Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Construction and validation of a nomogram for HBV-related hepatocellular carcino...
Journal Information
Vol. 28. Issue 4.
(July - August 2023)
Share
Share
Download PDF
More article options
Visits
298
Vol. 28. Issue 4.
(July - August 2023)
Original article
Full text access
Construction and validation of a nomogram for HBV-related hepatocellular carcinoma: A large, multicenter study
Visits
298
Ke Sua,1, Qiuni Shenb,1, Jian Tongc,1, Tao Gua,1, Ke Xud, Han Lia, Hao Chie, Yanlin Liua, Xueting Lif, Lianbin Weng, Yanqiong Songh, Qulian Guoi, Jiali Chena, Zhenying Wua, Yi Jianga, Kun Hej,
Corresponding author
hktongji@swmu.edu.cn

Corresponding authors.
, Lu Guok, Yunwei Hana,
Corresponding author
Lanpaoxiansheng@126.com

Corresponding authors.
a Department of Oncology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, China
b Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, China
c Department of Spinal Surgery, No.1 Orthopedics Hospital of Chengdu, 610000 Chengdu, China
d Department of Oncology, Chongqing General Hospital, 401147 Chongqing, China
e Clinical Medical College, Southwest Medical University, 646000 Luzhou, China
f Department of Oncology, 363 Hospital, 610041 Chengdu, China
g Department of Geriatric Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072 Chengdu, China
h Department of Radiotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042 Chengdu, China
i Department of Paediatrics, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, China
j Clinical Research Institute, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, China
k Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, China
Ver más
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (5)
Show moreShow less
Additional material (1)
Abstract
Introduction and Objectives

We initiated this multicenter study to integrate important risk factors to create a nomogram for hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) for clinician decision-making.

Patients and Methods

Between April 2011 and March 2022, 2281 HCC patients with an HBV-related diagnosis were included. All patients were randomly divided into two groups in a ratio of 7:3 (training cohort, n = 1597; validation cohort, n = 684). The nomogram was built in the training cohort via Cox regression model and validated in the validation cohort.

Results

Multivariate Cox analyses revealed that the portal vein tumor thrombus, Child–Pugh class, tumor diameter, alanine aminotransferase level, tumor number, extrahepatic metastases, and therapy were independent predictive variables impacting overall survival. We constructed a new nomogram to predict 1-, 2-, and 3-year survival rates based on these factors. The nomogram-related receiver operating characteristics (ROC) curves indicated that the area under the curve (AUC) values were 0.809, 0.806, and 0.764 in predicting 1-, 2-, and 3-year survival rates, respectively. Furthermore, the calibration curves revealed good agreement between real measurements and nomogram predictions. The decision curve analyses (DCA) curves demonstrated excellent therapeutic application potential. In addition, stratified by risk scores, low-risk groups had longer median OS than medium–high-risk groups (p < 0.001).

Conclusions

The nomogram we constructed showed good performance in predicting the 1-year survival rate for HBV- related HCC.

Keywords:
Hepatitis B virus
Hepatocellular carcinoma
Nomogram
Overall survival
Abbreviations:
HBV
HCC
mOS
ROC
C-index
DCA
PVTT
ALT
AUC
PD-1
PD-L1
SIRT
TACE
AJCC
BCLC
ALBI
RFA
ICIs
Full Text
1Introduction

Hepatocellular carcinoma (HCC) carries a high malignancy and death rate [1]. At the time of diagnosis, the majority of HCC is in an advanced stage with a poor prognosis [2]. Therefore, current research is focused on enhancing the prognosis and efficacy monitoring of HCC.

Today, the development of programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors provides HCC patients with new daybreak. With a median overall survival (mOS) of 19.2 months, the combination of atezolizumab and bevacizumab has emerged as the standard of care for HCC [3]. Our prior research also indicated that radiotherapy combined with tislelizumab and lenvatinib could increase the survival of patients with advanced HCC (median progression-free survival, mPFS = 10.2 months; mOS = 20.3 months) [4]. Furthermore, selective internal radiation therapy (SIRT), surgery, transcatheter arterial chemoembolization (TACE), and other treatment techniques have promising therapeutic outcomes in HCC [5–7].

The prognosis of HCC has been improved, but unfortunately, the patients are still prone to short- term recurrence even after undergoing radical surgery. In addition, HBV infection is the major cause of HCC in the Asia-Pacific region, including China, Japan, and South Korea [8,9]. Therefore, it is important to develop new treatment strategies and regular monitoring to improve the long-term survival rate of HBV-related HCC patients.

At present, several staging systems, such as the American Joint Committee on Cancer (AJCC) [10], Okuda staging systems [11], Barcelona Clinic Liver Cancer (BCLC) [12], and albumin–bilirubin (ALBI) [13], are being utilized to forecast the prognosis and direct the treatment of HCC. However, the predictive ability of these methods is restricted due to the fact that individual survival prognosis is dependent on several variables, including age, pathological characteristics, tumor diameter, clinical characteristics, radiotherapy, surgery, and chemotherapy [14,15]. Therefore, these parameters should be examined for improved HCC management.

A nomogram is a clinical prediction model based on multivariate regression analysis, which can integrate multiple predictors to better evaluate prognosis [16]. In addition, the nomogram can be customized to calculate an individual's survival rate, therefore it has potential clinical utility. Currently, HBV-related HCC has generated a tremendous global burden of disease. However, there is a scarcity of large-sample-size research to build nomograms of HBV-related HCC.

We initiated this multicenter study to integrate important risk factors to create a nomogram for HBV-related HCC for clinician decision-making.

2Patients and Methods2.1Patients

From April 2011 to March 2022, we consecutively enrolled a total of 2281 HBV-related HCC patients from 4 tertiary hospitals in China. The inclusion criteria were as follows: a) complete clinical data; b) age ≥ 18 years; and c) clinically or pathologically diagnosed HCC. Patients with hepatitis C virus (HCV), liver transplantation, and other types of malignancies were excluded.

2.2Data collection

We gathered patient information from the patient's case file. These data included age, sex, HBV, smoking, alcohol, alanine aminotransferase (ALT), diabetes, hypertension, cirrhosis, portal vein tumor thrombus (PVTT), portal hypertension, Child-Pugh class, albumin–bilirubin, AFP, leukocyte, BCLC stage, tumor size, extrahepatic metastasis, tumor number, lymph node metastasis, and treatments. OS was defined as the time from the first day of treatment initiation to the last follow-up or death.

2.3Statistical analysis

Categorical variables are expressed as percentages and absolute values and analyzed by the chi-square (χ2) test. We divided all patients into two groups at random in a 7:3 ratio (training cohort, n = 1597; validation cohort, n = 684). We first performed univariate Cox analysis in the validation cohort to identify prognostic indicators (p < 0.05) and then we introduced them into multivariate Cox analysis to identify independent influencing factors. Following that, we developed a new nomogram to predict 1-, 2-, and 3-year survival rates based on independent prognostic factors influencing OS. To validate the nomogram, concordance index (C-index), calibration curves (1000 bootstrap resamples), decision curve analyses (DCA), receiver operating characteristics (ROC) curve, and nomogram risk score were used. Patients were divided into three groups (high risk, moderate risk, and low risk) based on the x- tile risk score with the best cut-off value (Yale University, New Haven, CT). R 3.3.2 software and SPSS (version 26.0) were used for all statistical analyses. Two-sided P < 0.05 were considered statistically significant.

2.4Ethical statements

This retrospective study was approved by the Ethics Committee of The Affiliated Hospital of Southwest Medical University (approval number KY2020254) and complied with the standards of the Declaration of Helsinki. The Ethics Committee waived the requirement for informed consent because of the retrospective study.

3Results3.1Patient characteristics and overall survival

Our study included 2281 HBV-related HCC patients (training cohort, n = 1597; validation cohort, n = 684). The percentages of age ≥ 65 years, child A, male, AFP < 200 ng/ml, and ALBI-2 grade were 35.1%, 73.2%, 86.7%, 50.3%, and 60.8%, respectively. Moreover, the majority of patients have multiple tumors (69.9%) and are at stage C of BCLC (63.1%). The baseline characteristics of the training and validation cohort are shown in Table 1. As of May 2022, a total of 1145 (50.2%) patients died in this study. The median follow-up time was 28.6 (27.1–30.1) months, and the mOS was 24.9 (22.2–28.9) months (Fig. 1).

Table 1.

Baseline characteristics of the training cohort and validation cohort.

Variable  Total  Training cohort  Validation cohort  p 
Patients  2281  1597  684   
Male sex  1977 (86.7%)  1381 (86.5%)  596 (87.1%)  0.671 
Age ≥ 65 years  801 (35.1%)  559 (35.0%)  242 (35.4%)  0.863 
Smoking  1143 (50.1%)  807 (50.5%)  336 (49.1%)  0.537 
Alcoholism  960 (42.1%)  673 (42.1%)  287 (42.0%)  0.936 
Diabetes mellitus  215 (9.4%)  149 (9.3%)  66 (9.6%)  0.811 
Hypertension  311 (13.6%)  227 (14.2%)  84 (12.3%)  0.218 
Cirrhosis  1352 (59.3%)  960 (60.1%)  392 (57.3%)  0.212 
Portal hypertension  727 (31.9%)  507 (31.7%)  220 (32.2%)  0.845 
Child–Pugh class        0.480 
1670 (73.2%)  1180 (73.9%)  490 (71.6%)   
550 (24.1%)  377 (23.6%)  173 (25.3%)   
61 (2.7%)  40 (2.5%)  21 (3.1%)   
ALBI grade        0.027 
715 (31.3%)  527 (33%)  188 (27.5%)   
1386 (60.8%)  951 (59.5%)  435 (63.6%)   
180 (7.9%)  119 (7.5%)  61 (8.9%)   
Serum AFP, ng/ml        0.797 
< 200  1148 (50.3%)  811 (50.8%)  337 (49.3%)   
≥ 200, < 400  149 (6.5%)  104 (6.5%)  45 (6.6%)   
≥ 400  984 (43.1%)  682 (42.7%)  302 (44.2%)   
Leukocyte ≥ 4 × 109/L  1818 (79.7%)  1270 (79.5%)  548 (80.1%)  0.747 
Platelet count ≥ 100 × 109/L  1528 (67.0%)  1073 (67.2%)  455 (66.5%)  0.756 
ALT levels ≥ 40 U/L  1194 (52.3%)  837 (52.4%)  357 (52.2%)  0.924 
BCLC stage        0.661 
0/A  428 (18.8%)  292 (18.3%)  136 (19.9%)   
353 (15.5%)  247 (15.5%)  106 (15.5%)   
1439 (63.1%)  1018 (63.7%)  421 (61.5%)   
61 (2.7%)  40 (2.5%)  21 (3.1%)   
Number of tumors ≥ 2  1595 (69.9%)  1138 (71.3%)  457 (66.8%)  0.034 
Tumor diameter, cm        0.398 
< 3  407 (17.8%)  298 (18.7%)  109 (15.9%)   
≥ 3, < 5  535 (23.5%)  365 (22.9%)  170 (24.9%)   
≥ 5, < 10  819 (35.9%)  574 (35.9%)  245 (35.8%)   
≥ 10  520 (22.8%)  360 (22.5%)  160 (23.4%)   
PVTT  910 (39.9%)  645 (40.4%)  265 (38.7%)  0.462 
Lymph node metastasis  951 (41.7%)  666 (41.7%)  285 (41.7%)  0.987 
Extrahepatic metastases  491 (21.5%)  346 (21.7%)  145 (21.2%)  0.804 
Lung  308 (13.5%)  209 (13.1%)  99 (14.5%)   
Bone  133 (5.8%)  90 (5.6%)  43 (6.3%)   
Other  157 (6.9%)  119 (7.5%)  38 (5.6%)   
Treatments         
Supportive care  506 (22.2%)  349 (21.9%)  157 (23.0%)  0.562 
Liver resection  461 (20.2%)  324 (20.3%)  137 (20.0%)  0.888 
Radiotherapy  209 (9.2%)  142 (8.9%)  67 (9.8%)  0.493 
TACE  988 (43.3%)  707 (44.3%)  281 (41.1%)  0.159 
RFA  159 (7.0%)  111 (7.0%)  48 (7.0%)  0.954 
ICI  263 (11.5%)  177 (11.1%)  86 (12.6%)  0.307 
Targeted therapy  213 (9.3%)  144 (9.0%)  69 (10.1%)  0.421 
Chemotherapy  110 (4.8%)  80 (5.0%)  30 (4.4%)  0.524 

ALBI, albumin-bilirubin; AFP, alpha-fetoprotein; ALT, alanine aminotransferase; BCLC, Barcelona Clinic Liver Cancer; PVTT, portal vein tumor thrombus; TACE, transcatheter arterial chemoembolization; RFA, radiofrequency ablation; ICI, immune checkpoint inhibitor.

Fig. 1.

The median overall survival of all patients was 24.9 (22.2–28.9) months.

(0.2MB).
3.2Univariate and multivariate Cox analyses

In the training cohort, univariate and multivariate Cox analyses revealed that Child–Pugh class, ALT level, tumor number, tumor diameter, PVTT, extrahepatic metastases, and treatment (all p < 0.05) were independent prognostic factors affecting OS (Table 2).

Table 2.

Univariate and multivariate Cox analysis of overall survival in the training cohort Univariable Cox regression Multivariable Cox regression.

  HR  95%CI  p  HR  95%CI  p 
Sex (male/female)  1.384  1.110–1.726  0.004  1.171  0.925–1.483  0.190 
Age (≥60/<60 years)  0.848  0.732–0.983  0.029  0.931  0.798–1.087  0.366 
Smoking (positive/negative)  1.203  1.047–1.382  0.009  1.035  0.891–1.202  0.655 
Alcoholism (positive/negative)  1.137  0.989–1.306  0.071       
Diabetes mellitus (positive/negative)  1.124  0.893–1.417  0.319       
Hypertension (positive/negative)  0.754  0.608–0.934  0.010  0.933  0.749–1.163  0.536 
Cirrhosis (positive/negative)  1.147  0.994–1.323  0.061       
Portal hypertension (positive/negative)  1.391  1.204–1.608  < 0.001  1.098  0.942–1.281  0.232 
Child-Pugh class      < 0.001      < 0.001 
1.000      1.000     
2.018  1.732–2.351  < 0.001  1.477  1.226–1.780  < 0.001 
4.655  3.299–6.569  < 0.001  4.567  2.615–7.978  < 0.001 
ALBI grade      < 0.001      0.284 
1.000      1.000     
1.500  1.280–1.759  < 0.001  0.977  0.819–1.166  0.797 
2.994  2.333–3.843  < 0.001  0.770  0.546–1.084  0.134 
Serum AFP, ng/ml      < 0.001      0.645 
< 200  1.000      1.000     
≥ 200, < 400  1.049  0.777–1.418  0.754  1.108  0.817–1.503  0.510 
≥ 400  1.526  1.322–1.761  < 0.001  1.064  0.914–1.240  0.423 
Leukocyte (<4000/≥4000/μL)  1.125  0.946–1.338  0.182       
Platelet (<100,000/≥100,000/μL)  1.140  0.982–1.323  0.084       
ALT (≥40/<40U/L)  1.690  1.467–1.948  < 0.001  1.282  1.105–1.487  0.001 
BCLC stage      < 0.001      0.288 
0/A  1.000      1.000     
1.800  1.315–2.464  < 0.001  0.949  0.660–1.364  0.776 
3.745  2.904–4.830  < 0.001  1.189  0.843–1.679  0.324 
10.825  7.166–16.352  < 0.001  1.189  0.843–1.679  0.324 
Number of tumor (≥2/<2)  2.103  1.763–2.510  < 0.001  1.522  1.241–1.867  < 0.001 
Tumor diameter, cm      < 0.001      < 0.001 
< 3  1.000      1.000     
≥ 3, < 5  1.301  1.008–1.680  0.043  1.123  0.863–1.462  0.387 
≥ 5, < 10  2.125  1.696–2.663  < 0.001  1.539  1.209–1.959  < 0.001 
≥ 10  3.124  2.468–3.953  < 0.001  1.734  1.336–2.25  < 0.001 
PVTT (positive/negative)  2.347  2.042–2.698  < 0.001  1.340  1.120–1.602  0.001 
Lymph node metastasis (yes/no)  1.972  1.716–2.266  < 0.001  1.183  0.995–1.407  0.057 
Extrahepatic metastases (yes/no)  1.755  1.503–2.049  < 0.001  1.240  1.045–1.472  0.014 
Treatment      < 0.001      < 0.001 
Supportive care  1.000      1.000     
Surgery  0.223  0.164–0.303  < 0.001  0.348  0.253–0.477  < 0.001 
Radiotherapy  0.396  0.276–0.566  < 0.001  0.547  0.378–0.792  0.001 
RFA  0.075  0.031–0.183  < 0.001  0.139  0.057–0.340  < 0.001 
TACE  0.649  0.545–0.772  < 0.001  0.621  0.518–0.746  < 0.001 
ICI plus targeted therapy  0.962  0.681–1.357  0.824  0.912  0.635–1.309  0.616 
TACE plus ICI  0.549  0.363–0.830  0.004  0.448  0.294–0.684  < 0.001 
Adjuvant TACE  0.156  0.091–0.267  < 0.001  0.232  0.134–0.401  < 0.001 
Other  0.414  0.332–0.516  < 0.001  0.454  0.363–0.570  < 0.001 

ALBI, albumin-bilirubin; AFP, alpha-fetoprotein; ALT, alanine aminotransferase; BCLC, Barcelona Clinic Liver Cancer; PVTT, portal vein tumor thrombus; TACE, transcatheter arterial chemoembolization; RFA, radiofrequency ablation; ICI, immune checkpoint inhibitor.

3.3Construction and validation of a nomogram

We constructed a nomogram based on independent influencing factors identified in the multivariate analysis to predict 1-, 2-, and 3-year survival rates (Fig. 2). The C-index is 0.743 and 0.754 in the training and validation cohort, respectively. The calibration curves indicated good consistency in predicting 1-, 2-, and 3-year survival rates between actual observations and nomogram predictions in the training (Fig. 3A) and validation cohorts (Fig. 3B). The DCA curves of 1, 2, and 3-year survival rates also expressed promising potential for clinical application in the training (Fig. 3C) and validation cohorts (Fig. 3D). As shown by the DCA [17], the "all" and "none" represented the assumption that all patients were alive and dead, respectively. The net benefit of the nomogram was higher than that of treat none or treat all strategies for the threshold probability within a range of 0.1–0.7.

Fig. 2.

Nomogram based on independent prognostic factors affecting survival. TACE, transcatheter arterial chemoembolization; ICI, immune checkpoint inhibitor; RFA, radiofrequency ablation; PVTT, portal vein tumor thrombus; ALT, alanine aminotransferase.

(0.18MB).
Fig. 3.

Calibration curves for predicting 1-, 2-, and 3-year survival rates in the training (A) and validation cohorts (B). Decision curve analyses (DCA) curves of 1-, 2-, and 3-year survival rates in the training (C) and validation cohorts (D).

(0.33MB).

The time ROC package of R software is used to draw ROC curve and calculate AUC value [15]. The nomogram-related ROC curves of the training cohort indicated that the area under the curve (AUC) values were 0.784, 0.788, and 0.779 in predicting 1-, 2-, and 3-year survival rates, respectively (Fig. 4A). In the validation cohort, the AUC values of 1-, 2-, and 3-year survival rates were 0.809 (0.719–0.901), 0.806 (0.706–0.855), and 0.764 (0.629–0.836), respectively, which were higher than the single significant factor affecting the prognosis of HBV-related HCC (Fig. 4B, Supplementary Fig. 1A–G).

Fig. 4.

Nomogram-related operating characteristics curve (ROC) curves for survival in the training (A) and validation cohorts (B).

(0.18MB).
3.4Risk stratification

We used nomograms to calculate the total point for each patient and then used x-tile software to stratify the data. We divided all HBV-related HCC patients into high-risk (> 211.68), moderate-risk (141.19–211.68), and low-risk (< 141.19) groups based on the optimal cut-off score determined by x- tile. Furthermore, in all cohorts, low-risk groups stratified by risk scores exhibited longer mOS than medium–high-risk groups (6.167 vs. 1.233 vs. 0.317 years, p < 0.001, Fig. 5). Furthermore, similar results were achieved in the training and validation cohorts (Supplementary Fig. 2A-B).

Fig. 5.

Survival curve stratified based on risk scores.

(0.25MB).
4Discussion

Currently, HBV is the leading cause of HCC in the Asia-Pacific region [8,9]. We created and validated a prediction model for HBV-related HCC in this large, multicenter, retrospective investigation. The model, which included tumor burden, laboratory testing, and treatment, performed well in terms of prediction. This assists doctors in making clinical decisions on the management of HBV-related HCC.

At the moment, the introduction of immune checkpoint inhibitors has created new opportunities for cancer patients. Various immunotherapy regimens, including atezolizumab + bevacizumab [3], lenvatinib + pembrolizumab [18], camrelizumab + apatinib [19], and TACE + camrelizumab [20], were found to prolong the life of advanced HCC patients. However, predicting the efficacy of HCC still faces great challenges.

Unlike earlier prognostic indicators such as BCLC, ALBI, and Child–Pugh grade, for example, the model we developed estimates each patient's survival rate rather than merely separating patients into different risk categories, so better decreases the impacts of heterogeneity [13,21]. In the nomogram of HBV-related HCC established by Mo [22], C-index was 0.680 (0.645–0.715). And in Zheng's [23] nomogram for HBV-related HCC, the AUC values for predicting 1-, 2-, and 3-year disease-free survival were 0.761, 0.716, and 0.715, respectively. In addition, Huang et al. [24] reported that in the model of advanced HCC receiving stereotactic body radiation therapy, the AUC value for predicting a 1-year survival rate was 0.77. Furthermore, in the HCC nomogram of Zhang et al. [25], the AUC values for predicting 1-, 3-, and 5-year postoperative survival rates were 0.645, 0.671, and 0.635, respectively. In our study, the model we developed includes a wider range of treatment regimens and a bigger number of data, reducing sampling bias. The C-index was 0.754 and the AUC values for the 1-, 2-, and 3-year survival rates were 0.809, 0.806, and 0.764, respectively, which were superior to previous studies [22–25].

In addition, the calibration curves revealed excellent consistency in forecasting 1-, 2-, and 3-year survival rates based on real observations and nomogram predictions. Further risk classification based on the individual's overall score revealed that the low-risk group had a superior mOS than the moderate-to-high-risk group (p < 0.001). Based on these findings, this comprehensive model has potential prediction performance and can be employed in clinical research as a stratification criterion for HBV-related HCC.

In our study, we gathered information such as laboratory tests, past medical history, tumor burden, liver function, staging, demographic information, and treatment, among other things. The Child–Pugh class, ALT, PVTT, tumor number, tumor diameter, extrahepatic metastasis, and treatment were all found to be independent predictors of survival, prompting the development of a new nomogram. Previous research has also found that these indicators are related to prognosis [5,13,21,26,27]. Moreover, the AUC values of models constructed by combining tumor burden, laboratory tests, and treatment were greater than those of the individual predictors in our study. This further demonstrates that the model we developed is an intuitive clinical tool with good predictive performance to assist physicians in making rational HBV-related HCC treatment decisions. The constructed nomogram also illustrated the impact of various treatments on prognosis. Patients receiving only supportive care had the highest risk scores, whereas patients receiving postoperative adjuvant TACE had a higher survival rate than those receiving only surgery. Similar phenomena have been described in prior research [28,29]. Patients receiving radiofrequency ablation (RFA) had the lowest risk scores, which was a striking anomaly. Numerous studies have reported comparable efficacy between RFA and surgery [6,30,31]. This may be due to the small number of training cohort patients who received RFA (n = 53). In addition, risk scores for various treatments are not recommended as a direct guide for physicians to make treatment decisions because different treatments have different indications. Clinical decisions should be based on tumor burden and patient comorbidities, among other factors.

Although some nomograms for predicting the prognosis of HBV-related HCC have been developed [22,32], the model we developed has a larger sample size and more comprehensive data. Furthermore, this is the first multicenter, large-sample study to include multiple treatment modalities in predicting the prognosis of HBV-related HCC. It provides a theoretical foundation for the current comprehensive HCC treatment. This helps clinicians make clinical decisions about individualized HBV-related HCC treatment and lays the groundwork for the clinical management of high-risk patients.

In spite of the aforementioned benefits, the limitations of our study cannot be overlooked. First, because this is a retrospective study, we cannot disregard the possibility of selective bias. Nonetheless, the large number of samples increases the model's reliability. The heterogeneity generated by different immune checkpoint inhibitors (ICIs) also affects the predictive performance of our model, despite the inclusion of immunotherapy in our study. Besides, the lack of external validation also affects our interpretation of the results. In addition, we cannot provide data on the causes of death and perform competing risks analysis. Finally, we cannot directly compare the different available nomograms with their prediction rule in the same cohort of patients due to the lack of relevant data. Future experiments will require prospective studies and more specific drugs to further validate the model we constructed.

5Conclusions

The nomogram we constructed showed good performance in predicting a 1-year survival rate for HBV- related HCC. For HBV-related HCC patients with a high risk of death, a close surveillance program and adjuvant therapy should be considered.

Funding

This work was supported by Sichuan Science and Technology Program (2022YFS0622).

Author Contributions

Ke Su, Han Li, Lu Guo, Ke Xu, Tao Gu, Hao Chi, Yanlin Liu, Xueting Li, Lianbin Wen, Yanqiong Song, Kun He, Qulian Guo, Jiali Chen, Zhenying Wu, Yi Jiang, and Yunwei Han collected the data. Yunwei Han and Kun He designed the research study. Ke Su, Lu Guo, Jian Tong, Tao Gu, and Ke Xu wrote the manuscript and analyzed the data. Qiuni Shen participated in the revision of the manuscript. All authors approved the final version of the manuscript.

References
[1]
Sung H, Ferlay J, Siegel R, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. 2021;71(3):209-49. https://doi.org/10.3322/caac.21660.
[2]
Zhang Z, Tan J, Wang F, Dao F, Zhang Z, Lin HJFib, et al. Early diagnosis of hepatocellular carcinoma using machine learning method. 2020;8:254. https://doi.org/10.3389/fbioe.2020.00254.
[3]
Finn R, Qin S, Ikeda M, Galle P, Ducreux M, Kim T, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. 2020;382(20):1894-905. https://doi.org/10.1056/NEJMoa1915745.
[4]
Su K, Guo L, He K, Rao M, Zhang J, Yang X, et al. PD-L1 expression on circulating tumor cells can be a predictive biomarker to PD-1 inhibitors combined with radiotherapy and antiangiogenic therapy in advanced hepatocellular carcinoma. 2022;12:873830. https://doi.org/10.3389/fonc.2022.873830.
[5]
Su K, Gu T, Xu K, Wang J, Liao H, Li X, et al. Gamma knife radiosurgery versus transcatheter arterial chemoembolization for hepatocellular carcinoma with portal vein tumor thrombus: a propensity score matching study. 2022;16(4):858-67. https://doi.org/10.1007/s12072-022-10339-2.
[6]
Takayama T, Hasegawa K, Izumi N, Kudo M, Shimada M, Yamanaka N, et al. Surgery versus radiofrequency ablation for small hepatocellular carcinoma: a randomized controlled trial (SURF Trial). 2022;11(3):209-18. https://doi.org/10.1159/000521665.
[7]
Ricke J, Klümpen H, Amthauer H, Bargellini I, Bartenstein P, de Toni E, et al. Impact of combined selective internal radiation therapy and sorafenib on survival in advanced hepatocellular carcinoma. 2019;71(6):1164-74. https://doi.org/10.1016/j.jhep.2019.08.006.
[8]
Chen W, Zheng R, Baade P, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. 2016;66(2):115-32. https://doi.org/10.3322/caac.21338.
[9]
Zhu R, Seto W, Lai C, Yuen MJG, liver. Epidemiology of hepatocellular carcinoma in the Asia-Pacific region. 2016;10(3):332-9. https://doi.org/10.5009/gnl15257.
[10]
Chun Y, Kim S, Park J, Kim D, Han K, Chon C, et al. Prognostic value of the 7th edition of the AJCC staging system as a clinical staging system in patients with hepatocellular carcinoma. 2011;47(17):2568-75. https://doi.org/10.1016/j.ejca.2011.07.002.
[11]
Meram E, Longhurst C, Vardar B, Karaoglu K, Laeseke P, Ozkan OJH. Evaluation of staging systems to predict prognosis in hepatocellular carcinoma patients treated with radioembolization. 2022;8(1):e08770. https://doi.org/10.1016/j.heliyon.2022.e08770.
[12]
Forner A, Reig M, de Lope C, Bruix JJSild. Current strategy for staging and treatment: the BCLC update and future prospects. 2010;30(1):61-74. https://doi.org/10.1055/s-0030-1247133.
[13]
Kumada T, Toyoda H, Tada T, Yasuda S, Tanaka JJLc. Changes in background liver function in patients with hepatocellular carcinoma over 30 years: comparison of child-pugh classification and albumin bilirubin grade. 2020;9(5):518-28. https://doi.org/10.1159/000507933.
[14]
Zhang J, Wang T, Xu L, Wang P, Zhang M, Xu MJCca, et al. Development and validation of a prognostic model based on the albumin-to-fibrinogen ratio (AFR) and gamma-glutamyl transpeptidase-to-platelet ratio (GPR) in hepatocellular carcinoma patients. 2020;511:107-16. https://doi.org/10.1016/j.cca.2020.09.038.
[15]
Yan B, Su B, Bai D, Qian J, Zhang C, Jin S, et al. A practical nomogram and risk stratification system predicting the cancer-specific survival for patients with early hepatocellular carcinoma. 2021;10(2):496-506. https://doi.org/10.1002/cam4.3613.
[16]
Balachandran V, Gonen M, Smith J, DeMatteo RJTLO. Nomograms in oncology: more than meets the eye. 2015;16(4):e173-80. https://doi.org/10.1016/S1470-2045(14)71116-7.
[17]
Vickers A, van Calster B, Steyerberg EJD, research p. A simple, step-by-step guide to interpreting decision curve analysis. 2019;3:18. https://doi.org/10.1186/s41512-019-0064-7.
[18]
Finn R, Ikeda M, Zhu A, Sung M, Baron A, Kudo M, et al. Phase Ib study of lenvatinib plus pembrolizumab in patients with unresectable hepatocellular carcinoma. 2020;38(26):2960-70. https://doi.org/10.1200/JCO.20.00808.
[19]
Xu J, Shen J, Gu S, Zhang Y, Wu L, Wu J, et al. Camrelizumab in combination with apatinib in patients with advanced hepatocellular carcinoma (RESCUE): a nonrandomized, open-label, phase II trial. 2021;27(4):1003-11. https://doi.org/10.1158/1078-0432.CCR-20-2571.
[20]
Guo Y, Ren Y, Chen L, Sun T, Zhang W, Sun B, et al. Transarterial chemoembolization combined with camrelizumab for recurrent hepatocellular carcinoma. 2022;22(1):270. https://doi.org/10.1186/s12885-022-09325-6.
[21]
Reig M, Forner A, Rimola J, Ferrer-Fàbrega J, Burrel M, Garcia-Criado Á, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. 2022;76(3):681-93. https://doi.org/10.1016/j.jhep.2021.11.018.
[22]
Mo Q, Liu Y, Zhou Z, Li R, Gong W, Xiang B, et al. Prognostic value of aspartate transaminase/alanine transaminase ratio in patients with hepatitis B virus-related hepatocellular carcinoma undergoing hepatectomy. 2022;12:876900. https://doi.org/10.3389/fonc.2022.876900.
[23]
Zheng Z, Guan R, Zou Y, Jian Z, Lin Y, Guo R, et al. Nomogram based on inflammatory biomarkers to predict the recurrence of hepatocellular carcinoma-A multicentre experience. 2022;15:5089-102. https://doi.org/10.2147/JIR.S378099.
[24]
Huang W, Tsai C, Que J, Lo C, Lin Y, Dai Y, et al. Development and validation of a nomogram for patients with nonmetastatic BCLC stage C hepatocellular carcinoma after stereotactic body radiotherapy. 2020;9(3):326-37. https://doi.org/10.1159/000505693.
[25]
Zhang L, Luo P, Chen L, Song D, Xu A, Xu P, et al. Model to predict overall survival in patients with hepatocellular carcinoma after curative hepatectomy. 2020;10:537526. https://doi.org/10.3389/fonc.2020.537526.
[26]
Qian X, Liu Y, Wu F, Zhang S, Gong J, Nan Y, et al. The performance of serum alpha- fetoprotein for detecting early-stage hepatocellular carcinoma is influenced by antiviral therapy and serum aspartate aminotransferase: a study in a large cohort of hepatitis B virus-infected patients. 2022;14(8). https://doi.org/10.3390/v14081669.
[27]
Zheng Z, Liu Z, Zhang H, Guo X, Jia X, Wang J, et al. Efficacy and safety of apatinib in advanced hepatocellular carcinoma: a multicenter real world retrospective Study. 2022;13:894016. https://doi.org/10.3389/fphar.2022.894016.
[28]
Esagian S, Kakos C, Giorgakis E, Burdine L, Barreto J, Mavros MJC. Adjuvant transarterial chemoembolization following curative-intent hepatectomy versus hepatectomy alone for hepatocellular carcinoma: a systematic review and meta-analysis of randomized controlled trials. 2021;13(12). https://doi.org/10.3390/cancers13122984.
[29]
Wang Z, Ren Z, Chen Y, Hu J, Yang G, Yu L, et al. Adjuvant transarterial chemoembolization for HBV-related hepatocellular carcinoma after resection: a randomized controlled study. 2018;24(9):2074-81. https://doi.org/10.1158/1078-0432.CCR-17-2899.
[30]
Lee J, Jin Y, Shin S, Kwon J, Kim S, Suh Y, et al. Surgery versus radiofrequency ablation in patients with Child- Pugh class-A/single small (≤3 cm) hepatocellular carcinoma. 2022;28(2):207-18. https://doi.org/10.3350/cmh.2021.0294.
[31]
Hur M, Lee J, Kim J, Hong J, Park M, Cho H, et al. Comparison of overall survival between surgical resection and radiofrequency ablation for hepatitis B-related hepatocellular carcinoma. 2021;13(23). https://doi.org/10.3390/cancers13236009.
[32]
Zhang J, Wang Z, Guo X, Zhang J, Ni JJJogo. A nomogram for predicting the risk of postoperative recurrence of hepatitis B virus-related hepatocellular carcinoma in patients with high preoperative serum glutamyl transpeptidase. 2022;13(1):298-310. https://doi.org/10.21037/jgo-21-450.

These authors contributed equally to this work

Copyright © 2023. Fundación Clínica Médica Sur, A.C.
Article options
Tools
Supplemental materials
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

Quizás le interese:
10.1016/j.aohep.2023.101185
No mostrar más