Preoperative and postoperative nomograms in predicting early recurrence of hepatocellular carcinoma without macrovascular invasion after curative resection

DOI: https://doi.org/10.21203/rs.3.rs-891054/v2

Abstract

Background: Postoperative early recurrence (ER) is a major obstacle for long-term survival in patients with hepatocellular carcinoma (HCC) after curative liver resection (LR). This study aimed to establish pre- and postoperative nomograms in predicting ER for HCC without macrovascular invasion.

Methods: The patients who underwent curative LR for HCC from January 2012 to December 2016 were divided into training and internal prospective validation cohorts. Nomograms were constructed based on the independent risk factors derived from multivariate logistic regression analyses in training cohort. The predictive performance of nomograms was validated by internal prospective validation cohort.

Results: A total of 698 patients fulfilled with eligible criteria. Among them, 265 out of 482 patients (55.0%) in training cohort and 120 out 216 (55.6%) patients in validation cohort developed ER. The preoperative risk factors associated with ER were age, alpha fetoprotein (AFP), tumor diameter, tumor number; the postoperative risk factors associated with ER were age, tumor diameter, tumor number, microvasular invasion (MVI) and differentiation. The pre- and postoperative nomograms based on these factors showed good accuracy with C-indices of 0.712 and 0.850 in training cohort, and 0.754 and 0.857 in validation cohort, respectively. The calibration curves showed optimal agreement between the prediction by the nomograms and actual observation. The area under the receiver operating characteristic curves of pre- and postoperative nomograms were 0.721 and 0.848 in training cohort, and 0.754 and 0.844 in validation cohort, respectively.

Conclusions: Present nomograms showed good performance in predicting ER for HCC without macrovascular invasion before and after surgery, which were helpful for doctors in designation of treatments and selection of patients for regularly surveillance or administration of neoadjuvant therapies.

Background

Hepatocellular carcinoma (HCC) is the sixth most frequency cancer and the third leading cause of cancer-related death all over the world.[1, 2] Due to the prevalence of hepatitis C virus and alcohol intake, the incidence of HCC in western countries is increasing.[3, 4] Liver transplantation (LT), liver resection (LR) and radiofrequency ablation (RFA) were considered as three main curative modalities for patients with HCC.[2, 5] In clinical practice, owing to the donor shortage for LT and tumor location or diameter limitation for RFA, LR is the most common therapy for early and partial intermediate stage HCC.[5] In recent years, with the remarkable improvement of surgical techniques and perioperative management, selected patients with advanced stage HCC were also could benefit from LR.[6–9] Unfortunately, the dramatically high incidence of postoperative recurrence significantly decreased the survival expectancy for patients with HCC after curative LR.[2, 10, 11]

There are two common pattern of postoperative recurrence of HCC: early recurrence (ER) (< or = 2 years ) deriving from occult metastasis from the initial tumor and late recurrence or de novo HCC (> 2 years after surgery) arising from underlying liver diseases.[12–15] Since ER accounts for up to 70% of all postoperative recurrence and indicates poor long-term survival, more attention on ER has been drawn from researchers.[13–15] On the one hand, multiple risk factors associated with ER such as microvascular invasion (MVI), high preoperative alpha fetoprotein (AFP) level, chronic active hepatitis, the absence of tumor capsule and large tumor size have been revealed.[12–16] On the other hand, accumulate evidence showed that various postoperative adjuvant therapies such as TACE,[17–19] adoptive immunotherapy,[20] iodine-131-labled lipodol,[21, 22] interferon,[23] or cancer vaccines[24] could effectively delay the postoperative recurrence of HCC, although these results need to be further verified. Therefore, indentifying patients with high risk of ER to receive appropriate adjuvant therapies might be a promising avenue in prolonging the overall survival time after curative LR.

Nomogram has been accepted by abundant investigators in predicting outcomes of various diseases.[25–29] It is constructed based on the independent risk factors of special endpoints and showed more accurate than commonly used staging systems.[30] Recently, Zhang et al.[31] established a nomogram which was used to predict the incidence of ER in HCC with portal vein tumor thrombus after R0 LR. However, most curative LR was performed on the patients without macrovascular invasion, and the nomograms for predicting ER of this subgroup of patients is lacking. Additionally, to the best of our knowledge, no studies separately analyzed the relationship between preoperative clinical parameters and ER. And we believe a preoperative model for prediction of ER is helpful for surgeons in selecting optimal therapies for patients with high risk of postoperative ER.

In this study, independent preoperative and postoperative risk factors of ER was revealed using a large cohort of patients with HCC without macrovascular invasion. Then two nomograms were generated to predict ER before and after surgery based on these factors, respectively. The performance of these nomograms was validated by an internal prospective cohorts and receiver operating characteristic (ROC) curves.

Methods

Patients

To delete the heterogeneity on treatment of HCC from treatment concept and surgical techniques, this study only included the patients who underwent curative-intent resection for HCC from January 2012 to December 2016 in West China hospital, Sichuan University. The inclusion criteria included following items: (1) pathologically diagnosed as HCC without lymph node metastasis; (2) absence of tumor thrombus in major branches of the portal or hepatic veins; (3) receiving curative LR initially which was defined as removal of all recognizable tumor with a clear margin; (4) age not less than 18 years; (5) Child-Pugh A liver function. The excluding criteria were (1) patients with other type of tumors; (2) loss of follow-up within 2 years; (3) poor function of other major organ (heart, lung and kidney) and (4) incomplete data. Written informed consent was obtained from each patient for data to be used for research analysis. This study was approved by ethics committee of West China Hospital, Sichuan University.

Surgery

For preparation of surgery, imaging examinations included contrast-enhanced computerized tomography (CT), and/or magnetic resonance imaging (MRI) were performed to help evaluate the characteristics of the tumor. Routine blood tests included blood cell analysis, liver/renal/coagulation function tests, hepatitis B/C virus (HBV/HCV) screening tests, HBV deoxyribonucleic acid (HBV-DNA) measurement, serum tumor markers including alpha-fetal protein (AFP), carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA 19-9) and CA 125 were performed. The heart and lung function were primarily evaluated by electrocardiogram and chest x-ray, echocardiography and pulmonary function tests were added if necessary. Before surgery, a multidisciplinary team (MDT) consultation was routinely carried out to design the individual treatment for all patients. Surgical decisions were made based on tumor characteristics, reserve liver function, American Society of Anesthesiologists score[32] and technological in feasibility of LR.

All eligible patients were subjected to open surgery. A right subcostal incision with midline extension was normally obtained. Intraoperative ultrasound (US) was routinely performed to seek the presence of additional nodules which were not revealed preoperatively and determine a tumor-free margin of at least 1 cm. Anatomic resection was the first option for patients with ideal tumor location and no obvious liver cirrhosis. To decrease surgical blood loss, intermittent Pringle’s maneuver was performed at a cycle of 15/5 minutes of champ/unclamp time. After removal from the body, the tumor specimens were fixed with 4% paraformaldehyde within 15 minutes and then delivered to histological department for histological examination. Finally, surgical information including surgery duration, resection type, blood loss and transfusion were recorded carefully.

Follow-up

The postoperative follow-up program has been described in our previous study.[33] In brief, all patients were regularly followed up at the first postoperative month, at 3-months intervals for the subsequent 3 years, and every half year thereafter. Abdominal ultrasonography, serum AFP level, HBV-DNA load and liver function were routinely examined at each follow-up. Enhanced CT or MRI scan would be performed when suspicious lesions were found or persistent elevated AFP were measured. If necessary, bone scintigraphy or positron emission tomography was conducted to confirm bone or distant metastasis. Tumor recurrence was diagnosed based on the typical appearance of a new lesion in at least two radiological examinations, with or without elevated AFP levels. Once HCC recurrence was diagnosed, most appropriate treatments such as rehepatectomy, RFA, salvage LT, TACE, sorafenib, or best care support were recommended according to the characteristics of recurrent tumors and liver function. 

Recurrence time was defined as the time interval between the operation and the first diagnosis of recurrence. In line with previous studies, we classified the patients with recurrence time no more than 2 years into ER group, otherwise, late recurrence was considered.[12-15] The overall survival (OS) was defined as the time interval between the operation and death or last follow-up. The follow-up was censored in March 2019. 

Statistical analysis

All statistical analyses were performed in SPSS version 24.0 (IBM SPSS Inc, Chicago, IL) and R software version 3.5.0 with the rms package (http://www.r-project.org/). Continuous variables with a normal distribution were expressed as mean (standard deviation, SD) and analyzed using Student’s test or Mann-Whitney U test. Categorical variables were expressed as number or percentage and compared using Pearson Chi-Square or Fisher’s exact test. For all included patients, the cases who underwent curative LR between January 2012 and December 2014 were allocated into the training cohort, the cases who underwent curative LR between January 2014 and December 2016 were stratified into internal prospective validation cohort. Univariate and stepwise multivariate analyses were performed using logistic regression methods to identify the independent risk factors related to ER in the training cohort. Nomograms for preoperative and postoperative prediction of ER were generated based on the results of multivariate logistic regression analyses, respectively. The predictive performance of nomograms was measured by concordance index (C-index) and calibration curves. Validation of the model performance was performed using a internal prospective validation cohort. For clinical use of present nomograms, the total pre- and postoperative risk scores of each patient were calculated using the nomograms. Receiver operating characteristic (ROC) curve analysis was performed to calculate the optimal cut off values that were determined by maximizing the Youden index (sensitivity + specificity - 1). The predictive ability of the optimal cut off values was assessed by the sensitivity, specificity, predictive values, and likelihood ratios. All analyses were two tailed, p < 0.05 was considered statistically significant.

Results

The clinical characteristics of included patients

From January 2012 to December 2016, a total of 698 patients who fulfilled with our eligible criteria were included in this study. Among them, 482 patients who underwent curative LR before January 2015 were allocated into the training cohort, the later 216 patients were stratified into internal prospective validation cohort. The detailed information of these two cohorts were listed in Table 1. Except the neutrophil-to-lymphocyte ratio (NLR) (P = 0.007), platelet-to-lymphocyte ratio (PLR) (P = 0.011) and the presence of cirrhosis (P = 0.016), other baseline and clinicopathologic data were comparable between the training and validation cohorts. The median follow-up for all included patients was 36 months (range, 1–78 months). The incidence of ER was observed in 265 (55.0%) and 120 (55.6%) patients in the training and validation cohorts, respectively. For all included patients, the 1, 3 and 5-year OS rates in ER group were 66.47%, 23.85% and 13.18%, which was dramatically lower than that in late recurrence group with the 1, 3 and 5-year survival rate of 99.68%, 96.75% and 83.30%, respectively (P < 0.0001) (Fig. 1).

Table 1

The baseline and clinical characteristics of HCC patients in the training and validation cohorts

Clinical parameters

Training cohort (n = 482)

Validation cohort (n = 216)

P

Gender (male/famale)

404 (83.8%)/78 (16.2%)

185 (85.6%)/31 (14.4%)

0.538

Age, mean (SD)

49.58 (12.52)

51.68 (10.83)

0.698

HBsAg (positive/negative)

417 (86.5%)/65 (13.5%)

187 (87.0%)/29 (13.0%)

0.868

HBV-DNA (≥ 103/<103 copies/ml)

261 (54.2%)/221 (45.8%)

107 (49.5%)/109 (50.5%)

0.282

HBeAg (positive/negative)

97 (20.1%)/385 (79.9%)

45 (21.0%)/171 (79.0%)

0.323

AFP (< 20/20–400/>400 ng/mL)

157 (32.6%)/119 (24.7%)/206 (42.7%)

68 (31.6%)/54 (25.1%)/94 (43.3%)

0.966

NEU (> 3.56/≤3.56×109/L)

228 (47.4%)/254 (52.6%)

99 (46.0%)/117 (54.0%)

0.741

LYM (> 1.1/≤1.1×109/L)

362 (75.1%)/120 (24.9%)

171 (79.1%)/45 (20.9%)

0.255

PLT (> 100/≤100×109/L)

351 (72.8%)/131 (27.2%)

143 (66.2%)/73 (33.8%)

0.076

NLR (> 3/≤3)

153 (31.8%)/329 (68.2%)

47 (21.9%)/169 (78.1%)

0.007

PLR (> 111/≤111)

180 (37.3%)/302 (62.7%)

59 (27.4%)/157 (72.6%)

0.011

TBIL (> 28/≤28 umol/L)

15 (3.1%)/467 (96.9%)

8 (3.7%)/208 (96.3%)

0.689

ALT (> 50/≤50 IU/L)

161 (33.4%)/321 (66.6%)

76 (35.2%)/140 (64.8%)

0.646

AST (> 40/≤40 IU/L)

216 (44.8%)/266 (55.2%)

101 (46.8%)/115 (53.2%)

0.633

ALB (> 40/≤40g/L)

308 (63.9%)/174 (36.1%)

131 (60.6%)/85 (39.4%)

0.411

GGT (> 60/≤60IU/L)

240 (49.8%)/242 (50.2%)

114 (52.8%)/102 (47.2%)

0.466

PT (> 12.8/≤12.8 s)

119 (24.7%)/363 (75.3%)

39 (17.9%)/177 (82.1%)

0.344

INR (> 1.15/≤1.15)

101 (21.0%)/381 (79.0%)

50 (23.1%)/166 (76.9%)

0.515

Fib (> 2/≤2 g/L)

401 (83.2%)/81 (16.8%)

173 (79.9%)/43 (20.1%)

0.338

Tumor diameter (≤ 5/5–10/>10 cm)

219 (45.4%)/199 (41.2%)/65 (13.4%)

105 (48.4%)/89 (41.2%)/29 (13.4%)

0.531

Tumor number (1/2/3)

402 (83.4%)/58 (12.0%)/22 (4.6%)

170 (78.6%)/37 (17.2%)/9 (4.2%)

0.284

BCLC stage (A/B)

409 (84.9%)/73 (15.1%)

178 (82.3%)/38 (17.7%)

0.399

Cirrhosis (present/absent)

296 (61.4%)/186 (38.6%)

153 (70.8%)/63 (29.2%)

0.016

Differentiation (I + II/III + IV)

279 (57.9%)/203 (42.1%)

123 (56.9%)/93 (43.1%)

0.816

MVI (present/absent)

204 (42.3%)/278 (57.7%)

86 (39.8%)/130 (60.2%)

0.534

Statelite lesion (present/absent)

72 (14.9%)/410 (85.1%)

31 (14.4%)/185 (85.6%)

0.840

Resection (anatomic/non-anatomic)

250 (51.9%)/232 (48.1%)

110 (50.9%)/106 (49.1%)

0.818

Bold numbers indicate statistical significance
SD, standard deviation;HBsAg, hepatitis B surface antigen; HBV-DNA, hepatitis B virus deoxyribonucleic acid; HBeAg, hepatitis B e antigen; AFP, alpha-fetoprotein; NEU, neutrophil; LYM, lymphocyte; PLT, platelet; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; TBIL, total bilirubin; ALT, alanine transaminase; AST, aspartate aminotransferase; ALB, albumin; GGT, gamma-glutamyl transpeptidase; PT, prothrombin time; s, second; INR, international normalized ratio; Fib, fibrinogen; BCLC, Barcelona Clinic Liver Cancer staging system; MVI, microvascular invasion.


The independent predictors for early recurrence 

As shown in Table 2, univariate logistic analyses revealed that numerous variables including gender, age, hepatitis B surface antigen (HBsAg), HBV-DNA, hepatitis B e antigen (HBeAg), AFP, neutrophil (NEU), platelet (PLT), NLR, PLR, aspartate aminotransferase (AST), gamma-glutamyl transpeptidase (GGT), tumor diameter, tumor number, Barcelona Clinic Liver Cancer (BCLC) stage, differentiation, MVI, satellite lesions and resection type were significantly associated with the ER in training cohort. Subsequent multivariate analyses further revealed that four preoperative risk factors including age (odds ratio (OR) = 0.894, 95% confidence interval (CI): 0.987–0.990, P < 0.001), AFP level (OR = 1.328, 95% CI: 1.077–1.639, P = 0.008), tumor diameter (OR = 2.209, 95% CI: 1.645–2.967, P < 0.001) and tumor number (OR = 2.556, 95% CI: 1.474–4.434, P = 0.001), and five postoperative risk factors including age (OR = 0.981, 95% CI: 0.975–0.987, P < 0.001), tumor diameter (OR = 1.943, 95% CI: 1.438–2.624, P < 0.001), tumor number (OR = 1.826, 95% CI: 1.024–3.255, P = 0.041), differentiation (OR = 1.508, 95% CI: 1.059–2.358, P = 0.025) and MVI (OR = 2.904, 95% CI: 1.914–4.405, P < 0.001) were significantly associated with ER of HCC patients without macrovascular invasion after curative LR (Table 3).

Table 2

Univariate logistic analysis on clinical parameters in predicting early recurrence in the training cohort.

Clinical parameters

OR (95% CI)

P

Gender (male/famale)

1.27 (1.043–1.545)

0.017

Age (> 60/≤60 years)

0.596 (0.480–0.740)

< 0.001

HBsAg (positive/negative)

1.262 (1.075–1.482)

0.004

HBV-DNA (≥ 103/<103 copies/ml)

1.529 (1.164–2.010)

0.002

HBeAg (positive/negative)

1.771 (1.170–2.681)

0.007

AFP (< 20/20–400/>400 ng/ml)

1.337 (1.171–1.527)

< 0.001

NEU (> 3.56/≤3.56×109/L)

1.621 (1.241–2.117)

< 0.001

LYM (> 1.1/≤1.1×109/L)

1.221 (0.993–1.502)

0.059

PLT (> 100/≤100×109/L)

1.472 (1.189–1.821)

< 0.001

NLR (> 3/≤3)

2.000 (1.429–2.799)

< 0.001

PLR (> 111/≤111)

1.687 (1.247–2.282)

0.001

TB (> 28/≤28 umol/L)

2.000 (0.684–5.581)

0.206

ALT (> 50/≤50 IU/L)

1.368 (1.000-1.870)

0.05

AST (> 40/≤40 IU/L)

1.602 (1.218–2.108)

0.001

ALB (> 40/≤40g/L)

1.184 (0.947–1.482)

0.139

GGT (> 60/≤60IU/L)

1.697 (1.306–2.205)

< 0.001

PT (> 12.8/≤12.8 s)

1.164 (0.812–1.668)

0.41

INR (> 1.15/≤1.15)

1.244 (0.841–1.842)

0.275

Fib (> 2/≤2 g/L)

1.079 (0.873–1.335)

0.482

Tumor size (≤ 5/5–10/>10 cm)

1.791 (1.460–2.198)

< 0.001

Tumor number (1/2/3)

1.275 (1.108–1.466)

0.001

BCLC stage (A/B)

3.056 (1.795–5.203)

< 0.001

Cirrhosis (present/absent)

1.193 (0.949–1.499)

0.131

Differentiation (I + II/III + IV)

1.819 (1.365–2.426)

< 0.001

MVI (present/absent)

2.923 (2.133–4.005)

< 0.001

Statelite lesion (present/absent)

2.429 (1.461–4.037)

0.001

Resection (anatomic/non-anatomic)

1.475 (1.146–1.899)

0.003

Bold numbers indicate statistical significance
OR, odds ratio; CI, confidence interval; HBsAg, hepatitis B surface antigen; HBV-DNA, hepatitis B virus deoxyribonucleic acid; HBeAg, hepatitis B e antigen; AFP, alpha-fetoprotein; NEU, neutrophil; LYM, lymphocyte; PLT, platelet; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; TBIL, total bilirubin; ALT, alanine transaminase; AST, aspartate aminotransferase; ALB, albumin; GGT, gamma-glutamyl transpeptidase; PT, prothrombin time; s, second; INR, international normalized ratio; Fib, fibrinogen; BCLC, Barcelona Clinic Liver Cancer staging system; MVI, microvascular invasion.

 

Table 3

Multivariate logistic analysis on clinical parameters in predicting early recurrence in the training cohort.

Clinical parameters

OR (95% CI)

P

Preoperative

   

Age (> 60/≤60 years)

0.984 (0.987–0.990)

< 0.001

AFP (< 20/20–400/>400 ng/ml)

1.328 (1.077–1.639)

0.008

Tumor diameter (≤ 5/5–10/>10 cm)

2.209 (1.645–2.967)

< 0.001

Tumor number (1/2/3)

2.556 (1.474–4.434)

0.001

Postoperative

   

Age (> 60/≤60 years)

0.981 (0.975–0.987)

< 0.001

Tumor diameter (≤ 5/5–10/>10 cm)

1.943 (1.438–2.624)

< 0.001

Tumor number (1/2/3)

1.826 (1.024–3.255)

0.041

Differentiation (I + II/III + IV)

1.580 (1.059–2.358)

0.025

MVI (present/absent)

2.904 (1.914–4.405)

< 0.001

OR, odds ratio; CI, confidence interval; AFP, alpha-fetoprotein; MVI, microvascular invasion.

 

Construction of pre- and postoperative nomograms for predicting the early recurrence

Based on the results of multivariate analyses in training cohort, two nomograms for pre- or postoperatively predicting the ER were generated, respectively (Fig. 2). The C-indices of pre- and postoperative nomograms in training cohort were 0.712 (95% CI: 0.666–0.758, P < 0.001) and 0.850 (95% CI: 0.781–0.919, P < 0.001), respectively. The calibration plots showed ideal agreement on the incidence of ER between the prediction by the nomograms and actual observation in follow-up (Fig. 2).

For clinical use of present nomograms, as shown in Fig. 1, the projection of each variable on the point scale meant unique score of each variable. After summing the scores of all variables, total points for each patient could be calculated. Then the projection of total points on the probability scale represented the individual probability for ER.

Validation of the prediction models

Firstly, the performance of pre- and postoperative nomograms was validated by an internal prospective validation cohort. The pre- and postoperative total points for each patient in validation cohort were formulated using above two nomograms, respectively. Then the pre- or postoperative total points was treated as a new risk factor to calculate the C-indices and produce calibration curves of ER. The results showed that the C-indices for pre- and postoperative prediction of ER in validation cohort were 0.754 (95% CI: 0.690–0.818, P < 0.001) and 0.857 (95% CI: 0.750–0.949, P < 0.001), respectively. The calibration curves also showed ideal consistency between prediction and observation in the probability of ER (Fig. 3).

In addition, the predictive performance of present nomograms was evaluated by ROC curves (Fig. 4). In training cohort, the area under the ROC curves (AUC) of pre- and postoperative nomograms were 0.721 (95% CI: 0.684–0.759, P < 0.001) and 0.848 (95% CI: 0.814–0.883, P < 0.001) respectively; in internal prospective validation cohort, the AUC of pre- and postoperative nomograms were 0.754 (95% CI: 0.690–0.817, P < 0.001) and 0.844 (95% CI: 0.790–0.897, P < 0.001), respectively, which were comparable with the C-indices of nomograms. These results indicated that present nomograms have good performance in predicting the ER for patients with HCC without macrovascular invasion after curative LR.

The predictive ability of nomograms

As shown in Table 4, the optimal cut-off values of the pre- and postoperative total nomogram scores were 88 (range: 4-284) and 110 (range: 5-356), respectively. For preoperative model, the sensitivity, specificity, positive predictive value, negative predictive value, positive likelihood ratio, and negative likelihood ratio in distinguishing ER were 0.611, 0.716, 0.704, 0.587, 2,151 and 0.543 in the training cohort, and 0.730, 0.677, 0.733, 0.674, 2.260 and 0.399 in the validation cohort. With respect for postoperative model, the sensitivity, specificity, positive predictive value, negative predictive value, positive likelihood ratio, and negative likelihood ratio in distinguishing ER were 0.706, 0.802, 0.793, 0.724, 3.564 and 0.367 in the training cohort, and 0.679, 0.800, 0.764, 0.699, 3.394 and 0.401 in the validation cohort.

Table 4

Predictive ability of the optimal cut off values on the risk of early recurrence

variables

Preoperative nomogram

Postoperative nomogram

Training cohort (482)

Validation cohort (216)

Training cohort (482)

Validation cohort (216)

AUC

0.721 (0.684–0.759)

0.754 (0.690–0.817)

0.848 (0.814–0.883)

0.844 (0.790–0.897)

Cut-off score

88

88

110

110

Sensitivity

0.611 (0.567–0.654)

0.642 (0.578–0.706)

0.706 (0.665–0.747)

0.679 (0.617–0.741)

Specificity

0.716 (0.676–0.756)

0.76 (0.703–0.817)

0.802 (0.766–0.838)

0.800 (0.746–0.853)

Positive predictive value

0.704 (0.663–0.745)

0.77 (0.714–0.826)

0.793 (0.756–0.829)

0.764 (0.707–0.821)

Negative predictive value

0.587 (0.543–0.631)

0.629 (0.565–0.693)

0.724 (0.684–0.764)

0.699 (0.638–0.760)

Positive likelihood ratio

2.151 (2.011–2.291)

2.675 (2.393–2.957)

3.564 (3.294–3.834)

3.394 (3.014–3.774)

Negative likelihood ratio

0.543 (0.499–0.587)

0.471 (0.404–0.538)

0.367 (0.324–0.411)

0.401 (0.336–0.466)

CI, confidence interval.

Discussion

With the development of surgical techniques and perioperative management, LR has been become more and more safe for early, most intermediate and even selected advanced stage HCC.[34] And prognostic analysis showed that LR was a more effective therapy when compared with other therapies for HCC patients with advanced tumor burden.[35, 36] However, postoperative recurrence, especially ER, significantly shorten the survival expectancy for patients received curative LR.[13, 15, 16] In this study, the postoperative survival time in ER group was significantly shorter than that in late recurrence group. In addition, the repeated treatments after recurrence not only seriously impacted on the living quality of patients but also heavily strengthened the medical burden.

The risk predictors for ER have been investigated by numerous studies. Imamura et al.[12] found that non-anatomical resection, presence of microscopic vascular invasion and serum AFP level ≥ 32 ng/mL were significantly associated with the ER of HCC after hepatectomy. Portolani et al.[13] reported that cirrhosis, chronic active hepatitis, HCV positivity, vascular infiltration and transaminases values were significantly associated with ER of HCC patients after hepatectomy. Cheng et al.[16] observed that tumor diameter > 5 cm, the absence of a tumor capsule and the presence of microvascular invasion were correlated with ER of solitary HCC after curative resection. A recent study conducted by Zhang et al.[31] revealed that HBV positivity, advanced type of PVTT, high HBV-DNA load, presence of satellite nodules, elevated AFP and large tumor diameter were significantly associated with ER of HCC with PVTT after R0 LR. In present study, using a large cohort of HCC patients without macrovascular invasion, four preoperative and five postoperative independent risk factors of ER were identified. The nomograms based on these factors showed good predictive ability for ER with the C-indices of 0.721 and 0.850 for pre- and postoperative models in training cohort, and 0.754 and 0.857 for pre- and postoperative models in validation cohort, respectively. Meanwhile, the calibration curves in training and validation cohorts showed ideal agreement between prediction and actual observation.

All risk factors which were incorporated in present nomograms were easily available and have been demonstrated associated with prognosis of HCC patients after curative LR. In this study, we found that the age was negatively associated with the incidence of postoperative ER. Compared with younger patients, the elderly HCC patients normally had lower AFP level, lower rate of HBsAg positivity and less tumor burden.[37–39] Furthermore, the expression level of some oncogenes were significantly lower in elderly patients when compared with younger patients.[40] Tumor size and numbers were commonly used as elements in various HCC staging systems.[41–43] Larger tumor size and more numbers meant higher probability of intrahepatic metastasis and indicated poorer prognosis.[44–47] Serum AFP level was not only a significant prognostic predictor for HCC patients[48–50] but also associated with many metastasis characteristics of HCC such as MVI,[51] incomplete tumor capsule[52, 53] and satellite lesions.[54] MVI is a discourage signal of intrahepatic vessel dissemination,[55] which was repeatedly demonstrated as an independent risk factor for ER and poor OS of HCC patients who underwent curative LR.[56–58] Tumors with poor pathological differentiation represents worse instinct of tumor cells, which had more powerful ability in proliferation and metastasis than tumors with well differentiation.[59, 60]

The ROC curves showed that the optimal cut-off values for pre- and postoperative nomograms were 88 and 110, respectively. Patients with a score equal or more than cut-off values meant high risk of ER. In clinical practice, present preoperative nomogram might helpful for surgeons in designation of therapy for HCC patients. The postoperative nomogram might serve as a tool to select patients for neoadjuvant therapies and more frequent surveillance.

This study had some limitations. First of all, the models of this study were constructed based on retrospective data, the performance of them need to be validated prospectively. Secondly, this study only included patients from a single center, future external validations are necessary. Thirdly, the main etiology of HCC in present study was HBV infection, the performance of present models for other etiologies-related HCC needs to be validated. Finally, other recurrence-related molecules are necessary to further improve the predictive accuracy of these nomograms.

Conclusion

Present study revealed that four preoperative and five postoperative clinical variables were significantly associated with the ER of HCC patients without macrovascular invasion after curative LR. Two nomograms based on these predictors showed ideal predictive performance. These prediction models were meaningful for doctors in designation of treatments before surgery and selection of patients for regular surveillance and administration of neoadjuvant therapies after surgery.

Abbreviations

HCC: Hepatocellular carcinoma; ER: Early recurrence; LR: Liver resection; RFA: Radiofrequency ablation; HBV: Hepatitis B virus; HBV-DNA: Hepatitis B virus deoxyribonucleic acid; HBeAg: Hepatitis B e antigen; PLR: Platelet-to-lymphocyte ratio; NLR: Neutrophil-to-lymphocyte ratio; AFP: Alpha-fetoprotein; MVI: Microvascular invasion; OS: Overall survival; ROC: Receiver operating characteristic; AUC: Area under the ROC curve.

Declarations

Acknowledgments

None

Authors’ contributions

TH and XMQ conceived and designed the study; ZYF, XLL and LXZ performed all experiments; XLL responsible for the analysis and interpretation of data. ZYF wrote the paper. All authors read and approved the final manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (Nos. 81802468 and 81772193), the 1.3.5 project for disciplines of excellence, West China Hospital, Sichuan University (Nos. ZYGD20009 and ZYJC18008), the Sichuan Province Key Technologies R&D Program (19ZDYF), the Sichuan Science and Technology Program 2019YFS0207.

Availability of data and materials

The datasets generated and analysed during the current study are not publicly available due to patient privacy but are available from the corresponding author on reasonable request.

Ethics approval and consent to participate

This study was approved by The Clinical Medical Research Ethics Committee of the West China Hospital of the Sichuan University (IRB number: FWA00009482IRBIORG0004190) and was performed in accordance with the Declaration of Helsinki. All patients signed a preoperative informed consent form and agreed to participate in the accompanying scientific research.

Consent for publication

The manuscript is approved for publication by all the authors. Written informed consent was obtained from the patients and/or their legal guardians for publication, and any accompanying images, sex, age of these patients.

Competing interests

The authors declare no competing interests.

References

  1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A: Global cancer statistics, 2012. CA: a cancer journal for clinicians 2015, 65(2):87-108.
  2. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, Gores G: Hepatocellular carcinoma. Nature reviews Disease primers 2016, 2:16018.
  3. Bertuccio P, Turati F, Carioli G, Rodriguez T, La Vecchia C, Malvezzi M, Negri E: Global trends and predictions in hepatocellular carcinoma mortality. Journal of hepatology 2017, 67(2):302-309.
  4. Siegel RL, Miller KD, Jemal A: Cancer statistics, 2018. 2018, 68(1):7-30.
  5. Hartke J, Johnson M, Ghabril M: The diagnosis and treatment of hepatocellular carcinoma. Seminars in diagnostic pathology 2017, 34(2):153-159.
  6. Chok KS, Cheung TT, Chan SC, Poon RT, Fan ST, Lo CM: Surgical outcomes in hepatocellular carcinoma patients with portal vein tumor thrombosis. World journal of surgery 2014, 38(2):490-496.
  7. Ye JZ, Zhang YQ, Ye HH, Bai T, Ma L, Xiang BD, Li LQ: Appropriate treatment strategies improve survival of hepatocellular carcinoma patients with portal vein tumor thrombus. World journal of gastroenterology 2014, 20(45):17141-17147.
  8. Kokudo T, Hasegawa K, Matsuyama Y, Takayama T, Izumi N, Kadoya M, Kudo M, Ku Y, Sakamoto M, Nakashima O et al: Survival benefit of liver resection for hepatocellular carcinoma associated with portal vein invasion. Journal of hepatology 2016, 65(5):938-943.
  9. Sakamoto K, Nagano H: Surgical treatment for advanced hepatocellular carcinoma with portal vein tumor thrombus. Hepatology research : the official journal of the Japan Society of Hepatology 2017, 47(10):957-962.
  10. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. Journal of hepatology 2012, 56(4):908-943.
  11. Belghiti J, Panis Y, Farges O, Benhamou JP, Fekete F: Intrahepatic recurrence after resection of hepatocellular carcinoma complicating cirrhosis. Annals of surgery 1991, 214(2):114-117.
  12. Imamura H, Matsuyama Y, Tanaka E, Ohkubo T, Hasegawa K, Miyagawa S, Sugawara Y, Minagawa M, Takayama T, Kawasaki S et al: Risk factors contributing to early and late phase intrahepatic recurrence of hepatocellular carcinoma after hepatectomy. Journal of hepatology 2003, 38(2):200-207.
  13. Portolani N, Coniglio A, Ghidoni S, Giovanelli M, Benetti A, Tiberio GA, Giulini SM: Early and late recurrence after liver resection for hepatocellular carcinoma: prognostic and therapeutic implications. Annals of surgery 2006, 243(2):229-235.
  14. Cucchetti A, Piscaglia F, Caturelli E, Benvegnu L, Vivarelli M, Ercolani G, Cescon M, Ravaioli M, Grazi GL, Bolondi L et al: Comparison of recurrence of hepatocellular carcinoma after resection in patients with cirrhosis to its occurrence in a surveilled cirrhotic population. Annals of surgical oncology 2009, 16(2):413-422.
  15. Poon RT: Differentiating early and late recurrences after resection of HCC in cirrhotic patients: implications on surveillance, prevention, and treatment strategies. Annals of surgical oncology 2009, 16(4):792-794.
  16. Cheng Z, Yang P, Qu S, Zhou J, Yang J, Yang X, Xia Y, Li J, Wang K, Yan Z et al: Risk factors and management for early and late intrahepatic recurrence of solitary hepatocellular carcinoma after curative resection. HPB : the official journal of the International Hepato Pancreato Biliary Association 2015, 17(5):422-427.
  17. Sun JJ, Wang K, Zhang CZ, Guo WX, Shi J, Cong WM, Wu MC, Lau WY, Cheng SQ: Postoperative Adjuvant Transcatheter Arterial Chemoembolization After R0 Hepatectomy Improves Outcomes of Patients Who have Hepatocellular Carcinoma with Microvascular Invasion. Annals of surgical oncology 2016, 23(4):1344-1351.
  18. Liu S, Guo L, Li H, Zhang B, Sun J, Zhou C, Zhou J, Fan J, Ye Q: Postoperative Adjuvant Trans-Arterial Chemoembolization for Patients with Hepatocellular Carcinoma and Portal Vein Tumor Thrombus. Annals of surgical oncology 2018, 25(7):2098-2104.
  19. Zhang XP, Liu YC, Chen ZH, Sun JX, Wang K, Chai ZT, Shi J, Guo WX, Wu MC, Lau WY et al: Postoperative Adjuvant Transarterial Chemoembolization Improves Outcomes of Hepatocellular Carcinoma Associated with Hepatic Vein Invasion: A Propensity Score Matching Analysis. Annals of surgical oncology 2019, 26(5):1465-1473.
  20. Takayama T, Sekine T, Makuuchi M, Yamasaki S, Kosuge T, Yamamoto J, Shimada K, Sakamoto M, Hirohashi S, Ohashi Y et al: Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet (London, England) 2000, 356(9232):802-807.
  21. Lau WY, Lai EC, Leung TW, Yu SC: Adjuvant intra-arterial iodine-131-labeled lipiodol for resectable hepatocellular carcinoma: a prospective randomized trial-update on 5-year and 10-year survival. Annals of surgery 2008, 247(1):43-48.
  22. Boucher E, Corbinais S, Rolland Y, Bourguet P, Guyader D, Boudjema K, Meunier B, Raoul JL: Adjuvant intra-arterial injection of iodine-131-labeled lipiodol after resection of hepatocellular carcinoma. Hepatology (Baltimore, Md) 2003, 38(5):1237-1241.
  23. Sun HC, Tang ZY, Wang L, Qin LX, Ma ZC, Ye QH, Zhang BH, Qian YB, Wu ZQ, Fan J et al: Postoperative interferon alpha treatment postponed recurrence and improved overall survival in patients after curative resection of HBV-related hepatocellular carcinoma: a randomized clinical trial. Journal of cancer research and clinical oncology 2006, 132(7):458-465.
  24. Peng BG, Liang LJ, He Q, Kuang M, Lia JM, Lu MD, Huang JF: Tumor vaccine against recurrence of hepatocellular carcinoma. World journal of gastroenterology 2005, 11(5):700-704.
  25. He Y, Zhu Z, Chen Y, Chen F, Wang Y, Ouyang C, Yang H, Huang M, Zhuang X, Mao R et al: Development and Validation of a Novel Diagnostic Nomogram to Differentiate Between Intestinal Tuberculosis and Crohn's Disease: A 6-year Prospective Multicenter Study. The American journal of gastroenterology 2019, 114(3):490-499.
  26. Wang Y, Li J, Xia Y, Gong R, Wang K, Yan Z, Wan X, Liu G, Wu D, Shi L et al: Prognostic nomogram for intrahepatic cholangiocarcinoma after partial hepatectomy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2013, 31(9):1188-1195.
  27. Gross JP, Whelan TJ, Parulekar WR, Chen BE, Rademaker AW, Helenowski IB, Donnelly ED, Strauss JB: Development and validation of a nomogram to predict lymphedema following axillary surgery and radiotherapy in women with breast cancer from the NCIC CTG MA.20 randomized trial. International journal of radiation oncology, biology, physics 2019.
  28. Chon YE, Park H, Hyun HK, Ha Y, Kim MN, Kim BK, Lee JH, Kim SU: Development of a New Nomogram Including Neutrophil-to-Lymphocyte Ratio to Predict Survival in Patients with Hepatocellular Carcinoma Undergoing Transarterial Chemoembolization. 2019, 11(4).
  29. Nassiri F, Mamatjan Y, Suppiah S, Badhiwala JH, Mansouri S, Karimi S, Saarela O, Poisson L, Gepfner-Tuma I, Schittenhelm J et al: DNA methylation profiling to predict recurrence risk in meningioma: development and validation of a nomogram to optimize clinical management. Neuro-oncology 2019.
  30. Balachandran VP, Gonen M, Smith JJ, DeMatteo RP: Nomograms in oncology: more than meets the eye. The Lancet Oncology 2015, 16(4):e173-180.
  31. Zhang XP, Chen ZH, Zhou TF, Li LQ, Chen MS, Wen TF, Shi J, Guo WX, Wu MC, Lau WY et al: A nomogram to predict early postoperative recurrence of hepatocellular carcinoma with portal vein tumour thrombus after R0 liver resection: A large-scale, multicenter study. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology 2019.
  32. Owens WD, Felts JA, Spitznagel EL, Jr.: ASA physical status classifications: a study of consistency of ratings. Anesthesiology 1978, 49(4):239-243.
  33. Xu L, Li L, Wang P, Zhang M, Zhang Y, Hao X, Yan L, Li B, Wen T, Xu M: Novel Prognostic Nomograms for Hepatocellular Carcinoma Patients with Microvascular Invasion: Experience from a Single Center. Gut and liver 2019.
  34. Vitale A, Burra P, Frigo AC, Trevisani F, Farinati F, Spolverato G, Volk M, Giannini EG, Ciccarese F, Piscaglia F et al: Survival benefit of liver resection for patients with hepatocellular carcinoma across different Barcelona Clinic Liver Cancer stages: a multicentre study. Journal of hepatology 2015, 62(3):617-624.
  35. Zhong JH, Ke Y, Gong WF, Xiang BD, Ma L, Ye XP, Peng T, Xie GS, Li LQ: Hepatic resection associated with good survival for selected patients with intermediate and advanced-stage hepatocellular carcinoma. Annals of surgery 2014, 260(2):329-340.
  36. Hsu CY, Liu PH, Hsia CY, Lee YH, Nagaria TS, Lee RC, Lin HC, Huo TI: Surgical Resection is Better than Transarterial Chemoembolization for Patients with Hepatocellular Carcinoma Beyond the Milan Criteria: A Prognostic Nomogram Study. Annals of surgical oncology 2016, 23(3):994-1002.
  37. Faber W, Stockmann M, Schirmer C, Mollerarnd A, Denecke T, Bahra M, Klein F, Schott E, Neuhaus P, Seehofer D: Significant impact of patient age on outcome after liver resection for HCC in cirrhosis. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology 2014, 40(2):208-213.
  38. Huang J, Li BK, Chen GH, Li JQ, Zhang YQ, Li GH, Yuan YF: Long-term outcomes and prognostic factors of elderly patients with hepatocellular carcinoma undergoing hepatectomy. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract 2009, 13(9):1627-1635.
  39. Motoyama H, Kobayashi A, Yokoyama T, Shimizu A, Sakai H, Furusawa N, Notake T, Kitagawa N, Arai T, Yokoi K et al: Impact of advanced age on the short- and long-term outcomes in patients undergoing hepatectomy for hepatocellular carcinoma: a single-center analysis over a 20-year period. American journal of surgery 2015, 209(4):733-741.
  40. Yan H, Yang Y, Zhang L, Tang G, Wang Y, Xue G, Zhou W, Sun S: Characterization of the genotype and integration patterns of hepatitis B virus in early- and late-onset hepatocellular carcinoma. Hepatology (Baltimore, Md) 2015, 61(6):1821-1831.
  41. Llovet JM, Bru C, Bruix J: Prognosis of hepatocellular carcinoma: the BCLC staging classification. Seminars in liver disease 1999, 19(3):329-338.
  42. Minagawa M, Ikai I, Matsuyama Y, Yamaoka Y, Makuuchi M: Staging of hepatocellular carcinoma: assessment of the Japanese TNM and AJCC/UICC TNM systems in a cohort of 13,772 patients in Japan. Annals of surgery 2007, 245(6):909-922.
  43. Yau T, Tang VY, Yao TJ, Fan ST, Lo CM, Poon RT: Development of Hong Kong Liver Cancer staging system with treatment stratification for patients with hepatocellular carcinoma. Gastroenterology 2014, 146(7):1691-1700.e1693.
  44. Chiappa A, Zbar AP, Audisio RA, Leone BE, Biella F, Staudacher C: Factors affecting survival and long-term outcome in the cirrhotic patient undergoing hepatic resection for hepatocellular carcinoma. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology 2000, 26(4):387-392.
  45. Fu YP, Yi Y, Huang JL, Jing CY, Sun J, Ni XC, Lu ZF, Cao Y, Zhou J, Fan J et al: Prognostic Nomograms Stratify Survival of Patients with Hepatocellular Carcinoma Without Portal Vein Tumor Thrombosis After Curative Resection. The oncologist 2017, 22(5):561-569.
  46. Izumi R, Shimizu K, Ii T, Yagi M, Matsui O, Nonomura A, Miyazaki I: Prognostic factors of hepatocellular carcinoma in patients undergoing hepatic resection. Gastroenterology 1994, 106(3):720-727.
  47. Nathan H, Schulick RD, Choti MA, Pawlik TM: Predictors of survival after resection of early hepatocellular carcinoma. Annals of surgery 2009, 249(5):799-805.
  48. Ma WJ, Wang HY, Teng LS: Correlation analysis of preoperative serum alpha-fetoprotein (AFP) level and prognosis of hepatocellular carcinoma (HCC) after hepatectomy. World journal of surgical oncology 2013, 11:212.
  49. Allard MA, Sa Cunha A, Ruiz A, Vibert E, Sebagh M, Castaing D, Adam R: The postresection alpha-fetoprotein in cirrhotic patients with hepatocellular carcinoma. An independent predictor of outcome. Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract 2014, 18(4):701-708.
  50. Zhang XF, Yin ZF, Wang K, Zhang ZQ, Qian HH, Shi LH: Changes of serum alpha-fetoprotein and alpha-fetoprotein-L3 after hepatectomy for hepatocellular carcinoma: prognostic significance. Hepatobiliary & pancreatic diseases international : HBPD INT 2012, 11(6):618-623.
  51. Lei Z, Li J, Wu D, Xia Y, Wang Q, Si A, Wang K, Wan X, Lau WY, Wu M et al: Nomogram for Preoperative Estimation of Microvascular Invasion Risk in Hepatitis B Virus-Related Hepatocellular Carcinoma Within the Milan Criteria. JAMA surgery 2016, 151(4):356-363.
  52. Xu J, Liu C, Zhou L, Tian F, Tai MH, Wei JC, Qu K, Meng FD, Zhang LQ, Wang ZX et al: Distinctions between clinicopathological factors and prognosis of alpha-fetoprotein negative and positive hepatocelluar carcinoma patients. Asian Pacific journal of cancer prevention : APJCP 2012, 13(2):559-562.
  53. Tada T, Kumada T, Toyoda H, Kiriyama S, Sone Y, Tanikawa M, Hisanaga Y, Kitabatake S, Kuzuya T, Nonogaki K et al: Relationship between Lens culinaris agglutinin-reactive alpha-fetoprotein and pathologic features of hepatocellular carcinoma. Liver international : official journal of the International Association for the Study of the Liver 2005, 25(4):848-853.
  54. Maeda T, Takenaka K, Taguchi K, Kajiyama K, Shirabe K, Shimada M, Honda H, Sugimachi K: Small hepatocellular carcinoma with minute satellite nodules. Hepato-gastroenterology 2000, 47(34):1063-1066.
  55. Rodriguez-Peralvarez M, Luong TV, Andreana L, Meyer T, Dhillon AP, Burroughs AK: A systematic review of microvascular invasion in hepatocellular carcinoma: diagnostic and prognostic variability. Annals of surgical oncology 2013, 20(1):325-339.
  56. Fan ST, Poon RT, Yeung C, Lam CM, Lo CM, Yuen WK, Ng KK, Liu CL, Chan SC: Outcome after partial hepatectomy for hepatocellular cancer within the Milan criteria. The British journal of surgery 2011, 98(9):1292-1300.
  57. Kaibori M, Ishizaki M, Matsui K, Kwon AH: Predictors of microvascular invasion before hepatectomy for hepatocellular carcinoma. Journal of surgical oncology 2010, 102(5):462-468.
  58. Wang CC, Iyer SG, Low JK, Lin CY, Wang SH, Lu SN, Chen CL: Perioperative factors affecting long-term outcomes of 473 consecutive patients undergoing hepatectomy for hepatocellular carcinoma. Annals of surgical oncology 2009, 16(7):1832-1842.
  59. Han DH, Choi GH, Kim KS, Choi JS, Park YN, Kim SU, Park JY, Ahn SH, Han KH: Prognostic significance of the worst grade in hepatocellular carcinoma with heterogeneous histologic grades of differentiation. Journal of gastroenterology and hepatology 2013, 28(8):1384-1390.
  60. Sasaki K, Matsuda M, Ohkura Y, Kawamura Y, Inoue M, Hashimoto M, Ikeda K, Kumada H, Watanabe G: In hepatocellular carcinomas, any proportion of poorly differentiated components is associated with poor prognosis after hepatectomy. World journal of surgery 2014, 38(5):1147-1153.