Next Article in Journal
Understanding the Clinical Significance of MUC5AC in Biliary Tract Cancers
Next Article in Special Issue
Development and Validation of a Nomogram to Predict Overall Survival in Stage I–III Colorectal Cancer Patients after Radical Resection with Normal Preoperative Serum Carcinoembryonic Antigen
Previous Article in Journal
Locus-Specific Bisulfate NGS Sequencing of GSTP1, RNF219, and KIAA1539 Genes in the Total Pool of Cell-Free and Cell-Surface-Bound DNA in Prostate Cancer: A Novel Approach for Prostate Cancer Diagnostics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiomics Approaches for the Prediction of Pathological Complete Response after Neoadjuvant Treatment in Locally Advanced Rectal Cancer: Ready for Prime Time?

1
Radiation Oncology Department, University Hospital, 29200 Brest, France
2
LaTIM, INSERM, UMR 1101, University of Western Brittany, 29238 Brest, France
3
Medical Oncology Department, University Hospital, 29200 Brest, France
4
Digestive Surgery Department, University Hospital, 29200 Brest, France
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(2), 432; https://doi.org/10.3390/cancers15020432
Submission received: 29 November 2022 / Revised: 3 January 2023 / Accepted: 5 January 2023 / Published: 9 January 2023
(This article belongs to the Special Issue Advances in Colorectal Cancer Survival Analysis)

Abstract

:

Simple Summary

Therapeutic management of locally advanced rectal cancer has seen profound modifications in the latest years, mainly with the advent of total neoadjuvant therapy. Adding neoadjuvant chemotherapy to neoadjuvant chemoradiotherapy leads to an improvement in locoregional disease-free survival. Organ preservation rises as an option for patients with complete response after neoadjuvant therapy without compromising overall survival. A current challenge that remains is the ever-growing need for selection tools to enable a personalized therapeutic approach for each patient. In this article, we review the place of different biomarkers (clinical, biological, genomics, transcriptomics, proteomics, and radiomics) as well as their clinical implementation and discuss the most recent trends for future steps in prediction modeling in patients with locally advanced rectal cancer.

Abstract

In recent years, neoadjuvant therapy of locally advanced rectal cancer has seen tremendous modifications. Adding neoadjuvant chemotherapy before or after chemoradiotherapy significantly increases loco-regional disease-free survival, negative surgical margin rates, and complete response rates. The higher complete rate is particularly clinically meaningful given the possibility of organ preservation in this specific sub-population, without compromising overall survival. However, all locally advanced rectal cancer most likely does not benefit from total neoadjuvant therapy (TNT), but experiences higher toxicity rates. Diagnosis of complete response after neoadjuvant therapy is a real challenge, with a risk of false negatives and possible under-treatment. These new therapeutic approaches thus raise the need for better selection tools, enabling a personalized therapeutic approach for each patient. These tools mostly focus on the prediction of the pathological complete response given the clinical impact. In this article, we review the place of different biomarkers (clinical, biological, genomics, transcriptomics, proteomics, and radiomics) as well as their clinical implementation and discuss the most recent trends for future steps in prediction modeling in patients with locally advanced rectal cancer.

1. Introduction

Colorectal cancer is the third most frequent cancer, with approximately two million cases per year, and it represents the second leading cause of death by cancer [1]. Rectal cancer accounts for a third of all colorectal cancers. Therapeutic management of rectal cancer is based on the tumor level as well as the tumor staging. Locally advanced rectal cancer (LARC) is defined by a T3-T4 tumor and/or a nodal involvement, with no metastatic sites. In recent years, therapeutic management of LARCs has seen tremendous modifications, especially with the advent of total neoadjuvant therapy (TNT) and non-operative management (NOM). Adding neoadjuvant chemoradiotherapy (CRT) with a total dose of 50–56 Gy to the tumor before surgery was associated with increased loco-regional disease-free survival (LRDFS), higher pathological complete response rates (pCR), and higher negative surgical margin rates [2,3,4,5]. Neoadjuvant CRT did not improve metastasis-free survival (MFS) or overall survival (OS). Short course radiotherapy (SCRT) delivered in 25 Gy/5 fractions over a week is also associated with lower local recurrences [6,7,8]. Neoadjuvant CRT alone was thus the standard of care for all LARCs before being challenged by TNT. Indeed, the addition of neoadjuvant chemotherapy to neoadjuvant CRT was associated with improved LRDFS and pCR rates [9,10]. TNT did not impact negative surgical margins rates or OS [11,12,13,14].
For all LARCs, surgery has long been the cornerstone. Reaching pCR rates as high as 25% in group B of the CARO/ARO/AIO-12 trial [15] (consolidative CT after CRT) and 28% in the RAPIDO and PRODIGE-23 trials, total neoadjuvant therapy, compared to 12–14% rates with neoadjuvant CRT alone, is particularly interesting in the management of LARCs. Rates of 40–60% were even described. In patients presenting with pCR, careful monitoring and NOM was safe, with no impact on survival endpoints, and nearly half of the patients did not undergo surgery. Interestingly, if proposed, 83% of patients would opt for a NOM strategy [16] even if warned of potential worse outcomes in the case of local regrowth, as further discussed.
These pCR improvements come with the price of nonnegligible toxicity rates. In the PRODIGE-23 trial, for instance, grade 3–4 adverse events (AEs) occurred in 46% of patients during the neoadjuvant chemotherapy (CT) phase. Selection of patients is thus of paramount importance, with several treatment choices to be made between CRT alone, TNT with short or long CRT, induction or consolidative CT, and total mesorectal excision or NOM.
Assessment and prediction of pCR as well as other clinically meaningful endpoints have been extensively researched, with most studies focusing on the prediction of pCR. Despite the interesting results, none of the models are recommended in current guidelines. In this review, we review the place of different biomarkers (clinical, biological, genomics, transcriptomics, proteomics, and radiomics) as well as their clinical implementation and discuss the most recent trends for future steps in prediction modeling in patients with locally advanced rectal cancer.

2. Role of Clinical and Pathological Complete Response

Survival outcomes are significantly correlated with response to neoadjuvant CRT. In a cohort of 1089 patients [17], pCR patients harbored a significantly longer 5-year overall survival, 5-year disease-free survival, and 5-year distant recurrence-free survival. Pooled together, patients with a pCR and a near pCR response experienced an increased 5-year local recurrence-free survival.
Pooling data from 27 articles (17 different datasets), pCR was found in 484/3105 included patients (15.6%) [18]. Five-year disease-free survival was found in 83.3% (78.8–87.0%) of patients with pCR compared to 65.6% (63.6–68.0%) of non-pCR patients, resulting in an adjusted HR of 0.45. The role of pCR was also confirmed for local recurrence-free survival (HR 0.33 CI95% 0.19–0.60), distant metastasis-free survival (HR 0.40 CI95% 0.29–0.55), and overall survival (HR 0.51 CI95% 0.38–0.67). Of note, 8/17 studies were retrospective cohort studies. None were in the TNT setting.
The positive impact of pCR on survival outcomes leads to the development of non-surgical management, the main challenge then being the correlation between clinical complete response and pathological complete response.
Digital rectal examination (DRE), endoscopy, and magnetic resonance imaging (MRI) are the foundation for the local evaluation of tumor response following neoadjuvant therapy (NAT). The combined accuracy of all three modalities in predicting tumor absence is reported to be 98% [19]. A smooth, normal mucosa should be seen on DRE, though some slight abnormalities or a soft scar may be palpable. A flat white scar, telangiectasia, and the lack of an ulcer or nodule are the endoscopic features of cCR. In addition, there are no apparent lymph nodes or restricted diffusion [20].
One of the first reports of the watch-and-wait approach was made in 2011 by Maas et al. Within a cohort of 21 patients and a mean follow-up of 25 +/− 19 months, only one patient developed a local recurrence [21]. Similar reports were later published confirming the feasibility of the NOM. In a phase II randomized trial, 324 patients were compared with regard to the treatment sequence as well as survival endpoints with regard to the treatment strategy (watch-and-wait or surgery). No differences were found between groups in local recurrence-free survival, distant metastasis-free survival, and overall survival [22].
In a retrospective cohort of 113 patients, 22 patients experienced a local regrowth and were treated by salvage surgery. To be noted, a worse overall survival along with worse distant metastasis-free survival was noted in patients with local regrowth vs. those without local regrowth [23], confirming the absolute need for robust pCR prediction models to better personalize the treatments. Given the low rate of pCR, each model should be developed aiming for the highest balance between negative predictive value (NPV) and positive predictive value (PPV). The model should at first limit the risk of local relapse before opening the possibility of NOM. Each model should also be cost-effective, reproducible, and easily accessible. Given the place of MRI and other medical imaging modalities before CRT and the absence of additional costs, the radiomics model will be treated separately.

3. Prediction of pCR Using Non-Radiomics Model

Several approaches were proposed for the prediction of pCR and near-pathological complete response (nCR). In a systematic review summarizing 85 studies [24], several predictors (clinicopathological features, radiomics, gene expression, mutational and protein expression analyses) were associated with pCR. However, none appeared as a clear and robust biomarker. Indeed, only a fraction of the proposed models have been externally validated, and none have been incorporated into international guidelines. The advent of TNT, the development of NOM, and the recent advances in prediction modeling call for a new overview on this matter.

3.1. Clinical Predictors and Models

The vast majority of the published articles are based on either retrospective cohorts or secondary analysis of prospective trials. Prediction of pCR is often the main endpoint, clinical complete response (cCR) being used as a surrogate.
Assessment of cCR relies on DRE, post-CRT MRI, and endoscopy. In a recent meta-analysis, sensitivities/specificities of MRI, endorectal ultrasonography, and computed tomography (CT) were as high as 95%/31%, 97%/30%, and 96%/21%, respectively [25].
The role of endoscopy and especially post-CRT biopsy for the prediction of pCR was evaluated in a cohort of 198 patients, among which 186 underwent surgery [26]. Two features appeared as strong predictors of pCR: flattening of marginal swelling and negative post-CRT biopsies. Cancer-negative biopsy achieved the highest specificity (along with a low carcinoembryonic antigen), reaching 78.3%. However, sensitivity was the second lowest (65.0%). Consecutively, residual cancer was found in 76.0% of cases negative for cancer by endoscopic biopsy, proving the difficulty of pCR assessment.
Several clinical features are commonly accepted to increase the likelihood of pCR: low carcinoembryonic antigen (CEA) [27,28,29,30], small tumor size [31,32], low tumor/nodal stage [33], low histologic grade [33], small circumferential tumor extent [34], high hemoglobin levels [27,34], and a low neutrophil-to-lymphocyte ratio [35].
Using a propensity-matched cohort of 322 patients (161 in each group: cCR vs. no cCR), Mbanu et al. identified pre-treatment tumor diameter, tumor stage on magnetic resonance imaging (MRI), hemoglobin, alkaline phosphate, total radiotherapy depths, neutrophil-to-lymphocyte ratio (NLR), neutrophil-monocyte-to-lymphocyte ratio (NMLR), lymphocyte count, and albumin level as significant predictors of cCR [36]. Combining these parameters led to a nomogram with an area under the curve (AUC) of 0.75. Though the correlation between the fitted probability of cCR and observed cCR appeared to be interesting, no probability threshold was proposed.
In a wider cohort of 514 patients treated between 2004 and 2019 with mainly three-dimensional (3D) RT and two-dimensional (2D) RT with a total dose of 45 Gy to the whole pelvis and a tumor boost, longitudinal tumor diameter, extramural tumor invasion depth, CEA, hemoglobin levels, age, and interval between CRT and surgery were combined in a nomogram. A ROC of 0.72 (95%CI 0.68–0.77) was obtained for the prediction of good response defined by a Dworak tumor regression grade of 3 or 4 [37].
Among the identified clinical predictors, the delay between chemoradiotherapy and surgery appears to be the only one to be modifiable. The only prospective randomized trial to address the matter of timing remains the Lyon R90-01, in which pCR rates were analyzed with a 6–8-week interval compared with a shorter interval of 2 weeks [38]. To our knowledge, such a study was not conducted on the TNT era.
In one of the largest single-institution cohorts to date (1089 patients), 198 patients (18.2%) experienced a pCR [17]. No significant differences between the pCR and non-pCR patients were found regarding age, sex, vascular invasion, or CEA level. Tumor size, clinical node negativity, and differentiation were the only significant predictors. Of note, the delay between CRT and surgery was not available.
Despite the relative heterogeneity between these studies, several clinical features stood out. Tumor stage, the interval between CRT and surgery, nodal stage, histological differentiation, as well as biological parameters (CEA and hemoglobin) appeared to be consensual between most studies. Post-CRT negative biopsies reached a high specificity but remained more invasive.

3.2. Role of MRI and PET/CT

The size of the tumor and the nodes as well as tumor regression grade defined by qualitative assessment of change in signal intensity based on MRI are routinely used for pre-CRT tumor staging and for the assessment of response to therapy [39,40,41]. Where pre-CRT tumor volume does not seem effective for the prediction of pCR in most studies, post-CRT tumor evaluation was confirmed in several studies [31,42,43,44,45,46]. In a cohort of 64 patients, Sathyakumar et al. identified a complete response (ydwiT0) on post-CRT diffusion-weighted imaging (DWI) tumor volume with a reduction rate of >95% as the best predictor of pCR, with AUC values of 0.88 (CI 95% 0.74–1) and 0.84 (CI 95% 0.7–0.98), respectively [47]. Agreement for ydwiT0 was scored as moderate to high among three observers, with kappa values ranging from 0.79 to 0.90. The value of tumor shrinkage was also confirmed by other teams [44,48], highlighting the value of pre-/post-CRT evaluation and comparison. Regarding the choice of sequences to focus on, though the T2 morphological sequence has the advantage of availability and reproducibility, DWI and apparent diffusion coefficient (ADC) maps could help in differentiating residual tumor from fibrosis, but with conflicting results depending on the studies [49,50,51,52,53,54]. Similar results were obtained regarding dynamic contrast-enhanced imaging (DCE) [55,56].
MRI longitudinal monitoring in the context of MR-guided RT could also help in the stratification of patients between fast and slow regressors, but with no significant impact on 2-year DFS [57]. The distribution of response rates was not affected by the regression type, with only one complete response rate on an overall cohort of 20 patients limiting the power of the study. Similarly, in a cohort of 43 patients, MRI was conducted before, during, and after CRT. Variations in T2 and Diffusion B1000 values were predictors of response to treatment [58].
In a systematic review published in 2014, Memon et al. identified 17 series assessing PET prediction of pCR [59]. In cohorts varying from 20 to 151 patients, the most reliable predictors appeared to be a maximum standard uptake value at the time of post-CRT evaluation (SUV2Max), reduction in SUVMax (RISUVMax), and visual complete metabolic response. Of note, though a metabolic complete response was achieved for 23–60% of the patients, the pCR rate ranged from 6 to 35%.

3.3. Genomics, Transcriptomics

Based on a cohort of 38 patients, Gonçalves-Ribeiro et al. developed a two-protein immunohistochemical score that was further validated on an external cohort of 36 patients. Combining FN1 and COL3A1 with a logistic regression approach, the model was significantly associated with the tumor regression grade and pathological nodal stage and a statistical trend towards pCR [60]. Interestingly, only very few differences were observed between responders and non-responders with regard to tumor cells themselves, whereas the stroma displayed a profound transcriptomic change, as previously suggested [61,62]. The value of peri-tumoral tissues, notably in a radiomics workflow, should be further studied.
In a prospective cohort of 80 patients treated with CRT and subsequent surgery, four biomarkers (p53, p21, Ki67, and CD133) were identified as significantly correlated with TRG and pCR. A model based on the value of these four biomarkers significantly stratified patients according to their chance of pCR (14.5% vs. 83.3%, p < 0.001), with a positive predictive value (PPV) of 83.3% and a NPV of 85.5%. No test or validation sets were available [63]. The value of gene expression levels for the prediction of pCR was previously explored with different biomarkers being identified (LRR1Q3, FRMD3, SAMD5, and TMC7), but on smaller cohorts with, again, no validation sets [64].
The value of microsatellite instability (MSI) to predict pCR remains controversial. In a recent meta-analysis of 5086 patients (636 MSI-positive), MSI-positive tumors were associated with a reduction in pCR [65], whereas in a second meta-analysis, MSI status had no impact on pCR rate [66]. Similarly, due to low-volume cohorts, the role of the EGFR, MAPK/AKT, and pi3K/AKT/mTOR pathways should be further explored, especially given the recent development of specific pathway inhibitors such as ipatasertib [67].

3.4. Circulating Tumor Cells, Tissue, and Circulating miRNAs

There is great interest in pre- and post-treatment circulating DNA (ctDNA) monitoring, especially in multimodal and interdisciplinary treatment settings. Although results regarding baseline ctDNA levels are conflicting, pre-operative ctDNA appears to be predictive of pCR, as demonstrated in several retrospective or prospective cohort studies [68,69]. Combining ctDNA detection with an analysis of potentially detectable mutations could improve the prediction of recurrence and lead to substantial treatment adaptations [69].
Regarding baseline ctDNA levels, in a cohort of 36 patients, baseline ctDNA detection was a negative prognostic biomarker for OS [70]. The reduction in ctDNA levels during CRT limited the value of pre-operative ctDNA for prediction modeling.
In a cohort of 104 patients (29% pCR rate), preoperative ctDNA-positive rates were considerably lower in the ypCR than non-ypCR group, with a pathologic T stage (ypT) of 0–2 versus the ypT 3–4 group, and well-responding patients had a pathologic tumor regression grade of ypCAP 0–1 vs. the ypCAP 2–3 group [71]. Of note, pre-CRT ctDNA level was not correlated with response to treatment, as suggested in other studies [72,73]. However, all patients who experienced pCR had a negative pre-operative ctDNA, and a significant correlation was found between ctDNA detection and pCR (p = 0.02).
Post-hoc analysis of the prospective GEMCAD 1402 trial (180 patients) evaluated the addition of aflibercept to modified FOLFOX before CRT and subsequent surgery. ctDNA data were available for 72 patients; ctDNA was detectable in 83% of patients at baseline and only 15% of patients before surgery. No correlation was found between ctDNA and pCR, though ctDNA appeared as a prognostic variable for DFS and OS [74].
The monitoring of micro-RNAs (miRNAs) certainly has value in predicting response to NAT [75] and could be evaluated in both on tissue and blood samples. Several tissue miRNAs were associated with TRG, among which miR-630 and miR-622 reached sensitivities and specificities of 100% and were upregulated in responders [76]. miR-622 itself was further evaluated, but with conflicting results, as it appeared as highly expressed in pretreatment cells of non-responders (TRG4) [77]. Despite an unclear physiopathology of circulative miRNAs [78], several miRNAs have been identified as predictors of response after CRT. For example, miR-18b and miR-20a [79], as well as miR-125b [80] and miR-143 [81], were correlated with a response to CRT. To our knowledge, very few of these circulating miRNAs were externally validated. Wada et al. developed a blood-based model using a panel of eight different miRNAs, resulting in an AUC of 0.81 and a PPV of 51.4% for pCR prediction in an external cohort of 65 patients [82].
Single nucleotide polymorphisms (SNPs) are caused by a single nucleotide variation in the DNA sequence [83] and could have an impact on gene expression. Very few studies have evaluated the value of SNPs in LARC and in particular the response to treatment [75,84]. Nevertheless, promising results were shown with the role of rs61764370 SNP as a predictor of pCR, with increased 5-year PFS and OS when compared to the wild genotype patients [85]. In a cohort of 265 patients, five SNPS (rs744910, rs745103, rs6088619, rs10719, and rs17228212) had a predictive value for pCR.
Metabolic biomarkers, such as paraoxonase-1 (PON1) and α-ketoglutarate, could possibly be used for stratification between responders and non-responders or even pCR/non-pCR patients, as suggested by a single cohort study. Despite the small number of patients, this approach could be explored more thoroughly [86].

3.5. Pathomics

Analysis of anatomopathological images brings a new insight into prediction modeling. Using a cohort of 783 patients recruited from three different centers, Lou et al. developed a ResNet-18 network-based model [87]. On the external validation cohort (102 patients), the deep-learning model achieved an AUC of 0.72 (CI95% 0.59–0.84). The pathomics signature remained a significant predictor of pCR even after multivariate analysis. Of note, in the incorporated confounding features, only sex was a significant predictor (apart from the deep-learning model).
A combination of the radiomics and pathomics approaches was previously analyzed in a retrospective study conducted by Feng et al. [88]. With a training cohort of 303 patients, validation cohorts of 630 patients in total, and a prospective validation cohort of 100 patients, a radiopathomics model resulted in an AUC of 0.81 (CI95% 71.7–90.7%). Interestingly, the negative predictive value (NPV) reached 92.9% (86.2–99.5%), comforting clinicians in the case of a low predicted probability of pCR. Nevertheless, this high NPV should be put in balance with a pCR rate of 23%. This study remains one of the most well-conducted radiomics-based studies, with extensive validation and clinical impact, which could lead to treatment adaptations, such as dose escalation in non-pCR-predicted patients or NOM for pCR-predicted patients.

3.6. Microbiome

According to preclinical and clinical investigations, the gut microbiota may actively participate in various biochemical and pathophysiologic processes in the human body, including those involving inflammation and immunomodulation. However, data regarding response to CRT are particularly scarce in LARC patients. In a prospective longitudinal study, a decrease in LARC-related pathogens and an increase in Lactobacillus and Streptococcus was observed during CRT. Moreover, responders and non-responders exhibited differential microbiota, with Coriobacteriaceae and Fusobacterium being overrepresented in nonresponders. Combining ten biomarkers resulted in an AUC of 0.74 in a validation cohort (47 patients) [89]. Although the role of the microbiome on colorectal cancer tumorigenesis [90,91,92,93] was previously evaluated, the significance of these microbes regarding tumor response in LARC patients was unknown before the presented study. Given the latest data regarding the correlation between microbiota and tumor response in various localizations, the role of microbiota as predictors of response after CRT should definitely be investigated further. Fusobacterium, for instance, could be an indicator of tumor aggressiveness, as suggested by several studies [94,95].

4. Prediction of pCR Using Radiomics Model

A PubMed research query performed on the 14th of November 2022 using the terms ”Radiomics”, “Prediction”, “Complete Response”, and “Rectal Cancer” resulted in 61 studies published between 2016 and 2022, 37 of them having been published in 2021–2022, highlighting the large interest in this topic in the era of TNT and NOM. An overview of these studies with details on the type of NAT, population size, pCR rate, and existence of training/validation cohorts (external/internal) is further provided depending on the type of studied imaging modality.
Of the 61 identified studies, 13 studies were excluded for the following reasons: five were systematic, comprehensive, or narrative reviews; three were not radiomics-based studies; one had a primary endpoint of the prediction of MSI status; one focused on cervical cancer; one was a study protocol; one was a correction to the initial study; and one was a duplicate. As a result, 48 studies were further considered, among which the full article was available for 43 of them. Among the 48 studies, the vast majority (36/48: 75.0%) concerned MRI-only radiomics, and 4/48 (8.3%) and 8/48 (16.7%) concerned MRI-combined and non-MRI-based prediction models, respectively. Overall, a validation set was available for 30/48 (62.5%) of the studies, validation being prospectively conducted in only 2 studies (4.2%) [88,96]. The majority of prediction models were based on pre-CRT-only imaging (37/48: 77.1%), with a subset of models focusing on longitudinal (14.6%) and post-CRT (8.4%) imaging, when all studies were conducted in patients treated with CRT and not TNT.
Ranging from 16 to 592 patients, training cohort size did not significantly change when stratified by image modality (p = 0.81), timing of the imaging (p = 0.80), or when combining the image modality and the timing (p = 0.88).
As mentioned, only two trials had a prospective validation. Dinapoli et al. externally validated an MRI-based radiomics prediction model on two validation cohorts, one of which was prospectively conducted (THUNDER Trial). Despite having been prospectively included, analysis and validation were retrospectively performed. With a training cohort of 303 patients, 2 validation cohorts of respectively 480 and 150 patients, and a prospective validation cohort of 100 patients, the RAPIDS model presented by Feng et al. appears to be the only model to be fully validated. Combining baseline MRI radiomics features with pathomics nucleus and microenvironment features, the model achieved an AUC of 0.81 and a PPV of 0.51 in the prospective cohort. Combining the three sets of features (MRI, pathomics nucleus, and microenvironment features) significantly enhanced the model’s performance when compared to other single-modality models. Furthermore, in their effort of external validation, the authors developed a web-based platform that enables the widespread use of this model.
The low PPV values for most of the models must be put in balance with the low prevalence of pCR. When available with a range of 28.0–100.0, such non-invasive models show interesting results and should be further explored.
As mentioned, only 14.6% of the studies (7/48) used imaging with multiple time points (either pre- and mid-/post-CRT or pre- and during CRT). Although not significant, it should be acknowledged that the median training cohort size was smaller (35, CI95% 16–101.6) compared to the baseline (100.0, CI95% 86.0–130.8) and post-CRT (129.0, CI95% not available) cohorts only. The results of the longitudinally built radiomics models did not significantly outperform baseline prediction models in the validation cohorts. This could probably be explained by the low number of studies in which validation results were available in a longitudinal radiomics model (n = 5).
Delineation data were not retrieved for 4/48 studies, and most volumes were manually delineated (40/48–83.3%). When performed by multiple readers, multiple delineation was used as a feature selection step for only 12 of the studies (25.0%). Automatic or semi-automatic segmentation was used in four studies. The approach of Defeudis et al. should be highlighted because of the innovative pipeline it proposes. In addition to using manual segmentations defined by three different readers, an automatic segmentation was also evaluated. Interestingly, whereas the model using manual segmentation-based features performed highly on the training cohort (AUC 0.90), the model using the automatic segmentation features outperformed the other model on the validation cohort, with respective AUCs of 0.81 and 0.61 and accuracies of 75.0% and 65.0%.
These results highlight the need for broader external validation as well as quality assurance regarding the target definition, as further discussed in the limitations and perspectives paragraph.
Data regarding the 48 retrieved studies is hereby presented in Table 1, Table 2 and Table 3 for studies that focused on MRI-only, MRI-combined, and non-MRI imaging, respectively.

5. Limitations/Perspectives

Despite multiple studies and robust results, radiomics models fail to convince clinicians and are absent from current guidelines and clinical workflows.
The majority of models were tested on validation cohorts, despite only being prospectively evaluated.
Apart from a low level of evidence, several other concerns could explain the low acceptance of clinicians regarding radiomics-based models.
The majority of studies focused on GTV-extracted features, not taking into account variability in clinical target volumes (CTVs) and heterogeneity in the dosimetry planning and delivery. Given the complementary association between tumoral and peri-tumoral features, one might think that the definition of the CTV is important. Moreover, none of the studies evaluated the value of dose map-extracted features (dosiomics). Substantial differences in dose distribution can be observed depending on the treatment modality (3D-conformational, intensity-modulated RT, or volumetric arctherapy).
A positive impact was seen with automatically segmented GTVs [109], especially in the validation cohort. Such results highlight the need for quality assurance and possibly fully automatic workflows that would help standardize models’ development and validation and probably their generalization. In this aspect, the workflow proposed by Feng et al. would allow for such learning and application.
Previously presented models were mostly based on baseline MRIs, with several studies evaluating, with smaller samples, the value of multiple timepoint evaluations. Though this approach seems promising, the use of different imaging modalities provides new perspectives. For instance, the TEP 64Cu-ATSM-Rectum evaluates 64Cu-ATSM PET/CT as a new imaging biomarker in patients treated with CRT (NCT03951337).
Trans-omic analysis connects multi-omic layers and enables a better understanding of the physiopathology underlying, in our case, response to treatment. Though radiomics could be considered to be a macroscopic reflection of changes happening at the microscopic level, trans-omics could connect the information provided by genomics, transcriptomics, and proteomics using computational data. A complete understanding of the mechanisms of response to neo-adjuvant therapy would not only allow for prediction, but most likely for patient’s selection before treatment is even performed. However, to this day, and despite several teams working on this matter, no studies focusing on the prediction of pCR using a trans-omics approach have been published.
Finally, the recent change in LARC management and the advent of TNT limit the validity of previously presented studies, as none was performed or evaluated in TNT-treated patients. This lack of data is explained by the current workflow that most radiomics studies use. Whereas genomic data are often acquired prospectively, radiomics studies mostly consist of post-hoc retrospective analyses. Given the impact of TNT on pCR rates, one can expect the prediction from previous models to be inaccurate. New radiomics models must thus be developed. Promising results regarding the use of neoadjuvant immune checkpoint inhibitors (ICIs) could revolutionize the management of LARC patients. Though the addition of pembrolizumab to TNT did not modify the pCR rate in unselected LARCs patients [143], the use of nivolumab plus ipilimumab seems to be very promising in dMMR (mismatch repair-deficient) tumors. Combining PD-1 and CTLA-4 blockage led to a 60% pCR rate in dMMR patients, and no patients with a pMMR tumor experienced a pCR [144].

6. Conclusions

In recent years, neoadjuvant therapy for locally advanced rectal cancer has seen tremendous modifications. Neoadjuvant CRT in combination with pre-operative chemotherapy improves LRFS and pCR rates, opening the possibility of non-operative management and organ preservation without compromising overall survival. Given the challenge of pCR prediction, multiple approaches have been developed, and radiomics-based models especially have achieved promising results but lack external validation. Substantial efforts have been made regarding the quality of published radiomics models. Such approaches could be combined and/or compared to other promising approaches, such as circulating DNA.

Author Contributions

Conceptualization, V.B.; methodology, V.B. and B.B.; validation, V.B., U.S., O.P., J.-P.M., and B.B.; formal analysis, V.B.; writing—original draft preparation, V.B.; writing—review and editing, V.B., U.S., O.P., J.-P.M., D.V., and B.B.; supervision, B.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Kennecke, H.F.; Bahnson, H.T.; Lin, B.; O’Rourke, C.; Kaplan, J.; Pham, H.; Suen, A.; Simianu, V.V. Patterns of Practice and Improvements in Survival Among Patients With Stage 2/3 Rectal Cancer Treated With Trimodality Therapy. JAMA Oncol. 2022, 8, 1466–1470. [Google Scholar] [CrossRef] [PubMed]
  3. Sauer, R.; Becker, H.; Hohenberger, W.; Rodel, C.; Wittekind, C.; Fietkau, R.; Martus, P.; Tschmelitsch, J.; Hager, E.; Hess, C.F.; et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N. Engl. J. Med. 2004, 351, 1731–1740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Roh, M.S.; Colangelo, L.H.; O’Connell, M.J.; Yothers, G.; Deutsch, M.; Allegra, C.J.; Kahlenberg, M.S.; Baez-Diaz, L.; Ursiny, C.S.; Petrelli, N.J.; et al. Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 5124–5130. [Google Scholar] [CrossRef] [PubMed]
  5. Park, J.H.; Yoon, S.M.; Yu, C.S.; Kim, J.H.; Kim, T.W.; Kim, J.C. Randomized phase 3 trial comparing preoperative and postoperative chemoradiotherapy with capecitabine for locally advanced rectal cancer. Cancer 2011, 117, 3703–3712. [Google Scholar] [CrossRef]
  6. Swedish Rectal Cancer, T.; Cedermark, B.; Dahlberg, M.; Glimelius, B.; Pahlman, L.; Rutqvist, L.E.; Wilking, N. Improved survival with preoperative radiotherapy in resectable rectal cancer. N. Engl. J. Med. 1997, 336, 980–987. [Google Scholar] [CrossRef]
  7. Kapiteijn, E.; Marijnen, C.A.; Nagtegaal, I.D.; Putter, H.; Steup, W.H.; Wiggers, T.; Rutten, H.J.; Pahlman, L.; Glimelius, B.; van Krieken, J.H.; et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N. Engl. J. Med. 2001, 345, 638–646. [Google Scholar] [CrossRef] [Green Version]
  8. Sebag-Montefiore, D.; Stephens, R.J.; Steele, R.; Monson, J.; Grieve, R.; Khanna, S.; Quirke, P.; Couture, J.; de Metz, C.; Myint, A.S.; et al. Preoperative radiotherapy versus selective postoperative chemoradiotherapy in patients with rectal cancer (MRC CR07 and NCIC-CTG C016): A multicentre, randomised trial. Lancet 2009, 373, 811–820. [Google Scholar] [CrossRef] [Green Version]
  9. Conroy, T.; Bosset, J.F.; Etienne, P.L.; Rio, E.; Francois, E.; Mesgouez-Nebout, N.; Vendrely, V.; Artignan, X.; Bouche, 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. 2021, 22, 702–715. [Google Scholar] [CrossRef]
  10. Bahadoer, R.R.; Dijkstra, E.A.; van Etten, B.; Marijnen, C.A.M.; Putter, H.; Kranenbarg, E.M.; Roodvoets, A.G.H.; Nagtegaal, I.D.; Beets-Tan, R.G.H.; Blomqvist, L.K.; 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. 2021, 22, 29–42. [Google Scholar] [CrossRef]
  11. Kasi, A.; Abbasi, S.; Handa, S.; Al-Rajabi, R.; Saeed, A.; Baranda, J.; Sun, W. Total Neoadjuvant Therapy vs. Standard Therapy in Locally Advanced Rectal Cancer: A Systematic Review and Meta-analysis. JAMA Netw. Open 2020, 3, e2030097. [Google Scholar] [CrossRef] [PubMed]
  12. Ma, Z.; Tan, L.; Liu, Z.L.; Xiao, J.W. Total neoadjuvant therapy or standard chemoradiotherapy for locally advanced rectal cancer: A systematic review and meta-analysis. Front. Surg. 2022, 9, 911538. [Google Scholar] [CrossRef] [PubMed]
  13. Lin, H.; Wang, L.; Zhong, X.; Zhang, X.; Shao, L.; Wu, J. Meta-analysis of neoadjuvant chemotherapy versus neoadjuvant chemoradiotherapy for locally advanced rectal cancer. World J. Surg. Oncol. 2021, 19, 141. [Google Scholar] [CrossRef]
  14. Liu, S.; Jiang, T.; Xiao, L.; Yang, S.; Liu, Q.; Gao, Y.; Chen, G.; Xiao, W. Total Neoadjuvant Therapy (TNT) versus Standard Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancer: A Systematic Review and Meta-Analysis. Oncologist 2021, 26, e1555–e1566. [Google Scholar] [CrossRef] [PubMed]
  15. Fokas, E.; Allgauer, M.; Polat, B.; Klautke, G.; Grabenbauer, G.G.; Fietkau, R.; Kuhnt, T.; Staib, L.; Brunner, T.; Grosu, A.L.; et al. Randomized Phase II Trial of Chemoradiotherapy Plus Induction or Consolidation Chemotherapy as Total Neoadjuvant Therapy for Locally Advanced Rectal Cancer: CAO/ARO/AIO-12. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 3212–3222. [Google Scholar] [CrossRef]
  16. Gani, C.; Gani, N.; Zschaeck, S.; Eberle, F.; Schaeffeler, N.; Hehr, T.; Berger, B.; Fischer, S.G.; Classen, J.; Zipfel, S.; et al. Organ Preservation in Rectal Cancer: The Patients’ Perspective. Front. Oncol. 2019, 9, 318. [Google Scholar] [CrossRef]
  17. Shin, J.K.; Huh, J.W.; Lee, W.Y.; Yun, S.H.; Kim, H.C.; Cho, Y.B.; Park, Y.A. Clinical prediction model of pathological response following neoadjuvant chemoradiotherapy for rectal cancer. Sci. Rep. 2022, 12, 7145. [Google Scholar] [CrossRef]
  18. Maas, M.; Nelemans, P.J.; Valentini, V.; Das, P.; Rodel, C.; Kuo, L.J.; Calvo, F.A.; Garcia-Aguilar, J.; Glynne-Jones, R.; Haustermans, K.; et al. Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: A pooled analysis of individual patient data. Lancet Oncol. 2010, 11, 835–844. [Google Scholar] [CrossRef]
  19. Maas, M.; Lambregts, D.M.; Nelemans, P.J.; Heijnen, L.A.; Martens, M.H.; Leijtens, J.W.; Sosef, M.; Hulsewe, K.W.; Hoff, C.; Breukink, S.O.; et al. Assessment of Clinical Complete Response After Chemoradiation for Rectal Cancer with Digital Rectal Examination, Endoscopy, and MRI: Selection for Organ-Saving Treatment. Ann. Surg. Oncol. 2015, 22, 3873–3880. [Google Scholar] [CrossRef] [Green Version]
  20. Habr-Gama, A.; Perez, R.O.; Wynn, G.; Marks, J.; Kessler, H.; Gama-Rodrigues, J. Complete clinical response after neoadjuvant chemoradiation therapy for distal rectal cancer: Characterization of clinical and endoscopic findings for standardization. Dis. Colon Rectum 2010, 53, 1692–1698. [Google Scholar] [CrossRef]
  21. Maas, M.; Beets-Tan, R.G.; Lambregts, D.M.; Lammering, G.; Nelemans, P.J.; Engelen, S.M.; van Dam, R.M.; Jansen, R.L.; Sosef, M.; Leijtens, J.W.; et al. Wait-and-see policy for clinical complete responders after chemoradiation for rectal cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2011, 29, 4633–4640. [Google Scholar] [CrossRef] [PubMed]
  22. Garcia-Aguilar, J.; Patil, S.; Gollub, M.J.; Kim, J.K.; Yuval, J.B.; Thompson, H.M.; Verheij, F.S.; Omer, D.M.; Lee, M.; Dunne, R.F.; et al. Organ Preservation in Patients With Rectal Adenocarcinoma Treated With Total Neoadjuvant Therapy. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 2546–2556. [Google Scholar] [CrossRef] [PubMed]
  23. Smith, J.J.; Strombom, P.; Chow, O.S.; Roxburgh, C.S.; Lynn, P.; Eaton, A.; Widmar, M.; Ganesh, K.; Yaeger, R.; Cercek, A.; et al. Assessment of a Watch-and-Wait Strategy for Rectal Cancer in Patients With a Complete Response After Neoadjuvant Therapy. JAMA Oncol. 2019, 5, e185896. [Google Scholar] [CrossRef] [PubMed]
  24. Ryan, J.E.; Warrier, S.K.; Lynch, A.C.; Ramsay, R.G.; Phillips, W.A.; Heriot, A.G. Predicting pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer: A systematic review. Color. Dis. Off. J. Assoc. Coloproctology Great Br. Irel. 2016, 18, 234–246. [Google Scholar] [CrossRef]
  25. de Jong, E.A.; ten Berge, J.C.; Dwarkasing, R.S.; Rijkers, A.P.; van Eijck, C.H. The accuracy of MRI, endorectal ultrasonography, and computed tomography in predicting the response of locally advanced rectal cancer after preoperative therapy: A metaanalysis. Surgery 2016, 159, 688–699. [Google Scholar] [CrossRef]
  26. Kawai, K.; Ishihara, S.; Nozawa, H.; Hata, K.; Kiyomatsu, T.; Morikawa, T.; Fukayama, M.; Watanabe, T. Prediction of Pathological Complete Response Using Endoscopic Findings and Outcomes of Patients Who Underwent Watchful Waiting After Chemoradiotherapy for Rectal Cancer. Dis. Colon Rectum 2017, 60, 368–375. [Google Scholar] [CrossRef]
  27. Yoon, S.M.; Kim, D.Y.; Kim, T.H.; Jung, K.H.; Chang, H.J.; Koom, W.S.; Lim, S.B.; Choi, H.S.; Jeong, S.Y.; Park, J.G. Clinical parameters predicting pathologic tumor response after preoperative chemoradiotherapy for rectal cancer. Int. J. Radiat. Oncol. Biol. Phys. 2007, 69, 1167–1172. [Google Scholar] [CrossRef]
  28. Rodel, C.; Martus, P.; Papadoupolos, T.; Fuzesi, L.; Klimpfinger, M.; Fietkau, R.; Liersch, T.; Hohenberger, W.; Raab, R.; Sauer, R.; et al. Prognostic significance of tumor regression after preoperative chemoradiotherapy for rectal cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 8688–8696. [Google Scholar] [CrossRef]
  29. Park, Y.A.; Sohn, S.K.; Seong, J.; Baik, S.H.; Lee, K.Y.; Kim, N.K.; Cho, C.W. Serum CEA as a predictor for the response to preoperative chemoradiation in rectal cancer. J. Surg. Oncol. 2006, 93, 145–150. [Google Scholar] [CrossRef]
  30. Kleiman, A.; Al-Khamis, A.; Farsi, A.; Kezouh, A.; Vuong, T.; Gordon, P.H.; Vasilevsky, C.A.; Morin, N.; Faria, J.; Ghitulescu, G.; et al. Normalization of CEA Levels Post-Neoadjuvant Therapy is a Strong Predictor of Pathologic Complete Response in Rectal Cancer. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 2015, 19, 1106–1112. [Google Scholar] [CrossRef]
  31. Park, C.H.; Kim, H.C.; Cho, Y.B.; Yun, S.H.; Lee, W.Y.; Park, Y.S.; Choi, D.H.; Chun, H.K. Predicting tumor response after preoperative chemoradiation using clinical parameters in rectal cancer. World J. Gastroenterol. 2011, 17, 5310–5316. [Google Scholar] [CrossRef] [PubMed]
  32. Garland, M.L.; Vather, R.; Bunkley, N.; Pearse, M.; Bissett, I.P. Clinical tumour size and nodal status predict pathologic complete response following neoadjuvant chemoradiotherapy for rectal cancer. Int. J. Color. Dis. 2014, 29, 301–307. [Google Scholar] [CrossRef] [PubMed]
  33. Al-Sukhni, E.; Attwood, K.; Mattson, D.M.; Gabriel, E.; Nurkin, S.J. Predictors of Pathologic Complete Response Following Neoadjuvant Chemoradiotherapy for Rectal Cancer. Ann. Surg. Oncol. 2016, 23, 1177–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Huh, J.W.; Kim, H.R.; Kim, Y.J. Clinical prediction of pathological complete response after preoperative chemoradiotherapy for rectal cancer. Dis. Colon Rectum 2013, 56, 698–703. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, J.H.; Song, C.; Kang, S.B.; Lee, H.S.; Lee, K.W.; Kim, J.S. Predicting Pathological Complete Regression with Haematological Markers During Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancer. Anticancer. Res. 2018, 38, 6905–6910. [Google Scholar] [CrossRef] [PubMed]
  36. Mbanu, P.; Osorio, E.V.; Mistry, H.; Malcomson, L.; Yousif, S.; Aznar, M.; Kochhar, R.; Van Herk, M.; Renehan, A.G.; Saunders, M.P. Clinico-pathological predictors of clinical complete response in rectal cancer. Cancer Treat. Res. Commun. 2022, 31, 100540. [Google Scholar] [CrossRef]
  37. Kang, B.H.; Song, C.; Kang, S.B.; Lee, K.W.; Lee, H.S.; Kim, J.S. Nomogram for Predicting the Pathological Tumor Response from Pre-treatment Clinical Characteristics in Rectal Cancer. Anticancer. Res. 2020, 40, 2171–2177. [Google Scholar] [CrossRef]
  38. Francois, Y.; Nemoz, C.J.; Baulieux, J.; Vignal, J.; Grandjean, J.P.; Partensky, C.; Souquet, J.C.; Adeleine, P.; Gerard, J.P. Influence of the interval between preoperative radiation therapy and surgery on downstaging and on the rate of sphincter-sparing surgery for rectal cancer: The Lyon R90-01 randomized trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1999, 17, 2396. [Google Scholar] [CrossRef]
  39. Sun, Y.S.; Cui, Y.; Tang, L.; Qi, L.P.; Wang, N.; Zhang, X.Y.; Cao, K.; Zhang, X.P. Early evaluation of cancer response by a new functional biomarker: Apparent diffusion coefficient. AJR Am. J. Roentgenol. 2011, 197, W23–W29. [Google Scholar] [CrossRef]
  40. Nougaret, S.; Reinhold, C.; Mikhael, H.W.; Rouanet, P.; Bibeau, F.; Brown, G. The use of MR imaging in treatment planning for patients with rectal carcinoma: Have you checked the "DISTANCE"? Radiology 2013, 268, 330–344. [Google Scholar] [CrossRef]
  41. Xu, Q.; Xu, Y.; Sun, H.; Jiang, T.; Xie, S.; Ooi, B.Y.; Ding, Y. MRI Evaluation of Complete Response of Locally Advanced Rectal Cancer After Neoadjuvant Therapy: Current Status and Future Trends. Cancer Manag. Res. 2021, 13, 4317–4328. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, C.; Ye, F.; Liu, Y.; Ouyang, H.; Zhao, X.; Zhang, H. Morphologic predictors of pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Oncotarget 2018, 9, 4862–4874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Martens, M.H.; van Heeswijk, M.M.; van den Broek, J.J.; Rao, S.X.; Vandecaveye, V.; Vliegen, R.A.; Schreurs, W.H.; Beets, G.L.; Lambregts, D.M.; Beets-Tan, R.G. Prospective, Multicenter Validation Study of Magnetic Resonance Volumetry for Response Assessment After Preoperative Chemoradiation in Rectal Cancer: Can the Results in the Literature be Reproduced? Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 1005–1014. [Google Scholar] [CrossRef] [PubMed]
  44. Palmisano, A.; Esposito, A.; Di Chiara, A.; Ambrosi, A.; Passoni, P.; Slim, N.; Fiorino, C.; Albarello, L.; Di Muzio, N.; Calandrino, R.; et al. Could early tumour volume changes assessed on morphological MRI predict the response to chemoradiation therapy in locally-advanced rectal cancer? Clin. Radiol. 2018, 73, 555–563. [Google Scholar] [CrossRef]
  45. Kim, S.; Han, K.; Seo, N.; Kim, H.J.; Kim, M.J.; Koom, W.S.; Ahn, J.B.; Lim, J.S. T2-weighted signal intensity-selected volumetry for prediction of pathological complete response after preoperative chemoradiotherapy in locally advanced rectal cancer. Eur. Radiol. 2018, 28, 5231–5240. [Google Scholar] [CrossRef]
  46. Neri, E.; Guidi, E.; Pancrazi, F.; Castagna, M.; Castelluccio, E.; Balestri, R.; Buccianti, P.; Masi, L.; Falcone, A.; Manfredi, B.; et al. MRI tumor volume reduction rate vs. tumor regression grade in the pre-operative re-staging of locally advanced rectal cancer after chemo-radiotherapy. Eur. J. Radiol. 2015, 84, 2438–2443. [Google Scholar] [CrossRef]
  47. Sathyakumar, K.; Chandramohan, A.; Masih, D.; Jesudasan, M.R.; Pulimood, A.; Eapen, A. Best MRI predictors of complete response to neoadjuvant chemoradiation in locally advanced rectal cancer. Br. J. Radiol. 2016, 89, 20150328. [Google Scholar] [CrossRef] [Green Version]
  48. Kim, Y.C.; Lim, J.S.; Keum, K.C.; Kim, K.A.; Myoung, S.; Shin, S.J.; Kim, M.J.; Kim, N.K.; Suh, J.; Kim, K.W. Comparison of diffusion-weighted MRI and MR volumetry in the evaluation of early treatment outcomes after preoperative chemoradiotherapy for locally advanced rectal cancer. J. Magn. Reson. Imaging JMRI 2011, 34, 570–576. [Google Scholar] [CrossRef]
  49. Lambregts, D.M.; Rao, S.X.; Sassen, S.; Martens, M.H.; Heijnen, L.A.; Buijsen, J.; Sosef, M.; Beets, G.L.; Vliegen, R.A.; Beets-Tan, R.G. MRI and Diffusion-weighted MRI Volumetry for Identification of Complete Tumor Responders After Preoperative Chemoradiotherapy in Patients With Rectal Cancer: A Bi-institutional Validation Study. Ann. Surg. 2015, 262, 1034–1039. [Google Scholar] [CrossRef]
  50. Intven, M.; Reerink, O.; Philippens, M.E. Diffusion-weighted MRI in locally advanced rectal cancer: Pathological response prediction after neo-adjuvant radiochemotherapy. Strahlenther. Und Onkol. Organ Der Dtsch. Rontgenges. 2013, 189, 117–122. [Google Scholar] [CrossRef]
  51. Blazic, I.M.; Lilic, G.B.; Gajic, M.M. Quantitative Assessment of Rectal Cancer Response to Neoadjuvant Combined Chemotherapy and Radiation Therapy: Comparison of Three Methods of Positioning Region of Interest for ADC Measurements at Diffusion-weighted MR Imaging. Radiology 2017, 282, 615. [Google Scholar] [CrossRef] [PubMed]
  52. Enkhbaatar, N.E.; Inoue, S.; Yamamuro, H.; Kawada, S.; Miyaoka, M.; Nakamura, N.; Sadahiro, S.; Imai, Y. MR Imaging with Apparent Diffusion Coefficient Histogram Analysis: Evaluation of Locally Advanced Rectal Cancer after Chemotherapy and Radiation Therapy. Radiology 2018, 288, 129–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Lambrecht, M.; Vandecaveye, V.; De Keyzer, F.; Roels, S.; Penninckx, F.; Van Cutsem, E.; Filip, C.; Haustermans, K. Value of diffusion-weighted magnetic resonance imaging for prediction and early assessment of response to neoadjuvant radiochemotherapy in rectal cancer: Preliminary results. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 863–870. [Google Scholar] [CrossRef] [PubMed]
  54. Liang, C.Y.; Chen, M.D.; Zhao, X.X.; Yan, C.G.; Mei, Y.J.; Xu, Y.K. Multiple mathematical models of diffusion-weighted magnetic resonance imaging combined with prognostic factors for assessing the response to neoadjuvant chemotherapy and radiation therapy in locally advanced rectal cancer. Eur. J. Radiol. 2019, 110, 249–255. [Google Scholar] [CrossRef]
  55. Zou, H.H.; Yu, J.; Wei, Y.; Wu, J.F.; Xu, Q. Response to neoadjuvant chemoradiotherapy for locally advanced rectum cancer: Texture analysis of dynamic contrast-enhanced MRI. J. Magn. Reson. Imaging JMRI 2019, 49, 885–893. [Google Scholar] [CrossRef]
  56. Gollub, M.J.; Tong, T.; Weiser, M.; Zheng, J.; Gonen, M.; Zakian, K.L. Limited accuracy of DCE-MRI in identification of pathological complete responders after chemoradiotherapy treatment for rectal cancer. Eur. Radiol. 2017, 27, 1605–1612. [Google Scholar] [CrossRef] [Green Version]
  57. Sung, S.Y.; Lee, S.W.; Hong, J.H.; Kang, H.J.; Lee, S.J.; Kim, M.; Kim, J.H.; Kwak, Y.K. Linear Tumor Regression of Rectal Cancer in Daily MRI during Preoperative Chemoradiotherapy: An Insight of Tumor Regression Velocity for Personalized Cancer Therapy. Cancers 2022, 14, 3749. [Google Scholar] [CrossRef]
  58. Palmisano, A.; Di Chiara, A.; Esposito, A.; Rancoita, P.M.V.; Fiorino, C.; Passoni, P.; Albarello, L.; Rosati, R.; Del Maschio, A.; De Cobelli, F. MRI prediction of pathological response in locally advanced rectal cancer: When apparent diffusion coefficient radiomics meets conventional volumetry. Clin. Radiol. 2020, 75, 798.e1–798.e11. [Google Scholar] [CrossRef]
  59. Memon, S.; Lynch, A.C.; Akhurst, T.; Ngan, S.Y.; Warrier, S.K.; Michael, M.; Heriot, A.G. Systematic review of FDG-PET prediction of complete pathological response and survival in rectal cancer. Ann. Surg. Oncol. 2014, 21, 3598–3607. [Google Scholar] [CrossRef]
  60. Goncalves-Ribeiro, S.; Sanz-Pamplona, R.; Vidal, A.; Sanjuan, X.; Guillen Diaz-Maroto, N.; Soriano, A.; Guardiola, J.; Albert, N.; Martinez-Villacampa, M.; Lopez, I.; et al. Prediction of pathological response to neoadjuvant treatment in rectal cancer with a two-protein immunohistochemical score derived from stromal gene-profiling. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 2160–2168. [Google Scholar] [CrossRef]
  61. Calon, A.; Lonardo, E.; Berenguer-Llergo, A.; Espinet, E.; Hernando-Momblona, X.; Iglesias, M.; Sevillano, M.; Palomo-Ponce, S.; Tauriello, D.V.; Byrom, D.; et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet. 2015, 47, 320–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Isella, C.; Terrasi, A.; Bellomo, S.E.; Petti, C.; Galatola, G.; Muratore, A.; Mellano, A.; Senetta, R.; Cassenti, A.; Sonetto, C.; et al. Stromal contribution to the colorectal cancer transcriptome. Nat. Genet. 2015, 47, 312–319. [Google Scholar] [CrossRef] [PubMed]
  63. Hur, H.; Tulina, I.; Cho, M.S.; Min, B.S.; Koom, W.S.; Lim, J.S.; Ahn, J.B.; Kim, N.K. Biomarker-Based Scoring System for Prediction of Tumor Response After Preoperative Chemoradiotherapy in Rectal Cancer by Reverse Transcriptase Polymerase Chain Reaction Analysis. Dis. Colon Rectum 2016, 59, 1174–1182. [Google Scholar] [CrossRef] [PubMed]
  64. Watanabe, T.; Kobunai, T.; Akiyoshi, T.; Matsuda, K.; Ishihara, S.; Nozawa, K. Prediction of response to preoperative chemoradiotherapy in rectal cancer by using reverse transcriptase polymerase chain reaction analysis of four genes. Dis. Colon Rectum 2014, 57, 23–31. [Google Scholar] [CrossRef] [PubMed]
  65. Hasan, S.; Renz, P.; Wegner, R.E.; Finley, G.; Raj, M.; Monga, D.; McCormick, J.; Kirichenko, A. Microsatellite Instability (MSI) as an Independent Predictor of Pathologic Complete Response (PCR) in Locally Advanced Rectal Cancer: A National Cancer Database (NCDB) Analysis. Ann. Surg. 2020, 271, 716–723. [Google Scholar] [CrossRef]
  66. O’Connell, E.; Reynolds, I.S.; McNamara, D.A.; Prehn, J.H.M.; Burke, J.P. Microsatellite instability and response to neoadjuvant chemoradiotherapy in rectal cancer: A systematic review and meta-analysis. Surg. Oncol. 2020, 34, 57–62. [Google Scholar] [CrossRef]
  67. Sun, L.; Huang, Y.; Liu, Y.; Zhao, Y.; He, X.; Zhang, L.; Wang, F.; Zhang, Y. Ipatasertib, a novel Akt inhibitor, induces transcription factor FoxO3a and NF-kappaB directly regulates PUMA-dependent apoptosis. Cell Death Dis. 2018, 9, 911. [Google Scholar] [CrossRef] [Green Version]
  68. Wang, Y.; Yang, L.; Bao, H.; Fan, X.; Xia, F.; Wan, J.; Shen, L.; Guan, Y.; Bao, H.; Wu, X.; et al. Utility of ctDNA in predicting response to neoadjuvant chemoradiotherapy and prognosis assessment in locally advanced rectal cancer: A prospective cohort study. PLoS Med. 2021, 18, e1003741. [Google Scholar] [CrossRef]
  69. Liu, W.; Li, Y.; Tang, Y.; Song, Q.; Wang, J.; Li, N.; Chen, S.; Shi, J.; Wang, S.; Li, Y.; et al. Response prediction and risk stratification of patients with rectal cancer after neoadjuvant therapy through an analysis of circulating tumour DNA. EBioMedicine 2022, 78, 103945. [Google Scholar] [CrossRef]
  70. Pazdirek, F.; Minarik, M.; Benesova, L.; Halkova, T.; Belsanova, B.; Macek, M.; Stepanek, L.; Hoch, J. Monitoring of Early Changes of Circulating Tumor DNA in the Plasma of Rectal Cancer Patients Receiving Neoadjuvant Concomitant Chemoradiotherapy: Evaluation for Prognosis and Prediction of Therapeutic Response. Front. Oncol. 2020, 10, 1028. [Google Scholar] [CrossRef]
  71. Zhou, J.; Wang, C.; Lin, G.; Xiao, Y.; Jia, W.; Xiao, G.; Liu, Q.; Wu, B.; Wu, A.; Qiu, H.; et al. Serial Circulating Tumor DNA in Predicting and Monitoring the Effect of Neoadjuvant Chemoradiotherapy in Patients with Rectal Cancer: A Prospective Multicenter Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 301–310. [Google Scholar] [CrossRef] [PubMed]
  72. Tie, J.; Cohen, J.D.; Wang, Y.; Li, L.; Christie, M.; Simons, K.; Elsaleh, H.; Kosmider, S.; Wong, R.; Yip, D.; et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: A prospective biomarker study. Gut 2019, 68, 663–671. [Google Scholar] [CrossRef] [PubMed]
  73. Schou, J.V.; Larsen, F.O.; Sorensen, B.S.; Abrantes, R.; Boysen, A.K.; Johansen, J.S.; Jensen, B.V.; Nielsen, D.L.; Spindler, K.L. Circulating cell-free DNA as predictor of treatment failure after neoadjuvant chemo-radiotherapy before surgery in patients with locally advanced rectal cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 610–615. [Google Scholar] [CrossRef] [PubMed]
  74. Vidal, J.; Casadevall, D.; Bellosillo, B.; Pericay, C.; Garcia-Carbonero, R.; Losa, F.; Layos, L.; Alonso, V.; Capdevila, J.; Gallego, J.; et al. Clinical Impact of Presurgery Circulating Tumor DNA after Total Neoadjuvant Treatment in Locally Advanced Rectal Cancer: A Biomarker Study from the GEMCAD 1402 Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 2890–2898. [Google Scholar] [CrossRef] [PubMed]
  75. De Palma, F.D.E.; Luglio, G.; Tropeano, F.P.; Pagano, G.; D’Armiento, M.; Kroemer, G.; Maiuri, M.C.; De Palma, G.D. The Role of Micro-RNAs and Circulating Tumor Markers as Predictors of Response to Neoadjuvant Therapy in Locally Advanced Rectal Cancer. Int. J. Mol. Sci. 2020, 21, 7040. [Google Scholar] [CrossRef] [PubMed]
  76. Della Vittoria Scarpati, G.; Falcetta, F.; Carlomagno, C.; Ubezio, P.; Marchini, S.; De Stefano, A.; Singh, V.K.; D’Incalci, M.; De Placido, S.; Pepe, S. A specific miRNA signature correlates with complete pathological response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Int. J. Radiat. Oncol. Biol. Phys. 2012, 83, 1113–1119. [Google Scholar] [CrossRef]
  77. Ma, W.; Yu, J.; Qi, X.; Liang, L.; Zhang, Y.; Ding, Y.; Lin, X.; Li, G.; Ding, Y. Radiation-induced microRNA-622 causes radioresistance in colorectal cancer cells by down-regulating Rb. Oncotarget 2015, 6, 15984–15994. [Google Scholar] [CrossRef] [Green Version]
  78. Zhang, J.; Li, S.; Li, L.; Li, M.; Guo, C.; Yao, J.; Mi, S. Exosome and exosomal microRNA: Trafficking, sorting, and function. Genom. Proteom. Bioinform. 2015, 13, 17–24. [Google Scholar] [CrossRef] [Green Version]
  79. Azizian, A.; Kramer, F.; Jo, P.; Wolff, H.A.; Beissbarth, T.; Skarupke, R.; Bernhardt, M.; Grade, M.; Ghadimi, B.M.; Gaedcke, J. Preoperative Prediction of Lymph Node Status by Circulating Mir-18b and Mir-20a During Chemoradiotherapy in Patients with Rectal Cancer. World J. Surg. 2015, 39, 2329–2335. [Google Scholar] [CrossRef]
  80. D’Angelo, E.; Fassan, M.; Maretto, I.; Pucciarelli, S.; Zanon, C.; Digito, M.; Rugge, M.; Nitti, D.; Agostini, M. Serum miR-125b is a non-invasive predictive biomarker of the pre-operative chemoradiotherapy responsiveness in patients with rectal adenocarcinoma. Oncotarget 2016, 7, 28647–28657. [Google Scholar] [CrossRef] [PubMed]
  81. Hiyoshi, Y.; Akiyoshi, T.; Inoue, R.; Murofushi, K.; Yamamoto, N.; Fukunaga, Y.; Ueno, M.; Baba, H.; Mori, S.; Yamaguchi, T. Serum miR-143 levels predict the pathological response to neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer. Oncotarget 2017, 8, 79201–79211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wada, Y.; Shimada, M.; Morine, Y.; Ikemoto, T.; Saito, Y.; Zhu, Z.; Wang, X.; Etxart, A.; Park, Y.; Bujanda, L.; et al. Circulating miRNA Signature Predicts Response to Preoperative Chemoradiotherapy in Locally Advanced Rectal Cancer. JCO Precis. Oncol. 2021, 5, 1788–1801. [Google Scholar] [CrossRef] [PubMed]
  83. Haraksingh, R.R.; Snyder, M.P. Impacts of variation in the human genome on gene regulation. J. Mol. Biol. 2013, 425, 3970–3977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Agostini, M.; Crotti, S.; Bedin, C.; Cecchin, E.; Maretto, I.; D’Angelo, E.; Pucciarelli, S.; Nitti, D. Predictive response biomarkers in rectal cancer neoadjuvant treatment. Front. Biosci. 2014, 6, 110–119. [Google Scholar] [CrossRef] [Green Version]
  85. Sclafani, F.; Chau, I.; Cunningham, D.; Peckitt, C.; Lampis, A.; Hahne, J.C.; Braconi, C.; Tabernero, J.; Glimelius, B.; Cervantes, A.; et al. Prognostic role of the LCS6 KRAS variant in locally advanced rectal cancer: Results of the EXPERT-C trial. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2015, 26, 1936–1941. [Google Scholar] [CrossRef]
  86. Rodriguez-Tomas, E.; Arenas, M.; Gomez, J.; Acosta, J.; Trilla, J.; Lopez, Y.; Arquez, M.; Torres, L.; Araguas, P.; Hernandez-Aguilera, A.; et al. Identification of potential metabolic biomarkers of rectal cancer and of the effect of neoadjuvant radiochemotherapy. PLoS ONE 2021, 16, e0250453. [Google Scholar] [CrossRef]
  87. Lou, X.; Zhou, N.; Feng, L.; Li, Z.; Fang, Y.; Fan, X.; Ling, Y.; Liu, H.; Zou, X.; Wang, J.; et al. Deep Learning Model for Predicting the Pathological Complete Response to Neoadjuvant Chemoradiotherapy of Locally Advanced Rectal Cancer. Front. Oncol. 2022, 12, 807264. [Google Scholar] [CrossRef]
  88. Feng, L.; Liu, Z.; Li, C.; Li, Z.; Lou, X.; Shao, L.; Wang, Y.; Huang, Y.; Chen, H.; Pang, X.; et al. Development and validation of a radiopathomics model to predict pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer: A multicentre observational study. Lancet Digit. Health 2022, 4, e8–e17. [Google Scholar] [CrossRef]
  89. Yi, Y.; Shen, L.; Shi, W.; Xia, F.; Zhang, H.; Wang, Y.; Zhang, J.; Wang, Y.; Sun, X.; Zhang, Z.; et al. Gut Microbiome Components Predict Response to Neoadjuvant Chemoradiotherapy in Patients with Locally Advanced Rectal Cancer: A Prospective, Longitudinal Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 1329–1340. [Google Scholar] [CrossRef]
  90. Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86. [Google Scholar] [CrossRef]
  91. Almeida, A.; Mitchell, A.L.; Boland, M.; Forster, S.C.; Gloor, G.B.; Tarkowska, A.; Lawley, T.D.; Finn, R.D. A new genomic blueprint of the human gut microbiota. Nature 2019, 568, 499–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Ternes, D.; Karta, J.; Tsenkova, M.; Wilmes, P.; Haan, S.; Letellier, E. Microbiome in Colorectal Cancer: How to Get from Meta-omics to Mechanism? Trends Microbiol. 2020, 28, 401–423. [Google Scholar] [CrossRef] [PubMed]
  93. Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The microbiome, cancer, and cancer therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, S.; Wang, Q.; Zhou, C.; Chen, K.; Chang, H.; Xiao, W.; Gao, Y. Colorectal cancer, radiotherapy and gut microbiota. Chin. J. Cancer Res. = Chung-Kuo Yen Cheng Yen Chiu 2019, 31, 212–222. [Google Scholar] [CrossRef]
  95. Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef] [Green Version]
  96. Dinapoli, N.; Barbaro, B.; Gatta, R.; Chiloiro, G.; Casa, C.; Masciocchi, C.; Damiani, A.; Boldrini, L.; Gambacorta, M.A.; 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. 2018, 102, 765–774. [Google Scholar] [CrossRef]
  97. Antunes, J.T.; Ofshteyn, A.; Bera, K.; Wang, E.Y.; Brady, J.T.; Willis, J.E.; Friedman, K.A.; Marderstein, E.L.; Kalady, M.F.; Stein, S.L.; et al. Radiomic Features of Primary Rectal Cancers on Baseline T(2) -Weighted MRI Are Associated With Pathologic Complete Response to Neoadjuvant Chemoradiation: A Multisite Study. J. Magn. Reson. Imaging JMRI 2020, 52, 1531–1541. [Google Scholar] [CrossRef]
  98. Boldrini, L.; Cusumano, D.; Chiloiro, G.; Casa, C.; Masciocchi, C.; Lenkowicz, J.; Cellini, F.; Dinapoli, N.; Azario, L.; Teodoli, S.; et al. Delta radiomics for rectal cancer response prediction with hybrid 0.35 T magnetic resonance-guided radiotherapy (MRgRT): A hypothesis-generating study for an innovative personalized medicine approach. La Radiol. Med. 2019, 124, 145–153. [Google Scholar] [CrossRef] [Green Version]
  99. Boldrini, L.; Lenkowicz, J.; Orlandini, L.C.; Yin, G.; Cusumano, D.; Chiloiro, G.; Dinapoli, N.; Peng, Q.; Casa, C.; Gambacorta, M.A.; et al. Applicability of a pathological complete response magnetic resonance-based radiomics model for locally advanced rectal cancer in intercontinental cohort. Radiat. Oncol. 2022, 17, 78. [Google Scholar] [CrossRef]
  100. Bulens, P.; Couwenberg, A.; Intven, M.; Debucquoy, A.; Vandecaveye, V.; Van Cutsem, E.; D’Hoore, A.; Wolthuis, A.; Mukherjee, P.; Gevaert, O.; et al. Predicting the tumor response to chemoradiotherapy for rectal cancer: Model development and external validation using MRI radiomics. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 2020, 142, 246–252. [Google Scholar] [CrossRef]
  101. Chen, B.Y.; Xie, H.; Li, Y.; Jiang, X.H.; Xiong, L.; Tang, X.F.; Lin, X.F.; Li, L.; Cai, P.Q. MRI-Based Radiomics Features to Predict Treatment Response to Neoadjuvant Chemotherapy in Locally Advanced Rectal Cancer: A Single Center, Prospective Study. Front. Oncol. 2022, 12, 801743. [Google Scholar] [CrossRef] [PubMed]
  102. Chen, H.; Shi, L.; Nguyen, K.N.B.; Monjazeb, A.M.; Matsukuma, K.E.; Loehfelm, T.W.; Huang, H.; Qiu, J.; Rong, Y. MRI Radiomics for Prediction of Tumor Response and Downstaging in Rectal Cancer Patients after Preoperative Chemoradiation. Adv. Radiat. Oncol. 2020, 5, 1286–1295. [Google Scholar] [CrossRef]
  103. Cheng, Y.; Luo, Y.; Hu, Y.; Zhang, Z.; Wang, X.; Yu, Q.; Liu, G.; Cui, E.; Yu, T.; Jiang, X. Multiparametric MRI-based Radiomics approaches on predicting response to neoadjuvant chemoradiotherapy (nCRT) in patients with rectal cancer. Abdom. Radiol. 2021, 46, 5072–5085. [Google Scholar] [CrossRef] [PubMed]
  104. Chiloiro, G.; Cusumano, D.; de Franco, P.; Lenkowicz, J.; Boldrini, L.; Carano, D.; Barbaro, B.; Corvari, B.; Dinapoli, N.; Giraffa, M.; et al. Does restaging MRI radiomics analysis improve pathological complete response prediction in rectal cancer patients? A prognostic model development. La Radiol. Med. 2022, 127, 11–20. [Google Scholar] [CrossRef]
  105. Coppola, F.; Mottola, M.; Lo Monaco, S.; Cattabriga, A.; Cocozza, M.A.; Yuan, J.C.; De Benedittis, C.; Cuicchi, D.; Guido, A.; Rojas Llimpe, F.L.; et al. The Heterogeneity of Skewness in T2W-Based Radiomics Predicts the Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Diagnostics 2021, 11, 795. [Google Scholar] [CrossRef] [PubMed]
  106. Cui, Y.; Yang, X.; Shi, Z.; Yang, Z.; Du, X.; Zhao, Z.; Cheng, X. Radiomics analysis of multiparametric MRI for prediction of pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Eur. Radiol. 2019, 29, 1211–1220. [Google Scholar] [CrossRef]
  107. Cusumano, D.; Boldrini, L.; Yadav, P.; Yu, G.; Musurunu, B.; Chiloiro, G.; Piras, A.; Lenkowicz, J.; Placidi, L.; Romano, A.; et al. Delta radiomics for rectal cancer response prediction using low field magnetic resonance guided radiotherapy: An external validation. Phys. Med. PM Int. J. Devoted Appl. Phys. Med. Biol. Off. J. Ital. Assoc. Biomed. Phys. 2021, 84, 186–191. [Google Scholar] [CrossRef]
  108. Cusumano, D.; Meijer, G.; Lenkowicz, J.; Chiloiro, G.; Boldrini, L.; Masciocchi, C.; Dinapoli, N.; Gatta, R.; Casa, C.; Damiani, A.; et al. A field strength independent MR radiomics model to predict pathological complete response in locally advanced rectal cancer. La Radiol. Med. 2021, 126, 421–429. [Google Scholar] [CrossRef]
  109. Defeudis, A.; Mazzetti, S.; Panic, J.; Micilotta, M.; Vassallo, L.; Giannetto, G.; Gatti, M.; Faletti, R.; Cirillo, S.; Regge, D.; et al. MRI-based radiomics to predict response in locally advanced rectal cancer: Comparison of manual and automatic segmentation on external validation in a multicentre study. Eur. Radiol. Exp. 2022, 6, 19. [Google Scholar] [CrossRef]
  110. Delli Pizzi, A.; Chiarelli, A.M.; Chiacchiaretta, P.; d’Annibale, M.; Croce, P.; Rosa, C.; Mastrodicasa, D.; Trebeschi, S.; Lambregts, D.M.J.; Caposiena, D.; et al. MRI-based clinical-radiomics model predicts tumor response before treatment in locally advanced rectal cancer. Sci. Rep. 2021, 11, 5379. [Google Scholar] [CrossRef]
  111. Fu, J.; Zhong, X.; Li, N.; Van Dams, R.; Lewis, J.; Sung, K.; Raldow, A.C.; Jin, J.; Qi, X.S. Deep learning-based radiomic features for improving neoadjuvant chemoradiation response prediction in locally advanced rectal cancer. Phys. Med. Biol. 2020, 65, 075001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. van Griethuysen, J.J.M.; Lambregts, D.M.J.; Trebeschi, S.; Lahaye, M.J.; Bakers, F.C.H.; Vliegen, R.F.A.; Beets, G.L.; Aerts, H.; Beets-Tan, R.G.H. Radiomics performs comparable to morphologic assessment by expert radiologists for prediction of response to neoadjuvant chemoradiotherapy on baseline staging MRI in rectal cancer. Abdom. Radiol. 2020, 45, 632–643. [Google Scholar] [CrossRef] [PubMed]
  113. Horvat, N.; Veeraraghavan, H.; Khan, M.; Blazic, I.; Zheng, J.; Capanu, M.; Sala, E.; Garcia-Aguilar, J.; Gollub, M.J.; Petkovska, I. MR Imaging of Rectal Cancer: Radiomics Analysis to Assess Treatment Response after Neoadjuvant Therapy. Radiology 2018, 287, 833–843. [Google Scholar] [CrossRef] [Green Version]
  114. Horvat, N.; Veeraraghavan, H.; Nahas, C.S.R.; Bates, D.D.B.; Ferreira, F.R.; Zheng, J.; Capanu, M.; Fuqua, J.L., 3rd; Fernandes, M.C.; Sosa, R.E.; et al. Combined artificial intelligence and radiologist model for predicting rectal cancer treatment response from magnetic resonance imaging: An external validation study. Abdom. Radiol. 2022, 47, 2770–2782. [Google Scholar] [CrossRef] [PubMed]
  115. Jayaprakasam, V.S.; Paroder, V.; Gibbs, P.; Bajwa, R.; Gangai, N.; Sosa, R.E.; Petkovska, I.; Golia Pernicka, J.S.; Fuqua, J.L., 3rd; Bates, D.D.B.; 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. 2022, 32, 971–980. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Y.; Liu, W.; Pei, Q.; Zhao, L.; Gungor, C.; Zhu, H.; Song, X.; Li, C.; Zhou, Z.; Xu, Y.; et al. Predicting pathological complete response by comparing MRI-based radiomics pre- and postneoadjuvant radiotherapy for locally advanced rectal cancer. Cancer Med. 2019, 8, 7244–7252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Liu, Z.; Zhang, X.Y.; Shi, Y.J.; Wang, L.; Zhu, H.T.; Tang, Z.; Wang, S.; Li, X.T.; Tian, J.; Sun, Y.S. Radiomics Analysis for Evaluation of Pathological Complete Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 7253–7262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Mbanu, P.; Saunders, M.P.; Mistry, H.; Mercer, J.; Malcomson, L.; Yousif, S.; Price, G.; Kochhar, R.; Renehan, A.G.; van Herk, M.; et al. Clinical and radiomics prediction of complete response in rectal cancer pre-chemoradiotherapy. Phys. Imaging Radiat. Oncol. 2022, 23, 48–53. [Google Scholar] [CrossRef]
  119. Nardone, V.; Reginelli, A.; Grassi, R.; Vacca, G.; Giacobbe, G.; Angrisani, A.; Clemente, A.; Danti, G.; Correale, P.; Carbone, S.F.; et al. Ability of Delta Radiomics to Predict a Complete Pathological Response in Patients with Loco-Regional Rectal Cancer Addressed to Neoadjuvant Chemo-Radiation and Surgery. Cancers 2022, 14, 3004. [Google Scholar] [CrossRef]
  120. Nie, K.; Shi, L.; Chen, Q.; Hu, X.; Jabbour, S.K.; Yue, N.; Niu, T.; Sun, X. Rectal Cancer: Assessment of Neoadjuvant Chemoradiation Outcome based on Radiomics of Multiparametric MRI. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 5256–5264. [Google Scholar] [CrossRef]
  121. Pang, X.; Wang, F.; Zhang, Q.; Li, Y.; Huang, R.; Yin, X.; Fan, X. A Pipeline for Predicting the Treatment Response of Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancer Using Single MRI Modality: Combining Deep Segmentation Network and Radiomics Analysis Based on "Suspicious Region". Front. Oncol. 2021, 11, 711747. [Google Scholar] [CrossRef] [PubMed]
  122. Peterson, K.J.; Simpson, M.T.; Drezdzon, M.K.; Szabo, A.; Ausman, R.A.; Nencka, A.S.; Knechtges, P.M.; Peterson, C.Y.; Ludwig, K.A.; Ridolfi, T.J. Predicting Neoadjuvant Treatment Response in Rectal Cancer Using Machine Learning: Evaluation of MRI-Based Radiomic and Clinical Models. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 2022. [Google Scholar] [CrossRef] [PubMed]
  123. Petkovska, I.; Tixier, F.; Ortiz, E.J.; Golia Pernicka, J.S.; Paroder, V.; Bates, D.D.; Horvat, N.; Fuqua, J.; Schilsky, J.; Gollub, M.J.; et al. Clinical utility of radiomics at baseline rectal MRI to predict complete response of rectal cancer after chemoradiation therapy. Abdom. Radiol. 2020, 45, 3608–3617. [Google Scholar] [CrossRef] [PubMed]
  124. Shaish, H.; Aukerman, A.; Vanguri, R.; Spinelli, A.; Armenta, P.; Jambawalikar, S.; Makkar, J.; Bentley-Hibbert, S.; Del Portillo, A.; Kiran, R.; et al. Radiomics of MRI for pretreatment prediction of pathologic complete response, tumor regression grade, and neoadjuvant rectal score in patients with locally advanced rectal cancer undergoing neoadjuvant chemoradiation: An international multicenter study. Eur. Radiol. 2020, 30, 6263–6273. [Google Scholar] [CrossRef]
  125. Shi, L.; Zhang, Y.; Nie, K.; Sun, X.; Niu, T.; Yue, N.; Kwong, T.; Chang, P.; Chow, D.; Chen, J.H.; et al. Machine learning for prediction of chemoradiation therapy response in rectal cancer using pre-treatment and mid-radiation multi-parametric MRI. Magn. Reson. Imaging 2019, 61, 33–40. [Google Scholar] [CrossRef]
  126. Shin, J.; Seo, N.; Baek, S.E.; Son, N.H.; Lim, J.S.; Kim, N.K.; Koom, W.S.; Kim, S. MRI Radiomics Model Predicts Pathologic Complete Response of Rectal Cancer Following Chemoradiotherapy. Radiology 2022, 303, 351–358. [Google Scholar] [CrossRef]
  127. Song, M.; Li, S.; Wang, H.; Hu, K.; Wang, F.; Teng, H.; Wang, Z.; Liu, J.; Jia, A.Y.; Cai, Y.; et al. MRI radiomics independent of clinical baseline characteristics and neoadjuvant treatment modalities predicts response to neoadjuvant therapy in rectal cancer. Br. J. Cancer 2022, 127, 249–257. [Google Scholar] [CrossRef]
  128. Tang, B.; Lenkowicz, J.; Peng, Q.; Boldrini, L.; Hou, Q.; Dinapoli, N.; Valentini, V.; Diao, P.; Yin, G.; Orlandini, L.C. Local tuning of radiomics-based model for predicting pathological response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. BMC Med. Imaging 2022, 22, 44. [Google Scholar] [CrossRef]
  129. Wan, L.; Peng, W.; Zou, S.; Ye, F.; Geng, Y.; Ouyang, H.; Zhao, X.; Zhang, H. MRI-based delta-radiomics are predictive of pathological complete response after neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Acad. Radiol. 2021, 28 (Suppl. 1), S95–S104. [Google Scholar] [CrossRef]
  130. Wei, Q.; Chen, Z.; Tang, Y.; Chen, W.; Zhong, L.; Mao, L.; Hu, S.; Wu, Y.; Deng, K.; Yang, W.; et al. External validation and comparison of MR-based radiomics models for predicting pathological complete response in locally advanced rectal cancer: A two-centre, multi-vendor study. Eur. Radiol. 2022. [Google Scholar] [CrossRef]
  131. Yi, X.; Pei, Q.; Zhang, Y.; Zhu, H.; Wang, Z.; Chen, C.; Li, Q.; Long, X.; Tan, F.; Zhou, Z.; et al. MRI-Based Radiomics Predicts Tumor Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Front. Oncol. 2019, 9, 552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bordron, A.; Rio, E.; Badic, B.; Miranda, O.; Pradier, O.; Hatt, M.; Visvikis, D.; Lucia, F.; Schick, U.; Bourbonne, V. External Validation of a Radiomics Model for the Prediction of Complete Response to Neoadjuvant Chemoradiotherapy in Rectal Cancer. Cancers 2022, 14, 1079. [Google Scholar] [CrossRef] [PubMed]
  133. Capelli, G.; Campi, C.; Bao, Q.R.; Morra, F.; Lacognata, C.; Zucchetta, P.; Cecchin, D.; Pucciarelli, S.; Spolverato, G.; Crimi, F. 18F-FDG-PET/MRI texture analysis in rectal cancer after neoadjuvant chemoradiotherapy. Nucl. Med. Commun. 2022, 43, 815–822. [Google Scholar] [CrossRef] [PubMed]
  134. Giannini, V.; Mazzetti, S.; Bertotto, I.; Chiarenza, C.; Cauda, S.; Delmastro, E.; Bracco, C.; Di Dia, A.; Leone, F.; Medico, E.; et al. Predicting locally advanced rectal cancer response to neoadjuvant therapy with (18)F-FDG PET and MRI radiomics features. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 878–888. [Google Scholar] [CrossRef]
  135. Bibault, J.E.; Giraud, P.; Housset, M.; Durdux, C.; Taieb, J.; Berger, A.; Coriat, R.; Chaussade, S.; Dousset, B.; Nordlinger, B.; et al. Deep Learning and Radiomics predict complete response after neo-adjuvant chemoradiation for locally advanced rectal cancer. Sci. Rep. 2018, 8, 12611. [Google Scholar] [CrossRef]
  136. Hamerla, G.; Meyer, H.J.; Hambsch, P.; Wolf, U.; Kuhnt, T.; Hoffmann, K.T.; Surov, A. Radiomics Model Based on Non-Contrast CT Shows No Predictive Power for Complete Pathological Response in Locally Advanced Rectal Cancer. Cancers 2019, 11, 1680. [Google Scholar] [CrossRef] [Green Version]
  137. Lovinfosse, P.; Polus, M.; Van Daele, D.; Martinive, P.; Daenen, F.; Hatt, M.; Visvikis, D.; Koopmansch, B.; Lambert, F.; Coimbra, C.; et al. FDG PET/CT radiomics for predicting the outcome of locally advanced rectal cancer. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 365–375. [Google Scholar] [CrossRef]
  138. Lutsyk, M.; Gourevich, K.; Keidar, Z. Complete Pathologic Response Prediction by Radiomics Wavelets Features of Unenhanced CT Simulation Images in Locally Advanced Rectal Cancer Patients after Neoadjuvant Chemoradiation. Isr. Med. Assoc. J. IMAJ 2021, 23, 805–810. [Google Scholar] [CrossRef]
  139. Mao, Y.; Pei, Q.; Fu, Y.; Liu, H.; Chen, C.; Li, H.; Gong, G.; Yin, H.; Pang, P.; Lin, H.; et al. Pre-Treatment Computed Tomography Radiomics for Predicting the Response to Neoadjuvant Chemoradiation in Locally Advanced Rectal Cancer: A Retrospective Study. Front. Oncol. 2022, 12, 850774. [Google Scholar] [CrossRef]
  140. Shen, W.C.; Chen, S.W.; Wu, K.C.; Lee, P.Y.; Feng, C.L.; Hsieh, T.C.; Yen, K.Y.; Kao, C.H. Predicting pathological complete response in rectal cancer after chemoradiotherapy with a random forest using (18)F-fluorodeoxyglucose positron emission tomography and computed tomography radiomics. Ann. Transl. Med. 2020, 8, 207. [Google Scholar] [CrossRef]
  141. Yuan, Z.; Frazer, M.; Zhang, G.G.; Latifi, K.; Moros, E.G.; Feygelman, V.; Felder, S.; Sanchez, J.; Dessureault, S.; Imanirad, I.; et al. CT-based radiomic features to predict pathological response in rectal cancer: A retrospective cohort study. J. Med. Imaging Radiat. Oncol. 2020, 64, 444–449. [Google Scholar] [CrossRef] [PubMed]
  142. Zhuang, Z.; Liu, Z.; Li, J.; Wang, X.; Xie, P.; Xiong, F.; Hu, J.; Meng, X.; Huang, M.; Deng, Y.; et al. Radiomic signature of the FOWARC trial predicts pathological response to neoadjuvant treatment in rectal cancer. J. Transl. Med. 2021, 19, 256. [Google Scholar] [CrossRef] [PubMed]
  143. Rahma, O.E.; Yothers, G.; Hong, T.S.; Russell, M.M.; You, Y.N.; Parker, W.; Jacobs, S.A.; Colangelo, L.H.; Lucas, P.C.; Gollub, M.J.; et al. Use of Total Neoadjuvant Therapy for Locally Advanced Rectal Cancer: Initial Results From the Pembrolizumab Arm of a Phase 2 Randomized Clinical Trial. JAMA Oncol. 2021, 7, 1225–1230. [Google Scholar] [CrossRef] [PubMed]
  144. Chalabi, M.; Fanchi, L.F.; Dijkstra, K.K.; Van den Berg, J.G.; Aalbers, A.G.; Sikorska, K.; Lopez-Yurda, M.; Grootscholten, C.; Beets, G.L.; Snaebjornsson, P.; et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat. Med. 2020, 26, 566–576. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of MRI-only-based radiomics prediction model.
Table 1. Summary of MRI-only-based radiomics prediction model.
Study
Year
Studied Modality of Imaging
Timing
External Validation
-
Prospective Validation
Timing for Data Acquisition
-
Timing of Analysis: Prospective/Retrospective
Cohort SizeDelineationAssessment of RobustnessExtraction Software
-
Statistical Approach
Predicted OutcomeCorrection for Site HeterogeneityType of NAT: CRT vs. TNTpCR RateAUCAccuracyPPVAUCAccuracyPPV
TrainingValidation
Antunes et al. [97]
2020
MRI: T2
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 60, validation: 44Manual: single readerYIn-house
-
Random forest
pCRNCRTTraining: 21.7%, validation: 22.7%0.7070.5% for the overall cohortN/A0.71N/AN/A
Boldrini et al. [98]
2019
MRI: T2/T1S
longitudinal
N-4 cross fold validation
-
N
Retrospective
-
Retrospective
16Manual-Cooperation of 2 radiation oncologistsNModdicom (R)
-
Wilcoxon–Mann–Whitney
pCRN/ACRT12.5%0.93N/AN/AN/AN/AN/A
Boldrini et al. [99]
2022
MRI: T2
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 162 (see Dinapoli et al., 2018); testing: 59Manual: single reader, with review by an independent readerNModdicom (R)
-
GLM
pCRNTraining: see Dinapoli et al., 2018; validation: CapOX followed by CRTTraining 28%; validation cohort: 16.9%0.73N/AN/A0.8365.028.0
Bulens et al. [100]
2020
MRI: T2/DWI/ADC
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 70, validation: 55Manual consensus of 1 radiation oncologist and 1 radiologistNIn-house
-
LASSO logistic regression model
pCRNCRTTraining: 17%, validation: 15%0.8682.077.00.8784.0100.0
Chen et al. [101]
2022
MRI: T2
baseline
Y
-
N
Prospective
-
Retrospective
Training: 91, validation: 46N/ANPyRadiomics
-
Logistic regression
pCRNCRTTraining: 16.5%, validation: 19.5% *0.96N/AN/A0.87N/AN/A
Chen et al. [102]
2020
MRI: T2
longitudinal (pre/post)
Y
-
N
Retrospective
-
Retrospective
Training: 26, validation: 13Manual: single readerN/AIBEX
-
SVM
pCRN/ACRT25.6%N/A96.2%N/AN/A because of missing dataN/AN/A
Cheng et al. [103]
2021
MRI: T2/T2FS/T1
baseline
Y
-
N
Retrospective
-
Retrospective
Training = 128, validation: 65Manual: single reader (+ inter-reader variability assessment on 30 patients)YPyRadiomics
-
Logistic regression
pCRN/ACRTTraining: 15.6%, validation: 16.9%0.9693.8N/A0.9184.6N/A
Chiloiro et al. ** [104]
2022
MRI: N/A
post-CRT
N
-
N
Retrospective
-
Retrospective
144N/AN/AN/A
-
N/A
pCRN/ACRTN/A0.84N/AN/AN/AN/AN/A
Coppola et al. [105]
2021
MRI: T2/DWI
baseline
N
-
N
Retrospective
-
Retrospective
40Manual: Single readerN/AN/A
-
Random forest
near pCR + pCRN/ACRT37.5% *0.90N/A80.0N/AN/AN/A
Cui et al. [106]
2018
MRI: T2/T1/ADC
Baseline
Y
-
N
Retrospective
-
Retrospective
Training: 131; validation: 55ManualN/AAnalysis kit-GE
-
Logistic regression
pCRN/ACRTTraining: 16.8%, validation: 16.4%0.9587.8N/A0.9794.5N/A
Cusumano et al. [107]
2021
MRI (low field): T2/T1S
longitudinal
Y
-
N
Retrospective
-
Retrospective
Training: 16, validation: 43Manual-cooperation of 2 radiation oncologistsNModdicom (R)
-
Multiple logistic regression
pCRN/ACRTTraining: 12.5%, validation: 27%0.93N/AN/AN/A77.0N/A
Cusumano et al. [108]
2021
MRI: T2?
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective: 2 institutions
195 (institution1: 136; institution2: 59)Manual: nb readers?N/AModdicom (R)
-
Multiple logistic regression
pCRNCRTInstitution1: 22%; institution 2: 25%0.72 (after cross validation)N/AN/AN/AN/AN/A
Defeudis et al. [109]
2022
MRI: T2/ADC
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 47, testing: 20, validation: 28Manual: 3 readers + assessment by 2 supplementary readersNPyRadiomics
-
SVM
pCRNCRT16.8%0.9083.074.00.6168.075.0
Automatic segmentationNPyRadiomics
-
SVM
pCRNCRT16.8%0.8678.071.00.8175.075.0
Delli Pizzi et al. [110]
2021
MRI: T2
baseline
N
-
N
Retrospective
-
Retrospective
72Manual: 2 readersYPyRadiomics
-
Partial least-squares regression
near pCR + pCRN/ACRT47.2%*0.79N/AN/AN/AN/AN/A
Dinapoli et al. [96]
2018
MRI: T2
baseline
Y
-
N
Training: retrospective, validation 1: retrospective, validation 2: prospective
-
Retrospective
Training: 162, validation cohort 1: 34, validation cohort 2: 25Manual-training: cooperation of 2 radiation oncologists + 2 radiologists, validation 1: cooperation of 2 different radiation oncologists and radiologists, validation 2: same as trainingNModdicom (R)
-
GLM
pCRN/ACRTTraining 28%; validation cohort 1: 21%, validation cohort 2: 28%0.73N/AN/ACohort 1: 0.75, cohort 2: 0.79N/AN/A
Fu et al. [111]
2020
MRI: DWI
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective
43Manual: single readerN/APyRadiomics
-
Neural network
near pCR + pCRNCRT51.2%0.73 (after cross validation)N/AN/AN/AN/AN/A
Van Griethuysen et al. [112]
2020
MRI: T2/DWI/ADC
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 86, validation: 47Semi-automatic + manual refinementNPyRadiomics
-
Logistic regression
pCRNCRTTraining: 20.9%, validation: 21.3%0.71N/AN/A0.77N/AN/A
Horvat et al. [113]
2018
MRI: T2
post-CRT
N
-
N
Retrospective
-
Retrospective
114Manual: consensus of 2 readersN/AIn-house
-
Random forest
pCRNCRT18%0.93N/A72.0%N/AN/AN/A
Horvat et al. ** [114]
2022
MRI: T2
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 114? validation: 50Manual: 2 readersN/AN/A
-
N/A
pCRN/ACRTN/AN/AN/AN/A0.83N/A57.0
Jayaprakasam et al. ** [115]
2021
MRI: T2
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective
236Manual: 3 readersN/ACERR
-
SVM
pCRN/ACRTN/A0.8983.952.5N/AN/AN/A
Li et al. [116]
2019
MRI: T2
longitudinal (pre/post)
Y
-
N
Retrospective
-
Retrospective
Training: 87, validation: 44Manual: single reader, with review by an independent readerNIBEX
-
LASSO logistic regression model
pCRN/ACRTTraining: 20.7%, validation: 20.5%0.92N/AN/A0.87N/AN/A
Liu et al. [117]
2017
MRI: T2/DWI
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 16, validation: 43Manual: 2 readersYMATLAB workpackage
-
SVM
pCRN/ACRTTraining: 17.1%, validation: 17.1%0.9796.191.70.9894.390.0
Mbanu et al. [118]
2022
MRI: T2
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 200, validation: 104Manual: 2 readersYPyRadiomics
-
Logistic regression
cCRNCRT50% (selected patients for matching cohorts)0.76N/AN/A0.68N/AN/A
Nardone et al. [119]
2022
MRI: T2/DWI/ADC
longitudinal (pre/post)
Y
-
N
Retrospective
-
Retrospective
Training: 37, 2 validation cohorts: 33 + 30Manual: 2 readers + assessment by 2 supplementary readersYLife
X-
Logistic regression
pCRNCRTTraining: 27%; validation cohort 1: 18%, validation cohort 2: 17%0.8773.050.0Cohort 1: 0.92, cohort 2: 0.88Cohort 1: 72.7, cohort 2: 80.0Cohort 1: 40.0, cohort 2: 44.4
Nie et al. [120]
2016
MRI: T2/T1/DWI/DCE
Baseline
N-4 cross fold validation
-
N
Retrospective
-
Retrospective
48Manual: single readerN/AN/A
-
Neural network
pCRN/ACRT23%0.89N/AN/AN/AN/AN/A
Pang et al. [121]
2021
MRI: T2
post-CRT
Y
-
N
Retrospective
-
Retrospective
Training: 107, internal validation: 46, external validation: 34Automatic segmentationN/APyRadiomics
-
SVM
pCRNCRTTraining: 33.6%, internal validation: 17.4%, external validation: 17.6%0.9286.0N/AInternal validation: 0.83, external validation: 0.82Internal validation: 80.4, external validation: 85.3N/A
Peterson et al. ** [122]
2022
MRI: N/A
baseline
N
-
N
Retrospective
-
Retrospective
131N/AN/AN/A
-
Machine learning?
pCRN/ACRT26.7%0.73N/AN/AN/AN/AN/A
Petkovska et al. [123]
2020
MRI: T2
baseline
Y (sub-set of patients delineated by different readers)
-
N
Retrospective
-
Retrospective
Training: 102, validation: 66 (out of the training cohort but with different delineations)Manual: 1 reader (+ 2 readers for validation)NCERR
-
SVM
pCRNCRT19%0.75N/AN/AReader 1: 0.75, reader 2: 0.71N/AN/A
Shaish et al. [124]
2020
MRI: T2
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective
Training: 112, validation: 23Manual: single readerYPyRadiomics
-
Logistic regression
pCRNCRT15%0.80N/AN/AN/AN/AN/A
Shi et al. [125]
2019
MRI: T2/T1/ADC/DCE
longitudinal (pre/mid)
N-internal cross validation
-
N
Retrospective
-
Retrospective
35ManualNN/A
-
Neural network
pCRN/ACRT22.2%0.86N/AN/AN/AN/AN/A
Shin et al. [126]
2022
MRI: T2/ADC
post-CRT
Y
-
N
Retrospective
-
Retrospective
Training: 592, validation: 306Manual: single-reader assessment by 1 supplementary reader (+ inter-reader variability assessment on 40 patients)YPyRadiomics
-
LASSO logistic regression model
pCRNCRTTraining: 19.3%, validation: 24.5%0.89N/AN/A0.82N/A46.3
Song et al. [127]
2022
MRI: T2
baseline
Y
-
N
Retrospective
-
Retrospective
674 patientsManual: 4 readers and 2-step validationNN/A
-
SVM
pCRNCRT25.8%0.9996.496.10.7993.394.0
Tang et al. [128]
2022
MRI: T2
baseline
Y
-
N
Training: see Dinapoli et al. 2018; validation: retrospective
-
Training: see Dinapoli et al., 2018; validation: retrospective
Training: see Dinapoli et al., 2018; validation: 88Manual: single readerN/AModdicom (R)
-
Logistic regression
pCRNCRTTraining: see Dinapoli et al., 2018; validation: 13.6%0.93N/AN/A0.93N/AN/A
Wan et al. [129]
2020
MRI: T2/DWI
longitudinal (pre/post)
Y
-
N
Retrospective
-
Retrospective
Training: 116, validation: 49Manual: single-reader assessment by 1 supplementary readerN/ARadcloud
-
Logistic regression
pCRN/ACRTTraining: 16.4%, validation: 16.3%0.91N/AN/A0.91N/AN/A
Wei et al. ** [130]
2022
MRI: N/A
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 100, validation: 51ManualN/AN/A
-
Random forest
pCRN/ACRTN/A0.9176.0N/A0.8777.3N/A
Yi et al. [131]
2019
MRI: T2
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 93, validation: 40Manual: 2 readersYMaZda
-
SVM
pCRNCRT23.9%0.91N/AN/A0.87N/AN/A
Abbreviations: *: near pCR when indicated, **: full text not available, nb: number, NAT: neoadjuvant treatment, CRT: chemoradiotherapy, TNT: total neoadjuvant treatment, pCR: pathological complete response, AUC: area under the curve, PPV: positive predictive value, N/A: not available, Y: yes, N: no, MRI: magnetic resonance imaging, DWI: diffusion-weighted imaging, DCE: dynamic contrast enhanced, ADC: Apparent Diffusion Coefficient, T1S: T1-Star, T2FS: T2 fat saturated, SVM: support vector machine, LASSO: least absolute shrinkage and selection operator, GLM: generalized linear model.
Table 2. Summary of MRI-combined-based radiomics prediction model.
Table 2. Summary of MRI-combined-based radiomics prediction model.
Study
Year
Studied Modality of Imaging
Timing
External Validation
-
Prospective Validation
Timing for Data Acquisition
-
Timing of Analysis: Prospective/Retrospective
Cohort SizeDelineation: Manual/Auto, nb ReaderAssessment of RobustnessExtraction Software
-
Statistical Approach
Predicted OutcomeCorrection for Site HeterogeneityType of NAT: CRT vs. TNTpCR RateAUCAccuracyPPVAUCAccuracyPPV
TrainingValidation
Bordron et al. [132]
2022
MRI: T2/DWI
CE-CT
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 64, testing: 60Manual: single reader (+ inter-reader variability assessment on 25 patients)YMIRAS
-
Neural network: multilayer perceptron
pCRY (Combat)CRTTraining: 14%, validation: 8%0.9590.045.00.8185.544.4
Capelli et al. [133]
2022
MRI: T2/ADC
PET
baseline
N/A
-
N
Retrospective
-
Retrospective
50Manual:consensus of 2 radiation oncologistsN/APMOD
-
Logistic regression
pCRN/ACRT500.8674.0N/AN/AN/AN/A
Feng et al. [88]
2022
MRI: T2/T1/DWI
pathological slides
baseline
Y
-
Y
Retrospective: training + validation + prospective validation
-
Retrospective: training + validation + prospective validation
Training: 303, validation 1 + 2: 480 + 150, prospective: 100ManualN/APyRadiomics
-
Neural network: VGG-19
pCRNCRTTraining: 28%, validation 1/2: 22%/20%, prospective: 23%0.87N/A0.64Validation 1: 0.86, validation 2: 0.87, prospective: 0.81N/AValidation 1: 0.50, validation 2: 0.47, prospective: 0.51
Giannini et al. [134]
2019
MRI: T2/DWI
PET
baseline
N
-
N
Prospective
-
Retrospective
52Semi-automaticN/AMATLAB workpackage
-
Logistic regression
near pCR + pCRN/ACRT42.3%0.86N/AN/AN/AN/AN/A
Abbreviations: nb: number, NAT: neoadjuvant treatment, CRT: chemoradiotherapy, TNT: total neoadjuvant treatment, pCR: pathological complete response, AUC: area under the curve, PPV: positive predictive value, N/A: not available, Y: yes, N: no, MRI: magnetic resonance imaging, DWI: diffusion-weighted imaging, CE: contrast enhanced, CT: computed tomography, ADC: apparent diffusion coefficient, PET: positron emission tomography.
Table 3. Summary of non-MRI-based radiomics prediction model.
Table 3. Summary of non-MRI-based radiomics prediction model.
Study
Year
Studied Modality of Imaging
Timing
External Validation
-
Prospective Validation
Timing for Data Acquisition
-
Timing of Analysis: Prospective/Retrospective
Cohort SizeDelineation: Manual/Auto, nb ReaderAssessment of robustnessExtraction Software
-
Statistical Approach
Predicted OutcomeCorrection for Site HeterogeneityType of NAT: CRT vs. TNTpCR RateAUCAccuracyPPVAUCAccuracyPPV
TrainingValidation
Bibault et al. [135]
2018
NonCE-CT
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective
95Manual: 2 readersNIBEX
-
Neural network: tensor flow
pCRNCRT23.1%NaNNaNNaN0.7280.068.0
Hamerla et al. [136]
2019
NonCE-CT
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective
169Manual: consensus of 2 radiation oncologistsNPyRadiomics
-
Random forest
pCRN/ACRT13%NaN87.0 (before correction for imbalanced data); 50.0 (after correction for imbalanced data)NaNNaNNaNNaN
Lovinfosse et al. [137]
2017
PET
baseline
N
-
N
Retrospective
-
Retrospective
86Semi-automaticN/AIn-house
-
Logistic regression
pCRN/ACRT9.1%NaNNaNNaNNaNNaNNaN
Lutsyk et al. [138]
2021
NonCE-CT
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 98, validation: 42ManualNPyRadiomics
-
Neural network: multilayer perceptron
pCRNCRT27.1%0.8798.098.6NaN67.077.0
Mao et al. [139]
2022
NonCE-CT
baseline
Y
-
N
Retrospective
-
Retrospective
Training: 151, validation: 65Manual: 2 readers + assessment by 2 supplementary readersYMaZda
-
Logistic regression
pCRN/ACRTTraining: 19.9%, validation: 21.5%0.9387.060.50.8786.070.6
Shen et al. [140]
2020
PET
baseline
N-internal cross validation
-
N
Retrospective
-
Retrospective
169ManualNIn-house
-
Random forest
pCRN/ACRT13%0.87–0.97 (before cross validation)89.0 (after cross validation)67.0 (after cross validation)NaNNaNNaN
Yuan et al. [141]
2020
NonCE-CT
baseline
Y
-
N
Retrospective
-
Retrospective
Training = 60, validation: 31ManualNIn-house
-
Random forest > SVM
pCRNCRTTraining: 23.4%, validation: 19.4%NaNNaNNaNNaN83.9NaN
Zhuang et al. [142]
2021
CE-CT
baseline
Y
-
N
Training + validation: post-hoc analysis of the FORWAC trial
-
Retrospective
Training: 113, validation: 64Manual: 2 readersYPyRadiomics
-
GLM
pCRNFORWAC trial: 3 arms: arm A: LV5FU2 + RT, arm B: FOLFOX + RT, arm C: FOLFOX without RTTraining: 17.7%, validation: 17.2%1.000.97NaN0.8281.0NaN
Abbreviations: nb: number, NAT: neoadjuvant treatment, CRT: chemoradiotherapy, TNT: total neoadjuvant treatment, pCR: pathological complete response, AUC: area under the curve, PPV: positive predictive value, N/A: not available, Y: yes, N: no, MRI: magnetic resonance imaging, CE: contrast enhanced, CT: computed tomography, PET: positron emission tomography, GLM: generalized linear model.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bourbonne, V.; Schick, U.; Pradier, O.; Visvikis, D.; Metges, J.-P.; Badic, B. Radiomics Approaches for the Prediction of Pathological Complete Response after Neoadjuvant Treatment in Locally Advanced Rectal Cancer: Ready for Prime Time? Cancers 2023, 15, 432. https://doi.org/10.3390/cancers15020432

AMA Style

Bourbonne V, Schick U, Pradier O, Visvikis D, Metges J-P, Badic B. Radiomics Approaches for the Prediction of Pathological Complete Response after Neoadjuvant Treatment in Locally Advanced Rectal Cancer: Ready for Prime Time? Cancers. 2023; 15(2):432. https://doi.org/10.3390/cancers15020432

Chicago/Turabian Style

Bourbonne, Vincent, Ulrike Schick, Olivier Pradier, Dimitris Visvikis, Jean-Philippe Metges, and Bogdan Badic. 2023. "Radiomics Approaches for the Prediction of Pathological Complete Response after Neoadjuvant Treatment in Locally Advanced Rectal Cancer: Ready for Prime Time?" Cancers 15, no. 2: 432. https://doi.org/10.3390/cancers15020432

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

Article Metrics

Back to TopTop