International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Colorectal cancer (CRC) is the third most common malignant tumor worldwide and the second leading cause of cancer-related deaths (1). Epidemiological projections estimate that by 2040, the number of new CRC cases will reach 3.2 million, with deaths rising to 1.6 million, posing a major threat to global health (2). Rectal cancer (RC) accounts for approximately one-third of CRC cases, with 30–50% of patients diagnosed at a locally advanced stage (3). Locally advanced RC (LARC) is typically defined as RC with clinical or pathological stage T3-T4 and/or positive regional lymph nodes, without distant metastasis. It is typically treated with neoadjuvant chemoradiotherapy (nCRT) followed by total mesorectal excision (4,5). This approach reduces tumor volume, stage and local recurrence rate (LRR), and improves surgical resection and anal sphincter preservation rates (6,7). However, it has not significantly improved overall survival (OS) (8). Distant metastasis (DM), which occurs in 25–40% of cases (9,10), remains the primary cause of treatment failure and death, with current chemotherapy and immunotherapy regimens yielding suboptimal OS.
In recent years, groundbreaking advancements have been made in two therapeutic domains. First, for the microsatellite instability-high (MSI-H)/mismatch repair-deficient (dMMR) subtype, which constitutes ~3% of patients with LARC (11), immune checkpoint inhibitors targeting programmed cell death 1 (PD-1)/PD-1 ligand 1 have demonstrated remarkable efficacy (12). A phase II prospective study by Cercek et al (13) enrolled 16 patients with dMMR LARC. Among them, 12 patients achieved clinical complete response (cCR) following 6-month neoadjuvant dostarlimab therapy, with comprehensive imaging and endoscopic biopsy confirming the absence of residual tumor. During a median follow-up of 12 months (range, 6–25 months), none of the patients required chemoradiotherapy or surgical intervention, and no recurrence or disease progression was observed. These findings were further validated by a multicenter retrospective study by Yang et al (14), which included 20 patients with dMMR/MSI-H LARC treated with PD-1 inhibitor monotherapy. The overall CR rate reached 90% (18/20), comprising 11 cases with pathological CR (pCR) and 7 cases managed with watch-and-wait strategies for cCR/near-cCR. After a median follow-up of 25 months, all patients remained free from LR or DM, with 2-year disease-free survival (DFS) and OS rates both maintained at 100%. These results suggest that for patients with dMMR RC achieving cCR/near-cCR after neoadjuvant PD-1 inhibitor therapy, adopting a watch-and-wait strategy could enable non-surgical management without compromising survival outcomes. Second, the total neoadjuvant therapy (TNT) paradigm, which consolidates chemotherapy and radiotherapy into the preoperative phase, has significantly improved treatment completion rates and enabled early control of micrometastases, offering a novel approach to enhance prognosis. Multiple phase III trials (15,16) have confirmed that TNT outperforms conventional regimens by elevating pCR rates and improving long-term outcomes. For instance, the RAPIDO trial reported a pCR rate of 28.4% in the TNT group (vs. 14.3% in controls) (17), while the PRODIGE23 trial demonstrated superior 7-year DFS and OS rates of 67.6 and 81.9%, respectively, in the TNT cohort (vs. 62.5 and 76.1% in controls), with distant metastasis risk reduced to 73.6% (vs. 65.4%) (18). However, 5-year follow-up data from the RAPIDO trial (19) revealed an elevated LRR in the TNT group (10 vs. 6%, P<0.027), potentially attributable to dose limitations of short-course radiotherapy. These findings underscore the critical need for early prognostic prediction in patients with LARC to refine therapeutic decision-making.
The tumor-node-metastasis (TNM) staging system (20–22) is still one of the primary methods for predicting the prognosis of LARC. It has been shown that the pathological status of lymph nodes after nCRT is strongly linked to the risk of LR and DM (23). However, the predictive accuracy of the TNM system is limited by tumor heterogeneity and the subjectivity involved in manual assessments (24).
In addition to tumor staging, several clinicopathological factors, such as neurovascular invasion and tumor differentiation, play a role in prognostic evaluations. The tumor regression grade (TRG) is a key tool for assessing tumor response after nCRT (25) and can indirectly reflect prognosis. However, it is only applicable to postoperative pathological specimens, which limits its ability to assess prognosis before treatment in patients with LARC. KRAS and TP53 mutations are often considered markers of poor nCRT efficacy in LARC, correlating with a worse prognosis (26). However, determining the mutational status typically requires pathological examination of tumor tissue. Tumoral heterogeneity, both within the tumor and across different tumor sites, poses significant challenges to histological methods (27). The inability to monitor real-time genetic changes during treatment further limits the capacity of these methods to dynamically assess tumor biology. Additionally, commonly used tumor markers, such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9, have low sensitivity and specificity. These markers often result in high false-negative rates and have delays in reflecting patient prognosis (28). Many of the aforementioned methods rely on invasive procedures, which restrict their routine use in patient monitoring and surveillance.
Imaging plays a vital role in diagnosing and managing LARC, with computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT) being the primary modalities used. CT is essential for staging LARC due to its rapid acquisition, clear visualization of lesions and ability to assess peri-lesional involvement (29). Although there is insufficient evidence to confirm CT's ability to predict recurrent or metastatic lesions before the onset of other symptoms, it is invaluable for whole-body assessment in symptomatic patients or those with elevated CEA levels. However, its low soft-tissue resolution and associated radiation risk often necessitate combining it with other diagnostic tests to confirm a definitive diagnosis. PET/CT, which integrates functional and anatomical imaging, provides crucial information for accurately assessing treatment efficacy and predicting prognosis. This is achieved by revealing the metabolic activity and anatomical details of tumors. A meta-analysis comparing ultrasound, CT, MRI and PET/CT in the early detection of gastrointestinal tumor metastases found that PET/CT exhibited the highest sensitivity for diagnosing metastases (30). However, the high cost of PET/CT limits its widespread use in routine follow-up examinations.
MRI has emerged as the preferred modality for diagnosing, staging and prognosticating LARC, owing to its superior soft-tissue resolution and its ability to perform multi-sequence, multi-parameter imaging (31). MRI provides detailed and accurate assessments of lesion size, depth of infiltration, involvement of neighboring organs and distant metastasis. T2-weighted imaging (T2WI) is commonly used to evaluate tumor response to nCRT. It defines the TRG by observing the gradual homogenization of tumor signal intensity, changes in tumor volume and size, and the degree of fibrotic replacement (32). Diffusion-weighted imaging (DWI), a functional imaging sequence, assesses tumor response by measuring the diffusion of water molecules between tissues. This diffusion is positively correlated with tumor cell density. Sequences such as the apparent diffusion coefficient (ADC) and contrast-enhanced T1-weighted imaging (CE-TIWI) provide non-invasive insights into tumor cell density, degree of differentiation, microcirculatory perfusion and blood supply (33). Furthermore, imaging features such as extramural vascular invasion (EMVI) and mesorectal fascia (MRF) involvement are critical prognostic factors (34,35). To standardize MRI reporting and improve clinical decision-making consistency, international consensus guidelines recommend structured reporting templates. As proposed by Smith et al (36,37), a comprehensive MRI reporting framework for RC should incorporate essential anatomical descriptors, including tumor distance from the anal verge, MRF status and EMVI. Mnemonic systems such as DISTANCE (Distance, Involvement of Sphincter, T stage, Anal canal, Nodes, Circumferential Resection Margin, Extramural Venous Invasion) and its extended variant DISTANCED-DIS (incorporating Depth of invasion, Signal intensity, Spread) (38,39) facilitate systematic acquisition of key imaging parameters, thereby enhancing surgical planning and prognostic stratification. Despite reducing interobserver variability through standardized terminology, conventional MRI interpretation remains heavily reliant on subjective radiologic expertise, potentially compromising diagnostic comprehensiveness and accuracy. Consequently, there is an urgent need for new methods to provide prognostic information before nCRT, guiding more personalized and precise clinical decision-making.
With the ongoing advancements in precision medicine, radiomics and deep learning (DL) are increasingly applied in the diagnosis and treatment of various diseases (40,41). Radiomics extracts high-throughput, quantitative image data that are often imperceptible to the naked eye, and uses these data to build predictive models that enhance accurate diagnosis, treatment assessment and prognostic prediction. DL automates the extraction and selection of high-dimensional features, enabling comprehensive analysis of image data to identify and predict disease characteristics.
Several studies have explored the use of radiomics from CT and PET/CT imaging to predict the prognosis of LARC treated with nCRT, yielding promising results. A retrospective study involving 411 patients with LARC demonstrated that CT-based radiomics features could independently predict OS in patients with LARC undergoing nCRT (42). The study further showed that combining radiomics features with clinical data significantly enhanced the predictive performance of the model. In a study by Bundschuh et al (43), which analyzed data from 27 patients with LARC treated with nCRT, baseline PET/CT-derived texture parameters demonstrated strong predictive value for progression-free survival (PFS), though they showed limited predictive ability for OS. Similarly, Bang et al (44) found that texture parameters extracted from PET/CT images could serve as an indicator of tumor heterogeneity, useful for predicting the response to nCRT and LR in LARC. Despite these advances, studies using CT and PET/CT to predict the prognosis of nCRT in LARC are limited. This is due, in part, to CT's limitations in soft tissue discrimination and the low prevalence of PET/CT imaging.
Conventional imaging modalities, such as MRI and CT, predominantly rely on morphological indicators (e.g., tumor size, enhancement patterns), which are insufficient to distinguish viable tumor residues from post-therapeutic fibrosis during pretreatment or early treatment phases. Furthermore, existing prognostic biomarkers, such as TRG, require postoperative pathological confirmation and thus fail to dynamically guide therapeutic adjustments. Additional limitations include the lack of objective quantification methods for intratumoral heterogeneity, inadequate integration of multimodal data (imaging, genomic and clinical parameters), and challenges associated with the ‘black-box’ nature of DL models and their poor cross-center generalizability. These issues have substantially hindered the clinical translation of precision predictive frameworks. This review focuses on radiomics and DL technologies, delineating how they overcome the constraints of conventional approaches by addressing critical research gaps-specifically, the limited timeliness of early prediction, insufficient analysis of tumor heterogeneity and the absence of synergistic multidimensional data integration. By resolving these challenges, the present analysis provides innovative conceptual frameworks to advance precision oncology frameworks for LARC.
As a result, this paper will focus on recent research exploring MRI-based radiomics and DL for prognostic prediction in patients with LARC undergoing nCRT (Table I). The PubMed (https://pubmed.ncbi.nlm.nih.gov/), Web of Science (https://www.webofscience.com/) and Google Scholar (https://scholar.google.com/) databases were searched for studies conducted before August 31, 2024. During the search, keywords such as ‘locally advanced rectal cancer’, ‘neoadjuvant chemoradiotherapy’, ‘magnetic resonance imaging’, ‘survival’, ‘prognosis’, ‘metastasis’, ‘radiomics’ and ‘deep learning’ were used. The inclusion criteria were as follows: i) The study population was clearly defined as patients with LARC and all patients received nCRT; ii) MRI was used as the imaging modality; iii) radiomics or DL was employed to predict the prognosis of LARC; iv) an English abstract and a complete methodology description (including the number of cases, dataset partitioning strategy, image segmentation method, feature extraction process, statistical modeling and quantitative analysis indicators) were provided. The following exclusion criteria were applied: i) Studies aimed only at predicting the response to nCRT; ii) article types such as review article, conference abstract, editorial letter, case reports and non-original studies; iii) studies with incomplete methodological information or lacking quantitative validation; and iv) non-English literature. Since this study is not a systematic review, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines and tools for assessing methodological heterogeneity and selection bias were not applied (45).
Radiomics, first introduced by Lambin et al (46) in 2012, is a non-invasive technique that analyzes tumor heterogeneity by extracting and quantifying a range of shape, texture and other features from regions of interest (ROI) in medical images that are not visually detectable. The radiomics workflow generally includes several key stages: Image acquisition, ROI identification and segmentation, feature extraction and selection, and model building and validation (47). In the ROI segmentation step, either automated segmentation algorithms or manual techniques define the tumor region, from which high-throughput features are extracted (48). These features include first-order features, texture features, shape features, model-based features, transform-based features and higher-order features. Techniques such as least absolute shrinkage and selection operator (LASSO), principal component analysis and minimum redundancy maximum relevance are commonly applied to select the most relevant features and prevent model overfitting (49). The predictive power of radiomics models can be enhanced by integrating selected radiomic features with clinical data, pathological findings and treatment details. Logistic regression is frequently used due to its simplicity and effectiveness as a supervised classifier, while other widely used classifiers include support vector machines (SVM), random forests (RF), k-nearest neighbors, decision trees, neural networks and Bayesian classifiers (50–52). Finally, validating model performance is essential and typically involves assessing metrics such as the receiver operating characteristic curve, calibration curve, specificity, sensitivity and decision curve analysis. For robust reproducibility, models should undergo internal validation, and, if feasible, external validation across multiple centers (53).
Prior to nCRT in patients with LARC, baseline MRI can provide pivotal tumor characteristics, including tumor size, morphology, location, MRF invasion and lymph node metastasis. Such information is crucial for predicting tumor behavior, devising personalized treatment strategies and evaluating therapeutic outcomes. T2WI is often used for segmenting the tumor ROI due to its superior soft tissue contrast and multiplanar imaging capabilities. Accordingly, numerous studies extracted radiomics features based on pre-treatment T2WI to construct predictive models. For instance, in a retrospective study by Jayaprakasam et al (54), researchers explored the value of radiomics features extracted from mesorectum on pre-treatment T2WI in predicting outcomes for patients with LARC undergoing nCRT. They developed models to predict both LR and DM, achieving strong predictive performance. The area under the curve (AUC) for the LR and DM models was 0.79 and 0.87, respectively, with corresponding accuracies of 78.3 and 88.4%.
Several studies have enhanced predictive models by incorporating multiple MRI sequences, such as DWI, apparent ADC and CE-TIWI, alongside T2WI (55–57). Using multiparametric radiomics features derived from these sequences has shown promise in improving model accuracy. For instance, Cui et al (55) developed a model using pre-treatment T2WI, ADC and CE-TIWI to predict DFS in patients with LARC after nCRT. Their results demonstrated that the multiparametric model offered better DFS prediction accuracy than models using single sequence alone. In a larger multicenter retrospective study, Huang et al (56) extracted radiomics features from pre-treatment T2WI, DWI and CE-TIWI sequences to develop and validate a multiparametric MRI-based radiomics model for predicting LR or DM in patients with LARC treated with nCRT. The model achieved robust performance, with AUC values of 0.83 in the training set and 0.81 and 0.82 in the two validation sets, respectively.
These findings highlight the value of baseline MRI radiomics features, which provide extensive quantitative data to inform predictive models. The single-sequence models achieved AUC values ranging from 0.79 to 0.87 for predicting LR/DM (54), while multiparametric models exhibited improved predictive performance with AUC values of 0.81–0.83 (56). By capturing prognostic information prior to treatment, baseline MRI radiomics can help identify patients more likely to respond favorably to therapy, enabling the design of targeted treatment strategies. Furthermore, multiparametric imaging captures tumor characteristics more comprehensively than single-sequence methods, offering valuable insights for more accurate prognostic predictions (58).
Tumor heterogeneity in RC and its changes during treatment underscore the importance of post-treatment imaging analysis. Tibermacine et al (59) applied 2D manual segmentation (MS), 3D MS and bounding box methods to extract radiomics features from pre- and post-treatment T2WI following nCRT. Their model demonstrated strong predictive performance for both LR and DM, with AUC values ranging from 0.77 to 0.89 for predicting DFS. Further research by Jalil et al (60) explored the predictive value of textural features in pre- and post-treatment T2WI of patients with LARC treated with nCRT. The study found that both pre- and post-treatment texture features were significant predictors of recurrence-free survival (P<0.01), offering valuable insights for clinical prognostic assessments. These findings emphasize the predictive potential of both pre- and post-treatment imaging. They highlight the importance of comprehensive lesion analysis throughout the course of treatment to accurately assess prognosis and guide clinical decision-making.
Delta radiomics quantifies changes in tumor morphology and heterogeneity by analyzing differences between pre- and post-treatment images. It enables early prediction of tumor behavior, such as nCRT efficacy, LR and DM. As a result, it provides a foundation for personalized treatment strategies for patients (21,61). Jeon et al (62) extracted radiomics features from lesions in pre- and post-treatment T2WI of 101 patients with LARC. By performing phase subtraction, they derived Delta radiomics features and constructed nomograms to predict LR, DM and DFS after nCRT. These nomograms effectively risk-stratified the patients. Similarly, Chiloiro et al (63) developed a model to predict DM using an MRI-based Delta radiomics approach, achieving an accuracy of 80.9%, specificity of 85.7% and sensitivity of 71.4%. This demonstrated that Delta radiomics features are significantly associated with DM. In a subsequent study (64), they also confirmed that Delta radiomics features were valuable in predicting DFS in patients with LARC treated with nCRT using 0.35T MRI-guided radiotherapy. Current evidence demonstrates that delta radiomics captures the spatiotemporal heterogeneity of the tumor treatment response (predictive AUC range: 0.77–0.89), exhibiting unique advantages in clinical translation applications, including dynamic monitoring during nCRT, prognostic stratification and precision therapeutic interventions.
In the management of LARC, several factors beyond radiomic features influence patient prognosis, including tumor indicators, pathological T-stage, N-stage, EMVI and MRF status (34,35). Park et al (65) found that integrating imaging signs with texture features from baseline MRI could effectively predict the efficacy of nCRT and the risk of LR. In particular, their model, which utilized the long-stroke low grey emphasis (GLRLM_LRLGE) texture feature, significantly predicted LR (P=0.039). Texture features are thought to correlate with histological characteristics that reflect tumor heterogeneity (66,67), and increased heterogeneity is often associated with higher malignancy. Therefore, texture features that capture the varying levels of tumor heterogeneity in LARC could enhance the ability to predict individual responses to nCRT and survival outcomes.
Certain studies suggest that combining radiomics features with clinical parameters in a joint model could enhance predictive accuracy. In a multicenter retrospective study involving 629 patients, Liu et al (57) developed a column chart based on MRI-derived radiomics features, which successfully predicted DM-free survival (DMFS). Furthermore, their combined model, which incorporated radiomics features alongside clinicopathological ypT and ypN staging, significantly improved the classification accuracy of DMFS outcomes. A subsequent study (68) confirmed that integrating radiomics features with ypT-stage and ypN-stage in a combined clinicopathology-imaging histology model outperformed the clinicopathology-only model in predicting DMFS. Meng et al (69) constructed a joint model combining pre-treatment enhanced MRI radiomics features with ypN staging and MRF status, achieving enhanced prediction accuracy for DFS, with P-values of 0.001 and 0.005 in the training and validation sets, respectively. Cui et al (55) developed a joint model that integrated multiparametric radiomics features with clinicopathological variables, which outperformed the clinicopathological model alone in predicting DFS, with a concordance index (C-index) of 0.803 and 0.780 in the training and validation sets, respectively. Collectively, these studies demonstrate that combining radiomics features with clinicopathological data can substantially improve the accuracy of prognosis prediction in patients with LARC.
Although radiomics-based imaging methods have shown improved predictive power for assessing nCRT outcomes in patients with LARC, manual lesion outlining for radiomics analysis remains time-consuming and labor-intensive. As a result, DL approaches may offer valuable potential for advancing research in this area.
DL encompasses a broad range of machine learning algorithms based on multi-layer deep neural networks, rather than a specific model. Unlike radiomics, DL automatically learns to extract and select high-dimensional features through neural networks, enhancing the robustness of the model and enabling more comprehensive extraction of image information (70). Convolutional neural networks (CNNs), one of the most commonly used DL models in medical imaging, offer distinct advantages in tasks such as image segmentation and classification. A typical CNN consists of an input layer, a convolutional layer, a pooling layer, a fully connected layer and an output layer, with the convolutional layer serving as the core. CNN directly takes the image as input, extracts features through multiple convolutional and pooling layers, processes the extracted features in the fully connected layer and produces the output based on the specific task at hand.
DL offers several advantages over radiomics: First, DL automatically learns and extracts features from images, eliminating the need for expert-driven manual feature extraction. This reduces bias and increases efficiency. Second, while radiomics features are generally applicable, they often lack specificity. In tumors that tend to undergo significant transformations, such as colorectal cancer, traditional radiomics can be less effective. By contrast, DL can extract more disease-specific features tailored to the particular type or stage of disease. Additionally, DL models exhibit strong adaptability and excellent generalization, allowing them to be adjusted and optimized for different tasks and data types. As a result, they can maintain high performance across diverse datasets and scenarios (71).
Studies using DL have primarily focused on evaluating the efficacy of nCRT in patients with LARC (72,73). Recently, however, researchers have begun exploring its potential for predicting prognosis. In a multicenter retrospective study, Liu et al (74) developed a multiparametric MRI-based DL model to predict DM in patients with LARC undergoing nCRT. The results demonstrated that features extracted from pre-treatment MRI scans performed well in predicting DM, with AUCs of 0.938 and 0.894 in the training and validation sets, respectively. Furthermore, incorporating clinicopathological factors into a joint model enhanced prognostic prediction, outperforming the DL model and clinical model alone (P<0.001).
In a multinational, multicenter study with a large cohort, Jiang et al (75) developed a vision transformer (ViT)-based DL model to predict the prognosis of patients with LARC using pre-treatment T2WI. The ViT model demonstrated strong predictive performance for OS, achieving a C-index of 0.82. This result highlights its potential as a preoperative risk stratification tool. Additionally, the study found that ViT outperformed conventional CNN in predicting OS, with a C-index of 0.82 for ViT compared to 0.67 for CNN. This suggests that ViT can serve as a viable alternative to CNN for imaging analysis in LARC prognosis prediction. In a multicenter retrospective study with a large sample size, Zhang et al (76) developed a 3D CNN model (MuST), an imaging radiomics model based on pre-treatment T2WI, DWI and ADC to predict OS, DMFS and local recurrence-free survival (LRFS), respectively. The comparison revealed that the MuST model significantly outperformed the radiomics model, as evidenced by a higher C-index. This finding suggests that DL offers a more accurate and stable approach for predicting the prognosis of patients with LARC. Current studies indicate that the predictive performance of DL models for key endpoints (OS, DM and LRFS) is consistently stable, with C-index values ranging from 0.82 to 0.94.
These studies highlight that DL models, compared to traditional machine learning models (e.g., RF, logistic regression) and manual radiological assessment, have evolved with deeper algorithmic development and model training, resulting in superior performance in prognostic prediction. This progress offers clinicians the potential for early, non-invasive risk assessment of patients and could eventually replace the labor-intensive process of manual outlining. However, DL-based studies remain limited compared to traditional radiomics. Therefore, further research is necessary to validate model stability and explore their clinical potential.
Comparative analysis demonstrates distinct performance variations between radiomics and DL approaches in prognostic prediction. Radiomic models utilizing pre-T2WI imaging combined with clinical covariates exhibit moderate predictive capability for DM. For instance, the model developed by Wang et al (68) achieves AUCs of 0.83 (training) and 0.74 (validation), with the validation C-index of 0.788 (95% CI: 0.751–0.825). By contrast, the DL model integrating multiparametric MRI (T2WI + DWI) displays superior predictive performance. The DL-based DM prediction framework by Liu et al (74) significantly outperforms radiomic methods, attaining higher AUCs of 0.938 vs. 0.83 (training) and 0.892 vs. 0.74 (validation), alongside elevated C-index values of 0.851 (95% CI: 0.795–0.906) and 0.747 (95% CI: 0.665–0.830), respectively. This performance disparity underscores DL's inherent capacity to decode complex imaging patterns, particularly when synergizing multiparametric data with clinical features.
In predicting the prognosis of nCRT for LARC, radiomics and DL each show potential but differ in accuracy, usability and clinical feasibility. Radiomics demonstrates certain accuracy in predicting DFS, LR and DM. For instance, the radiomics model based on multiparametric MRI achieves an AUC of 0.83 for predicting LR or DM (56) and shows accuracies of 0.803 and 0.780 (55). However, its clinical application faces three major challenges: i) Variations in MRI scanning protocols across centers hinder data standardization and feature comparability; ii) manual ROI delineation is time-consuming and introduces observer-dependent bias, limiting usability; and iii) instability in feature extraction algorithms restricts model generalizability.
By contrast, DL exhibits superior predictive performance. A DL model based on pre-treatment MRI achieves AUC values of 0.938 and 0.894 in the training and validation sets, respectively, for predicting distant metastasis, and the MuST model shows a significantly higher C-index than radiomics models (74,76). The advantages of DL include automatic feature extraction to avoid human bias and adaptive learning to enhance model generalizability. However, its limitations are also evident: Complex network structures are prone to overfitting, reliance on large-scale annotated data and the ‘black-box’ nature that affects the interpretability of results.
Overall, both technologies face challenges related to data heterogeneity, insufficient generalizability and reproducibility. Current studies are mostly based on single-center, small-sample retrospective data and lack external validation. In radiomics workflows, subtle differences in image acquisition, ROI segmentation and feature extraction can lead to variability in results. DL models are significantly influenced by the quality of training data and the parameter optimization process is complex, making complete reproducibility across studies difficult. Future research should focus on constructing multi-center standardized databases, developing more robust feature extraction algorithms and addressing the ‘black-box’ issue of models through explainable technologies to enhance their clinical application value and feasibility.
Radiomics, as an emerging technology, still faces several challenges in clinical application. One major issue is data standardization and quality control. Variations in equipment and inconsistencies in scanning parameters can impact the accuracy of image feature extraction and the generalization of models. Additionally, the stability and reproducibility of radiomics features limit the broader applicability of these models. Another challenge is the manual delineation of lesion ROIs, which is not only time-consuming but also prone to subjective bias, potentially compromising model accuracy. Future advancements in fully or semi-automated segmentation technologies are expected to address this limitation. Currently, most radiomics studies rely on small, single-center datasets. To improve the accuracy and reliability of these models, further validation through large-scale, multicenter, prospective studies is essential.
Likewise, the performance of DL models is heavily contingent upon the availability of extensive, high-quality labeled image datasets. Nonetheless, in the domain of medical imaging, the assembly of such datasets poses considerable challenges, especially when dealing with lesions that exhibit substantial variability. Despite the growing trend towards automated data annotation, the incorporation of manual review and verification processes remains essential to guarantee the precision and reliability of the data. Additionally, DL models, particularly complex neural networks, are often criticized as ‘black boxes’ because their decision-making processes are opaque, hindering their interpretability and trustworthiness in clinical settings (77). To enhance the clinical applicability of these models, it is essential to uncover the causal mechanisms underlying them and improve the transparency and interpretability of their decision-making processes. Optimizing DL algorithms is crucial for enhancing model accuracy, efficiency and interpretability. However, the significant computational resources required for DL models may limit their use in resource-limited settings. Future research should focus on developing more efficient algorithms and exploring methods to effectively integrate domain knowledge, thereby improving the utility and reliability of DL models in medical image analysis.
The clinical implementation of artificial intelligence (AI)-based prognostic models faces critical ethical and technical challenges. First, data bias compromises model fairness, as training datasets with insufficient racial, age or gender representation (e.g., overrepresentation of specific populations) reduce predictive accuracy in minority groups. Second, technical biases from cross-institutional variations in MRI field strengths and scanning protocols introduce systematic feature extraction errors. Finally, the opacity of ‘black-box’ decision mechanisms in DL models undermines clinical credibility, where nonlinear decision pathways lack transparency and conflict with medical traceability requirements.
Current studies demonstrate that multimodal radiomics models, which combine preoperative CT and MRI data, offer substantial advantages in predicting the response to nCRT in patients with LARC, outperforming any single imaging modality (78). However, the fusion of multiple imaging techniques, such as MRI, CT or PET/CT, has not been extensively studied for predicting the prognosis of patients with LARC (79). Future research should explore this area further, aiming to enhance the accuracy and reliability of prognostic predictions by integrating data from various imaging modalities.
Future research should prioritize three key directions to address current limitations. First, a standardized multicenter validation framework should be established through international collaboration, including unified MRI protocols (e.g., field strength, sequence parameters, contrast agent use) and open-access LARC imaging databases. Additionally, prospective clinical trials are needed to evaluate model performance in real-world settings, providing evidence for translational practice. Second, an Explainable AI model should be developed by integrating Class Activation Mapping to visualize decision regions and Shapley Additive Explanations values to quantify imaging feature contributions (80). Third, multimodal omics integration should be pursued by merging radiomics with genomics and molecular biomarkers to comprehensively characterize tumor heterogeneity. For instance, Fathi Kazerooni et al (81) performed a radiogenomics analysis by integrating multiparametric MRI and whole-transcriptome sequencing in pediatric low-grade gliomas, enabling dual prediction of prognosis and therapy based on immune features.
Traditional medical imaging captures the morphological features of LARC from a macroscopic perspective, revealing overall structural and functional changes. By contrast, genetic and pathological analyses provide an in-depth view of the molecular mechanisms and histological alterations at the microscopic level, offering a more detailed understanding of tumor biology (82,83). Imaging genomics and multi-omics techniques bridge the gap between macroscopic imaging features and microscopic molecular events. These methods enable a comprehensive characterization of disease heterogeneity by extracting quantitative features from imaging data and integrating them with genomic, transcriptomic and pathological information. This integration enhances not only disease diagnosis and precise staging but also the prediction of treatment responses and patient prognosis, laying the foundation for personalized treatment strategies.
Looking ahead, improving data quality, fostering multicenter collaborations and applying advanced analytics will be crucial. The ongoing development of automated DL segmentation algorithms and end-to-end modeling approaches has the potential to overcome the limitations of current technologies. Additionally, the integration of multimodal models combining radiomics, pathomics and genomics will likely offer a more holistic view of prognosis, improving predictive accuracy and further advancing the field of precision medicine.
MRI-based radiomics and DL show promising predictive performance in assessing the prognosis of nCRT in LARC. These technologies hold substantial clinical potential as tools for early, non-invasive assessment, aiding clinicians in patient risk stratification and playing a pivotal role in the development of personalized treatment plans.
Not applicable.
This study was supported by the Major Program Co-sponsored by Province and Ministry (grant no. WKJ-ZJ-2210), the Fundamental Research Funds for the Central Universities (grant no. 226-2024-00059) and the ‘Pioneer’ and ‘Leading Goose’ R&D Program of Zhejiang (grant no. 2024C03047).
Not applicable.
JS and QH designed and conducted this review. JS critically revised the final version of the manuscript. YS, TD and JL conceived and drafted the manuscript. YS and TD prepared the table. JS provided funding for the research. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
ADC |
apparent diffusion coefficient |
|
AUC |
area under the curve |
|
CEA |
carcinoembryonic antigen |
|
CE-TIWI |
contrast-enhanced T1-weighted imaging |
|
C-index |
concordance index |
|
CNNs |
convolutional neural networks |
|
CRC |
colorectal cancer |
|
CT |
computed tomography |
|
DFS |
disease-free survival |
|
DL |
deep learning |
|
DM |
distant metastasis |
|
DMFS |
DM-free survival |
|
DWI |
diffusion-weighted imaging |
|
EMVI |
extramural vascular invasion |
|
LARC |
locally advanced rectal cancer |
|
LASSO |
least absolute shrinkage and selection operator |
|
LR |
local recurrence |
|
LRFS |
local recurrence-free survival |
|
LRR |
local recurrence rate |
|
MRF |
mesorectal fascia |
|
MRI |
magnetic resonance imaging |
|
MS |
manual segmentation |
|
MSI |
microsatellite instability |
|
nCRT |
neoadjuvant chemoradiotherapy |
|
OS |
overall survival |
|
PET/CT |
positron emission tomography/computed tomography |
|
PFS |
progression-free survival |
|
PRISMA |
Preferred Reporting Items for Systematic Reviews and Meta-Analyses |
|
RC |
rectal cancer |
|
RF |
random forests |
|
ROI |
regions of interest |
|
SVM |
support vector machines |
|
TNM |
tumor-node-metastasis |
|
TRG |
tumor regression grade |
|
T2WI |
T2-weighted imaging |
|
ViT |
vision transformer |
|
Siegel RL, Wagle NS, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2023. CA Cancer J Clin. 73:233–254. 2023.PubMed/NCBI | |
|
Morgan E, Arnold M, Gini A, Lorenzoni V, Cabasag CJ, Laversanne M, Vignat J, Ferlay J, Murphy N and Bray F: Global burden of colorectal cancer in 2020 and 2040: Incidence and mortality estimates from GLOBOCAN. Gut. 72:338–344. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Staal FCR, van der Reijd DJ, Taghavi M, Lambregts DMJ, Beets-Tan RGH and Maas M: Radiomics for the prediction of treatment outcome and survival in patients with colorectal cancer: A systematic review. Clin Colorectal Cancer. 20:52–71. 2021.PubMed/NCBI | |
|
Aklilu M and Eng C: The current landscape of locally advanced rectal cancer. Nat Rev Clin Oncol. 8:649–659. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Garrido-Laguna I, et al: NCCN guidelines insights: Rectal cancer, version 6.2020. J Natl Compr Canc Netw. 18:806–815. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
van der Valk MJM, Hilling DE, Bastiaannet E, Kranenbarg EMK, Beets GL, Figueiredo NL, Habr-Gama A, Perez RO, Renehan AG and van de Velde CJH; IWWD Consortium, : Long-term outcomes of clinical complete responders after neoadjuvant treatment for rectal cancer in the International watch & wait database (IWWD): An international multicentre registry study. Lancet. 391:2537–2545. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Gauci C, Ravindran P, Pillinger S and Lynch AC: Robotic surgery for multi-visceral resection in locally advanced colorectal cancer: Techniques, benefits and future directions. Laparoscopic, Endoscopic and Robotic Surgery. 6:123–126. 2023. View Article : Google Scholar | |
|
Valentini V, van Stiphout RG, Lammering G, Gambacorta MA, Barba MC, Bebenek M, Bonnetain F, Bosset JF, Bujko K, Cionini L, et al: Selection of appropriate end-points (pCR vs 2yDFS) for tailoring treatments with prediction models in locally advanced rectal cancer. Radiother Oncol. 114:302–309. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Fokas E, Liersch T, Fietkau R, Hohenberger W, Beissbarth T, Hess C, Becker H, Ghadimi M, Mrak K, Merkel S, et al: Tumor regression grading after preoperative chemoradiotherapy for locally advanced rectal carcinoma revisited: Updated results of the CAO/ARO/AIO-94 trial. J Clin Oncol. 32:1554–1562. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, Putter H, Wiggers T, Rutten HJ, Påhlman L, Glimelius B and van de Velde CJ; Dutch Colorectal Cancer Group, : Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol. 12:575–582. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Glynne-Jones R, Wyrwicz L, Tiret E, Brown G, Rödel C, Cervantes A and Arnold D; ESMO Guidelines Committee, : Rectal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 28:iv22–iv40. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Cabezón-Gutiérrez L, Custodio-Cabello S, Palka-Kotlowska M, Díaz-Pérez D, Mateos-Dominguez M and Galindo-Jara P: Neoadjuvant immunotherapy for dMMR/MSI-H locally advanced rectal cancer: The future new standard approach? Eur J Surg Oncol. 49:323–328. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, El Dika IH, Segal N, Shcherba M, Sugarman R, et al: PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N Engl J Med. 386:2363–2376. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang R, Wu T, Yu J, Cai X, Li G, Li X, Huang W, Zhang Y, Wang Y, Yang X, et al: Locally advanced rectal cancer with dMMR/MSI-H may be excused from surgery after neoadjuvant anti-PD-1 monotherapy: A multiple-center, cohort study. Front Immunol. 14:11822992023. View Article : Google Scholar : PubMed/NCBI | |
|
Bahadoer RR, Dijkstra EA, van Etten B, Marijnen CAM, Putter H, Kranenbarg EM, Roodvoets AGH, Nagtegaal ID, Beets-Tan RGH, Blomqvist LK, et al: Short-course radiotherapy followed by chemotherapy before total mesorectal excision (TME) versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer (RAPIDO): A randomised, open-label, phase 3 trial. Lancet Oncol. 22:29–42. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Conroy T, Bosset JF, Etienne PL, Rio E, François É, Mesgouez-Nebout N, Vendrely V, Artignan X, Bouché O, Gargot D, et al: Neoadjuvant chemotherapy with FOLFIRINOX and preoperative chemoradiotherapy for patients with locally advanced rectal cancer (UNICANCER-PRODIGE 23): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 22:702–715. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zwart WH, Temmink SJD, Hospers GAP, Marijnen CAM, Putter H, Nagtegaal ID, Blomqvist L, Kranenbarg EM, Roodvoets AGH, Martling A, et al: Oncological outcomes after a pathological complete response following total neoadjuvant therapy or chemoradiotherapy for high-risk locally advanced rectal cancer in the RAPIDO trial. Eur J Cancer. 204:1140442024. View Article : Google Scholar : PubMed/NCBI | |
|
Conroy T, Castan F, Etienne PL, Rio E, Mesgouez-Nebout N, Evesque L, Vendrely V, Artignan X, Bouché O, Gargot D, et al: Total neoadjuvant therapy with mFOLFIRINOX versus preoperative chemoradiotherapy in patients with locally advanced rectal cancer: Long-term results of the UNICANCER-PRODIGE 23 trial. Ann Oncol. 35:873–881. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Dijkstra EA, Nilsson PJ, Hospers GAP, Bahadoer RR, Kranenbarg EMK, Roodvoets AGH, Putter H, Berglund Å, Cervantes A, Crolla R, et al: Locoregional failure during and after short-course radiotherapy followed by chemotherapy and surgery compared with long-course chemoradiotherapy and surgery: A 5-year follow-up of the RAPIDO trial. Ann Surg. 278:e766–e772. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Valentini V, van Stiphout RG, Lammering G, Gambacorta MA, Barba MC, Bebenek M, Bonnetain F, Bosset JF, Bujko K, Cionini L, et al: Nomograms for predicting local recurrence, distant metastases, and overall survival for patients with locally advanced rectal cancer on the basis of European randomized clinical trials. J Clin Oncol. 29:3163–3172. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Lin H, Lu X, Huang Y, Xu Z, Huang S, Wang X and Chi P: A nomogram to predict distant metastasis after neoadjuvant chemoradiotherapy and radical surgery in patients with locally advanced rectal cancer. J Surg Oncol. 115:462–469. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Merkel S, Weber K, Schellerer V, Göhl J, Fietkau R, Agaimy A, Hohenberger W and Hermanek P: Prognostic subdivision of ypT3 rectal tumours according to extension beyond the muscularis propria. Br J Surg. 101:566–572. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Park IJ, You YN, Agarwal A, Skibber JM, Rodriguez-Bigas MA, Eng C, Feig BW, Das P, Krishnan S, Crane CH, et al: Neoadjuvant treatment response as an early response indicator for patients with rectal cancer. J Clin Oncol. 30:1770–1776. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Bera K, Katz I and Madabhushi A: Reimagining T staging through artificial intelligence and machine learning image processing approaches in digital pathology. JCO Clin Cancer Inform. 4:1039–1050. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Patel UB, Taylor F, Blomqvist L, George C, Evans H, Tekkis P, Quirke P, Sebag-Montefiore D, Moran B, Heald R, et al: Magnetic resonance imaging-detected tumor response for locally advanced rectal cancer predicts survival outcomes: MERCURY experience. J Clin Oncol. 29:3753–3760. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
De Mattia E, Polesel J, Mezzalira S, Palazzari E, Pollesel S, Toffoli G and Cecchin E: Predictive and prognostic value of oncogene mutations and microsatellite instability in locally-advanced rectal cancer treated with neoadjuvant radiation-based therapy: A systematic review and meta-analysis. Cancers (Basel). 15:14692023. View Article : Google Scholar : PubMed/NCBI | |
|
Watanabe T, Kobunai T, Yamamoto Y, Matsuda K, Ishihara S, Nozawa K, Iinuma H, Shibuya H and Eshima K: Heterogeneity of KRAS status may explain the subset of discordant KRAS status between primary and metastatic colorectal cancer. Dis Colon Rectum. 54:1170–1178. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ciocan RA, Ciocan A, Mihăileanu FV, Ursu CP, Ursu Ș, Bodea C, Cordoș AA, Chiș BA, Al Hajjar N, Dîrzu N and Dîrzu DS: Metabolic signatures: Pioneering the frontier of rectal cancer diagnosis and response to neoadjuvant treatment with biomarkers-a systematic review. Int J Mol Sci. 25:23812024. View Article : Google Scholar : PubMed/NCBI | |
|
Raman SP, Chen Y and Fishman EK: Evolution of imaging in rectal cancer: Multimodality imaging with MDCT, MRI, and PET. J Gastrointest Oncol. 6:172–184. 2015.PubMed/NCBI | |
|
Kinkel K, Lu Y, Both M, Warren RS and Thoeni RF: Detection of hepatic metastases from cancers of the gastrointestinal tract by using noninvasive imaging methods (US, CT, MR imaging, PET): A meta-analysis. Radiology. 224:748–756. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Horvat N, Rocha CC, Oliveira BC, Petkovska I and Gollub MJ: MRI of rectal cancer: Tumor staging, imaging techniques, and management. Radiographics. 39:367–387. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Nahas SC, Nahas CS, Marques CF, Ribeiro U Jr, Cotti GC, Imperiale AR, Capareli FC, Chen AT, Hoff PM and Cecconello I: Pathologic complete response in rectal cancer: Can we detect it? Lessons learned from a proposed randomized trial of watch-and-wait treatment of rectal cancer. Dis Colon Rectum. 59:255–263. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Smith JJ and Garcia-Aguilar J: Advances and challenges in treatment of locally advanced rectal cancer. J Clin Oncol. 33:1797–1808. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Park H: Predictive factors for early distant metastasis after neoadjuvant chemoradiotherapy in locally advanced rectal cancer. World J Gastrointest Oncol. 13:252–264. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Joo JI, Lim SW and Oh BY: Prognostic impact of carcinoembryonic antigen levels in rectal cancer patients who had received neoadjuvant chemoradiotherapy. Ann Coloproctol. 37:179–185. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Smith N and Brown G: Preoperative staging of rectal cancer. Acta Oncol. 47:20–31. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Smith NJ, Barbachano Y, Norman AR, Swift RI, Abulafi AM and Brown G: Prognostic significance of magnetic resonance imaging-detected extramural vascular invasion in rectal cancer. Br J Surg. 95:229–236. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Nougaret S, Reinhold C, Mikhael HW, Rouanet P, Bibeau F and Brown G: The use of MR imaging in treatment planning for patients with rectal carcinoma: have you checked the ‘DISTANCE’? Radiology. 268:330–344. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nougaret S, Gormly K, Lambregts DMJ, Reinhold C, Goh V, Korngold E, Denost Q and Brown G: MRI of the rectum: A decade into DISTANCE, moving to DISTANCED. Radiology. 314:e2328382025. View Article : Google Scholar : PubMed/NCBI | |
|
Hosny A, Parmar C, Quackenbush J, Schwartz LH and Aerts H: Artificial intelligence in radiology. Nat Rev Cancer. 18:500–510. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, et al: Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin. 69:127–157. 2019.PubMed/NCBI | |
|
Wang J, Shen L, Zhong H, Zhou Z, Hu P, Gan J, Luo R, Hu W and Zhang Z: Radiomics features on radiotherapy treatment planning CT can predict patient survival in locally advanced rectal cancer patients. Sci Rep. 9:153462019. View Article : Google Scholar : PubMed/NCBI | |
|
Bundschuh RA, Dinges J, Neumann L, Seyfried M, Zsótér N, Papp L, Rosenberg R, Becker K, Astner ST, Henninger M, et al: Textural parameters of tumor heterogeneity in 18F-FDG PET/CT for therapy response assessment and prognosis in patients with locally advanced rectal cancer. J Nucl Med. 55:891–897. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Bang JI, Ha S, Kang SB, Lee KW, Lee HS, Kim JS, Oh HK, Lee HY and Kim SE: Prediction of neoadjuvant radiation chemotherapy response and survival using pretreatment [(18)F]FDG PET/CT scans in locally advanced rectal cancer. Eur J Nucl Med Mol Imaging. 43:422–431. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 372:n712021. View Article : Google Scholar : PubMed/NCBI | |
|
Lambin P, Rios-Velazquez E, Leijenaar R, Carvalho S, van Stiphout RG, Granton P, Zegers CM, Gillies R, Boellard R, Dekker A and Aerts HJ: Radiomics: Extracting more information from medical images using advanced feature analysis. Eur J Cancer. 48:441–446. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Scapicchio C, Gabelloni M, Barucci A, Cioni D, Saba L and Neri E: A deep look into radiomics. Radiol Med. 126:1296–1311. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Floca R, Bohn J, Haux C, Wiestler B, Zöllner FG, Reinke A, Weiß J, Nolden M, Albert S, Persigehl T, et al: Radiomics workflow definition & challenges-German priority program 2177 consensus statement on clinically applied radiomics. Insights Imaging. 15:1242024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin M, Tang X, Cao L, Liao Y, Zhang Y and Zhou J: Using ultrasound radiomics analysis to diagnose cervical lymph node metastasis in patients with nasopharyngeal carcinoma. Eur Radiol. 33:774–783. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ghosh A, Yekeler E, Teixeira SR, Dalal D and States L: Role of MRI radiomics for the prediction of MYCN amplification in neuroblastomas. Eur Radiol. 33:6726–6735. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xie F, Zhao Q, Li S, Wu S, Li J, Li H, Chen S, Jiang W, Dong A, Wu L, et al: Establishment and validation of novel MRI radiomic feature-based prognostic models to predict progression-free survival in locally advanced rectal cancer. Front Oncol. 12:9012872022. View Article : Google Scholar : PubMed/NCBI | |
|
Chiloiro G, Cusumano D, Romano A, Boldrini L, Nicolì G, Votta C, Tran HE, Barbaro B, Carano D, Valentini V, et al: Delta radiomic analysis of mesorectum to predict treatment response and prognosis in locally advanced rectal cancer. Cancers (Basel). 15:30822023. View Article : Google Scholar : PubMed/NCBI | |
|
Steyerberg EW and Harrell FE Jr: Prediction models need appropriate internal, internal-external, and external validation. J Clin Epidemiol. 69:245–247. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Jayaprakasam VS, Paroder V, Gibbs P, Bajwa R, Gangai N, Sosa RE, Petkovska I, Pernicka JS, Fuqua JL III, Bates DDB, et al: MRI radiomics features of mesorectal fat can predict response to neoadjuvant chemoradiation therapy and tumor recurrence in patients with locally advanced rectal cancer. Eur Radiol. 32:971–980. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cui Y, Yang W, Ren J, Li D, Du X, Zhang J and Yang X: Prognostic value of multiparametric MRI-based radiomics model: Potential role for chemotherapeutic benefits in locally advanced rectal cancer. Radiother Oncol. 154:161–169. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang H, Han L, Guo J, Zhang Y, Lin S, Chen S, Lin X, Cheng C, Guo Z and Qiu Y: Pretreatment MRI-based radiomics for prediction of rectal cancer outcome: A discovery and validation study. Acad Radiol. 31:1878–1888. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Meng X, Zhang H, Li Z, Liu J, Sun K, Meng Y, Dai W, Xie P, Ding Y, et al: Predicting distant metastasis and chemotherapy benefit in locally advanced rectal cancer. Nat Commun. 11:43082020. View Article : Google Scholar : PubMed/NCBI | |
|
Wei Q, Chen L, Hou X, Lin Y, Xie R, Yu X, Zhang H, Wen Z, Wu Y, Liu X and Chen W: Multiparametric MRI-based radiomic model for predicting lymph node metastasis after neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Insights Imaging. 15:1632024. View Article : Google Scholar : PubMed/NCBI | |
|
Tibermacine H, Rouanet P, Sbarra M, Forghani R, Reinhold C and Nougaret S; GRECCAR Study Group, : Radiomics modelling in rectal cancer to predict disease-free survival: Evaluation of different approaches. Br J Surg. 108:1243–1250. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jalil O, Afaq A, Ganeshan B, Patel UB, Boone D, Endozo R, Groves A, Sizer B and Arulampalam T: Magnetic resonance based texture parameters as potential imaging biomarkers for predicting long-term survival in locally advanced rectal cancer treated by chemoradiotherapy. Colorectal Dis. 19:349–362. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dinapoli N, Barbaro B, Gatta R, Chiloiro G, Casà C, Masciocchi C, Damiani A, Boldrini L, Gambacorta MA, Dezio M, et al: Magnetic resonance, vendor-independent, intensity histogram analysis predicting pathologic complete response after radiochemotherapy of rectal cancer. Int J Radiat Oncol Biol Phys. 102:765–774. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon SH, Song C, Chie EK, Kim B, Kim YH, Chang W, Lee YJ, Chung JH, Chung JB, Lee KW, et al: Delta-radiomics signature predicts treatment outcomes after preoperative chemoradiotherapy and surgery in rectal cancer. Radiat Oncol. 14:432019. View Article : Google Scholar : PubMed/NCBI | |
|
Chiloiro G, Rodriguez-Carnero P, Lenkowicz J, Casà C, Masciocchi C, Boldrini L, Cusumano D, Dinapoli N, Meldolesi E, Carano D, et al: Delta radiomics can predict distant metastasis in locally advanced rectal cancer: The challenge to personalize the cure. Front Oncol. 10:5950122020. View Article : Google Scholar : PubMed/NCBI | |
|
Chiloiro G, Boldrini L, Preziosi F, Cusumano D, Yadav P, Romano A, Placidi L, Lenkowicz J, Dinapoli N, Bassetti MF, et al: A predictive model of 2yDFS during MR-Guided RT Neoadjuvant chemoradiotherapy in locally advanced rectal cancer patients. Front Oncol. 12:8317122022. View Article : Google Scholar : PubMed/NCBI | |
|
Park H, Kim KA, Jung JH, Rhie J and Choi SY: MRI features and texture analysis for the early prediction of therapeutic response to neoadjuvant chemoradiotherapy and tumor recurrence of locally advanced rectal cancer. Eur Radiol. 30:4201–4211. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Miles KA, Ganeshan B and Hayball MP: CT texture analysis using the filtration-histogram method: What do the measurements mean? Cancer Imaging. 13:400–406. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Hocquelet A, Auriac T, Perier C, Dromain C, Meyer M, Pinaquy JB, Denys A, Trillaud H, Senneville BD and Vendrely V: Pre-treatment magnetic resonance-based texture features as potential imaging biomarkers for predicting event free survival in anal cancer treated by chemoradiotherapy. Eur Radiol. 28:2801–2811. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Chen J, Zheng N, Zheng K, Zhou L, Zhang Q and Zhang W: Predicting the risk of distant metastasis in patients with locally advanced rectal cancer using model based on pre-treatment T2WI-based radiomic features plus postoperative pathological stage. Front Oncol. 13:11095882023. View Article : Google Scholar : PubMed/NCBI | |
|
Meng Y, Zhang Y, Dong D, Li C, Liang X, Zhang C, Wan L, Zhao X, Xu K, Zhou C, et al: Novel radiomic signature as a prognostic biomarker for locally advanced rectal cancer. J Magn Reson Imaging. 13:doi: 10.1002/jmri.25968. 2018. | |
|
Trebeschi S, van Griethuysen JJM, Lambregts DMJ, Lahaye MJ, Parmar C, Bakers FCH, Peters N, Beets-Tan RGH and Aerts H: Deep learning for fully-automated localization and segmentation of rectal cancer on multiparametric MR. Sci Rep. 7:53012017. View Article : Google Scholar : PubMed/NCBI | |
|
Shen D, Wu G and Suk HI: Deep learning in medical image analysis. Annu Rev Biomed Eng. 19:221–248. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shin J, Seo N, Baek SE, Son NH, Lim JS, Kim NK, Koom WS and Kim S: MRI radiomics model predicts pathologic complete response of rectal cancer following chemoradiotherapy. Radiology. 303:351–358. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Zhang XY, Shi YJ, Wang L, Zhu HT, Tang Z, Wang S, Li XT, Tian J and Sun YS: Radiomics analysis for evaluation of pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Clin Cancer Res. 23:7253–7262. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Zhang D, Liu Z, Li Z, Xie P, Sun K, Wei W, Dai W, Tang Z, Ding Y, et al: Deep learning radiomics-based prediction of distant metastasis in patients with locally advanced rectal cancer after neoadjuvant chemoradiotherapy: A multicentre study. EBioMedicine. 69:1034422021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Zhao H, Saldanha OL, Nebelung S, Kuhl C, Amygdalos I, Lang SA, Wu X, Meng X, Truhn D, et al: An mri deep learning model predicts outcome in rectal cancer. Radiology. 307:e2222232023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang S, Cai G, Xie P, Sun C, Li B, Dai W, Liu X, Qiu Q, Du Y, Li Z, et al: Improving prognosis and assessing adjuvant chemotherapy benefit in locally advanced rectal cancer with deep learning for MRI: A retrospective, multi-cohort study. Radiother Oncol. 188:1098992023. View Article : Google Scholar : PubMed/NCBI | |
|
Najjar R: Redefining radiology: A review of artificial intelligence integration in medical imaging. Diagnostics (Basel). 13:27602023. View Article : Google Scholar : PubMed/NCBI | |
|
Li ZY, Wang XD, Li M, Liu XJ, Ye Z, Song B, Yuan F, Yuan Y, Xia CC, Zhang X and Li Q: Multi-modal radiomics model to predict treatment response to neoadjuvant chemotherapy for locally advanced rectal cancer. World J Gastroenterol. 26:2388–2402. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Shahzadi I, Zwanenburg A, Lattermann A, Linge A, Baldus C, Peeken JC, Combs SE, Diefenhardt M, Rödel C, Kirste S, et al: Analysis of MRI and CT-based radiomics features for personalized treatment in locally advanced rectal cancer and external validation of published radiomics models. Sci Rep. 12:101922022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou B, Khosla A, Lapedriza A, Oliva A and Torralba A: Learning deep features for discriminative localization. Computer Vision and Pattern Recognition (cs.CV). 2015.https://doi.org/10.48550/arXiv.1512.04150 | |
|
Kazerooni AF, Kraya A, Rathi KS, Kim MC, Vossough A, Khalili N, Familiar AM, Gandhi D, Khalili N, Kesherwani V, et al: Multiparametric MRI along with machine learning predicts prognosis and treatment response in pediatric low-grade glioma. Nat Commun. 16:3402025. View Article : Google Scholar | |
|
Bando H, Ohtsu A and Yoshino T: Therapeutic landscape and future direction of metastatic colorectal cancer. Nat Rev Gastroenterol Hepatol. 20:306–322. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dayde D, Tanaka I, Jain R, Tai MC and Taguchi A: Predictive and prognostic molecular biomarkers for response to neoadjuvant chemoradiation in rectal cancer. Int J Mol Sci. 18:5732017. View Article : Google Scholar : PubMed/NCBI |