Skip to main content

A comparison of 2D and 3D magnetic resonance imaging-based intratumoral and peritumoral radiomics models for the prognostic prediction of endometrial cancer: a pilot study

Abstract

Background

Accurate prognostic assessment is vital for the personalized treatment of endometrial cancer (EC). Although radiomics models have demonstrated prognostic potential in EC, the impact of region of interest (ROI) delineation strategies and the clinical significance of peritumoral features remain uncertain. Our study thereby aimed to explore the predictive performance of varying radiomics models for the prediction of LVSI, DMI, and disease stage in EC.

Methods

Patients with 174 histopathology-confirmed EC were retrospectively reviewed. ROIs were manually delineated using the 2D and 3D approach on T2-weighted MRI images. Six radiomics models involving intratumoral (2Dintra and 3Dintra), peritumoral (2Dperi and 3Dperi), and combined models (2Dintra + peri and 3Dintra + peri) were developed. Models were constructed using the logistic regression method with five-fold cross-validation. Area under the receiver operating characteristic curve (AUC) was assessed, and was compared using the Delong’s test.

Results

No significant differences in AUC were observed between the 2Dintra and 3Dintra models, or the 2Dperi and 3Dperi models in all prediction tasks (P > 0.05). Significant difference was observed between the 3Dintra and 3Dperi models for LVSI (0.738 vs. 0.805) and DMI prediction (0.719 vs. 0.804). The 3Dintra + peri models demonstrated significantly better predictive performance in all 3 prediction tasks compared to the 3Dintra model in both the training and validation cohorts (P < 0.05).

Conclusions

Comparable predictive performance was observed between the 2D and 3D models. Combined models significantly improved predictive performance, especially with 3D delineation, suggesting that intra- and peritumoral features can provide complementary information for comprehensive prognostication of EC.

Background

Endometrial cancer (EC) is the most common gynecological malignancy in developed country and second most common in China. Its incidence and mortality rate were still rising in recent years [1, 2]. Approximately 3% of women will develop EC in their lifetime with a median age of 61 years at first diagnosis [3]. While EC has an encouraging overall 5-year survival rate of 81% [4], clinicians face significant challenges in treatment decisions of overtreatment for low-risk patients and undertreatment for high-risk patients. Accurate risk assessment rely heavily on important histopathological factors such as International Federation of Gynecology and Obstetrics (FIGO) stage, lymphovascular space invasion (LVSI), and depth of myometrial invasion (DMI) [5,6,7,8]. While precisely determining these prognostic metrics continues to be crucial for distant relapse assessment and of poor prognosis estimation. However, such prognostic biomarkers can only be confirmed by histopathology, and is the main challenge of current clinical practice.

Magnetic resonance imaging (MRI) is considered mainstay for the preoperative evaluation of EC, given its high soft tissue contrast resolution allowing for the delineation of tumor invasion [1, 9]. However, the subjective nature in visual interpretation remains a limitation of conventional MRI. As reported by Arnaiz et al. assessing its role in disease staging, accurate prediction was only achieved in 47.2% of cases [10]. Radiomics represent a novel technique enabling the extraction of large amounts of data from medical images in a high-throughput and quantitative manner [11]. Since its advent, it has demonstrated great potential as a diagnostic and prognostic tool for a wide range of cancers [12,13,14,15]. In the context of EC, radiomics have demonstrated the value in not only the characterization of tumors, but also the prediction of high-risk diseases and survival outcomes [16,17,18,19,20].

Region of interest (ROI) delineation strategies can influence feature extraction and hence, affect the downstream performance of a radiomics model. Two approaches currently exist — the two-dimensional (2D) approach, which involves the delineation of lesions from a single image layer at the largest cross-section, or simply the center slice, and the three-dimensional (3D) approach, which involves the use of all tumor-containing slices. While the latter approach has been widely advocated for its comprehensiveness in tumor characterization [17, 20, 21], the need for manual delineation renders it more time-consuming and labor-intensive. As of now, the optimal ROI delineation strategy for the radiomics profiling of EC remains unclear. Besides, the majority of previous MRI-based radiomics research has placed focused on the characterization of intratumoral regions [16, 20, 21]. However, the surrounding microenvironment has been increasingly shown to offer insight into the clinical behavior of primary lesions [22,23,24].

This study thereby aimed to compare the clinical value of 2D and 3D MRI-based radiomics features for the prediction of LVSI, DMI, and disease stage in EC, as well as contribute to the existing literature on the significance of peritumoral radiomics features for prognostication of the disease.

Methods

Patient selection

Patients diagnosed with endometrial cancer between January 2017 and December 2022 at Peking University Third Hospital were retrospectively reviewed. All patients who underwent pelvic magnetic resonance imaging (MRI) within 14 days prior to hysterectomy were considered. The exclusion criteria included the following: (1) tumors with a maximum diameter of < 1 cm, (2) inadequate or poor image quality, (3) absence of relevant clinical or pathological information, (4) prior treatment with adjuvant chemotherapy or radiotherapy, and (5) non-endometrial primary malignancies.

This study was approved by the Ethics Committee of the institute (M2023637), who waived the requirement for informed patient consent.

Clinicopathological data collection

Clinicopathological characteristics including age, DMI, LVSI, tumor stage, histopathological type and serum CA125 and CA199 levels were obtained from medical records. Histopathological assessment was performed on all surgical specimens. Tumor staging was performed according to the 2009 FIGO staging system, with stages I – II and III – IV were classified as early- and late-stage EC, respectively [25].

Image acquisition and segmentation

Preoperative pelvic MRI was performed using 1.5 T Avanto scanner and 3.0 T Skyra scanners, with protocol parameters detailed in Supplementary Table S1. Axial T2-weighted images were acquired and used for subsequent analysis due to their ability to effectively distinguish between tumor-tissue and normal surrounding myometrial-tissue [1]. All images were imported into uAI Research Portal (V1.1, United Imaging Intelligence, Co., Ltd., Shanghai, China).

Intratumoral ROIs (ROIintra) were manually delineated by a radiologist (R.Y.) with 5-year experience, which were then reviewed and refined where necessary by a second radiologist (X.X.) with 15-year experience. Both radiologists were blinded to the study. ROIintra was defined as the entire tumor volume across all tumor slices on 3D images, and as the maximum tumor cross-section on 2D images. Any cystic or hemorrhagic areas were avoided. Peritumoral ROIs (ROIperi) were automatically delineated by extending the tumor margin by 3 mm generated using the boundary “dilation” tool in the uAI platform [19, 24].

Image preprocessing and radiomics feature extraction

Image preprocessing and radiomics feature extraction were performed in accordance to the Image Biomarker Standardisation Initiative (IBSI [26]). All T2-weighted images were resampled to an isotropic pixel size of 1 × 1 × 1 mm using BSpline interpolation to eliminate resolution discrepancies between devices. Pixel intensities were subsequently standardized to a common scale to further minimize background noise.

Radiomics features were extracted using the PyRadiomics python package. A total of 1197 radiomics features were extracted for each ROI, and consisted of the following 7 classes: first-order statistics (19.5%), shape-based features (1.2%), texture-based features including gray level co-occurrence matrix (GLCM) (23.9%) capturing texture, gray level run length matrix (GLRLM) (17.4%) describing gray level runs, gray level size zone matrix (GLSZM) (17.4%) characterizing zone sizes, neighboring gray tone difference matrix (NGTDM) (5.4%) quantifying differences between tumor and neighborhood pixels, and gray level dependence matrix (GLDM) (15.2%) analyzing dependency of gray levels. Subsequently Combined feature (ROIintra+peri) was fused by merging the extracted features from intra-tumoral models and respective peritumoral models. All extracted features were standardized using the Z-score normalization method to standardize the different measurement units across features, preventing model results from being influenced by feature-scale variations.

Radiomic feature selection and machine learning model construction

First, features with correlation coefficients P-value less than 0.05 were remained. Least absolute shrinkage and selection operator regression (LASSO) was subsequently performed to select for those with non-zero coefficients. Five-fold cross-validation was applied, features appeared in more than two times were finally selected by “vote” function on the uAI platform to establish the radiomic models.

The models were eventually constructed using selected radiomics features using logistic regression. Patients were randomly divided into five equal size subsets, one subset served as the validation set. The remaining four-fifths were the training set. This process was then repeated five times with different subsets for validation and training. The performance was averaged across the five times process to obtain a robust estimate of generalizability. The radiomics workflow is illustrated in Fig. 1.

Fig. 1
figure 1

The radiomics workflow

Statistical analysis

All statistical analyses were conducted using the SPSS software (IBM, Chicago, IL, USA, R23.0.0.0). Diagnostic performance was assessed using the receiver operating characteristic (ROC) curve analysis in terms of areas under the ROC curve (AUC), sensitivity, and specificity and accuracy. AUCs were compared using the Delong test. and are illustrated in the form of heatmaps.

Results

Baseline clinicopathological characteristics

A total of 174 patients with EC were enrolled in our study. The patient selection process is illustrated in Fig. 2. The average age was 56.96 ± 11.43 years (range, 27–86 years). Among them, 37 (21.3%) presented with late-stage disease. LVSI was observed in 39 (22.4%) patients, while DMI was seen in 55 (31.6%). The most common EC cell type was observed to be endometrioid adenocarcinoma (88.5%). All baseline characteristics of the patients are shown in Table 1.

Fig. 2
figure 2

The patient selection process

Table 1 Baseline clinicopathological characteristics of the included patients

Radiomics feature selection

Six models, 2Dintra, 2Dperi, 2Dintra + peri, 3Dintra, 3Dperi, and 3Dintra + peri, were constructed for each task. The total number of radiomics features retained in each model were as follows: 11, 8, 11, 2, 7, and 6, respectively, for LVSI prediction; 8, 14, 13, 14, 10, and 13, respectively, for DMI prediction; and 6, 6, 7, 7, 7, and 11, respectively, for FIGO stage prediction. The distribution of radiomics feature classes of intra- and peri-tumoral region across all models is shown in Fig. 3a. The number of intra- and peritumoral features incorporated into each model is shown in Fig. 3b. The exact feature names and type are provided in Supplementary Tables S2S4.

Fig. 3
figure 3

Distribution and composition of (a) the three feature classes (first-order, shape, texture) across the models, and (b) the intra- and peritumoral features across the models

Predictive performance of the independent radiomics models

The ROC curve results of the six established models are shown in both Table 2; Fig. 4. The Delong test results are shown in Fig. 5 and Supplementary Tables S5S10.

Table 2 Logistic regression with five-fold cross-validation for predictive performance of the models
Fig. 4
figure 4

The receiver operating characteristic curve analysis results for (a, b) LVSI prediction, (c, d) DMI prediction, and (e, f) disease stage prediction

Fig. 5
figure 5

The Delong test results for (a, b) LVSI prediction, (c, d) DMI prediction, and (e, f) disease stage prediction between the training and validation cohorts, respectively

In terms of ROI delineation strategies, significant differences were observed between the 2Dintra and 3Dintra models for LVSI prediction in the training cohort [0.810 (0.718–0.903) vs. 0.738 (0.634–0.844), P = 0.001]. Additionally, significant differences were shown between the 2Dperi and 3Dperi models for both DMI [0.928 (0.889–0.972) vs. 0.833 (0.761–0.909), P = 0.000] and stage prediction [0.760 (0.658–0.860) vs. 0.824 (0.747–0.908), P = 0.000] in the training data. Such significant differences were not observed between all models in the validation cohort (P > 0.05).

In terms of intra-and peri-tumoral features, significant differences were observed between the 2Dintra and 2Dperi models for DMI prediction [0.784 (0.699–0.871) vs. 0.928 (0.889–0.972, P = 0.000] in the training cohort, which was not observed in the validation cohort (P > 0.05). Significant differences were observed between the 3Dintra versus 3Dperi models for LVSI [0.738 (0.634–0.844) vs. 0.813 (0.782–0.898), P = 0.000], DMI [0.761 (0.679–0.847) vs. 0.833 (0.761–0.909), P = 0.000], and stage prediction [0.782 (0.688–0.877) vs. 0.824 (0.747–0.908), P = 0.049)] in the training cohort. Such significance remained in the validation cohort for LVSI prediction [0.738 (0.634–0.844) vs. 0.805 (0.625–0.977), P = 0.049] and DMI prediction [0.719 (0.538–0.904) vs. 0.804 (0.654–0.953), P = 0.019], but not for stage prediction [0.738 (0.540–0.932) vs. 0.763 (0.581–0.942].

Predictive performance of the combined radiomics models

For LVSI prediction, the 2Dintra + peri and 3Dintra + peri models demonstrated AUC values of 0.853 (0.773–0.933) and 0.816 (0.730–0.901), respectively, in the training cohort and values of 0.720 (0.493–0.944) and 0.812 (0.633–0.979), respectively, in the validation cohort. For DMI prediction, the AUC values were 0.839 (0.768–0.913) and 0.845 (0.776–0.917), respectively, in the training cohort, and were 0.799 (0.638–0.963) and 0.807 (0.653–0.959), respectively, in the validation cohort. For stage prediction, significant differences were demonstrated in both the training cohort [0.814 (0.719–0.909) vs. [0.864 (0.795–0.933), P = 0.006] and the validation cohort [0.687 (95% CI, 0.472–0.896) vs. 0.804 (0.640–0.967), P = 0.023] in the validation cohort.

Compared to the 2Dintra model, significantly increase in predictive performance was observed with inclusion of peritumoral features for LVSI (+ 0.043, P = 0.006) and DMI (+ 0.055, (P = 0.000)) in the training cohort. Significant increase was observed for DMI prediction (+ 0.039, P = 0.045) in the validation cohort. Compared to the 3Dintra model, significant increase in prediction efficacy was observed for LVSI prediction (+ 0.078, P = 0.000), DMI prediction (+ 0.084, P = 0.000), and FIGO stage prediction (+ 0.066, P = 0.000) in the training cohort. Such significance was also observed for LVSI prediction (+ 0.074, P = 0.018), DMI prediction (+ 0.088, P = 0.003), and FIGO stage prediction (+ 0.066, P = 0.039) in the validation cohort.

Discussion

Six prognostic radiomics models of various ROI delineation strategies were constructed in this study, all of which yielded favorable diagnostic performance, demonstrating the potential as non-invasive tools for the prediction of LVSI, DMI, and FIGO stage to guide the management of EC. Our study further found comparable results between the 2D and 3D radiomics models, implying the lack of added value with the more labor-intensive 3D delineation approach. Our findings also demonstrated that assessment of combined intra- and peritumoral features can further enhance the predictive ability of the model.

There has been ongoing debate regarding the use of 2D and 3D radiomics features. Several studies have reported the superiority of 3D models in capturing tumor characteristics, Ortiz-Ramón et al. employed 2D and 3D MRI radiomics to differentiate the origins of brain metastases. Machado et al. utilized MRI radiomics to predict recurrence in clinically non-functioning pituitary macroadenomas. Watzenboeck et al. employed MRI radiomics to assess fetal lung development [27,28,29], but are often limited in sample size and in their overlooking of peritumoral regions for feature extraction. Our study found comparable performance between 2D and 3D approaches both for intra- and peritumoral ROIs, which is in line with the those of the study by Zhang et al. for tumor phenotype prediction in non-small cell lung carcinomas [30], and the study by Arefan et al. for axillary lymph node metastasis risk in breast cancer [31], which also reported similar performance between 2D and 3D radiomics approaches. However, it’s important to note that findings in EC have been mixed. Fasmer et al. [25], using a relatively small sample size, found that 3D whole-tumor radiomic signatures yielded comparable AUC in the training set but significantly lower AUC in the validation set for advanced FIGO stage prediction in EC. These varying results across studies highlight the need for further investigation into the optimal radiomics approach for EC, particularly with larger sample sizes and robust validation.

Such observations may be attributable to the inevitable amplification of noise as the whole tumor volume is delineated across multiple image slices. While 2D ROI delineation within a representative slicing plane may encapsulate key characteristics of the entire tumor volume. Our study highlighted that despite the potential for additional information provided in assessing the entire tumor volume, the marginal gains in predictive ability may not outweigh the practical demands associated with manual multi-slice annotation. This observation could provide support for the promising model performance reported in previous literature, where the utilization of a single slice yielded comparable results [28]. Nonetheless, large-sample studies are warranted for verification of our results.

The growing recognition of the role of the tumor microenvironment in cancer biology has resulted in the recent shift in focus from the primary tumor itself to the surrounding stroma for radiomics feature extraction [32, 33]. Such significance in prognostic value of surrounding tissues has been demonstrated in breast [34] and ovarian cancers [35]. In this study, we simultaneously developed predictive models through an automated 3 mm boundary expansion of the intratumoral ROI, aimed at capturing spatial characteristics beyond the primary tumor. To our knowledge, this is the first study to compare the effects of 2D versus 3D ROI delineation approaches on peritumoral region. Our results demonstrated useful information that combined models can indeed be attained by inclusion of peritumoral lesion, showed highest diagnostic performance in both the 2D and 3D models, especially 3D models. The obtained AUC results demonstrate reasonable performance and are consistent with those of the study by Yan et al., who demonstrated the complementary nature of intra- and peritumoral features for LVSI and DMI prediction with AUCs of 0.859 and 0.856 respectively [22, 23]. This indicates that, compared to intratumoral features alone, peritumoral regions provide supplementary prognostic information. When coupled with intratumoral radiomics, this minimally invasive multi-regional segmentation provided a balanced assessment of the local tumor-stroma interplay for improved prognostic modeling.

Interesting, the 3Dperi model was observed to significantly outperform the 3Dintra model in our study. This may suggest that tissues in direct contact with the tumor are likely to undergo microstructural alterations prior to histologic changes. 3D peritumoral radiomics may thus offer the sensitivity required for identifying such pre-invasive alterations, and enable a more comprehensive characterization of spatial heterogeneity in the tumor microenvironment. Based on our findings, we propose the potential clinical utility of sampling adjacent stromal tissues, in addition to the primary lesion, for the histopathological assessment of ECs.

Our study had several limitations. First, the retrospective design and relatively modest sample size may limit the generalizability of our findings. Larger prospective studies are thereby needed to validate our results. Second, only a narrow set of histopathological variables with unbalanced cohorts were included due to data availability constraints, these differences could influence the radiomics features extracted and potentially impact the performance of our models. While our study was predominantly endometrioid adenocarcinoma, the validation results demonstrated good stability across the iterations of cross-validation. This suggests that despite pathology variability not fully accounted for, the validation approach helped improve generalizability to these cases Third, the optimal extent of peritumoral expansion was not explored in this study. Fourth, manual delineation may have inevitably resulted in inter-observer variability, which could have been mitigated by utilization of a semi-automated or automated approach. Lastly, more advanced hyperparameter optimization and deep learning methods were not applied due to the sample size of this study.

Conclusions

This study developed and investigated the role of 2D and 3D intratumoral and peritumoral radiomics models in the prediction of LVSI, DMI, and disease stage in EC. A 3D ROI delineation approach did not achieve significant improvement in prediction performance. Inclusion of peritumoral features significantly enhanced prediction accuracy of the models, especially the 3D model. Further studies with standardized delineation strategies are warranted to validate our results and advance the clinical translation of radiomics in the field of gynecological oncology.

Data availability

The datasets used or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

EC:

Endometrial cancer

FIGO:

International Federation of Gynecology and Obstetrics

LVSI:

Lymphovascular space invasion

DMI:

Depth of myometrial invasion

MRI:

Magnetic resonance imaging

3D:

Three-dimensional

2D:

Two-dimensional

GLCM:

Gray level co-occurrence matrix

GLRLM:

Gray level run length matrix

GLSZM:

Gray level size zone matrix

NGTDM:

Gray tone difference matrix

GLDM:

Gray level dependence matrix

LASSO:

Least absolute shrinkage and selection operator regression

ROC:

Receiver operating characteristic

AUC:

Areas under the ROC curve

2Dintra :

Two-dimensional intratumoral model

2Dperi :

Two-dimensional peritumoral model

2Dintra + peri :

Two-dimensional intratumoral + peritumoral model

3Dintra :

Three-dimensional intratumoral model

3Dperi :

Three-dimensional peritumoral model

3Dintra + peri :

Three-dimensional intratumoral + peritumoral model

References

  1. Maheshwari E, Nougaret S, Stein EB, Rauch GM, Hwang K-P, Stafford RJ, et al. Update on MRI in evaluation and treatment of Endometrial Cancer. Radiographics. 2022;42:2112–30.

    Article  PubMed  Google Scholar 

  2. Feng J, Lin R, Li H, Wang J, He H. Global and regional trends in the incidence and mortality burden of endometrial cancer, 1990–2019: updated results from the global burden of Disease Study, 2019. Chin Med J. 2019;137(3):294–302.

    Article  Google Scholar 

  3. Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME, Singh N. Endometrial cancer. Lancet. 2022;399:1412–28.

    Article  PubMed  Google Scholar 

  4. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73:17–48.

    Article  PubMed  Google Scholar 

  5. Peters EEM, Bartosch C, McCluggage WG, Genestie C, Lax SF, Nout R, et al. Reproducibility of lymphovascular space invasion (LVSI) assessment in endometrial cancer. Histopathology. 2019;75:128–36.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Ueno Y, Forghani B, Forghani R, Dohan A, Zeng XZ, Chamming’s F, et al. Endometrial carcinoma: MR imaging–based texture model for preoperative risk Stratification—A preliminary analysis. Radiology. 2017;284:748–57.

    Article  PubMed  Google Scholar 

  7. Zhu X, Ying J, Yang H, Fu L, Li B, Jiang B. Detection of deep myometrial invasion in endometrial cancer MR imaging based on multi-feature fusion and probabilistic support vector machine ensemble. Comput Biol Med. 2021;134:104487.

    Article  PubMed  Google Scholar 

  8. Colombo N, Creutzberg C, Amant F, Bosse T, González-Martín A, Ledermann J, et al. ESMO-ESGO-ESTRO Consensus Conference on Endometrial Cancer: diagnosis, treatment and follow-up†. Ann Oncol. 2016;27:16–41.

    Article  CAS  PubMed  Google Scholar 

  9. Guo W, Wang T, Lv B, Jiang J, Liu Y, Zhao P. Advances in Radiomics Research for Endometrial Cancer: a Comprehensive Review. J Cancer. 2023;14:3523–31.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Arnaiz J, Muñoz A-B, Verna V, Gonzalez-Rodilla I, Schneider J. Magnetic resonance imaging for the Pre-surgical Assessment of Endometrial Cancer: results in a Routine Clinical setting, outside dedicated trials; a cross-sectional study. Anticancer Res. 2016;36:1891–4.

    PubMed  Google Scholar 

  11. Lambin P, Leijenaar RTH, Deist TM, Peerlings J, de Jong EEC, van Timmeren J, et al. Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol. 2017;14:749–62.

    Article  PubMed  Google Scholar 

  12. Bitencourt AGV, Gibbs P, Rossi Saccarelli C, Daimiel I, Lo Gullo R, Fox MJ, et al. MRI-based machine learning radiomics can predict HER2 expression level and pathologic response after neoadjuvant therapy in HER2 overexpressing breast cancer. EBioMedicine. 2020;61:103042.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Yu J, Shi Z, Lian Y, Li Z, Liu T, Gao Y, et al. Noninvasive IDH1 mutation estimation based on a quantitative radiomics approach for grade II glioma. Eur Radiol. 2017;27:3509–22.

    Article  PubMed  Google Scholar 

  14. Jiang X, Song J, Zhang A, Cheng W, Duan S, Liu X, et al. Preoperative Assessment of MRI-Invisible Early-Stage Endometrial Cancer with MRI-Based Radiomics Analysis. J Magn Reson Imaging. 2023;58:247–55.

    Article  PubMed  Google Scholar 

  15. Huang Y-Q, Liang C-H, He L, Tian J, Liang C-S, Chen X, et al. Development and validation of a Radiomics Nomogram for Preoperative Prediction of Lymph Node Metastasis in Colorectal Cancer. J Clin Oncol. 2016;34:2157–64.

    Article  PubMed  Google Scholar 

  16. Bereby-Kahane M, Dautry R, Matzner-Lober E, Cornelis F, Sebbag-Sfez D, Place V, et al. Prediction of tumor grade and lymphovascular space invasion in endometrial adenocarcinoma with MR imaging-based radiomic analysis. Diagn Interv Imaging. 2020;101:401–11.

    Article  CAS  PubMed  Google Scholar 

  17. Stanzione A, Cuocolo R, Del Grosso R, Nardiello A, Romeo V, Travaglino A, et al. Deep myometrial infiltration of Endometrial Cancer on MRI: a Radiomics-Powered Machine Learning Pilot Study. Acad Radiol. 2021;28:737–44.

    Article  PubMed  Google Scholar 

  18. Bc Y, Y L, Fh M, Gf Z, F F, Mh S, et al. Radiologists with MRI-based radiomics aids to predict the pelvic lymph node metastasis in endometrial cancer: a multicenter study. Eur Radiol. 2021;31(1):411–22.

    Article  Google Scholar 

  19. Veeraraghavan H, Friedman CF, DeLair DF, Ninčević J, Himoto Y, Bruni SG, et al. Machine learning-based prediction of microsatellite instability and high tumor mutation burden from contrast-enhanced computed tomography in endometrial cancers. Sci Rep. 2020;10:17769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yan BC, Li Y, Ma FH, Feng F, Sun MH, Lin GW, et al. Preoperative Assessment for High-Risk Endometrial Cancer by developing an MRI ‐ and clinical‐based Radiomics Nomogram: a Multicenter Study. Magn Reson Imaging. 2020;52:1872–82.

    Article  Google Scholar 

  21. Zhao M, Wen F, Shi J, Song J, Zhao J, Song Q, et al. MRI-based radiomics nomogram for the preoperative prediction of deep myometrial invasion of FIGO stage I endometrial carcinoma. Med Phys. 2022;49:6505–16.

    Article  CAS  PubMed  Google Scholar 

  22. Yan B, Zhao T, Li Z, Ren J, Zhang Y. An MR-based radiomics nomogram including information from the peritumoral region to predict deep myometrial invasion in stage I endometrioid adenocarcinoma: a preliminary study. BJR. 2023;96:20230026.

    Article  PubMed  Google Scholar 

  23. Yan B, Jia Y, Li Z, Ding C, Lu J, Liu J, et al. Preoperative prediction of lymphovascular space invasion in endometrioid adenocarcinoma: an MRI-based radiomics nomogram with consideration of the peritumoral region. Acta Radiol. 2023;64:2636–45.

    Article  PubMed  Google Scholar 

  24. Lin Z, Wang T, Li Q, Bi Q, Wang Y, Luo Y, et al. Development and validation of MRI-based radiomics model to predict recurrence risk in patients with endometrial cancer: a multicenter study. Eur Radiol. 2023;33:5814–24.

    Article  PubMed  Google Scholar 

  25. Fasmer KE, Hodneland E, Dybvik JA, Wagner-Larsen K, Trovik J, Salvesen Ø, et al. Whole‐volume Tumor MRI Radiomics for Prognostic modeling in Endometrial Cancer. J Magn Reson Imaging. 2021;53:928–37.

    Article  PubMed  Google Scholar 

  26. The image biomarker standardisation initiative. — IBSI 0.0.1dev documentation. https://ibsi.readthedocs.io/en/latest/#

  27. Ortiz-Ramon R, Larroza A, Arana E, Moratal D. A radiomics evaluation of 2D and 3D MRI texture features to classify brain metastases from lung cancer and melanoma. Annu Int Conf IEEE Eng Med Biol Soc. 2017;2017:493–6.

    PubMed  Google Scholar 

  28. Machado LF, Elias PCL, Moreira AC, Dos Santos AC, Murta Junior LO. MRI radiomics for the prediction of recurrence in patients with clinically non-functioning pituitary macroadenomas. Comput Biol Med. 2020;124:103966.

    Article  PubMed  Google Scholar 

  29. Watzenboeck ML, Heidinger BH, Rainer J, Schmidbauer V, Ulm B, Rubesova E, et al. Reproducibility of 2D versus 3D radiomics for quantitative assessment of fetal lung development: a retrospective fetal MRI study. Insights Imaging. 2023;14:31.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Zhang X, Zhang G, Qiu X, Yin J, Tan W, Yin X, et al. Radiomics under 2D regions, 3D regions, and peritumoral regions reveal tumor heterogeneity in non-small cell lung cancer: a multicenter study. Radiol med. 2023;128:1079–92.

    Article  PubMed  Google Scholar 

  31. Arefan D, Chai R, Sun M, Zuley ML, Wu S. Machine learning prediction of axillary lymph node metastasis in breast cancer: 2D versus 3D radiomic features. Med Phys. 2020;47:6334–42.

    Article  PubMed  Google Scholar 

  32. P K, A O. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res. 2010;316(8):1324–31.

  33. Li R. Peritumoral Radiomics and Predicting Treatment Response. JAMA Netw Open. 2020;3:e2016125.

  34. Xu H, Liu J, Chen Z, Wang C, Liu Y, Wang M, et al. Intratumoral and peritumoral radiomics based on dynamic contrast-enhanced MRI for preoperative prediction of intraductal component in invasive breast cancer. Eur Radiol. 2022;32:4845–56.

    Article  CAS  PubMed  Google Scholar 

  35. Wu Y, Jiang W, Fu L, Ren M, Ai H, Wang X. Intra- and peritumoral radiomics for predicting early recurrence in patients with high-grade serous ovarian cancer. Abdom Radiol. 2022;48:733–43.

    Article  Google Scholar 

Download references

Acknowledgements

We would like to thank the native English-speaking scientists of Elixigen Company (Huntington Beach, CA, USA) for editing our manuscript.

Funding

This study has received funding by National Natural Science Foundation of China (grant number 81971578, 81701648), the Proof of Concept Program of Zhongguancun Science City and Peking University Third Hospital (grant number HDCXZHKC2022202) and Beijing Natural Science Foundation (grant number Z190020).

Author information

Authors and Affiliations

Authors

Contributions

All authors have had access to the data and all drafts of the manuscript. Specifc contributions are as follows: design and supervision: RY, XX, NL; development of methodology: SQ, RY; collection and extraction of data: JX, RY, WZ, KL; analysis and interpretation of data (e.g., statistical analysis, algorithm and software development): SQ, RY, PJ; manuscript writing: NL, RY; final approval of manuscript: all authors.

Corresponding author

Correspondence to Ning Lang.

Ethics declarations

Ethics approval and consent to participate

Institutional Review Board approval was obtained.

Consent for publication

Written informed consent was obtained from the patient for publication of this research and any accompanying images.

Competing interests

The authors of this manuscript declare no relationships with any companies, whose products or services may be related to the subject matter of the article.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yan, R., Qin, S., Xu, J. et al. A comparison of 2D and 3D magnetic resonance imaging-based intratumoral and peritumoral radiomics models for the prognostic prediction of endometrial cancer: a pilot study. Cancer Imaging 24, 100 (2024). https://doi.org/10.1186/s40644-024-00743-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40644-024-00743-2

Keywords