radiomic models in these spaces, as robust gold standards for risk stratification are already present via pathologic examination or imaging. Furthermore, the exploration of radiomic-based models has thus far been independently sequestered within the fields of radiation oncology, radiology, and biomedical imaging. As the use of radiomics in PCa further evolves, multidisciplinary collaborations are necessary to place an indication to the technology.
A clear focus of PCa radiomics is GS discrimination, reflecting the need for improvements in risk stratification during an initial PCa diagnosis. In this regard, the Prostate Imaging Reporting and Data System (PI-RADS) was validated to identify clinically significant versus insignificant prostate cancer, with prediction rates of up to 82% [49]. However, while PI-RADS works well for the definition of benign versus malignant tissue, it does not differentiate between low-risk and high-risk PCa. Rather, this risk stratification is defaulted to the use of GS on direct prostate biopsy or surgical pathology.
While GS is the most established histologic biomarker, due to sampling methods and the high intratumoral heterogeneity associated with PCa, GS can be discordant between biopsy and final surgical pathology in 20% to 60% of patients [45,46]. Even further, the risk of upgrading between biopsy and surgery ranges from 5% to 65% in some patient populations [50]. To address this gap, radiomics-based GS prediction may serve as a surrogate measure of tumor heterogeneity and provide an opportunity for further risk stratification between biopsy and initial treatment via surgery or radiation. In this regard, several basic science studies have explored correlations between radiomic features and genetic characteristics of PCa. While these radiogenomic models have not been externally validated, McCann et al. [51] and Switlyk et al. [52] have demonstrated associations between radiomic features and the genetic marker phosphatase and tensin homolog. Similarly, a pilot study by Sun and colleagues correlated radiomic features with hypoxic gene expression—a gene signature identified as an independent risk factor for metastasis-free survival in patients with PCa [53]. With further confirmation correlating radiomic features and genetic drivers of PCa, tumor heterogeneity can be further addressed to facilitate treatment planning.
Parallel to explorations of GS discrimination are those predicting adverse pathology such as PSM, EPE, and LNI. While preoperative imaging may allow for identification of EPE and LNI, diagnostic performance varies widely, with studies reporting sensitivity and specificity as low as 47% to 56% [54,55,56]. Given that EPE and LNI are associated with significantly decreased likelihood of recurrence-free and progression-free survival [13], early stratification may facilitate conversations regarding multimodal therapy or prompt changes in patient management.
Finally, as is apparent in the distribution of articles included in this review, direct prediction of treatment outcomes via radiomic models is an investigation still in its infancy. Given the long natural life history unique to PCa, a lengthy follow-up time is required for studies predicting post-RP BCR or postradiation BF. Furthermore, while seven studies have developed models to predict BCR and BF, external validation was only pursued in three, with none utilized in postradiation populations. Prior to clinical integration, further validation efforts are required. A brief review of currently ongoing clinical trials reveals several trials utilizing MRI-derived radiomic signatures for the prediction of discontinuation of active surveillance [56,57], local control rates [58], and disease extension [59]. Furthermore, as explorations of combined imaging techniques utilizing prostate-specific membrane antigen (PSMA) PET/CT continue to evolve, risk stratification may extend further beyond that of the prostate MRI.
Overall, while most of the included studies in this review presented good-to-excellent ROC-AUC values in predicting GS, adverse pathology, and cancer control, these findings must be considered within the context of publication bias and variability of methodology. First, the development of a radiomic signature consists of several key steps surrounding image acquisition and preprocessing, feature extraction, data integration, and data analysis—each step of which can be modified in statistical methodology, segmentation of regions of interest, cross-validation, testing and validation, and reporting. Given that radiomic features and their corresponding models are highly sensitive to any modifications to these steps, investigations on radiomic feature variability, robustness of available datasets, and reproducibility in multiple cohorts are required prior to consideration of external validation [60,61]. Along similar lines, it is also clear that current studies on PCa radiomics lack comparison to currently clinically available prediction tools or clinical characteristics such as PSA at diagnosis, age, or PI-RADS score. While a few studies have compared their full radiomic models to the CAPRA and CAPRA-S score [39,42], for example, this is a feature in a small minority (<5%) of studies [62]. Even for those investigations integrating clinical characteristics with their radiomic models, there is no analysis to assess possible correlations between each radiomic feature versus already-available clinical information. Until there are adequate comparators between radiomics and information easily obtained during the PCa clinical care pathway, clinical applicability of this technology will be severely hampered.

5. Conclusions

Overall, MRI-derived PCa radiomics presents as an emerging research field with the potential to offer noninvasive, imaging-based biomarkers useful for risk stratification and prediction of treatment response. Furthermore, radiomics has the potential to facilitate quantitative characterization of tumor heterogeneity, thus enabling disease-tailored treatment planning. While radiomic models show promise in predicting high-risk GS and adverse pathology, direct application to prediction of treatment outcomes remains an ongoing investigation. As these studies mature and reach potential for clinical integration, concerted efforts to establish adequate comparators, standardize methodology, and systematically validate these models must be prioritized.

Author Contributions

Conceptualization: L.M.H. Methodology: L.M.H. Software: not applicable. Validation: not applicable. Formal analysis: L.M.H. Investigation: L.M.H. Resources: M.J.B. Data curation: L.M.H. and O.T. Writing—original draft preparation: L.M.H. Writing—review and editing: L.M.H., Y.H., O.T. and M.J.B. Visualization: not applicable. Supervision: M.J.B. Project administration: not applicable. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was a review of literature and did not require institutional review board approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Spohn, S.K.B.; Bettermann, A.S.; Bamberg, F.; Benndorf, M.; Mix, M.; Nicolay, N.H.; Fechter, T.; Hölscher, T.; Grosu, R.; Chiti, A.; et al. Radiomics in prostate cancer imaging for a personalized treatment approach—Current aspects of methodology and a systematic review on validated studies. Theranostics 2021, 16, 8027–8042. [Google Scholar] [CrossRef]
  2. Liberini, V.; Laudicella, R.; Balma, M.; Nicolotti, D.G.; Buschiazzo, A.; Grimaldi, S.; Lorenzon, L.; Bianchi, A.; Peano, S.; Bartolotta, T.V.; et al. Radiomics and artificial intelligence in prostate cancer: New tools for molecular hybrid imaging and theragnostics. Eur. Radiol. Exp. 2022, 6, 27. [Google Scholar] [CrossRef] [PubMed]
  3. Cuocolo, R.; Cipullo, M.B.; Stanzione, A.; Ugga, L.; Romeo, V.; Radice, L.; Brunetti, A.; Imbriaco, M. Machine learning applications in prostate cancer magnetic resonance imaging. Eur. Radiol. Exp. 2019, 3, 35–46. [Google Scholar] [PubMed]
  4. Mao, Q.; Zhou, M.T.; Zhao, Z.P.; Liu, N.; Yang, L.; Zhang, X.M. Role of radiomics in the diagnosis and treatment of gastrointestinal cancer. World J. Gastroenterol. 2022, 28, 6002–6016. [Google Scholar] [CrossRef] [PubMed]
  5. Yuan, H.; Xu, X.; Tu, S.; Chen, B.; Wei, Y.; Ma, Y. The CT-based intratumoral and peritumoral machine learning radiomics analysis in predicting lymph node metastasis in rectal carcinoma. BMC Gastroenterol. 2022, 22, 463. [Google Scholar] [CrossRef]
  6. Du, Q.; Baine, M.; Bavitz, K.; McAllister, J.; Liang, X.; Yu, H.; Ryckman, J.; Yu, L.; Jiang, H.; Zhou, S.; et al. Radiomic feature stability across 4D respiratory phases and its impact on lung tumor prognosis prediction. PLoS ONE 2019, 14, e0216480. [Google Scholar] [CrossRef] [Green Version]
  7. Sotoudeh, H.; Saadatpour, Z.; Rezaei, A.; Sotoudeh, M.; Wheeler, C.A.; Singhal, A.; Tanwar, M. Radiomics for differentiation of the posterior fossa pilocytic astrocytoma versus hemangioblastomas in adults. A pilot study. Clin. Imaging 2022, 93, 26–30. [Google Scholar] [CrossRef]
  8. Shi, Y.; Wahle, E.; Du, Q.; Krajewski, L.; Liang, X.; Zhou, S.; Zhang, C.; Baine, M.; Zheng, D. Associations between Statin/Omega3 Usage and MRI-Based Radiomics Signatures in Prostate Cancer. Diagnostics 2021, 11, 85. [Google Scholar] [CrossRef]
  9. Yu, Z.; Ding, J.; Pang, H.; Fang, H.; He, F.; Xu, C.; Li, X.; Ren, K. A triple-classification for differentiating renal oncocytoma from renal cell carcinoma subtypes and CK7 expression evaluation: A radiomics analysis. BMC Urol. 2022, 22, 147. [Google Scholar] [CrossRef] [PubMed]
  10. Somford, D.M.; Hamoen, E.H.; Fütterer, J.J.; van Basten, J.P.; Hulsbergen-van de Kaa, C.A.; Vreuls, W.; van Oort, I.M.; Vergunst, H.; Kiemeney, L.A.; Barentsz, J.O.; et al. The predictive value of endorectal 3 Tesla multiparametric magnetic resonance imaging for extraprostatic extension in patients with low, intermediate and high risk prostate cancer. J. Urol. 2013, 5, 1728–1734. [Google Scholar] [CrossRef]
  11. Eastham, J.A.; Auffenberg, G.B.; Barocas, D.A.; Chou, R.; Crispino, T.; Davis, J.W.; Eggener, S.; Horwitz, E.M.; Kane, C.J.; Kirkby, E.; et al. Clinically localized prostate cancer: AUA/ASTRO guideline part I: Introduction, risk assessment, staging and risk-based management. J. Urol. 2022, 208, 10–18. [Google Scholar] [CrossRef] [PubMed]
  12. Van Leenders, G.J.L.H.; van der Kwast, T.H.; Grignon, D.J.; Evans, A.J.; Kristiansen, G.; Kweldam, C.F.; Litjens, G.; McKenney, J.K.; Melamed, J.; Mottet, M.; et al. The 2019 International Society of Urological Pathology (ISUP) Consensus Conference on Grading of Prostatic Carcinoma. Am. J. Surg. Pathol. 2020, 44, e87–e99. [Google Scholar] [CrossRef] [PubMed]
  13. Kawachi, M.H.; Bahnson, R.R.; Barry, M.; Busby, J.E.; Carroll, P.R.; Carter, H.B.; Catalona, W.J.; Cookson, M.S.; Epstein, J.I.; Etzioni, R.B.; et al. NCCN clinical practice guidelines in oncology: Prostate cancer early detection. J. Natl. Compr. Cancer Netw. 2010, 8, 240–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Weinreb, J.C.; Barentz, J.O.; Choyke, P.L.; Cornud, F.; Haider, M.A.; Macura, K.J.; Margolis, D.; Schnall, M.D.; Shtern, F.; Tempany, C.M.; et al. PI-RADS Prostate Imaging-Reporting and Data System, v2.1. American College of Radiology; 2019. Available online: https://www.acr.org/-/media/ACR/Files/RADS/Pi-RADS/PIRADS-V2-1.pdf (accessed on 15 March 2021).
  15. Chaddad, A.; Niazi, T.; Probst, S.; Bladou, F.; Anidjar, M.; Bahoric, B. Predicting Gleason Score of Prostate Cancer Patients Using Radiomic Analysis. Front. Oncol. 2018, 8, 630. [Google Scholar] [CrossRef] [Green Version]
  16. Penzias, G.; Singanamalli, A.; Elliott, R.; Gollamudi, J.; Shih, N.; Feldman, M.; Stricker, P.D.; Delprado, W.; Tiwari, S.; Böhm, M.; et al. Identifying the morphologic basis for radiomic features in distinguishing different Gleason grades of prostate cancer on MRI: Preliminary findings. PLoS ONE 2018, 13, e0200730. [Google Scholar] [CrossRef] [Green Version]
  17. Toivonen, J.; Perez, I.M.; Movahedi, P.; Merisaari, H.; Pesola, M.; Taimen, P.; Boström, P.J.; Pohjankukka, J.; Kiviniemi, A.; Pahikkala, T.; et al. Radiomics and machine learning of multisequence multiparametric prostate MRI: Towards improved non-invasive prostate cancer characterization. PLoS ONE 2019, 14, e0217702. [Google Scholar] [CrossRef] [Green Version]
  18. Hectors, S.J.; Cherny, M.; Yadav, K.K.; Beksaç, A.T.; Thulasidass, H.; Lewis, S.; Davicioni, E.; Wang, P.; Tewari, A.K.; Taouli, B. Radiomics Features Measured with Multiparametric Magnetic Resonance Imaging Predict Prostate Cancer Aggressiveness. J. Urol. 2019, 202, 498–505. [Google Scholar] [CrossRef]
  19. Abdollahi, H.; Mofid, B.; Shiri, I.; Razzaghdoust, A.; Saadipoor, A.; Mahdavi, A.; Galandooz, H.M.; Mahdavi, S.R. Machine learning-based radiomic models to predict intensity-modulated radiation therapy response, Gleason score and stage in prostate cancer. Radiol. Med. 2019, 124, 555–567. [Google Scholar] [CrossRef]
  20. McGarry, S.D.; Bukowy, J.D.; Iczkowski, K.A.; Unteriner, J.G.; Duvnjak, P.; Lowman, A.K.; Jacobsohn, K.; Hohenwalter, M.; Grffin, M.O.; Barrington, A.W.; et al. Gleason Probability Maps: A Radiomics Tool for Mapping Prostate Cancer Likelihood in MRI Space. Tomography 2019, 5, 127–134. [Google Scholar] [CrossRef]
  21. Gong, L.; Xu, M.; Fang, M.; Zou, J.; Yang, S.; Yu, X.; Xu, D.; Zhou, L.; Li, H.; He, B.; et al. Noninvasive Prediction of High-Grade Prostate Cancer via Biparametric MRI Radiomics. J. Magn. Reson. Imaging 2020, 52, 1102–1109. [Google Scholar] [CrossRef]
  22. Zhang, G.; Han, Y.; Wei, J.; Qi, Y.F.; Gu, D.S.; Lei, J.; Yan, W.G.; Xiao, Y.; Xue, H.D.; Feng, F.; et al. Radiomics Based on MRI as a Biomarker to Guide Therapy by Predicting Upgrading of Prostate Cancer From Biopsy to Radical Prostatectomy. J. Magn. Reson. Imaging 2020, 52, 1239–1248. [Google Scholar] [CrossRef]
  23. Santone, A.; Brunese, M.C.; Donnarumma, F.; Guerriero, P.; Mercaldo, F.; Reginelli, A.; Miele, V.; Giovagnoni, A.; Brunese, L. Radiomic features for prostate cancer grade detection through formal verification. Radiol. Med. 2021, 126, 688–697. [Google Scholar] [CrossRef]
  24. Makowski, M.R.; Bressem, K.K.; Franz, L.; Kader, A.; Niehues, S.M.; Keller, S.; Rueckert, D.; Adams, L.C. De Novo Radiomics Approach Using Image Augmentation and Features From T1 Mapping to Predict Gleason Scores in Prostate Cancer. Investig. Radiol. 2021, 56, 661–668. [Google Scholar] [CrossRef] [PubMed]
  25. Damascelli, A.; Gallivanone, F.; Cristel, G.; Cava, C.; Interlenghi, M.; Esposito, A.; Brembilla, G.; Briganti, A.; Montorsi, F.; Castiglioni, I.; et al. Advanced Imaging Analysis in Prostate MRI: Building a Radiomic Signature to Predict Tumor Aggressiveness. Diagnostics 2021, 11, 594. [Google Scholar] [CrossRef] [PubMed]
  26. Gugliandolo, S.G.; Pepa, M.; Isaksson, L.J.; Marvaso, G.; Raimondi, S.; Botta, F.; Gandini, S.; Ciardo, D.; Volpe, S.; Riva, G.; et al. MRI-based radiomics signature for localized prostate cancer: A new clinical tool for cancer aggressiveness prediction? Sub-study of prospective phase II trial on ultra-hypofractionated radiotherapy (AIRC IG-13218). Eur. Radiol. 2021, 31, 716–728. [Google Scholar] [CrossRef] [PubMed]
  27. Castillo, T.J.M.; Starmans, M.P.A.; Arif, M.; Niessen, W.J.; Klein, S.; Bangma, C.H.; Schoots, I.G.; Veeland, J.F. A Multi-Center, Multi-Vendor Study to Evaluate the Generalizability of a Radiomics Model for Classifying Prostate cancer: High Grade vs. Low Grade. Diagnostics 2021, 11, 369. [Google Scholar] [CrossRef] [PubMed]
  28. Rodrigues, A.; Santinha, J.; Galvão, B.; Matos, C.; Couto, F.M.; Papanikolaou, N. Prediction of Prostate Cancer Disease Aggressiveness Using Bi-Parametric Mri Radiomics. Cancers 2021, 13, 6065. [Google Scholar] [CrossRef]
  29. Gong, L.; Xu, M.; Fang, M.; He, B.; Li, H.; Fang, X.; Dong, D.; Tian, J. The potential of prostate gland radiomic features in identifying the Gleason score. Comput. Biol. Med. 2022, 144, 105318. [Google Scholar] [CrossRef]
  30. Ma, S.; Xie, H.; Wang, H.; Yang, J.; Han, C.; Wang, X.; Zhang, X. Preoperative Prediction of Extracapsular Extension: Radiomics Signature Based on Magnetic Resonance Imaging to Stage Prostate Cancer. Mol. Imaging Biol. 2020, 22, 711–721. [Google Scholar] [CrossRef]
  31. Xu, L.; Zhang, G.; Zhao, L.; Mao, L.; Li, X.; Yan, W.; Xiao, Y.; Lei, J.; Sun, H.; Jin, Z. Radiomics Based on Multiparametric Magnetic Resonance Imaging to Predict Extraprostatic Extension of Prostate Cancer. Front. Oncol. 2020, 10, 940. [Google Scholar] [CrossRef]
  32. Bai, H.; Xia, W.; Ji, X.; He, D.; Zhao, X.; Bao, J.; Zhou, J.; Wei, X.; Huang, Y.; Li, Q.; et al. Multiparametric Magnetic Resonance Imaging-Based Peritumoral Radiomics for Preoperative Prediction of the Presence of Extracapsular Extension With Prostate Cancer. J. Magn. Reson. Imaging 2021, 54, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  33. Cuocolo, R.; Stanzione, A.; Faletti, R.; Gatti, M.; Calleris, G.; Fornari, A.; Gentile, F.; Motta, A.; Dell’Aversana, S.; Creta, M.; et al. MRI index lesion radiomics and machine learning for detection of extraprostatic extension of disease: A multicenter study. Eur. Radiol. 2021, 31, 7575–7583. [Google Scholar] [CrossRef] [PubMed]
  34. Hou, Y.; Bao, J.; Song, Y.; Bao, M.L.; Jiang, K.W.; Zhang, J.; Yang, G.; Hu, C.H.; Shi, H.B.; Wang, X.M.; et al. Integration of clinicopathologic identification and deep transferrable image feature representation improves predictions of lymph node metastasis in prostate cancer. EBioMedicine 2021, 68, 103395. [Google Scholar] [CrossRef] [PubMed]
  35. Bourbonne, V.; Jaouen, V.; Nguyen, T.A.; Tissot, V.; Doucet, L.; Hatt, M.; Visvikis, D.; Pradier, O.; Valéri, A.; Fournier, G.; et al. Development of a Radiomic-Based Model Predicting Lymph Node Involvement in Prostate Cancer Patients. Cancers 2021, 13, 5672. [Google Scholar] [CrossRef] [PubMed]
  36. Zheng, H.; Miao, Q.; Liu, Y.; Mirak, S.A.; Hosseiny, M.; Scalzo, F.; Raman, S.S.; Sung, K. Multiparametric MRI-based radiomics model to predict pelvic lymph node invasion for patients with prostate cancer. Eur. Radiol. 2022, 32, 5688–5699. [Google Scholar] [CrossRef]
  37. Liu, X.; Wang, X.; Zhang, Y.; Sun, Z.; Zhang, X.; Wang, X. Preoperative prediction of pelvic lymph nodes metastasis in prostate cancer using an ADC-based radiomics model: Comparison with clinical nomograms and PI-RADS assessment. Abdom. Radiol. 2022, 47, 3327–3337. [Google Scholar] [CrossRef]
  38. He, D.; Wang, X.; Fu, C.; Wei, X.; Bao, J.; Ji, X.; Bai, H.; Xia, W.; Gao, X.; Huang, Y.; et al. MRI-based radiomics models to assess prostate cancer, extracapsular extension and positive surgical margins. Cancer Imaging 2021, 21, 46. [Google Scholar] [CrossRef]
  39. Li, L.; Shiradkar, R.; Leo, P.; Algohary, A.; Fu, P.; Tirumani, S.H.; Mahran, A.; Buzzy, C.; Obmann, V.C.; Mansoori, B.; et al. A novel imaging based Nomogram for predicting post-surgical biochemical recurrence and adverse pathology of prostate cancer from pre-operative bi-parametric MRI. EBioMedicine 2021, 63, 103163. [Google Scholar] [CrossRef]
  40. Bourbonne, V.; Fournier, G.; Vallières, M.; Lucia, F.; Doucet, L.; Tissot, V.; Cuvelier, G.; Hue, S.; Le Penn Du, H.; Perdriel, L.; et al. External Validation of an MRI-Derived Radiomics Model to Predict Biochemical Recurrence after Surgery for High-Risk Prostate Cancer. Cancers 2020, 12, 814. [Google Scholar] [CrossRef] [Green Version]
  41. Yan, Y.; Shao, L.; Liu, Z.; He, W.; Yang, G.; Liu, J.; Xia, H.; Zhang, Y.; Chen, H.; Liu, C.; et al. Deep Learning with Quantitative Features of Magnetic Resonance Images to Predict Biochemical Recurrence of Radical Prostatectomy: A Multi-Center Study. Cancers 2021, 13, 3098. [Google Scholar]
  42. Shiradkar, R.; Ghose, S.; Mahran, A.; Li, L.; Hubbard, I.; Fu, P.; Tirumani, S.H.; Ponsky, L.; Purysko, A.; Madabhushi, A. Prostate Surface Distension and Tumor Texture Descriptors From Pre-Treatment MRI Are Associated With Biochemical Recurrence Following Radical Prostatectomy: Preliminary Findings. Front. Oncol. 2022, 12, 2055. [Google Scholar] [CrossRef] [PubMed]
  43. Gnep, K.; Fargeas, A.; Gutiérrez-Carvajal, R.E.; Commandeur, F.; Mathieu, R.; Ospina, J.D.; Rolland, Y.; Rohou, T.; Vincendeau, S.; Hatt, M.; et al. Haralick textural features on T2-weighted MRI are associated with biochemical recurrence following radiotherapy for peripheral zone prostate cancer. J. Magn. Reson. Imaging 2016, 45, 103–117. [Google Scholar] [CrossRef] [PubMed]
  44. Fernandes, C.D.; Dinh, C.V.; Walraven, I.; Heijmink, S.W.; Smolic, M.; van Griethuysen, J.J.M.; Simões, R.; Losnegård, A.; van der Poel, H.G.; Pos, F.J.; et al. Biochemical recurrence prediction after radiotherapy for prostate cancer with T2w magnetic resonance imaging radiomic features. Phys. Imaging Radiat. Oncol. 2018, 7, 9–15. [Google Scholar] [CrossRef] [Green Version]
  45. Zhong, Q.Z.; Long, L.H.; Liu, A.; Li, C.; Xiu, X.; Hou, X.; Wu, Q.; Gao, H.; Xu, Y.; Zhao, T.; et al. Radiomics of Multiparametric MRI to Predict Biochemical Recurrence of Localized Prostate Cancer After Radiation Therapy. Front. Oncol. 2020, 10, 731. [Google Scholar] [CrossRef]
  46. Lim, C.S.; Abreu-Gomez, J.; Thornhill, R.; James, N.; Al Kindi, A.; Lim, A.S.; Schieda, N. Utility of machine learning of apparent diffusion coefficient (ADC) and T2-weighted (T2W) radiomic features in PI-RADS version 2.1 category 3 lesions to predict prostate cancer diagnosis. Abdom. Radiol. 2021, 46, 5647–5658. [Google Scholar] [CrossRef]
  47. Michaely, H.J.; Aringhieri, G.; Cioni, D.; Neri, E. Current Value of Biparametric Prostate MRI with Machine-Learning or Deep-Learning in the Detection, Grading, and Characterization of Prostate Cancer: A Systematic Review. Diagnostics 2022, 12, 799. [Google Scholar] [CrossRef] [PubMed]
  48. Telecan, T.; Andras, I.; Crisan, N.; Giurgiu, L.; Căta, E.D.; Caraiani, C.; Lebovici, A.; Boca, B.; Balint, Z.; Diosan, L.; et al. More than Meets the Eye: Using Textural Analysis and Artificial Intelligence as Decision Support Tools in Prostate Cancer Diagnosis—A Systematic Review. J. Pers. Med. 2022, 12, 983. [Google Scholar] [CrossRef]
  49. Divrik, R.T.; Eroglu, A.; Sahin, A.; Zorlu, F.; Ozen, H. Increasing the number of biopsies increases the concordance of Gleason scores of needle biopsies and prostatectomy specimens. Urol. Oncol. 2007, 25, 376–382. [Google Scholar] [CrossRef]
  50. Chen, S.; Yang, Y.; Peng, T.; Yu, X.; Deng, H.; Guo, Z. The prediction value of PI-RADS v2 score in high-grade Prostate Cancer: A multicenter retrospective study. Int. J. Med. Sci. 2020, 17, 1366–1374. [Google Scholar] [CrossRef]
  51. San Francisco, I.F.; DeWolf, W.C.; Rosen, S.; Upton, M.; Olumi, A.F. Extended prostate needle biopsy improves concordance of Gleason grading between prostate needle biopsy and radical prostatectomy. J. Urol. 2003, 169, 136–140. [Google Scholar] [CrossRef]
  52. McCann, S.M.; Jiang, Y.; Fan, X.; Wang, J.; Antic, T.; Prior, F.; VanderWeele, D.; Oto, A. Quantitative multiparametric MRI features and PTEN expression of peripheral zone prostate cancer: A pilot study. AJR Am. J. Roentgenol. 2016, 206, 559–565. [Google Scholar] [CrossRef] [PubMed]
  53. Switlyk, M.D.; Salberg, U.B.; Geier, O.M.; Vlatkovic, L.; Lilleby, W.; Lyng, H.; Seierstad, T. PTEN Expression in Prostate Cancer: Relationship With Clinicopathologic Features and Multiparametric MRI Findings. AJR Am. J. Roentgenol. 2019, 212, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  54. Sun, Y.; Williams, S.; Byrne, D.; Keam, S.; Reynolds, H.M.; Mitchell, C.; Wraith, D.; Murphy, D.; Haworth, A. Association analysis between quantitative MRI features and hypoxia-related genetic profiles in prostate cancer: A pilot study. Br. J. Radiol. 2019, 92, 20190373. [Google Scholar] [CrossRef]
  55. Arslan, A.; Karaarslan, E.; Güner, A.L.; Sağlıcan, Y.; Tuna, M.B.; Kural, A.R. Comparing the Diagnostic Performance of Multiparametric Prostate MRI Versus 68Ga-PSMA PET-CT in the Evaluation Lymph Node Involvement and Extraprostatic Extension. Acad. Radiol. 2022, 29, 698–704. [Google Scholar] [CrossRef] [PubMed]
  56. Majchrzak, N.; Cieśliński, P.; Milecki, T.; Głyda, M.; Karmelita-Katulska, K. MRI utility in predicting extraprostatic extension of prostate cancer and biochemical recurrence after radical prostatectomy. Nowotw. J. Oncol. 2021, 71, 197–201. [Google Scholar] [CrossRef]
  57. Heinrich-Heine Univeristy Duesseldorf. Prospective Evaluation of Mp-MRI, MR-Guided Biopsy, and Molecular Markers for Active Surveillance of Prostate Cancer (PROMM-AS). NCT03979573. Updated 6 June 2019. Available online: https://beta.clinicaltrials.gov/study/NCT03979573 (accessed on 27 September 2022).
  58. Punnen, S.; MRI Based Active Selection for Treatment Trial (MAST). NCT02242773. Updated 14 September 2022. Available online: https://beta.clinicaltrials.gov/study/NCT02242773 (accessed on 27 September 2022).
  59. Henry Ford Health System. Multi-Parametric Magnetic Resonance Imaging for Prostate Cancer Patients. NCT03180398. Updated 17 February 2022. Available online: https://beta.clinicaltrials.gov/study/NCT03180398 (accessed on 27 September 2022).
  60. IRCCS San Raffaele. MR Radiomic Features in Prostate Cancer. NCT04219059. Updated 9 February 2021. Available online: https://beta.clinicaltrials.gov/study/NCT04219059 (accessed on 27 September 2022).
  61. Hatt, M.; Laurent, B.; Ouahabi, A.; Fayad, H.; Tan, S.; Li, L.; Lu, W.; Jaouen, V.; Tauber, C.; Czakon, J.; et al. The first MICCAI challenge on PET tumor segmentation. Med. Image Anal. 2018, 44, 177–195. [Google Scholar] [CrossRef] [Green Version]
  62. Zwanenburg, A.; Löck, S. Why validation of prognostic models matters? Radiother. Oncol. 2018, 127, 370–373. [Google Scholar] [CrossRef]
Table 1. Summary of studies utilizing magnetic resonance imaging (MRI)-derived radiomic models to predict Gleason score.
Table 1. Summary of studies utilizing magnetic resonance imaging (MRI)-derived radiomic models to predict Gleason score.
AuthorYearn=Outcome MeasureImages UsedFeature SelectionRegion of InterestNumber of FeaturesModel ValidationResults
Chaddad [15]201899GS ≤ 6, 3 + 4, ≥4 + 3T2WI
ADC
Random forest (RF)Lesion414:1 training:testing
5-fold cross-validation
Gleason score ≤ 6, AUC = 0.834
Gleason score = 3 + 4, AUC = 0.727
Gleason score ≥ 4 + 3, AUC = 0.774
Penzias [16]201836GS 6 vs. >6T2WIRandom forest (RF)Prostate Lesion20013-fold cross-validation
23:13 training:testing
Gabor texture features model, AUC = 0.69
Gland lumen shape feature model, AUC = 0.75
Toivonen [17]2019100GS 3 + 3 vs. ≥3 + 3T2WI
DWII
Logistic regression with L1 or L2 regularizationLesionADC 1281/7105
T2WI 1631/7105
Inner 10-fold cross-validation; Outer leave-pair-out cross-validation (LPOCV)AUC = 0.88
Hectors [18]201964GS ≥ 8T2WI ADCLeast absolute shrinkage and selection operator (LASSO)Lesion226 Nested cross-validationGleason score ≥ 8, AUC = 0.72
Decipher score ≥ 0.6, AUC = 0.84
Abdollahi [19]201933IMRT response
GS ≤ 7 or >7
T2WI ADCRadiomics module of 3D Slicer
SelectKBest_chi2
Variance threshold
Select from Model
Select Percentile
LesionIMRT response: 40/1135
GS score: NR
10-fold cross-validationPre-IMRT ADC, AUC = 0.77 for IMRT response
T2WI, AUC = 0.0739 for GS prediction
ADC, AUC = 0.675 for GS prediction
McGarry [20]201948GS < 4, GS 4–5T2WI
ADC
DCE
Custom algorithm programmed in MATLAB ProstateNR3:1 training:testing
3-fold cross-validation
AUC = 0.79
Gong [21]2020489GS > 7T2WI
DWI
Pyradiomics v3.6 in Python
Minimum redundancy maximum relevance (mRMR)
ProstateProstate: 1345Single-sequenceDWI, AUC = 0.801 in training
DWI, AUC = 0.787 in testing
T2WI, AUC = 0.712 in training
T2WI, AUC = 0.645 in testing
Zhang [22]2020166GS upgradingT2WI ADC
DCE
Mutual information maximation (MIM) criterion Lesion20/4404116:50 training:testingCombined T2WI, ADC, DCE training, AUC = 0.899
Combined T2WI, ADC, DCE testing, AUC = 0.868
Santone [23]2021112GGG 1–5T2WILanguage of Temporal Ordering Specification (LOTOS)Area of suspicionNRNRSensitivity, range 0.95–1.0
Specificity, 1.0
Makowski [24]202185GS 6, 7, and ≥8T2WI
ADC
R, package tidyverse
Random forest (RF)
Stochastic gradient boosting (SGB)
Support vector machine (SVM)
K-nearest neighbor (KNN)
Lesion
Peripheral/Transitional zones
322Cross-validationSVM model, AUC = 0.92
RF model, AUC = 0.83
SGB model, AUC = 0.75
KNN model, AUC = 0.50
Damascelli [25]202162GS 7 (3 + 4), 7 (4 + 3), 8, 9T2WI
DWI
DCE
ADC
Radiomics module of 3D Slicer v4.10.2LesionT2WI features: 44/93
ADC features: 61/93
10-fold cross-validation
Training set only
Combined T2WI & ADC model for GGG, AUC = 0.88
Combined T2WI & ADC model for LNI, AUC = 0.90
Combined T2WI & ADC model for EPE, AUC = 0.85
Gugliandolo [26]202149GS 3 + 4, GS 4 + 3T2WIHierarchical clustering procedure ProstateProstate: 1058/1702Leave-out cross-validationAUC = 0.80 in training
AUC = 0.75 in testing
Castillo [27]2021204GS ≤ 6, ≥7T2WI
DWI
ADC
Workflow for optimal radiomics classification (WORC)Lesion5404:1 training:testing
100× random-split cross-validation
Training: AUC = 0.72, sensitivity = 0.76, specificity = 0.55
Testing: AUC = 0.75, sensitivity = 0.88, specificity = 0.63
External validation: AUC = 0.75
Rodrigues [28]2021281GS > 7T2WI
DWI
ADC
Support vector machine weighing (SVM-RFE)
Boruta algorithm
Minimum redundancy maximum relevance (mRMR)
LASSO regularization
Lesion
Prostate
Lesion, 167/321
Prostate: 257/321
288 pipelines produced, producing Gland and Lesion datasetsSVM-RFE pipeline, F2 = 0.7226, Kappa = 0.3781
mRMR pipeline, F2 = 0.7071, Kappa = 0.4095
Whole gland, F2 = 0.7426, Kappa = 0.351
Lesion: F2 = 0.6682, Kappa = 0.3687
Gong [29]2022489GS > 7, =7, <7T2WI
DWI
ADC
Pyradiomics v3.6 in Python
Minimum redundancy maximum relevance (mRMR)
Prostate3D: 1409/4227
2D: 1046/3138
2:1 training:testing3D, AUC = 0.800, specificity = 0.781, sensitivity = 0.703
2D, AUC = 0.776, specificity = 0.798, sensitivity = 0.674
Table 2. Summary of studies utilizing MRI-derived radiomic models to predict adverse pathology (i.e., positive surgical margins, extraprostatic extension, and lymph node invasion).
Table 2. Summary of studies utilizing MRI-derived radiomic models to predict adverse pathology (i.e., positive surgical margins, extraprostatic extension, and lymph node invasion).
AuthorYearn=Outcome MeasureImages UsedFeature SelectionRegion of InterestNumber of FeaturesModel ValidationResults
Ma [30]2020119EPET2WILeast absolute shrinkage and selection operator (LASSO)Prostate + 1 mm margin156/161910-fold cross-validation
2:1 training:testing
74 training
45 validation
Training: AUC = 0.906, sensitivity = 0.946; specificity = 0.770
Validation: AUC = 0.821, sensitivity = 0.846, specificity = 0.703
Xu [31]2020115EPET2WI
DWI
DCE
ADC
Least absolute shrinkage and selection operator (LASSO)
Minimum redundancy maximum relevance (mRMR)
Lesions3082 training
33 testing
Training AUC = 0.919
Testing AUC = 0.865
Validation AUC = 0.857, sensitivity = 0.714, specificity = 0.895
Bai [32]2021284EPET2WI
ADC
Least absolute shrinkage and selection operator (LASSO)
Minimum redundancy maximum relevance (mRMR)
Peritumoral
Intratumoral
19/1595
19/1595
158 internal training
68 internal testing
58 external validation
Peritumoral model AUC = 0.674
Intratumoral model AUC = 0.554
Final model on internal testing AUC = 0.803
Final model on external validation AUC = 0.598
Cuocolo [33]2021193EPET2WI
DWI
ADC
Scikit-learn Python3 package
Weka data mining software
Synthetic Minority Oversampling Technique
Index lesion14/243610-fold cross-validation
104 training set
43 external test set
46 external validation
Training set AUC = 0.83
External test set AUC = 0.80
External validation set 2 AUC = 0.73
Hou [34]2021401LNIT2WI
DWI
ADC
Pyradiomics v3.6 in Python
Random forest (RF)
ProstateNR/2553280 training
71 internal testing
50 external tests
Training, AUC = 0.93
Internal testing, AUC = 0.92
External testing, AUC = 0.76
Bourbonne [35]2021280LNIADC
T2WI
Multilayer Perceptron Network, SPSS v24.0Lesions6/86406:4 training:testingCombined clinical and radiomic model training AUC = 1.0
Combined clinical and radiomic model testing AUC = 0.87
Zheng [36]2022244LNIT2WI
ADC
PyRadiomics v3.0.1 in Python 3.6
Sequential Floating Forwarding Selection
Index lesion115-fold cross validation
160 training
84 testing
AUC = 0.915; sensitivity = 0.786; specificity = 0.90; NPV = 0.955; PPV = 0.611
Liu [37]2022602LNIT2WI
DWI
ADC
DCE
ANOVA
Recursive feature elimination
Prostate
Lesions
19/755 (model 1); 11/829 (model 2); 17/646 (model 3); 16/650 (model 4)332 training
142 testing
Training cohort:
Model 1 (AUC = 0.85, sensitivity = 0.74, specificity = 0.86)
Model 2 (AUC = 0.86, sensitivity = 0.71, specificity = 0.91)
Model 3 (AUC = 0.94, sensitivity = 0.89, specificity = 0.93)
Model 4 (AUC = 0.82, sensitivity = 0.67, specificity = 0.81)
Testing cohort:
Model 1 (AUC = 0.63, sensitivity = 0.55, specificity = 0.76)
Model 2 (AUC = 0.70, sensitivity = 0.69, specificity = 0.67)
Model 3 (AUC = 0.73, sensitivity = 0.81, specificity = 0.62)
Model 4 (AUC = 0.56, sensitivity = 0.93, specificity = 0.29)
Damascelli [25]202162LNI
EPE
T2WI
DWI
DCE
ADC
Radiomics module of 3D Slicer v4.10.2
Support vector machine model
Index lesionT2WI features: 44/93
ADC features: 61/93
10-fold cross-validation
Training set only
Combined T2WI and ADC model for LNI, AUC = 0.90
Combined T2WI and ADC model for EPE, AUC = 0.85
He [38]2021459PSM
EPE
T2WI
DWI
ADC
Minimum redundancy maximum relevance (mRMR)
Least absolute shrinkage and selection operator (LASSO)
Lesions278 (T2WI)
293 (ADC)
EPE prediction
268 (T2WI)
295 (ADC)
PSM prediction
7:3 training:testingECE, ADC AUC = 0.625
ECE, ADC integrated clinical model AUC = 0.728
PSM, T2WI AUC = 0.614
PSM, ADC AUC = 0.733
PSM, T2WI integrated clinical model AUC = 0.680
PSM, ADC Integrated clinical model AUC = 0.766
Li [39]2021198Adverse pathology, including EPE, SVI, and LNIT2WI
DWI
ADC
Minimum redundancy maximum relevance (mRMR)Prostate55-fold, 10-run cross-validation
71 training
127 testing
Adverse pathology AUC = 0.71
Table 3. Summary of studies utilizing MRI-derived radiomic models to predict biochemical recurrence following radical prostatectomy.
Table 3. Summary of studies utilizing MRI-derived radiomic models to predict biochemical recurrence following radical prostatectomy.
AuthorYearn=Outcome MeasureImages UsedFeature SelectionRegion of InterestNumber of FeaturesModel ValidationResults
Bourbonne [40]2020195BCRT2WI
ADC
Multilayer Perceptron Network, SPSS v24.0LesionNR107 training
88 external testing
Clinical AUC = 0.68
Radiomic AUC = 0.82
Clinical and radiomic AUC = 0.86
Li [39]2021198BCRT2WI
DWI
ADC
Minimum redundancy maximum relevance (mRMR)Prostate55-fold, 10-run cross-validation
71 training
127 testing
Training model HR = 7.01, 95% CI: 1.21–40.68, p < 0.05, independent of preoperative and clinicopathologic parameters in multivariable analysis
Testing model HR = 1.9, 95% CI: 1.4–2.7, p < 0.05
Yan [41]2021485BCRT2WIDeep survival radiomic neural network
Dense box: three hidden layers with 48 neurons
Auto-coding: two hidden layers with 48 neurons + a hidden layer with 24 neurons
Lesions155/702368 training
34 external testing
83 external validation
3-year BCR training AUC = 0.84
3-year BCR external testing AUC = 0.85
3-year BCR external validation AUC = 0.84
5-year BCR training AUC = 0.83
5-year BCR external testing AUC = 0.88
5-year BCR external validation AUC = 0.88

Significantly improved accuracy compared to GG-RP, CAPRA-S, NCCN, and CAPRA clinical models
Shiradkar [42]2022133BCRT2WI
ADC
Random forest (RF)
Cox proportional hazards regression model
Prostate
Lesions
10Cross-validation
2:1 training:testing
3-year BCR HR: 2.91, 95% CI: 1.45–11.51, p = 0.02

Significantly improved accuracy compared to clinical characteristics, CAPRA, CAPRA-S, and Decipher scores
Table 4. Summary of studies utilizing MRI-derived radiomic models to predict biochemical failure following radiation therapy.
Table 4. Summary of studies utilizing MRI-derived radiomic models to predict biochemical failure following radiation therapy.
AuthorYearn=Outcome MeasureImages UsedFeature SelectionRegion of InterestNumber of FeaturesModel ValidationResults
Gnep [43]201774BFT2WI
ADC
Random forest (RF)Lesion
Prostate
Peripheral zone
Transitional zone
144Training onlyFive radiomic features C-index 0.90 ± 0.09 (tumor volume, T2W difference variance mean, T2W contrast mean, maximal tumor area, ADC median)
Fernandes [44]2018120BFT2WIMinimum redundancy maximum relevance (mRMR)
Random forest (RF)
Prostate + 2 mm margin25410-fold cross-validationAUC = 0.63
Zhong [45]202091BFT1WI
T2WI DWI
Inception-Resnet-v2 networkProstate45/15363-fold cross-validation
4:1 training:testing
Mean training AUC = 0.99
Mean testing AUC = 0.73
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Huynh, L.M.; Hwang, Y.; Taylor, O.; Baine, M.J. The Use of MRI-Derived Radiomic Models in Prostate Cancer Risk Stratification: A Critical Review of Contemporary Literature. Diagnostics 2023, 13, 1128. https://doi.org/10.3390/diagnostics13061128

AMA Style

Huynh LM, Hwang Y, Taylor O, Baine MJ. The Use of MRI-Derived Radiomic Models in Prostate Cancer Risk Stratification: A Critical Review of Contemporary Literature. Diagnostics. 2023; 13(6):1128. https://doi.org/10.3390/diagnostics13061128

Chicago/Turabian Style

Huynh, Linda My, Yeagyeong Hwang, Olivia Taylor, and Michael J. Baine. 2023. "The Use of MRI-Derived Radiomic Models in Prostate Cancer Risk Stratification: A Critical Review of Contemporary Literature" Diagnostics 13, no. 6: 1128. https://doi.org/10.3390/diagnostics13061128

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop