Skip to main content
  • Primary research
  • Open access
  • Published:

Application of artificial intelligence in a real-world research for predicting the risk of liver metastasis in T1 colorectal cancer

Abstract

Background

Liver is the most common metastatic site of colorectal cancer (CRC) and liver metastasis (LM) determines subsequent treatment as well as prognosis of patients, especially in T1 patients. T1 CRC patients with LM are recommended to adopt surgery and systematic treatments rather than endoscopic therapy alone. Nevertheless, there is still no effective model to predict the risk of LM in T1 CRC patients. Hence, we aim to construct an accurate predictive model and an easy-to-use tool clinically.

Methods

We integrated two independent CRC cohorts from Surveillance Epidemiology and End Results database (SEER, training dataset) and Xijing hospital (testing dataset). Artificial intelligence (AI) and machine learning (ML) methods were adopted to establish the predictive model.

Results

A total of 16,785 and 326 T1 CRC patients from SEER database and Xijing hospital were incorporated respectively into the study. Every single ML model demonstrated great predictive capability, with an area under the curve (AUC) close to 0.95 and a stacking bagging model displaying the best performance (AUC = 0.9631). Expectedly, the stacking model exhibited a favorable discriminative ability and precisely screened out all eight LM cases from 326 T1 patients in the outer validation cohort. In the subgroup analysis, the stacking model also demonstrated a splendid predictive ability for patients with tumor size ranging from one to50mm (AUC = 0.956).

Conclusion

We successfully established an innovative and convenient AI model for predicting LM in T1 CRC patients, which was further verified in the external dataset. Ultimately, we designed a novel and easy-to-use decision tree, which only incorporated four fundamental parameters and could be successfully applied in clinical practice.

Introduction

Colorectal cancer (CRC) is universally acknowledged as one of the most prevalent gastrointestinal tract malignancies with considerably high morbidity and mortality, drawing more and more attention annually [1,2,3]. In 2/3 of CRC patients, metastasis is commonly recognized as both a pivotal clinical feature and a risk factor of high mortality for intractable CRC [4]. During the progression of CRC, over 50% of patients tend to develop liver metastasis (LM) which is the predominant contributor to unfavorable prognosis of CRC [4, 5]. Synchronous LM is determined at the time of diagnosis and 15–25% CRC patients had synchronous LM [6, 7].

Endoscopic therapy is a widely accepted and adopted as a valid therapeutic method for T1 CRC patients. Nonetheless, for early CRC patients with LM, conventional surgical excision, neoadjuvant chemotherapy and radiofrequency ablation are the most effective and recommended treatments, which significantly prolong the 5-year overall survival (OS) rate of CRC patients [8, 9]. However, considering the inferior early screening methods, approximately 90% of CRC patients with LM fail to be diagnosed precisely in the early stage and thus undergo incomplete endoscopic resection, which ultimately gives rise to undesirable clinical outcomes [10, 11]. Although scholars and academicians have conducted abundant in-depth researches on metastasis-related signatures in vivo and vitro, a satisfactory predictive model of LM for CRC in the early stage is still lacking [12,13,14]. Consequently, we aimed at developing an easy-to-use model to predict the risk of LM for patients in the early stage of CRC accurately and robustly.

Currently, there exists an upregulating and irreversible tendency of discipline integration between medical science and artificial intelligence (AI) [15,16,17]. Besides, both depth and breadth of the discipline integration have been significantly enhanced [14, 15]. Researchers employed machine learning (ML) as the breaking point in solving the complicated issue of CRC clinical prediction and acquired plentiful significant breakthroughs [18,19,20]. Nevertheless, these findings simply shed light on the intriguing area of T1 CRC with lymph node metastasis which resembles a virgin land to be further explored by utilizing ML. Given that the majority of previous investigations merely concentrated on the public database when studying the apparent discrepancy among diverse populations, limitations ineluctably appeared. Consequently, clinical data involving the real outer validation is of vital significance to construct a superior prediction model.

In the study, we developed a comprehensive recognition model via adopting AI and ML algorithms, which could remarkably promote the identification of T1 CRC with LM and improve the prognosis of these patients in clinical practice. In addition, the predictive model was constructed via utilizing clinically common and accessible parameters, and further validated in an independent CRC cohort.

Materials and methods

Clinical sample collection

An open-access and publicly available CRC cohort was retrieved from Surveillance, Epidemiology, and End Results (SEER) Program database in the U.S. National Cancer Institute. The CRC cohort functioned as a powerful resource for investigators to comprehensively comprehend the natural history of CRC and significantly ameliorated the healthcare quality for CRC patients [21, 22]. An additional outer validation cohort of CRC patients who underwent surgery from 2010 to 2021 was obtained from Xijing hospital. The CRC cohort's inclusive criteria were demonstrated as follows: (1) the primary diagnosis was CRC; (2) patients were diagnosed with T1 CRC; (3) liver reexamination was completed within six months of diagnosis; (4) patients with sufficient clinical data. Additionally, exclusive criteria were exhibited as follows: (1) patients who have undergone neoadjuvant radiotherapy; (2) metachronous liver metastases (after diagnosis); (3) comorbidity with other tumors; (4) comorbidity with serious cardiopulmonary disease. Written and informed consent was obtained from all participants. All aspects of the clinical cohort study were evaluated by and included in the Institutional Ethics Committee of Xijing Hospital.

Study population

T1 CRC is defined as a category of tumor that invades only the submucosa, regardless of the presence or absence of lymph node metastasis (LNM). Utilizing the SEER database which employed the 7th cancer TNM stages of the American Joint Committee, we analyzed the data of all patients diagnosed with T1 CRC from 2010 to 2016. Primary demographic data, tumor information and laboratory indexes were extracted by utilizing SEER disease codes and then employed for model construction. Fundamental demographic data included age at diagnosis, gender, race, and marital status. Tumor information contained primary site, size, grade, histologic category and TNM stage. Laboratory indexes involved carcinoembryonic antigen (CEA) prior to surgery, tumor deposits, and perineural invasion (PNI). Survival time and status were collected for further clinical estimation of the predictive model. Furthermore, the information of our validation cohort was normalized via following the criteria of the SEER database (Additional file 1: Table S1). And all clinical information underwent data transformation for the sake of further application in model construction (Additional file 2: Table S2).

Construction of the predictive model

In our research, seven ML models were employed to predict LM in patients with T1 stage CRC. To build up tree decision models, we adopted Light Gradient Boosting Decision (LGBM), Random Forest (RF), and Classification and Regression Trees (CART). LGBM is a gradient boosting framework that utilizes the tree-based learning algorithm, which has been successfully applied in the construction of medical models in recent years [23, 24]. RF is a universally employed ML algorithm to deal with classification and regression issues via the multiple decision trees approach [25]. CART is a classical decision tree algorithm applied in either classification or regression predictive models [26]. The K-Nearest Neighbor (KNN) algorithm was utilized in basic prediction technique. KNN is identified as a vital classification algorithm in the supervised ML domain and is extensively applied in pattern recognition, data mining and intrusion detection [27]. To construct the kernel-based model, the Support Vector Machine (SVM) was selected and put into use. SVM is a supervised ML model that employs classification algorithms for the two-group categorization [28]. Gaussian Naive Bayesian (GNB) algorithm was included in the linear model for specific utilization under the circumstance where the features manifested continuous values [29]. Multilayer Perceptron (MLP) is a feed-forward neural network supplement and has been extensively applied in distinct prediction models [30]. In the wake of employing the Bootstrap aggregating (Bagging) algorithm to optimize the performance of established models, stacked regression was utilized to obtain a stacking model via integrating seven models to output a desirable outcome [31, 32].

To polish up performance of the model and retain maximum authenticity of the data, we strictly employed the Synthetic Minority Over-sampling technique in the inner training dataset to solve the issue of data imbalance [33]. To begin with, patients in the SEER database were randomly assigned to the training set (80%) and testing set (20%) respectively while the proportion of LM ( +) (patients with LM) subgroup was approximately identical to that of the LM (−) (patients without LM) subgroup (Additional files 12 and 13). In the training set, k-fold cross validation (k = 10) was performed, and grid search was adopted to figure out the best combination of parameters. For each set of parameters, the model was in turn fitted and validated with 8/10 and 2/10 of data respectively. Subsequently, our T1 CRC cohort in the Chinese population was utilized as an extra outer validation set further to examine both applicability and efficiency of the model (Additional file 14). The overall workflow is elaborately demonstrated in Fig. 1.

Fig. 1
figure 1

The workflow of selection procedure for colorectal cancer patients

Assessment of model performance

To ensure rational comparison of the models and assess their performance, a multitude of indicators were employed involving confusion matrix, the area under the curve (AUC), sensitivity, specificity, precision, negative predictive value (NPV), false discovery rate (FDR), accuracy, and average precision (AP). In addition, the area under receiver operating characteristic curves (AU-ROC) was utilized as a performance index while the AP value was employed as the criterion for the precision-recall (PR) curve [34]. The average value of parameters was ultimately executed on the testing set and additional outer validation one. Survival analysis was further adopted in the model to evaluate whether it was capable of accurately predicting CRC patients’ outcomes.

In light of the fact that neoplastic size was widely recognized as an effective predictor of CRC outcome, we tested nonlinearity of the model via analysis of 5-knot restricted cubic splines (RCS) and evaluated potential correlation of model with the hazard of LM [35]. In order to estimate the performance of models in patients with small CRC sizes, we stratified the testing set into 4 subgroups, tumor sizes of which being 1–10 mm, 1–20 mm, 1–50 mm and > 50 mm respectively. Their AUC and AP values were then calculated.

Moreover, to make the real clinical decision process more reliable, training samples were adopted prior to utilization of over-sampling strategy. Subsequently, to exhibit the specific decision process of how CRC patients with LM were discriminated from the model, regression tree analysis was conducted via CART algorithm.

Statistical analysis

SEER*Stat software (8.3.6 version) was adopted to acquire targeted CRC patients from the SEER database. Python (version 3.6.9) and R software (version 4.0.5) were utilized to perform statistical analyzes. Python packages were listed: ‘imblearn’, ‘sklearn’, ‘lightgbm’, and ‘mlxtend’. R packages were vividly demonstrated as follows: ‘tableone’, ‘survival’, ‘mice’, and ‘dplyr’. Demographic differences between the two subgroups were tested utilizing either Student’s t-test or Pearson chi‐square test. Results were considered statistically significant when P ≤ 0.05.

Results

Case structures and clinical baselines

Included CRC data in our study from SEER database ranged from 2010 to 2016. In the aggregate, 262,285 CRC patients were initially enrolled. According to the inclusive and exclusive criteria, a totality of 16,785 patients were enrolled in the inner dataset and 326 out of 8226 CRC patients in Xijing hospital were recruited ultimately (Fig. 1). Baseline clinical characteristics of the SEER CRC cohort (Training dataset) and Xijing CRC cohort (Validation dataset) were exhibited in detail (Table 1).

Table 1 Clinical baseline features of SEER and Xijing hospital database

Eleven independent clinical factors were included in our established model, incorporating age at diagnosis, gender, marital status at diagnosis, primary site, tumor size, tumor grade, tumor type, N stage, CEA level, tumor deposits, and PNI. Patients from the SEER database were categorized into LM (−) subgroup (16,023 patients without LM, 95.5%) and LM (+) (762 patients with LM, 4.5%) subgroup respectively. For diagnosed age, we found that the proportion of patients under 60 years of age in LM (+) subgroup (333/762; 43.7%) significantly surpassed that in LM (−) subgroup (6553/16,023; 40.9%; P < 0.001). Notably, the ratio of male CRC was significantly higher in LM (+) subgroup than in its counterpart (P = 0.001). Intriguing, there demonstrated no statistical difference in terms of race between the two subgroups. In line with our anticipation, an upregulated occurrence rate was observed in the single (167/2611, 6.4%) than the married (376/8918, 4.2%; P < 0.001). Regarding tumor sites, rectum was the most common primary site in both subgroups, and the proportion is comparatively higher than other T stages CRC patients (P < 0.001). In respect to progression of CRC, the average tumor size of LM (+) subgroup (52.1 mm) was considerably larger than that of LM (−) one (17.5 mm; P < 0.001). Analogously, LM (+) subgroup demonstrated significantly higher proportions of both Grade II-IV (92.8% vs 68%; P < 0.001) and advanced N stage CRC than LM (−) subgroup (P < 0.001). Furthermore, we observed upregulated levels of tumor deposits, PNI and positive rate of CEA in LM (+) subgroup than its counterpart (P < 0.001). As for tumor differentiation, Adenocarcinoma (Adenocarcinoma, NOS, Adenocarcinoma in tubulovillous adenoma and Adenocarcinoma in adenomatous polyp; 12714/16785, 75.7%) was confirmed as the most common neoplastic category among T1 patients (Table 2).

Table 2 Distributions of clinicopathological characteristics in two groups

Parameters tuning in our models

We trained the LGBM with a depth of five, a learning rate of 0.01, basic learners of 240, leaves of 16, and max bins of 128. For RF and CART, we also elected 5 as the maximum depth of the basic trees. The number of neighbors 200 for KNN was the best. In MLP, we ultimately selected the learning rate of 0.01, epochs of 300, hidden layer of 1, and utilized the Adam Optimizer and ReLU activation function. For SVM, a combination of a C value of 0.01 and kernel smoothing parameters of 0.0001 was determined as the ultimate choice. Additionally, every Bagging model, in possession of 10 basic models, was trained with identical algorithms but various data. The ultimate stacking model incorporated seven bagging models, probability and GNB output by which were recognized as meta classifier.

Evaluation of models

Via internal verifying, all models were observed to reveal superior predictive abilities (AUC values > 0.94). Moreover, by incorporating seven other single models, the stacking model demonstrated a favorable AUC of up to 0.9631 (Fig. 2a). Except for GNB models, AP values of approximately all models attain comparatively preferable levels. Noticeably, the ultimate AP of the stacking mode reached 0.693 (Additional file 3: Figure S1a). Expectedly, the external validation set demonstrated satisfying performance. All models exhibited dramatically high predictive value except the MLP model, and the stacking model contained a final AUC value of 0.992 and an ultimate AP value of 0.811 (Fig. 2b and Additional file 3: Fig. S1b). Additionally, via employing the confusion matrix to appraise the value of models, predictive outcomes of both the inner testing set and outer validation set were displayed in Table 3. LGBM produced fewer quantities of FN (False Negative) and FP (False Positive) than other models in both testing sets. The stacking model was capable of screening approximately all LM (+) patients in both sets. Detailed values of AUC, sensitivity, specificity, precision, NPV, FDR, accuracy, AP, F1-values, and Matthews correlation coefficient of each model in inner and outer validation sets were listed respectively in Additional file 4: Table S3 and Additional file 5: Table S4. The accuracy of five single models reached 0.95, among which LGBM displayed the highest precision (0.9657). The specificity of MLP and sensitivity of GNB were the highest among seven single models. Taken together, the stacking model consistently outperformed other single ML models.

Fig. 2
figure 2

Predictive value of overall models after optimization. Inner validation in SEER database: a ROC curves of seven individual models and stacking model. Outer validation in our Chinese cohort: b ROC curves of seven individual models and stacking model. SEER: Surveillance, Epidemiology, and End Results; and ROC: receiver operating characteristic

Table 3 Confusion matrices of developed models

To further assess comprehensive performance of the AI model, we made comparisons between previous models and logistic regression ones based on our data. Corresponding results testified that the stack-bagging model outperformed other models (Additional file 6: Table S5).

Furthermore, by means of employing survival status and time from the SEER database, we plotted the Kaplan Meier (K–M) curves of the testing set. It was universally acknowledged that LM functioned as an unfavorable prognostic indicator for CRC patients (Additional file 7: Figure S2a). Likewise, we found that the stacking model resembled LM in predicting T1 CRC patients’ outcomes (Additional file 7: Figure S2b).

Comparison of significance of each factor

In all single models, tumor size, preoperative CEA levels, tumor deposits, N stage, histology, and PNI all revealed equally fundamental significance in predicting for LM in T1 CRC. Despite the fact that the AI model manifested desirable performance, the individualized influence of each factor on the result and underlying relationships between these factors remained largely unknown. Hence, we calculated and digitized the significance of each factor used in the built-up AI models (Additional file 8: Figure S3 and Additional file 9: Table S6). Coinciding with previous anticipation, we found that tumor size, CEA level prior to surgery, tumor deposits, and N stage were the top four crucial predictors among all models. Particularly worth mentioning is the fact that tumor size standed out as the most critical one amidst nearly all models.

Subgroup analysis

On account of the reality that tumor size might play a dominant role in prediction while other parameters made relatively less contributions in terms of forecasting model performance, we determined to further investigate the association of tumor size with LM hazard. Firstly, RCS function of tumor size in the training set exhibited a non-linear profile (non-linearity P value < 0.001; Fig. 3a), indicating that this clinical feature should be encoded as a categorical factor and was inappropriate for being employed in canonical logistic regression analysis. Notably, the 50 mm tumor size demonstrated an optimal cut-off value for subgroup analysis (Fig. 3a). Therefore, we utilized the representative AUC and AP value to further explore the model performance in disparate subgroups. Analysis results indicated that AUC values of 1–50 mm and > 50 mm subgroups reached 0.956 and 0.8772 respectively (Fig. 3b).

Fig. 3
figure 3

Estimation of models’ discriminant capability for T1 CRC patients with different tumor sizes. a Restricted cubic spline of tumor size. b ROC curves of seven individual models and stacking model for patients with different tumor sizes (1–50 mm and > 50 mm). CRC: colorectal cancer; and ROC: receiver operating characteristic

In light of the fact that patients with tumor size larger than 50 mm accounted for a lower percentage than the 1–50 mm subgroup, we further divided patients into 1–10 mm and 1–20 mm subgroups. The AUC values (Bagging Stacking model) of 1–10 mm and 1–20 mm subgroups reached 0.8212 and 0.8608 respectively (Additional file 10: Figure S4a and b). Generally speaking, the stacking model was triumphantly verified to possess a favorable prediction capacity in T1 CRC patients with small tumor sizes.

Clinical application

Although the stacking model manifested both desirable and robust predictive power for LM in T1 CRC, the model was intricate in nature which could not be easily apprehended by clinicians. As a consequence, we developed an easy-to-use instrument (clinical decision tree) for the sake of supplementing clinical decision-making process with pragmatic guidance (Fig. 4). In this decision tree, target population were categorized into five groups according to the following four most crucial factors namely CEA level, tumor size, tumor deposits and age. The ROC of clinical decision tree archived 0.949 (Additional file 11: Figure S5), undoubtedly a demonstration of its remarkable discriminative and predictive ability. The population harboring such characteristics as CEA Positive or Borderline, positive tumor deposits, age ≤ 83 and tumor size > 10 manifested high proportion of LM (32.4%) and could be categorized into the high-risk subgroup of LM. On the contrary, remanent three types of patients uniformly demonstrated low occurrence of LM.

Fig. 4
figure 4

Decision tree tool to discriminate liver metastasis in T1 colorectal cancer patients

Discussion

Liver is generally identified as one of the most commonly seen metastasis sites for CRC while LM is universally recognized as the most lethal factor of CRC patients [36, 37]. Early diagnosis of LM could assist clinicians in taking prompt and timely intervention to improve the prognosis of patients, especially for CRC T1 patients [38, 39]. CRC patients in T1 stage could select either surgical or endoscopic treatment, partly depending on the status of distant metastasis. Hence, a convenient and accurate predictive model of LM is urgently demanded to offer guidance on personalized therapeutic strategies.

In the study, we established an innovative and convenient model to predict early LM by incorporating 11 clinicopathologic parameters in T1 CRC utilizing seven AI methods. We firstly combined our real-world researches with public data online on a large scale to comprehensively construct and assess LM predictive models in T1 CRC. Given that the AUC of these models was more extensive than 0.94 and model accuracy was approximately as 100% as possible, we came to the conclusion that above-established models were desirable and robust in yielding favorable clinical benefits, which might be of tremendous assistance to clinicians in the selection process of underlying LM CRC patients. More intriguingly, our model manifested extraordinary competence indiscriminating the LM in T1 CRC patients with small tumor size (1–50 mm) from others. Ultimately, to develop an easy-to-use instrument in clinical practice, we plotted a decision tree to screen out the high-risk population of LM. The visualized decision tree was not only precise but also easy to comprehend for clinicians.

Our real-world research incorporated 326 cases of T1 CRC, amidst which LM occurred in merely eight patients (8/326), significantly lower than that of the SEER database (762/16785, P < 0.001). The discrepancy in the LM ratio might be attributed to low diagnostic efficacy in developing countries [40, 41]. Interestingly, compared with more advanced T stage CRC patients (169/326), PNI was more frequently appeared in T1 CRC patients of our hospital (1266/8226), consistent with results of the SEER database (11350/16785). Abundant evidence has demonstrated that the percentage of PNI occurring in all T stages is approximately 10–15%. Moreover, PNI is an independent biomarker that indicates aggressive behavior and unfavorable prognosis of CRC [42,43,44,45]. Nonetheless, scarcely explained by published literature were underlying causes behind the high ratio of PNI in T1 CRC which deserved further investigation. In addition, serum CEA was confirmed to have a positive relationship with LM. Accumulating evidence has suggested that the expression level of CEA could function as an independent indicator for the prognosis of CRC patients [46]. Therefore, it was not surprising that the concentration of preoperative plasma CEA was significantly higher in CRC patients with LM compared with those with primary CRC [47,48,49]. Besides, among all indicators, tumor size has been regarded as one of the most significant biomarkers in predicting LM status. It has been reported that tumor size was intimately associated with both lymph and hepatic metastases of CRC [50]. Furthermore, scientists have verified that age might play a nonnegligible role in the advancement and prognosis of CRC [51]. Despite increment in young CRC patients, compelling evidence revealed that the young tended to have more favorable outcomes than the old [51]. Contradictorily, our research indicated that CRC patients younger than 60 years of age were more apt to experience risk of LM than their counterparts, which was consistent with several other researchers [52,53,54]. The probable reason might have something to do with frequently occurred mismatch repair gene mutation and upregulated aggressive neoplastic biology in younger patients [55].

To date, multitudes of investigators have constructed diverse models to predict the metastatic capability of CRC. For instance, Tang et al. [14] built up a novel nomogram to forecast LM in all T stages CRC patients via utilizing multivariable Cox regression. They also found that synchronous LM was an independent prognostic factor for CRC patients. Analogously, Li et al. [56] employed the SEER database to construct a T1 CRC all distant metastasis model by virtue of the conventional logistic regression. Howbeit, due to the limitation of the algorithm and the approach to process data, they acquired a passable model (AUC = 0.879) with ineluctable overfitting. Recently, with enormous technical advancement of AI, the application of ML model in neoplastic diagnosis and prognostic assessment has become increasingly prevalent [57, 58]. Numerous novel ML algorithms have remedied deficiencies of canonical statistical methods, such as overfitting, unbalanced data distribution and so on. Ji Hyun Ahn et al. [19] developed an innovative model (AUC = 0.96) to predict LNM in the early stage of CRC patients via utilizing the SEER database and adopting seven AI methods. Nevertheless, these studies were retrospective, single-center, and with small quantities of patients. Additionally, Ichimasa et al. [59] testified that AI could downregulate unnecessary surgery after endoscopic resection of LNM (−) T1 CRC compared with current guidelines. Nonetheless, few models for predicting the incidence of LM in T1 CRC patients were developed and assessed utilizing AI methods. In the current study, we established nine models and then validated them in our own dataset. Besides, their efficacy of predicting LM in early CRC was also compared by dint of easily available clinical and histopathological features. Moreover, we found that our constructed AI models could not only assist clinicians in selecting patients with a high risk of LM, but also resemble LM in accurately predicting T1 CRC patients’ outcomes. Our models still exhibited a superior ability to discriminate the LM in T1 CRC patients with small tumor size from others (1–50 mm).

So far, only surgical resection has been verified as a curative therapeutic approach for CRC patients with early and resectable LM [60, 61]. For patients with untestable LM, early application of systemic chemotherapy might ameliorate the prognosis and enhance the median survival ratio [62]. Integrating entire above-mentioned results, we believed that further utilization of T1 CRC LM models would contribute to the clinical decision making and improve the present therapeutic status.

Admittedly, there still exists several limitations and weaknesses in the study. Firstly, in light that the SEER database is an open and available national program of America, these newly established models might not work in other countries. Secondly, quantities of enrolled patients in our hospital were far from sufficient, and merely eight patients manifested LM status. These shortcomings might lead to a limited verification outcome. In the future, more in-depth and extensive studies will be urgently needed. In addition, we intend to package the stacking model and decision tree to a novel software or website and validate them clinically afterwards in our next work.

Conclusions

In the present study, we successfully established an innovative and stacking bagging model which incorporates 11 clinicopathologic features to predict the incidence of LM in T1 CRC. Our findings indicated that age, gender, married status, primary site, tumor size, CEA, tumor type, grade, N stage and PNI were crucial factors for forecasting LM, amidst which tumor size mattered most. As expected, the stacking bagging model, which integrated strengths of seven single models, demonstrated the strongest predictive power in both databases of SEER and our hospital. Moreover, we found that the stacking model resembled LM when it came to accurate prediction of T1 CRC patients’ outcomes. A novel easy-to-use tool (decision tree) was developed to guide clinicians in screening out high-risk patients of LM and exposing them to more aggressive therapeutic strategies.

Availability of data and materials

The datasets used and/or analyzed during the current study are included in this published article and its additional files.

Abbreviations

CRC:

Colorectal cancer

LM:

Liver metastasis

OS:

Overall survival

AI:

Artificial intelligence

ML:

Machine learning

SEER:

Surveillance, Epidemiology, and End Results

LNM:

Lymph node metastasis

CEA:

Carcinoembryonic antigen

PNI:

Perineural invasion

LGBM:

Light Gradient Boosting Decision

RF:

Random Forest

CART:

Classification and Regression Trees

KNN:

K-Nearest Neighbor

SVM:

Support Vector Machine

GNB:

Gaussian Naive Bayesian

MLP:

Multilayer Perceptron

Bagging:

Bootstrap aggregating

AUC:

Area under the curve

NPV:

Negative predictive value

FDR:

False discovery rate

AP:

Average precision

AU-ROC:

Area under receiver operating characteristic curves

PR:

Precision-recall

RCS:

Restricted cubic splines

FN:

False Negative

FP:

False Positive

K-M:

Kaplan Miere

References

  1. Sung H, Ferlay J, Siegel RL, 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 Cancer J Clin. 2021;71(3):209–49.

    PubMed  Google Scholar 

  2. Bray F, Soerjomataram I. The changing global burden of cancer: transitions in human development and implications for cancer prevention and control. In: Gelband H, Jha P, Sankaranarayanan R, Horton S, editors. Cancer: disease control priorities, vol. 3. Washington (DC): The International Bank for Reconstruction and Development/The World Bank © 2015 International Bank for Reconstruction and Development/The World Bank; 2015.

    Google Scholar 

  3. Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, Bray F. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 2020;159(1):335-349.e315.

    PubMed  Google Scholar 

  4. Kow AWC. Hepatic metastasis from colorectal cancer. J Gastrointest Oncol. 2019;10(6):1274–98.

    PubMed  PubMed Central  Google Scholar 

  5. Helling TS, Martin M. Cause of death from liver metastases in colorectal cancer. Ann Surg Oncol. 2014;21(2):501–6.

    PubMed  Google Scholar 

  6. Cirocchi R, Trastulli S, Boselli C, Montedori A, Cavaliere D, Parisi A, Noya G, Abraha I. Radiofrequency ablation in the treatment of liver metastases from colorectal cancer. Cochrane Database Syst Rev. 2012;6:Cd006317.

    Google Scholar 

  7. Adam R, de Gramont A, Figueras J, Kokudo N, Kunstlinger F, Loyer E, Poston G, Rougier P, Rubbia-Brandt L, Sobrero A, et al. Managing synchronous liver metastases from colorectal cancer: a multidisciplinary international consensus. Cancer Treat Rev. 2015;41(9):729–41.

    PubMed  Google Scholar 

  8. Kopetz S, Chang GJ, Overman MJ, Eng C, Sargent DJ, Larson DW, Grothey A, Vauthey JN, Nagorney DM, McWilliams RR. Improved survival in metastatic colorectal cancer is associated with adoption of hepatic resection and improved chemotherapy. J Clin Oncol. 2009;27(22):3677–83.

    PubMed  PubMed Central  Google Scholar 

  9. Chakedis J, Schmidt CR. Surgical treatment of metastatic colorectal cancer. Surg Oncol Clin N Am. 2018;27(2):377–99.

    PubMed  Google Scholar 

  10. Giannis D, Sideris G, Kakos CD, Katsaros I, Ziogas IA. The role of liver transplantation for colorectal liver metastases: a systematic review and pooled analysis. Transplant Rev. 2020;34(4):100570.

    Google Scholar 

  11. Arru M, Aldrighetti L, Castoldi R, Di Palo S, Orsenigo E, Stella M, Pulitanò C, Gavazzi F, Ferla G, Di Carlo V, et al. Analysis of prognostic factors influencing long-term survival after hepatic resection for metastatic colorectal cancer. World J Surg. 2008;32(1):93–103.

    PubMed  Google Scholar 

  12. Xu H, Wang C, Song H, Xu Y, Ji G. RNA-Seq profiling of circular RNAs in human colorectal Cancer liver metastasis and the potential biomarkers. Mol Cancer. 2019;18(1):8.

    PubMed  PubMed Central  Google Scholar 

  13. Li H, Dai W, Xia X, Wang R, Zhao J, Han L, Mo S, Xiang W, Du L, Zhu G, et al. Modeling tumor development and metastasis using paired organoids derived from patients with colorectal cancer liver metastases. J Hematol Oncol. 2020;13(1):119.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Tang M, Wang H, Cao Y, Zeng Z, Shan X, Wang L. Nomogram for predicting occurrence and prognosis of liver metastasis in colorectal cancer: a population-based study. Int J Colorectal Dis. 2021;36(2):271–82.

    PubMed  Google Scholar 

  15. Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019;25(1):44–56.

    CAS  PubMed  Google Scholar 

  16. Hamet P, Tremblay J. Artificial intelligence in medicine. Metab Clin Exp. 2017;69s:S36–40.

    PubMed  Google Scholar 

  17. Iqbal MJ, Javed Z, Sadia H, Qureshi IA, Irshad A, Ahmed R, Malik K, Raza S, Abbas A, Pezzani R, et al. Clinical applications of artificial intelligence and machine learning in cancer diagnosis: looking into the future. Cancer Cell Int. 2021;21(1):270.

    PubMed  PubMed Central  Google Scholar 

  18. Wang Y, He X, Nie H, Zhou J, Cao P, Ou C. Application of artificial intelligence to the diagnosis and therapy of colorectal cancer. Am J Cancer Res. 2020;10(11):3575–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Ahn JH, Kwak MS, Lee HH, Cha JM, Shin HP, Jeon JW, Yoon JY. Development of a novel prognostic model for predicting lymph node metastasis in early colorectal cancer: analysis based on the surveillance, epidemiology, and end results database. Front Oncol. 2021;11:614398.

    PubMed  PubMed Central  Google Scholar 

  20. Kudo SE, Ichimasa K, Villard B, Mori Y, Misawa M, Saito S, Hotta K, Saito Y, Matsuda T, Yamada K, et al. Artificial intelligence system to determine risk of T1 colorectal cancer metastasis to lymph node. Gastroenterology. 2021;160(4):1075-1084.e1072.

    CAS  PubMed  Google Scholar 

  21. Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell. 2007;128(4):707–19.

    CAS  PubMed  Google Scholar 

  22. Daly MC, Paquette IM. Surveillance, Epidemiology, and End Results (SEER) and SEER-medicare databases: use in clinical research for improving colorectal cancer outcomes. Clin Colon Rectal Surg. 2019;32(1):61–8.

    PubMed  PubMed Central  Google Scholar 

  23. Ke G, Meng Q, Finley T, Wang T, Chen W, Ma W, Ye Q, Liu T-Y. Lightgbm: a highly efficient gradient boosting decision tree. Adv Neural Inf Process Syst. 2017;30:3146–54.

    Google Scholar 

  24. Létinier L, Jouganous J, Benkebil M, Bel-Létoile A, Goehrs C, Singier A, Rouby F, Lacroix C, Miremont G, Micallef J, et al. Artificial intelligence for unstructured healthcare data: application to coding of patient reporting of adverse drug reactions. Clin Pharmacol Therapeutics. 2021;110:392–400.

    Google Scholar 

  25. Breiman L. Random forests—random features. Machine learning 1999.

  26. Fearn T. Classification and regression trees (CART). J Near Infrared Spectrosc. 2006;17(1):13.

    Google Scholar 

  27. Keller JM, Gray MR, Givens JA. A fuzzy K-nearest neighbor algorithm. IEEE Trans Syst Man Cybern. 2012. https://doi.org/10.1109/TSMC.1985.6313426.

    Article  Google Scholar 

  28. Joachims T. Text categorization with support vector machines: learning with many relevant features. In: Proc Conference on Machine Learning: 1998; 1998.

  29. Chickering DM, Heckerman D. Efficient approximations for the marginal likelihood of bayesian networks with hidden variables. Mach Learn. 1997;29(2):181–212.

    Google Scholar 

  30. Ruck DW. Feature selection using a multilayer perceptron. Neural Network Comput. 1990;2:40–8.

    Google Scholar 

  31. Leo B. Stacked regressions. Mach Learn. 1996. https://doi.org/10.1007/BF00117832.

    Article  Google Scholar 

  32. Breiman L. Bagging prediction. Mach Learn. 1996;24:123–40.

    Google Scholar 

  33. Chawla NV, Bowyer KW, Hall LO, Kegelmeyer WP. SMOTE: synthetic minority over-sampling technique. J Artif Intell Res. 2002;16(1):321–57.

    Google Scholar 

  34. Davis JJ, Goadrich MH. The relationship between precision-recall and ROC curves. In: Proceedings of the 23rd international conference on machine learning: 2006; 2006.

  35. Harrell FE Jr, Lee KL, Pollock BG. Regression models in clinical studies: determining relationships between predictors and response. J Natl Cancer Inst. 1988;80(15):1198–202.

    PubMed  Google Scholar 

  36. Engstrand J, Nilsson H, Strömberg C, Jonas E, Freedman J. Colorectal cancer liver metastases—a population-based study on incidence, management and survival. BMC Cancer. 2018;18(1):78.

    PubMed  PubMed Central  Google Scholar 

  37. van der Geest LG, Lam-Boer J, Koopman M, Verhoef C, Elferink MA, de Wilt JH. Nationwide trends in incidence, treatment and survival of colorectal cancer patients with synchronous metastases. Clin Exp Metastasis. 2015;32(5):457–65.

    PubMed  Google Scholar 

  38. Yin J, Bai Z, Song J, Yang Y, Wang J, Han W, Zhang J, Meng H, Ma X, Yang Y, et al. Differential expression of serum miR-126, miR-141 and miR-21 as novel biomarkers for early detection of liver metastasis in colorectal cancer. Chin J Cancer Res. 2014;26(1):95–103.

    PubMed  PubMed Central  Google Scholar 

  39. Lv Y, Feng QY, Wei Y, Ren L, Ye Q, Wang X, Cui Y, Liu T, Zhou B, Wang M, et al. Benefits of multi-disciplinary treatment strategy on survival of patients with colorectal cancer liver metastasis. Clin Transl Med. 2020;10(3):e121.

    PubMed  PubMed Central  Google Scholar 

  40. Yao T, Shiono S. Differences in the pathological diagnosis of colorectal neoplasia between the East and the West: Present status and future perspectives from Japan. Dig Endosc. 2016;28(3):306–11.

    PubMed  Google Scholar 

  41. Schlemper RJ, Itabashi M, Kato Y, Lewin KJ, Riddell RH, Shimoda T, Sipponen P, Stolte M, Watanabe H. Differences in the diagnostic criteria used by Japanese and Western pathologists to diagnose colorectal carcinoma. Cancer. 1998;82(1):60–9.

    CAS  PubMed  Google Scholar 

  42. Alotaibi AM, Lee JL, Kim J, Lim SB, Yu CS, Kim TW, Kim JH, Kim JC. Prognostic and oncologic significance of perineural invasion in sporadic colorectal cancer. Ann Surg Oncol. 2017;24(6):1626–34.

    PubMed  Google Scholar 

  43. Al-Sukhni E, Attwood K, Gabriel EM, LeVea CM, Kanehira K, Nurkin SJ. Lymphovascular and perineural invasion are associated with poor prognostic features and outcomes in colorectal cancer: a retrospective cohort study. Int J Surg. 2017;37:42–9.

    PubMed  Google Scholar 

  44. Yang Y, Huang X, Sun J, Gao P, Song Y, Chen X, Zhao J, Wang Z. Prognostic value of perineural invasion in colorectal cancer: a meta-analysis. J Gastrointest Surg. 2015;19(6):1113–22.

    PubMed  Google Scholar 

  45. Knijn N, Mogk SC, Teerenstra S, Simmer F, Nagtegaal ID. Perineural invasion is a strong prognostic factor in colorectal cancer: a systematic review. Am J Surg Pathol. 2016;40(1):103–12.

    PubMed  Google Scholar 

  46. Zhu J, Hao J, Ma Q, Shi T, Wang S, Yan J, Chen R, Xu D, Jiang Y, Zhang J, et al. A novel prognostic model and practical nomogram for predicting the outcomes of colorectal cancer: based on tumor biomarkers and log odds of positive lymph node scheme. Front Oncol. 2021;11:661040.

    PubMed  PubMed Central  Google Scholar 

  47. Pakdel A, Malekzadeh M, Naghibalhossaini F. The association between preoperative serum CEA concentrations and synchronous liver metastasis in colorectal cancer patients. Cancer Biomark. 2016;16(2):245–52.

    CAS  PubMed  Google Scholar 

  48. Polivka J, Windrichova J, Pesta M, Houfkova K, Rezackova H, Macanova T, Vycital O, Kucera R, Slouka D, Topolcan O. The level of preoperative plasma KRAS mutations and CEA predict survival of patients undergoing surgery for colorectal cancer liver metastases. Cancers (Basel). 2020;12(9):2434.

    CAS  Google Scholar 

  49. Lou Z, Meng RG, Zhang W, Yu ED, Fu CG. Preoperative carcinoembryonic antibody is predictive of distant metastasis in pathologically T1 colorectal cancer after radical surgery. World J Gastroenterol. 2013;19(3):389–93.

    PubMed  PubMed Central  Google Scholar 

  50. Guo K, Feng Y, Yuan L, Wasan HS, Sun L, Shen M, Ruan S. Risk factors and predictors of lymph nodes metastasis and distant metastasis in newly diagnosed T1 colorectal cancer. Cancer Med. 2020;9(14):5095–113.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Abasse Kassim S, Tang W, Abbas M, Wu S, Meng Q, Zhang C, Li X, Chen R. Clinicopathologic and epidemiological characteristics of prognostic factors in post-surgical survival of colorectal cancer patients in Jiangsu Province, China. Cancer Epidemiol. 2019;62:101565.

    PubMed  Google Scholar 

  52. Mo S, Cai X, Zhou Z, Li Y, Hu X, Ma X, Zhang L, Cai S, Peng J. Nomograms for predicting specific distant metastatic sites and overall survival of colorectal cancer patients: a large population-based real-world study. Clin Transl Med. 2020;10(1):169–81.

    PubMed  PubMed Central  Google Scholar 

  53. Luo D, Liu Q, Yu W, Ma Y, Zhu J, Lian P, Cai S, Li Q, Li X. Prognostic value of distant metastasis sites and surgery in stage IV colorectal cancer: a population-based study. Int J Colorectal Dis. 2018;33(9):1241–9.

    PubMed  Google Scholar 

  54. Tohmé C, Labaki M, Hajj G, Abboud B, Noun R, Sarkis R. Colorectal cancer in young patients: presentation, clinicopathological characteristics and outcome. Lebanese Med J. 2008;56(4):208–14.

    Google Scholar 

  55. Law JH, Koh FH, Tan KK. Young colorectal cancer patients often present too late. Int J Colorectal Dis. 2017;32(8):1165–9.

    PubMed  Google Scholar 

  56. Li Q, Wang G, Luo J, Li B, Chen W. Clinicopathological factors associated with synchronous distant metastasis and prognosis of stage T1 colorectal cancer patients. Sci Rep. 2021;11(1):8722.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Kourou K, Exarchos TP, Exarchos KP, Karamouzis MV, Fotiadis DI. Machine learning applications in cancer prognosis and prediction. Comput Struct Biotechnol J. 2015;13:8–17.

    CAS  PubMed  Google Scholar 

  58. Xiao Y, Wu J, Lin Z, Zhao X. A deep learning-based multi-model ensemble method for cancer prediction. Comput Methods Programs Biomed. 2018;153:1–9.

    PubMed  Google Scholar 

  59. Ichimasa K, Kudo SE, Mori Y, Misawa M, Matsudaira S, Kouyama Y, Baba T, Hidaka E, Wakamura K, Hayashi T, et al. Artificial intelligence may help in predicting the need for additional surgery after endoscopic resection of T1 colorectal cancer. Endoscopy. 2018;50(3):230–40.

    PubMed  Google Scholar 

  60. Ito K, Govindarajan A, Ito H, Fong Y. Surgical treatment of hepatic colorectal metastasis: evolving role in the setting of improving systemic therapies and ablative treatments in the 21st century. Cancer J. 2010;16(2):103–10.

    PubMed  Google Scholar 

  61. Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. Clinical score for predicting recurrence after hepatic resection for metastatic colorectal cancer: analysis of 1001 consecutive cases. Ann Surg. 1999;230(3):309–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Gallagher DJ, Kemeny N. Metastatic colorectal cancer: from improved survival to potential cure. Oncology. 2010;78(3–4):237–48.

    PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

CX, JZ, XC designed the study; TH, JZ, DX contributed to the conception of the study and completed the manuscript together; RC and YJ contributed significantly to statistical analysis and manuscript preparation; SW and SG helped perform the analysis with constructive discussions. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jianyong Zheng or Chunsheng Xu.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Ethics Committee of the Xijing Hospital of Air Force Medical University. The approval number of current research is No. KY20203269-1. All participants gave written, informed consent.

Consent for publication

Not applicable.

Conflicts of interest

The authors have no conflicts of interest to declare.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1: Table S1.

Normalization standards of clinical data in outer validation set.

Additional file 2: Table S2.

References for property values of clinical features in models.

Additional file 3: Figure S1.

PR curves for overall models. Inner validation in SEER database: (a) PR curves, indicating the tradeoff between precision and recall. Outer validation in our Chinese cohort: (b) PR curves, indicating the tradeoff between precision and recall. SEER: Surveillance, Epidemiology, and End Results; and PR: precision-recall.

Additional file 4: Table S3.

Performance of developed models in inner datasets.

Additional file 5: Table S4.

Performance of developed models in our real-world dataset.

Additional file 6: Table S5.

Comparison of AI algorithms and logistic regression algorithm.

Additional file 7: Figure S2.

Evaluation of the prognostic value for stacking-bagging model. (a) The survival curve based real data. (b) The survival curve based on predictive outcomes.

Additional file 8: Figure S3.

Factor importance of the developed models. Bar graphs describe the proportion of importance of the different predictors in models. The top ten factor importance were exhibited in models: (a) Average of factor importance in seven models, (b) LGBM, (c) RF, (d) GNB, (e) KNN, (f) MLP, (g) CART, and (h) SVM. LGBM: Light Gradient Boosting Decision; RF: Random Forest; GNB: Gaussian Naive Bayesian; KNN: k-nearest neighbor algorithm; MLP: Multilayer Perceptron; CART: Classification and Regression Trees; and SVM: Support Vector Machine.

Additional file 9: Table S6.

Significances of clinical features in AI models.

Additional file 10: Figure S4.

Models’ prediction value for T1 CRC patients with small tumor sizes. (a) ROC curves of seven individual models and stacking model in tumor size (1–10 mm). (b) ROC curves of seven individual models and stacking model in tumor size (1–20 mm).

Additional file 11: Figure S5.

Performance of decision tree model. (a) ROC curves of seven individual models and stacking model. (b) PR curves, indicating the tradeoff between precision and recall. ROC: receiver operating characteristic; and PR: precision-recall.

Additional file 12:

Original data of inner testing set of SEER.

Additional file 13:

Original data of inner training set of SEER.

Additional file 14:

Original data of outer validation set from Xijing hospital.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Han, T., Zhu, J., Chen, X. et al. Application of artificial intelligence in a real-world research for predicting the risk of liver metastasis in T1 colorectal cancer. Cancer Cell Int 22, 28 (2022). https://doi.org/10.1186/s12935-021-02424-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-021-02424-7

Keywords