Open Access

Novel predictive nomograms based on aspartate aminotransferase‑to‑platelet ratio index for hepatocellular carcinoma with post‑operative adjuvant transarterial chemoembolization

  • Authors:
    • Qinghua Shu
    • Nannan Zhang
    • Jianbo Han
    • Xiaopeng Yan
    • Bowen Sha
    • Liang Zhao
    • Yongxiang Yi
    • Yufeng Zhang
  • View Affiliations

  • Published online on: November 7, 2023     https://doi.org/10.3892/ol.2023.14137
  • Article Number: 3
  • Copyright: © Shu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The clinical significance of systemic inflammation assessed with laboratory analysis of blood samples has been validated in a variety of cancers. The present study was conducted to investigate prognostic significance of preoperative aspartate aminotransferase‑to‑platelet ratio index (APRI) for the outcome of hepatocellular carcinoma (HCC) patients receiving post‑operative adjuvant transarterial chemoembolization (PATACE). A total of 201 patients who underwent PATACE were retrospectively analyzed. A nomogram for HCC was developed using predictors based on multivariate Cox models, and bootstrapping was performed for validation. According to the ROC curve, which was used to divide patients into two cohorts: High APRI group (APRI>1.02) and Low APRI group (APRI≤1.02). In subgroup survival analysis, patients with a relatively low APRI had significantly longer disease‑free survival (DFS) and overall survival (OS) than patients with a relatively high APRI, regardless of Barcelona Clinic Liver Cancer (BCLC) stages (BCLC 0/A or BCLC B/C, both P<0.05); while in China liver cancer staging I/II and TNM I/II stage patients, relatively low APRI was associated with improved DFS and OS (both P<0.05). Multivariate Cox models demonstrated that APRI and BCLC stages were independent prognostic factors of DFS and OS (both P<0.05). Nomograms for DFS and OS were constructed, respectively. Calibration curve analysis showed that the standard curve fitted well with the predicted curve. Time‑receiver operating characteristic curve analysis revealed that the nomogram had high efficiency. Decision curve analysis demonstrated the high clinical value of the nomogram. APRI is an independent prognostic factor of DFS and OS in HCC patients receiving PATACE, and the combination of APRI with the HCC staging system can refine risk stratification to provide a more accurate prognostic assessment for the outcome of patients receiving PATACE.

Introduction

Primary liver cancer is the fourth most common malignancy and the second cause of cancer-related death in China, posing a severe threat to the life and health of Chinese patients. Besides, the morbidity and mortality of primary liver cancer are increasing on a global scale (1,2). In pathology, liver cancers mainly include hepatocellular carcinoma (HCC, 75–85%), intrahepatic cholangiocarcinoma (10–15%), and combined hepatocellular-cholangiocarcinoma (3). The present study focused on HCC for analysis. For the past few years, treatment for HCC has been significantly advanced, and surgery remains the first choice (4). However, the incidence of complications after hepatectomy remains high, especially in patients complicated by primary liver fibrosis and cirrhosis (5). According to the Barcelona Clinic Liver Cancer (BCLC) Staging System, transarterial chemoembolization (TACE) is recommended as the standard therapy for patients with advanced or unresectable tumor (hepatic compensation, Child B) (6,7). Some clinical trials have proved the effectiveness of the TACE as an adjuvant therapy for HCC (6,8,9). For example, a randomized controlled trial revealed that post-operative adjuvant TACE (PATACE) was highly effective in HCC patients but at a high risk of recurrence, such as those with multiple tumor nodules, macroscopic vascular invasion, or large tumor diameter (>5 cm) (8). Multiple studies recommended PATACE for HCC patients with multiple tumor nodules, a tumor of a large size, or vascular invasion (1012). The efficacy of PATACE can be attributed to its effect to improve the prognosis of HCC patients, as traditional imaging fails to discover the microscopic tumor foci and concealed multiple foci in the liver before surgery (10). Therefore, PATACE has been extensively applied in post-operative HCC patients who have multiple risk factors of recurrence, such as large tumor diameter, multiple tumor nodules, microvascular invasion (MVI) and satellite lesions (6,10).

Nevertheless, the long-term survival of this population remains unclear (4,13). Even though TACE and liver resection are considered the most effective therapies, this population may likely not receive benefits from surgery and therefore have poor survival outcomes (14). Assessment for the hepatic functional reserve is critical for HCC treatment, as liver cirrhosis potentially is a major cause of post-operative death (15) and it is also a leading cause of 70–90% HCC cases. Liver reserve is markedly more important in early HCC than that in advanced disease (16,17). Different from other malignancies, the survival outcome of HCC patients is largely dependent on the baseline liver function and the dissemination of the primary tumor. In this context, there is an urgent need to look for non-invasive, accurate biomarkers that are indicative of liver function and inflammation. In the meantime, a prognostic model based on the biomarker, liver function and tumor stage is also in demand. Hematological parameters following systemic inflammation have been assembled as an inflammation-based prognostic scoring system to predict cancer survival. For example, peripheral blood subpopulations, including lymphocytes, monocytes and platelets (PLT), are prognostic for the outcome of varying cancers. A previous study found that anti-PLT therapy could prevent HCC and improve the survival of mice with chronic hepatitis B (CHB) (18). Moreover, numerous studies have reported the relationship between PLT and HCC diagnosis, post-operative complications, and survival (1924). A previous study found that preoperative PLT decline predicted an increased incidence of complications, liver insufficiency and death in HCC patients after tumor resection (22). The preoperative aspartate aminotransferase (AST)-to-PLT ratio index (APRI) has been identified as an independent prognostic index for liver failure in HCC patients after hepatectomy (20). However, it has been controversial how the preoperative PLT increase affects the survival outcome of HCC patients. Amano et al (21) reported that a PLT count lower than 105/mm3 was unfavorable for the overall survival (OS) and disease-free survival (DFS) of HCC patients. They also found that patients beyond the Milan criteria could benefit from hepatectomy, and patients with a sufficiently high PLT count enjoyed an improved survival outcome. By contrast, Hwang et al (23) pointed out that HCC patients with PLT increase reversely had significantly shorter survival times. Combining the studies, it is of paramount importance to identify the prognostic significance of PLT-related indicators, including PLT count, PLT-to-lymphocyte ratio (PLR), and APRI. AST, a spectrum ranging from hepatitis to fatty liver, is commonly used to assess liver injury. Previous studies have proved that inflammation-based prognostic factors, including PLR, Glasgow outcome scale score, neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), and AST/alanine aminotransferase ratio (AST/ALT), are prognostic for the outcome of HCC. APRI is reported as a convenient indicator that can predict the post-operative outcome of HBV-related HCC patients (16,2527). It is applicable in clinical practice as a tool for fast and reliable assessment of the severity of liver function and cirrhosis, and it even can be used as an alternative to liver biopsy. However, it remains unclear whether APRI remains applicable for identifying the patients responsive to PATACE.

Clinical staging is critical for patient prognosis and treatment selection. Motivated by this fact, multiple staging systems have been developed all over the world for the past few years, including TNM staging (28), Okuda staging (29), BCLC (30), Japan-integrated staging (JIS) (31), Cancer of the Liver Italian Program (CLIP) (32), China liver cancer staging (CNLC) and Hong Kong liver cancer (HKLC). Okuda staging is the first to consider liver function as a tumor-influencing factor, but it does not include certain important prognostic factors such as vascular invasion, tumor number and metastasis. JIS is an assembly of the TNM staging and Child-Pugh scoring systems released by the Liver Cancer Study Group of Japan (LCSGJ), while the patient's personal situation is not considered. CLIP fails to identify patients with early-stage disease and assess cancer-related symptoms that are essential for patient prognosis. HKLC was developed in 2014 based on a large group of patients with HBV-related HCC patients. It is more sensitive than the BCLC staging system to patients requiring aggressive treatment. Despite several studies (33,34) reporting the superior capability of HKLC to BCLC in predicting survival outcomes, validation in different cohorts is still required. Presently, the BCLC and AJCC-TNM remain the most effective and reliable staging systems (28). Since the present study mainly focused on HCC patients in China, the CNLC staging system was also applied here.

Nomogram is devised as a prognostic tool combining known prognostic markers to quantify the prognostic risk as precisely as possible. The present study attempted to discuss the prognostic significance of the APRI for OS and DFS of HCC patients receiving radical hepatectomy and PATACE. In addition, a potent nomogram of high clinical prognostic value was established based on the APRI and HCC staging systems. The present study was in accordance with the TRIPOD reporting checklist.

Materials and methods

Patients

The current study retrospectively analyzed the data from 201 HCC patients, who received radical hepatectomy in the Second Hospital of Nanjing from January 2012 to December 2018 (Fig. 1). Data on follow-up were collected from outpatient medical records, telephone interviews, or WeChat conversations. Inclusion criteria were as follows: i) pathological diagnosis of HCC; ii) presence of any of the following risk factors, such as multiple tumor nodules, macroscopic vascular invasion, tumor diameter >5 cm, and microsatellite lesions; iii) no preoperative interventional, targeted, or immune therapy; iv) without or complicated by other malignancies; v) complete tumor resection; vi) first PATACE (lobaplatin and iodized oil) within 4–6 weeks after hepatectomy; and vii) long-term treatment with oral antivirals.

All participants were reviewed 4 weeks after hepatectomy, and PATACE was recommended in cases without recurrent lesions in the liver. Recurrence was considered in the presence of a new tumor focus within 4 weeks after radical hepatectomy, and the cases were excluded from the present study. Patients who underwent preoperative anti-cancer treatment, had not received a PATACE, had a history of other malignancy or had incomplete follow-up data were excluded from the present study. The present study was approved (approval no. B2021-322) by the Second Hospital of Nanjing ethics committee (Nanjing, China) and was conducted in line with the standards of the Declaration of Helsinki (as revised in 2013). As part of the consent process, written informed consent was provided by all participants.

Data collection and definitions

Clinical data were collected within one week preceding surgery, including age, sex, pathology report, serum albumin, total bilirubin, ALT, AST, alpha-fetoprotein (AFP), PLT, tumor parameters and globulin. Staging systems adopted the AJCC-TNM (8th edition), BCLC and CNLC staging systems. APRI was calculated as follows: APRI={[AST (IU/l)/upper limit of normal]/PLT(×109/l)} ×100 (35). Any discrepancy was resolved by discussion.

Follow-up

Follow-up data as of December 2021 were acquired. OS was defined by a period from the beginning of PATACE to patient death. Patients surviving the last follow-up were censored. DFS was defined by a period from the beginning of PATACE to recurrence or the last follow-up. Follow-up examinations included HCC markers (AFP, AFP-L3, DCP and GP73), abdominal ultrasound, upper abdominal contrast-enhanced CT or Gd-EOB-DTPA MRI, and liver function test. All participants were followed up every 3–4 months within 2 years after surgery and every 6 months after 2 years. Recurrence was diagnosed in the context of post-operative serum AFP >20 ng/ml and the presence of a new focus on abdominal ultrasound, upper abdominal contrast-enhanced CT, or Gd-EOB-DTPA MRI. Patient death, time to recurrence, imaging results and HCC markers during the follow-up period were recorded in detail. Follow-up and outcome assessments were fulfilled by two researchers to minimize bias.

Treatment process

Radical surgery a right subcostal ‘inverted-L’ incision was created, and it was extended to the left subcostal region if necessary. Careful examination for peritoneal cavity was needed to confirm the tumor extent, extrahepatic metastasis or possible intraperitoneal dissemination. Anatomical hepatectomy was preferred for the tumor occupying half of the liver, or in the liver lobe or segment. For the tumor with a large size, a deep position, or next to blood vessel, preoperative 3D CT image was obtained to confirm the tumor border and the extent of resection. Liver parenchyma devascularization was performed with a cavitron ultrasonic surgical aspirator (CUSA) and bipolar electrocoagulation, during which corresponding blood vessels were ligated or sutured by silk or 3–0 Proline. The Pringle maneuver was used to control hepatic portal blood flow if necessary, with the time strictly controlled to be less than 15 min.

PATACE

After 4–6 weeks of hepatectomy, TACE was conducted for the remaining liver when the liver function recovered. The Seldinger technique was applied to gain vascular access in the femoral artery (or the radial artery), and the catheter was extended to the celiac trunk or common hepatic artery to complete DSA. Subtraction images were acquired in the arterial, parenchymal and venous phases. The catheter head was selectively inserted into the right or left hepatic artery, and the emulsion mixed with 2–10 ml Lipiodol Ultra Fluide (Guerbet) and 10–20 mg Lobaplatin for Injection (Hainan Changan International Pharmaceutical Co., Ltd.) was injected through a microcatheter (Progreat; Terumo Corp.). A combination of liver function and area of the body was used to determine the doses of chemotherapeutics and iodized oil to be given.

Statistical analysis

Statistical analysis was fulfilled with the SPSS 26.0 (IBM Corp.) and GraphPad Prism 8 (Dotmatics). The optimal cut-off value for APRI was determined by drawing a receiver operating characteristic (ROC) curve. Chi-square test, Fisher's exact test, and Mann-Whitney U test was applied to analyze the relationship between clinical pathological features and APRI. One-way analysis of variance was used for pairwise comparisons. Kaplan-Meier method together with the log-rank test was adopted to explore the effect of APRI on DFS and OS. Backward stepwise Cox regression modeling was applied to perform multivariate analysis for the variables with P<0.1 in univariate analysis. P<0.05 demonstrated a difference with statistical significance. Concordance index values (C-index) and calibration plots were employed to assess the performance of nomogram using the R package ‘rms’ (v4.2.1, http://www.r-project.org/). Time-dependent ROC curves and estimates of the area under the curve (AUC) were used to compare the predictive value of the nomogram with OS and DFS. Using the decision curve analysis (DCA), net benefits and threshold probabilities were quantified for the nomogram, and the clinical benefit was quantified.

Results

Clinicopathological characteristics of patients and optimal cut-off value of APRI

There were more male than female patients (83.6 vs. 16.4%). Among all patients, ages ranged from 11–81 years (median, 53.6±10.9 years). A total of 96% of the cases were caused by hepatitis B. Patients with HCV-RNA or HBV-DNA levels exceeding 500 IU/ml were 1.5 or 23.9% of the total, respectively, while 59.7% of the patients developed liver cirrhosis. The number of APRI exhibits non-significant difference among samples of different BCLC stage, TNM and CNLC stage (Fig. 2A-C). The time-dependent ROC curve was generated to identify the optimal cut-off value of APRI for OS prediction (Fig. 2D). The cut-off value of APRI was determined as 1.02, associated with an AUC of 0.771, sensitivity of 58.0%, specificity of 88.5, and 95% confidence interval (95% CI) of 0.703–0.838 (Table I).

Table I.

Analysis of ROC curves of APRI.

Table I.

Analysis of ROC curves of APRI.

VariableAUCOptimal cutoff valueYouden indexSensitivity (%)Specificity (%)P-value95% CI
APRI0.7711.020.46558.088.50.0000.703–0.838

[i] AUC, area under the curve; ROC, receiver operating characteristic; CI, confidence interval; APRI, aspartate aminotransferase-to-platelet ratio index.

Correlation between APRI and clinicopathological features

According to the cut-off value, patients were divided into the High-APRI group (APRI >1.02) and Low-APRI group (APRI ≤1.02). Correlational analysis demonstrated that APRI was significantly associated with AST, total bilirubin, prothrombin time (PT), ALT, albumin, HBV-DNA, PLT, WBC, CNLC stage, BCLC stage and AJCC-TNM stage (all P<0.05). The relationship between APRI and clinicopathological features of HCC patients was detailed in Table II.

Table II.

Relations of APRI with the clinicopathological characteristics of HCC patients.

Table II.

Relations of APRI with the clinicopathological characteristics of HCC patients.

APRI

VariablesNumber of patients, n (%)≤1.02 n (%)>1.02 n (%)χ2P-value
Age, years 0.748
  ≤4023 (11.4)15 (0.9)8 (12.5)0.104
  >40178 (88.6)122 (89.1)56 (87.5)
Sex 1.0500.305
  Male168 (83.6)112 (81.8)56 (87.5)
  Female33 (16.4)25 (18.2)8 (12.5)
Viral hepatitis -0.113
  HBV193 (96.0)134 (97.8)59 (92.2)
  HCV8 (4.0)3 (2.2)5 (7.8)
Liver cirrhosis 0.7420.389
  Yes120 (59.7)79 (57.7)41 (64.1)
  No81 (40.3)58 (42.3)23 (35.9)
HBV-DNA, IU/ml 3.9860.046
  ≤500144 (71.6)106 (77.4)38 (59.4)
  >50048 (23.9)28 (20.4)20 (31.3)
HCV-RNA, IU/ml -0.226
  ≤5006 (3.0)1 (0.7)5 (7.8)
  >5003 (1.5)2 (1.5)1 (1.5)
AFP, ng/ml 2.7140.099
  ≥20115 (57.2)73 (53.3)42 (65.6)
  <2086 (42.8)64 (46.7)22 (34.4)
AFP-L3, % 0.3650.546
  ≥1088 (43.8)58 (42.3)30 (46.9)
  <10113 (56.2)79 (57.7)34 (53.1)
Tumor size, cm 1.3880.239
  ≤5140 (69.7)99 (72.3)41 (64.1)
  >561 (30.3)38 (27.7)23 (35.9)
AST level, U/l 60.208<0.0001
  <40149 (74.1)124 (90.5)25 (39.1)
  ≥4052 (25.9)13 (9.5)39 (60.9)
TB, µmol/l 7.2990.007
  <34.2187 (93.0)132 (96.4)55 (85.9)
  ≥34.214 (7.0)5 (3.6)9 (14.1)
Prothrombin time, sec 34.280<0.0001
  <14157 (78.1)123 (89.8)34 (53.1)
  ≥1444 (21.9)14 (10.2)30 (46.9)
ALT level, U/l 21.083<0.0001
  <40133 (66.2)105 (76.6)28 (43.8)
  ≥4068 (33.8)32 (23.4)36 (56.2)
Albumin, g/dl 23.660<0.0001
  <3541 (20.4)15 (10.9)26 (40.6)
  ≥35160 (79.6)122 (89.1)38 (59.4)
PLT, ×109/l 60.5520.0001
  <10081 (40.3)30 (21.9)51 (79.7)
  ≥100120 (59.7)107 (78.1)13 (20.3)
WBC, ×109/l 17.702<0.0001
  <5104 (51.7)57 (41.6)47 (73.4)
  ≥597 (48.3)80 (58.4)17 (26.6)
Tumour capsule 5.6420.018
  Complete161 (80.1)116 (84.7)45 (70.3)
  Incomplete40 (19.9)21 (15.3)19 (29.7)
Vascular invasiona 3.1010.078
  Present53 (26.4)31 (22.6)22 (34.4)
  Absent148 (73.6)106 (77.4)42 (65.6)
Nerve invasion 2.6410.104
  Present16 (8.0)8 (5.8)8 (12.5)
  Absent185 (92.0)129 (94.2)56 (87.5)
Edmondson-Steiner grade 1.4750.224
  I–II113 (56.2)81 (59.1)32 (50.0)
  III–IV88 (43.7)56 (40.9)32 (50.0)
Tumor number 2.6410.104
  ≤3185 (92.0)129 (94.2)56 (87.5)
  >316 (8.0)8 (5.8)8 (12.5)
BCLC stage 5.4660.019
  0-A166 (82.6)119 (86.9)47 (73.4)
  B-C35 (17.4)18 (13.1)17 (26.6)
CNLC stage 11.2520.001
  I–II181 (90.0)130 (94.9)51 (79.7)
  III–IV20 (10.0)7 (5.1)13 (20.3)
TNM stage 6.9170.009
  I–II180 (89.6)128 (93.4)52 (81.3)
  III–IV21 (10.4)9 (6.6)12 (18.7)

a Microscopic and macroscopic tumour thrombus. BCLC, Barcelona Clinic Liver Cancer; CNLC, China liver cancer staging; TNM, Tumor-Node-Metastasis system of the American Joint Committee on cancer; AFP, alpha-fetoprotein; ALT, alanine aminotransferase; AST, aspartate aminotransferase; HBV-DNA, hepatitis B virus deoxyribonucleic acid; HCV-RNA, hepatitis C virus ribonucleic acid; HCC, hepatocellular carcinoma; PLT, platelets; TB, total bilirubin; APRI, aspartate aminotransferase-to-platelet ratio index; WBC, white blood cell.

Survival analysis of APRI

The relationship between the APRI and survival outcomes (OS and DFS) of HCC patients was then analyzed in the High-APRI and Low-APRI groups using the Kaplan-Meier method followed by log-rank test. The results revealed that the DFS (P<0.0001) and OS (P<0.0001) of patients in the low-APRI group were significantly improved than those of patients in the High-APRI group (Fig. 3A and B). Subsequently, stratification analysis was performed based on CNLC, BCLC and AJCC-TNM staging systems. In the present study, early-stage tumor was classified as CNLC I/II, BCLC 0/A, and TNM I/II, while late-stage tumor was classified as CNLC III/IV, BCLC B/C, and TNM III/IV. It was revealed that either in BCLC 0/A or BCLC B/C stage, patients with a relatively low APRI had significantly longer DFS and OS than patients with a relatively high APRI (both P<0.05) (Fig. 3C-F). While in CNLC I/II (Fig. 3G and H) and TNM I/II (Fig. 3K and L) stage patients, relatively low APRI was associated with improved DFS and OS (both P<0.05). There were no distinct differences in DFS and OS between the High-APRI and Low-APRI groups in CNLC III/IV (DFS, P=0.502; OS, P=0.775; Fig. 3I and J) and TNM III/IV (DFS, P=0.184; OS, P=0.212; Fig. 3M and N) patients.

Figure 3.

Kaplan-Meier survival curves of HCC patients in high APRI group and low APRI group after BCLC, CNLC and TNM stage stratification. Patients were divided into two groups, APRI ≤1.02 and >1.02, by optimal cutoff value of APRI. (A) DFS of patients with APRI ≤1.02 was higher than those with APRI >1.02 (P<0.0001, log-rank test). (B) OS of patients with APRI ≤1.02 was also higher than those with APRI >1.02 (P<0.0001, log-rank test). (C) DFS curves of patients classified as BCLC 0-A stage (P<0.0001). (D) OS curves of patients classified as BCLC 0-A stage (P<0.0001). (E). DFS curves of patients classified as BCLC B-C stage (P=0.042). (F) OS curves of patients classified as BCLC B-C stage (P=0.012). (G) DFS curves of patients classified as CNLC I–II stage (P<0.0001). (H) OS curves of patients classified as CNLC I–II stage (P<0.0001). (I) DFS curves of patients classified as CNLC III–IV stage (P=0.502). (J) OS curves of patients classified as CNLC III–IV stage (P=0.775). (K) DFS curves of patients classified as TNM I–II stage (P<0.0001). (L) OS curves of patients classified as TNM I–II stage (P<0.0001). (M) DFS curves of patients classified as TNM III–IV stage (P=0.184). (N) OS curves of patients classified as TNM III–IV stage (P=0.212). HCC, hepatocellular carcinoma; APRI, preoperative aspartate aminotransferase-to-platelet ratio index; BCLC, Barcelona Clinic Liver Cancer; CNLC, China liver cancer staging; TNM, Tumor Node Metastasis classification; DFS, disease-free survival; OS, overall survival.

Risk factors influencing the survival of HCC patients

The uni- and multi-variate Cox regression analyses on the prognostic significance of APRI for DFS in patients with HCC patients are shown in Table III. Univariate analysis demonstrated that AFP (P=0.017), PLT (P=0.022), ALT (P=0.004), AST (P<0.0001), total bilirubin (P=0.001), PT (P=0.010), tumor size (P=0.025), tumor number (P=0.002), tumor capsule (P<0.0001), vascular invasion (P<0.0001), neural invasion (P=0.005), Edmondson-Steiner grade (P=0.002), TNM stage (P<0.0001), CNLC stage (P<0.0001), BCLC stage (P<0.0001) and APRI (P<0.0001) were prognostic for the DFS of patients. The variables with P<0.1 were further included in the backward stepwise Cox regression model. The result showed that tumor capsule (P=0.040), Edmondson-Steiner grade (P=0.038), BCLC stage (P<0.0001) and APRI (P<0.0001) were independent prognostic factors of DFS in HCC patients.

Table III.

Univariate and multivariate Cox proportional hazards regression analysis of factors for DFS of HCC patients.

Table III.

Univariate and multivariate Cox proportional hazards regression analysis of factors for DFS of HCC patients.

UnivariateMultivariate


VariableHR95% CIP-valueHR95% CIP-value
Sex (male vs. female)1.2560.761–2.0720.373
Age (≤40 vs. >40, years)0.6850.410–1.1450.148
Types of viral hepatitis (HBV vs. HCV)1.3090.610–2.8110.489
Livers cirrhosis (Yes vs. No)1.0920.757–1.5750.639
AFP (<20 vs. ≥20, ng/ml)1.5741.086–2.2810.0171.2030.799–1.8120.376
AFP-L3 (<10 vs. ≥10, %)1.3360.935–1.9110.112
HBV-DNA (≤500 vs. >500, IU/ml)0.8950.576–1.3920.623
HCV-DNA (≤500 vs. >500, IU/ml)0.7070.135–3.6970.681
WBC (<5 vs. ≥5, ×109/l)1.0970.768–1.5670.610
PLT (<100 vs. ≥100, ×109/l)0.6600.461–0.9430.0220.9840.607–1.5940.947
Albumin (<35 vs. ≥35, g/dl)0.8240.540–1.2580.370
ALT level (<40 vs. ≥40, U/l)1.7051.188–2.4470.0041.0560.688–1.6200.804
AST level (<40 vs. ≥40, U/l)2.4771.710–3.5890.0000.9310.523–1.6590.810
TB (<34.2 vs. ≥34.2, µmol/l)2.6041.459–4.6460.0011.7170.931–3.1660.083
Prothrombin time (≤14 vs. >14, sec)1.6751.130–2.4830.0101.1280.716–1.7780.603
Tumour size (≤5 vs. >5, cm)1.5411.056–2.2480.0251.3810.928–2.0560.112
Tumour number (≤3 vs. >3)2.4681.382–4.4080.0020.9600.432–2.1340.920
Tumour capsule (Complete vs. Incomplete)2.6161.759–3.8890.0001.5951.022–2.4900.040
Vascular invasion (Present vs. Absent)2.1701.485–3.1700.0000.8750.512–1.4960.626
Nerve invasion (Present vs. Absent)2.2861.281–4.0770.0051.1650.595–2.2790.656
Edmondson-Steiner grade (I–II vs. III–IV)1.7831.247–2.5490.0021.4781.021–2.1400.038
BCLC stage (0-A vs. B-C)3.2142.122–4.8690.0002.3101.467–3.638<0.001
CNLC stage (I–II vs. III–IV)5.4663.322–8.9930.0001.6310.731–3.6410.232
TNM stage (I–II vs. III–IV)3.4222.082–5.6240.0000.7420.312–1.7620.499
APRI (≤1.02 vs. >1.02)2.6411.842–3.7880.0002.1591.480–3.150<0.001

[i] CI, confidence interval; HR, hazard ratio; BCLC, Barcelona Clinic Liver Cancer; CNLC, China liver cancer staging; TNM, Tumor Node Metastasis classification; AFP, alpha-fetoprotein; ALT, alanine aminotransferase; AST, aspartate aminotransferase; HBV-DNA, hepatitis B virus deoxyribonucleic acid; HCV-RNA, hepatitis C virus ribonucleic acid; HCC, hepatocellular carcinoma; PLT, platelets; TB, total bilirubin; APRI, aspartate aminotransferase-to-platelet ratio index; WBC, white blood cell.

The uni- and multi-variate Cox regression analyses on the prognostic significance of APRI for OS in HCC patients are demonstrated in Table IV. The result of univariate analysis revealed significant prognostic value of AFP (P=0.015), PLT (P<0.0001), ALT (P=0.003), AST (P<0.0001), total bilirubin (P<0.0001), PT (P=0.003), tumor size (P=0.003), tumor number, tumor capsule, vascular invasion, neural invasion, Edmondson-Steiner grade, TNM, CNLC and BCLC stage and APRI (all P<0.0001). In the backward stepwise Cox regression model, tumor size (P=0.003), Edmondson-Steiner grade (P=0.007), BCLC stage (P<0.0001) and APRI (P<0.0001) were independent prognostic factors of OS in HCC patients.

Table IV.

Univariate and multivariate Cox proportional hazards regression analysis of factors for OS of HCC patients.

Table IV.

Univariate and multivariate Cox proportional hazards regression analysis of factors for OS of HCC patients.

UnivariateMultivariate


VariablesHR95% CIP-valueHR95% CIP-value
Sex (male vs. female)0.9800.562–1.7080.943
Age (≤40 vs. >40, years)0.7210.399–1.3020.278
Viral hepatitis (HBV vs. HCV)0.9130.335–2.4910.859
Livers cirrhosis (Yes vs. No)0.9220.604–1.4080.707
AFP (<20 vs. ≥20, ng/ml)1.7341.111–2.7050.0151.1710.70–1.9540.546
AFP-L3 (<10 vs. ≥10, %)1.2460.819–1.8940.304
HBV-DNA (≤500 vs. >500, IU/ml)0.9580.575–1.5970.869
HCV-DNA (≤500 vs. >500, IU/ml)0.8440.152–4.6850.846
WBC (<5 vs. ≥5, ×109/l)0.8100.531–1.2330.325
PLT (<100 vs. ≥100, ×109/l)0.4460.292–0.6810.0000.8550.492–1.4860.579
Albumin (<35 vs. ≥35, g/dl)0.7000.432–1.1360.149
ALT level (<40 vs. ≥40, U/l)1.9001.247–2.8950.0030.9070.532–1.5470.720
AST level (<40 vs. ≥40,U/l)3.4922.292–5.3210.0000.9400.468–1.8870.862
TB (<34.2 vs. ≥34.2, µmol/l)3.1631.675–5.9740.0001.8570.943–3.6580.073
Prothrombin time (≤14 vs. >14, sec)1.9751.260–3.0940.0031.2760.779–2.0910.333
Tumour size (≤5 vs. >5, cm)1.9071.244–2.9260.0031.9821.271–3.0890.003
Tumour number (≤3 vs. >3)3.1491.664–5.9590.0001.3160.571–3.0330.519
Tumour capsule (Complete vs. Incomplete)2.9141.870–4.5410.0001.1630.620–2.1800.638
Vascular invasion (Present vs. Absent)3.0401.983–4.6600.0000.9470.490–1.8300.872
Nerve invasion (Present vs. Absent)3.5811.976–6.4890.0001.4540.711–2.9720.305
Edmondson-Steiner grade (I–II vs. III–IV)2.3551.533–3.6190.0001.8381.182–2.8600.007
BCLC stage (0-A vs. B-C)4.1982.675–6.5850.0003.3702.102–5.403<0.001
CNLC stage (I–II vs. III–IV)8.2724.915–13.9230.0001.9180.816–4.5080.135
TNM stage (I–II vs. III–IV)5.1003.047–8.5360.0000.8090.295–2.2210.681
APRI (≤1.02 vs. >1.02)4.3812.859–6.7140.0003.5902.300–5.604<0.001

[i] CI, confidence interval; HR, hazard ratio; BCLC, Barcelona Clinic Liver Cancer; CNLC, China liver cancer staging; TNM, Tumor Node Metastasis classification; AFP, α-fetoprotein; ALT, alanine aminotransferase; AST, aspartate aminotransferase; HBV-DNA, hepatitis B virus deoxyribonucleic acid; HCV-RNA, hepatitis C virus ribonucleic acid; HCC, hepatocellular carcinoma; PLT, platelets; TB, total bilirubin; APRI, aspartate aminotransferase-to-platelet ratio index; WBC, white blood cell.

Nomogram construction and validation

To improve stratification of patients with different prognoses, a nomogram was constructed based on the multivariate Cox regression analysis of the DFS and OS (Fig. 4). Scores were assigned to each risk factor, and each included patient's grade was determined by the sum of these scores. In this nomogram, a higher score predicts an improved survival outcome. Internal validation was performed with 500 re-samplings. The calibration curves plotted with the 1-, 3- and 5-year DFS (Fig. 5A-C, respectively) were well matched with the idealized 45 line (the x-axis represents the nomogram-predicted probability of DFS, whereas the y-axis shows the actual DFS). The calibration curves plotted with 1-, 3- and 5-year OS were well matched with the idealized 45 line [Fig. 5D-F, (the x-axis represents the nomogram-predicted probability of OS, whereas the y-axis shows the actual OS). The C-index of the nomogram for predicting DFS and OS probability were 0.700 (95% CI: 0.650–0.750) and 0.775 (95% CI: 0.726–0.824) (Tables V and VI). The C-index for 1-, 3- and 5-year DFS was 0.735 (95% CI: 0.636–0.834), 0.735 (95% CI: 0.659–0.810), and 0.686 (95% CI: 0.599–0.774), respectively; while that for 1, 3, and 5-year OS was 0.864 (95% CI: 0.792–0.936), 0.812 (95% CI: 0.731–0.893), and 0.783 (95% CI: 0.704–0.862), respectively (Table VII). The time-dependent AUC (t-AUC) for predicting the 1-, 3- and 5-year and all DFS was 0.734 (95% CI: 0.657–0.810) (Fig. 6A), 0.722 (95% CI: 0.651–0.793) (Fig. 6B) and 0.756 (95% CI: 0.690–0.821) (Fig. 6C), and 0.776 (95% CI: 0.712–0.839; Fig. 6D), respectively (Table VIII). The t-AUC for predicting the 1-, 3-, 5-year and all OS was 0.815 (95% CI: 0.729–0.900) (Fig. 7A), 0.807 (95% CI: 0.739–0.874) (Fig. 7B), 0.842 (95% CI: 0.784–0.899) (Fig. 7C), and 0.843 (95%CI: 0.787–0.900) (Fig. 7D), respectively (Table VIII). The result demonstrated high consistency between the predicted and actual outcome, suggesting favorable performance of the nomogram.

Table V.

C-index of nomogram and other predictors in OS.

Table V.

C-index of nomogram and other predictors in OS.

OS
PredictorsC-index95% CIP-value
Nomogram0.7750.726–0.824<0.001
APRI0.6730.623–0.723<0.001
Tumour size0.6040.550–0.6580.003
Edmondson-Steiner grade0.6070.553–0.661<0.001
BCLC stage0.6240.576–0.672<0.001

[i] C-index, concordance index; OS, overall survival; CI, confidence interval; APRI, aspartate aminotransferase-to-platelet ratio index; BCLC, Barcelona Clinic Liver Cancer.

Table VI.

C-index of nomogram and other predictors in DFS.

Table VI.

C-index of nomogram and other predictors in DFS.

DFS

PredictorsC-index95% CIP-value
Nomogram0.7000.650–0.750<0.001
APRI0.6070.564–0.650<0.001
Tumour capsule0.5920.553–0.631<0.001
Edmondson-Steiner grade0.5930.547–0.639<0.001
BCLC stage0.5980.558–0.637<0.001

[i] C-index, concordance index; DFS, Disease-Free Survival; CI, confidence interval; APRI, aspartate aminotransferase-to-platelet ratio index; BCLC, Barcelona Clinic Liver Cancer.

Table VII.

The C-index of nomogram and other predictors in 1-year, 3-year, 5-year DFS and OS.

Table VII.

The C-index of nomogram and other predictors in 1-year, 3-year, 5-year DFS and OS.

Overall survival Disease-free survival


Predictors1-year C-index (95% CI)P-value3-year C-index (95% CI)P-value5-year C-index (95% CI)P-valuePredictors1-year C-index (95% CI)P-value3-year C-index (95% CI)P-value5-year C-index (95% CI)P-value
Nomogram0.864<0.0010.812<0.0010.783<0.001Nomogram0.735<0.0010.735<0.0010.686<0.001
(0.792- (0.731- (0.704- (0.636- (0.659- (0.599-
0.936) 0.893) 0.862) 0.834) 0.810) 0.774)
APRI0.679<0.0010.671<0.0010.673<0.001APRI0.602<0.0010.638<0.0010.605<0.001
(0.585- (0.610- (0.622- (0.541- (0.561- (0.562-
0.773) 0.732) 0.724) 0.663) 0.715) 0.648)
Tumour size0.709<0.0010.628<0.0010.6060.001Tumour0.597<0.0010.595<0.0010.592<0.001
(0.618–0.799) (0.567–0.689) (0.551–0.661) capsule(0.540–0.649) (0.556–0.636) (0.553–0.631)
Edmondson-0.6550.0040.6000.0040.607<0.001Edmondson-0.625<0.0010.595<0.0010.595<0.001
Steiner grade(0.564–0.745) (0.537–0.663) (0.554–0.660) Steiner grade(0.566–0.683) (0.546–0.644) (0.548–0.642)
BCLC stage0.733<0.0010.636<0.0010.624<0.001BCLC stage0.626<0.0010.600<0.0010.598<0.001
(0.639–0.827) (0.577–0.695) (0.575–0.673) (0.571–0.681) (0.557–0.643) (0.559–0.637)

[i] C-index, concordance index; OS, overall survival; DFS, Disease-Free Survival; CI, confidence interval; APRI, aspartate aminotransferase-to-platelet ratio index; BCLC, Barcelona Clinic Liver Cancer.

Table VIII.

The AUC of nomogram and other predictors in 1-year, 3-year, 5-year DFS and OS.

Table VIII.

The AUC of nomogram and other predictors in 1-year, 3-year, 5-year DFS and OS.

Overall survival Disease-free survival


Predictors1-year AUC (95% CI)P-value3-year AUC (95% CI)P-value5-year AUC (95% CI)P-valuePredictors1-year AUC (95% CI)P-value3-year AUC (95% CI)P-value5-year AUC (95% CI)P-value
Nomogram0.815<0.0010.807<0.0010.842<0.001Nomogram0.734<0.0010.722<0.0010.756<0.001
(0.729–0.900) (0.739–0.874) (0.784–0.899) (0.657–0.810) (0.651–0.793) (0.690–0.821)
APRI0.6450.0090.706<0.0010.724<0.001APRI0.6100.0020.638<0.0010.651<0.001
(0.537–0.753) (0.623–0.790) (0.649–0.798) (0.525–0.696) (0.561–0.715) (0.575–0.726)
Tumour size0.710<0.0010.6330.0030.5940.023Tumour0.6060.0150.6180.0040.620.004
(0.607–0.813) (0.546–0.721) (0.513–0.674) capsule(0.519–0.694) (0.541–0.696) (0.543–0.696)
Edmondson-0.6110.0460.5980.0290.6510.040Edmondson-0.6370.0020.5970.0170.6200.004
Steiner grade(0.505–0.717) (0.512–0.684) (0.573–0.728) Steiner grade(0.555–0.720) (0.519–0.676) (0.542–0.699)
BCLC stage0.6940.0010.6410.0020.635<0.001BCLC stage0.6360.0020.6040.0110.5980.017
(0.582–0.806) (0.551–0.730) (0.554–0.715) (0.549–0.723) (0.561–0.715) (0.521–0.676)

[i] AUC, area under the curve; OS, overall survival; DFS, Disease-Free Survival; CI, confidence interval; APRI, aspartate aminotransferase-to-platelet ratio index; BCLC, Barcelona Clinic Liver Cancer.

To gain insight into the performance of the nomogram, the nomogram with other significant prognostic factors was compared (Tables V and VI). For DFS, the C-index of nomogram (0.700) was markedly higher than that of tumor capsule (0.592), Edmondson-Steiner grade (0.593), APRI (0.607) and BCLC stage (0.598); while for OS, the C-index of nomogram (0.775) was also improved as compared with that of tumor size (0.604), Edmondson-Steiner grade (0.607), APRI (0.673) and BCLC stage (0.624). For DFS, the t-AUC of nomogram (0.776) was markedly higher than that of tumor capsule (0.613), Edmondson-Steiner grade (0.604), APRI (0.681), and BCLC stage (0.603); while for OS, the t-AUC of nomogram (0.843) was also improved as compared with that of tumor size (0.584), Edmondson-Steiner grade (0.656), APRI (0.732) and BCLC stage (0.638) (Table IX).

Table IX.

The AUC of nomogram and other predictors in DFS and OS.

Table IX.

The AUC of nomogram and other predictors in DFS and OS.

Overall survival Disease-free survival


PredictorsAUC95% CIP-valuePredictorsAUC95% CIP-value
Nomogram0.8430.787–0.900<0.001Nomogram0.7760.712–0.839<0.001
APRI0.7320.659–0.805<0.001APRI0.6810.608–0.755<0.001
Tumour size0.5840.504–0.6640.042Tumour capsule0.6130.536–0.6900.007
Edmondson-Steiner grade0.6560.580–0.7330.001Edmondson-Steiner grade0.6040.525–0.6830.013
BCLC stage0.6380.559–0.717<0.001BCLC stage0.6030.526–0.6810.013

[i] AUC, area under the curve; OS, overall survival; DFS, Disease-Free Survival; CI, confidence interval; APRI, aspartate aminotransferase-to-platelet ratio index; BCLC, Barcelona Clinic Liver Cancer.

DCA is commonly used to evaluate the clinical net benefit of a nomogram. In the present study, DCA showed that the nomogram increased net benefits and exhibited a wider range of threshold probabilities in predicting 1-, 3-, and 5-year OS and DFS (Fig. 8A-D). Collectively, the combination of APRI and HCC staging systems can improve stratification of patients with different prognoses.

Discussion

The present study identified preoperative APRI and BCLC staging as independent prognostic factors for OS and DFS of HCC patients receiving PATACE. In addition, APRI was also significantly associated with the DFS and OS of patients stratified by BCLC, CNLC and TNM stages. It was also found that absence of tumor capsule and tumor size >5 cm were independently prognostic for the DFS and OS of patients, consistent with the previous literature (36). Moreover, the present study constructed a nomogram by combining the APRI and HCC staging systems (CNLC, BCLC and TNM) to help quantify the prognostic risk and then provide more prognostic information. It is known that HCC patients are usually accompanied by liver cirrhosis and fibrosis, which are great concerns for patients (17). Besides, hepatectomy can lead to post-operative alterations of neuroendocrine, metabolism and immune systems, resulting in immune dysfunction and increasing the risk of complications after PATACE. Theoretically, factors such as malnutrition, poor immune status and liver cirrhosis, may affect the prognosis of HCC patients after PATACE. Previous studies proved the prognostic value of APRI in liver malignancy (3739), but its significance in prognosis of HCC patients undergoing PATACE is less studied (36). Therefore, more independent data are in demand to validate the prognostic value of APRI in HCC.

Growing evidence has suggested that host inflammatory responses are predictive of the clinical outcome of HCC patients given their significant implications for tumorigenesis and metastasis. In addition, HCC patients receiving PATACE are at a high risk of recurrence and post-operative alterations in the immune system. A recent study revealed that APRI is an effective and non-invasive indicator that can be used to assess the risk of liver fibrosis in patients with CHB and chronic hepatitis C (CHC). APRI was first reported by Wai et al (35) as a biochemical alternative to predict the advanced fibrosis and liver cirrhosis in patients with CHC. In the following years, the effect of APRI was further explored in identifying HIV infection and differentiating liver cirrhosis in hepatitis B cohorts. While in previous years, APRI has been proven as capable of predicting the risk of developing HCC in patients with liver cirrhosis and the death rate associated with tumor resection (16,20,40). It was also revealed that APRI can be used as a valuable prognostic indicator for HCC patients undergoing radiofrequency ablation and hepatectomy (26,41).

The present study applied ROC analysis to determine the optimal cut-off value of APRI as 1.02 for OS of HCC patients, and classified patients into the High- and Low-APRI groups. It was found that the DFS (P<0.0001) and OS (P<0.0001) of patients in the Low-APRI group were significantly improved than those of patients in the High-APRI group. In the multivariate Cox regression analysis, tumor capsule (P=0.040), Edmondson-Steiner grade (P=0.038), BCLC stage (P<0.0001) and APRI (P<0.0001) were identified as significantly prognostic for the DFS of patients; while tumor size (P=0.003), Edmondson-Steiner grade (P=0.007), BCLC stage (P<0.0001) and APRI (P<0.0001) were independent prognostic factors for OS of patients. It was previously reported that in patients with single HCC, APRI<0.47 was associated with a significantly higher OS rate and a remarkably lower recurrence rate, as compared with the control group (26). Hung et al (40) and Shen et al (26) reported a cut-off value of APRI of 0.47 and 0.62, respectively. The discrepancy may be attributed to the different methods used to calculate APRI cut-off value and the different etiologies of HCC. In addition, the differences in tumor status, sample size, and patient inclusion criteria can also make an effect. In a previous study by Hung et al (40) involving 76 HBV-mono-infected HCC patients from 12 trials, all the tumors were <5 cm in diameter with the median value of 2.5 cm, which is significantly smaller than the 4.8 cm (median) in the present study. In another study made by Shen et al (26) included 332 HCC patients with a mean tumor size of 8.76 cm. Among them, 25.6% had large vessels, and 20.8% were complicated by portal vein tumor thrombosis. All these patients with such characteristics were not considered in the present study. In spite of the different cut-off values of APRI, a lower value of APRI generally predicts an improved OS rate.

However, the relationship between APRI increase and poor prognosis remains unclear. It is hypothesized that numerous HCC patients with APRI increase have a low preoperative PLT, which may be a result of spleen enlargement that leads to PLT destruction or progressive liver fibrosis that leads to decreased production of thrombopoietin. In addition, the low preoperative PLT may also correlate to major post-operative complications, liver failure and mortality. It has also been reported that PLT increase is associated with the poor prognosis of nasopharyngeal carcinoma, gastric, colorectal and endometrial cancer (23,4244). Similarly, there is no specific explanation for the relationship between PLT increase and poor prognosis. The current potential explanations are as follows: First, PLT increase can promote tumor growth by advancing angiogenesis. As cancer patients usually present with coagulation abnormalities leading to disturbed PLT function, increase in PLT secrets more angiogenic factors to stimulate tumor angiogenesis. Second, PLT can interact with tumor cells via receptor-ligand pairs, thereby promoting tumor cell growth and invasion (4547). Third, PLT are actively involved in host immune attack to tumor (48,49). Previous studies theorized that PLT decrease the cytolytic activity of NK cells to protect tumor cells from NK attack (48). Inflammation of the liver caused by viral infection and alcohol consumption leads to HCC onset (36), and AST from the mitochondria in hepatocytes is a reliable and sensitive biomarker of liver inflammation. Disorders in the liver result in mitochondrial damage and subsequent release of intrahepatic AST into the blood, suggesting heavily impaired liver function. It is known that hepatocellular injury is tightly associated with the occurrence of HCC (50). Moreover, a large group of HCC patients with APRI elevation may also have an increase in AST, indicating stress response or injury of the liver, such as reactivation of HBV replication or progressive liver fibrosis (51). These events are associated with a poor survival rate in HCC patients. This also supports the finding of the present study that patients with APRI >1.02 had poorer survival outcomes and a higher risk of recurrence. Therefore, APRI elevation indicates severely impaired liver function and poor tumor prognosis.

CNLC, BCLC and AJCC-TNM (8th edition) staging systems were found to be capable of stratifying HCC patients based on their risk categories in the present study. BCLC and AJCC-TNM (8th edition) staging systems are the most widely used tools currently. Multivariate Cox regression analysis demonstrated that the BCLC stage was an independent risk factor of prognosis in HCC patients undergoing PATACE. The nomogram established by significant independent prognostic factors is intuitive and can maximize the predictive accuracy in assessment for prognosis in individuals. The present study constructed a nomogram based on the multivariate Cox regression analysis for DFS and OS of HCC patients. It was identified that tumor size, tumor capsule and BCLC stage were correlated with the prognosis of HCC patients receiving PATACE. The multivariate analysis of the present study also identified that APRI was superior to AST and PLT in predicting survival outcomes in this population. Referring to literature, there was only one study reporting the use of an APRI-based nomogram in predicting the prognosis of HCC patients receiving radical surgery followed by adjuvant TACE. However, the previous study did not further explore the relationship between the APRI and HCC stage and OS of patients (39,16,52). Besides, the case data in that study were limited and not sufficient for external validation, and certain bias existed in partial data. The present study generated a nomogram that combined tumor status, tumor stage and APRI. This nomogram can be used for individualized survival estimation in patients with HCC patients receiving radical surgery followed by adjuvant TACE and can also help surgeons make appropriate post-operative decisions for treatment and individualized monitoring.

Furthermore, there are some considerations when building a nomogram. The number of surviving and succumbed patients should be 10-fold greater than the number of variables used to construct the nomogram in order to reduce the prediction error in the predicted probability <10%. The number of deaths was 88, which is 22-fold larger than the number of variables in the present study. Due to the insufficient number of cases in the external validation group, an internal validation was conducted with 500 sets of bootstrap samples with a calibration curve and well verified the nomogram. Collectively, the present study proved the favorable performance of the nomogram in predicting survival outcome in these patients using the calibration plot and C-index, which was superior to other prognostic factors.

At present, multiple prognostic indicators for post-operative HCC patients have been identified, but there is a paucity of indicators for patients receiving PATACE. This may be attributed to some factors that affect the clinical outcome of patients with TACE, such as liver function, tumor characteristics and treatment modalities. HCC patients who received PATACE were included in the present study as second-line treatment, in an attempt to avoid the confounders caused by other treatments. In addition, 1.02 as the cut-off value was used of APRI for analysis, which is not in line with the previous studies. For example, the cut-off value of APRI reported by Shen et al (26) and Tang et al (38) were 0.40 and 0.62, respectively, which were obtained from the ROC curve for patients receiving TACE or liver operations. Moreover, it was also identified that a larger tumor size and a higher BCLC grade were predictive of poorer survival outcomes of patients with HCC patients, consistent with previous studies (17,53,54). It has been established that tumor and clinical features, such as tumor size, tumor number, BCLC stage and MVI, have implications for survival and recurrence of HCC patients, while non-tumor parameters, such as inflammation, viral infection and liver fibrosis, are important in HCC recurrence.

There are certain limitations to the present study. For example, this was a single-center retrospective study, and the majority of participants had an HBV infection background (96%). Therefore, a large-cohort, multi-center study with stratification analysis based on etiology is in demand to validate the findings of the present study. In addition, bias may not be completely avoided given that all participants were treated in the same medical center. Moreover, further prospective study is required to validate the results of the present study, in other centers. A control group may also be warranted to reinforce these results. Finally, the present study adopted baseline APRI for analysis, but APRI values were dynamic during follow-up. Thus, it may lead to loss of patient data during follow-up. In conclusion to sum up, the present study identified that APRI has certain clinical value as an independent prognostic factor for DFS and OS of HCC patients receiving PATACE. Combining the APRI and HCC stages, surgeons can improve stratifying of patients into different risk categories, thereby efficiently identifying the high-risk group for improved treatment.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Nanjing Medical University Science and Technology Development Fund (grant no. NNUB20210165).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

QHS, NNZ, JBH, YXY and YFZ conceptualized and designed the study. XPY, BWS and LZ acquired, analyzed and interpreted the data. QHS, NNZ and JBH drafted the manuscript. QHS, YXY and YFZ critically revised the manuscript for important intellectual content. QHS and YFZ confirm the authenticity of all the raw data. All authors have made a significant contribution to the present study, and have read and approved the final version of the manuscript.

Ethics approval and consent to participate

The present study was conducted in accordance with the Declaration of Helsinki (as revised in 2013) and was approved (approval no. B2021-322) by the Second Hospital of Nanjing ethics committee (Nanjing, China). All patients enrolled in this study signed an informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Zhou M, Wang H, Zeng X, Yin P, Zhu J, Chen W, Li X, Wang L, Wang L, Liu Y, et al: Mortality, morbidity, and risk factors in China and its provinces, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet. 394:1145–1158. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Llovet JM, Villanueva A, Marrero JA, Schwartz M, Meyer T, Galle PR, Lencioni R, Greten TF, Kudo M, Mandrekar SJ, et al: Trial design and endpoints in hepatocellular carcinoma: AASLD Consensus Conference. Hepatology. 73 (Suppl 1):S158–S191. 2021. View Article : Google Scholar

5 

Cescon M, Vetrone G, Grazi GL, Ramacciato G, Ercolani G, Ravaioli M, Del GM and Pinna AD: Trends in perioperative outcome after hepatic resection: Analysis of 1500 consecutive unselected cases over 20 years. Ann Surg. 249:995–1002. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Bruix J and Sherman M: Management of hepatocellular carcinoma: An update. Hepatology. 53:1020–1022. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Forner A, Gilabert M, Bruix J and Raoul JL: Treatment of intermediate-stage hepatocellular carcinoma. Nat Rev Clin Oncol. 11:525–535. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Tournoux C, Paoletti X and Barbare JC: Treatment outcomes for hepatocellular carcinoma using chemoembolization in combination with other therapies. Cancer Treat Rev. 33:762–763. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Lau WY, Lai EC, Leung TW and 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. Ann Surg. 247:43–48. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Jiang JH, Guo Z, Lu HF, Wang XB, Yang HJ, Yang FQ, Bao SY, Zhong JH, Li LQ, Yang RR and Xiang BD: Adjuvant transarterial chemoembolization after curative resection of hepatocellular carcinoma: Propensity score analysis. World J Gastroenterol. 21:4627–4634. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Paik KY and Kim EK: Pathologic response to preoperative transarterial chemoembolization for resectable hepatocellular carcinoma may not predict recurrence after liver resection. Hepatobiliary Pancreat Dis Int. 15:158–164. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Wang Z, Ren Z, Chen Y, Hu J, Yang G, Yu L, Yang X, Huang A, Zhang X, Zhou S, et al: Adjuvant transarterial chemoembolization for HBV-related hepatocellular carcinoma after resection: A randomized controlled study. Clin Cancer Res. 24:2074–2081. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, Pracht M, Yokosuka O, Rosmorduc O, Breder V, et al: Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 389:56–66. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Tsurusaki M and Murakami T: Surgical and Locoregional therapy of HCC: TACE. Liver Cancer. 4:165–175. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Wu CY, Lin JT, Ho HJ, Su CW, Lee TY, Wang SY, Wu C and Wu JC: Association of nucleos(t)ide analogue therapy with reduced risk of hepatocellular carcinoma in patients with chronic hepatitis B: A nationwide cohort study. Gastroenterology. 147:143–151. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Hann HW, Wan S, Lai Y, Hann RS, Myers RE, Patel F, Zhang K, Ye Z, Wang C and Yang H: Aspartate aminotransferase to platelet ratio index as a prospective predictor of hepatocellular carcinoma risk in patients with chronic hepatitis B virus infection. J Gastroenterol Hepatol. 30:131–138. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Su CW, Chau GY, Hung HH, Yeh YC, Lei HJ, Hsia CY, Lai CR, Lin HC and Wu JC: Impact of Steatosis on prognosis of patients with early-stage hepatocellular carcinoma after hepatic resection. Ann Surg Oncol. 22:2253–2261. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Sitia G, Aiolfi R, Di Lucia P, Mainetti M, Fiocchi A, Mingozzi F, Esposito A, Ruggeri ZM, Chisari FV, Iannacone M and Guidotti LG: Antiplatelet therapy prevents hepatocellular carcinoma and improves survival in a mouse model of chronic hepatitis B. Proc Natl Acad Sci USA. 109:E2165–E2172. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Hwang SJ, Luo JC, Li CP, Chu CW, Wu JC, Lai CR, Chiang JH, Chau GY, Lui WY, Lee CC, et al: Thrombocytosis: A paraneoplastic syndrome in patients with hepatocellular carcinoma. World J Gastroenterol. 10:2472–2477. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Ichikawa T, Uenishi T, Takemura S, Oba K, Ogawa M, Kodai S, Shinkawa H, Tanaka H, Yamamoto T, Tanaka S, et al: A simple, noninvasively determined index predicting hepatic failure following liver resection for hepatocellular carcinoma. J Hepatobiliary Pancreat Surg. 16:42–48. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Amano H, Tashiro H, Oshita A, Kobayashi T, Tanimoto Y, Kuroda S, Tazawa H, Itamoto T, Asahara T and Ohdan H: Significance of platelet count in the outcomes of hepatectomized patients with hepatocellular carcinoma exceeding the Milan criteria. J Gastrointest Surg. 15:1173–1181. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Maithel SK, Kneuertz PJ, Kooby DA, Scoggins CR, Weber SM, Martin RC II, Mcmasters KM, Cho CS, Winslow ER, Wood WC and Staley CA III: Importance of low preoperative platelet count in selecting patients for resection of hepatocellular carcinoma: A multi-institutional analysis. J Am Coll Surg. 212:638–650. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Hwang SG, Kim KM, Cheong JH, Kim HI, An JY, Hyung WJ and Noh SH: Impact of pretreatment thrombocytosis on blood-borne metastasis and prognosis of gastric cancer. Eur J Surg Oncol. 38:562–567. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Kurt M, Onal IK, Sayilir AY, Beyazit Y, Oztas E, Kekilli M, Turhan N, Karaman K and Akdogan M: The role of mean platelet volume in the diagnosis of hepatocellular carcinoma in patients with chronic liver disease. Hepatogastroenterology. 59:1580–1582. 2012.PubMed/NCBI

25 

Murray KF and Carithers RJ: AASLD practice guidelines: Evaluation of the patient for liver transplantation. Hepatology. 41:1407–1432. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Shen SL, Fu SJ, Chen B, Kuang M, Li SQ, Hua YP, Liang LJ, Guo P, Hao Y and Peng BG: Preoperative aspartate aminotransferase to platelet ratio is an independent prognostic factor for hepatitis B-induced hepatocellular carcinoma after hepatic resection. Ann Surg Oncol. 21:3802–3809. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zhang C, Wu J, Xu J, Xu J, Xian J, Xue S and Ye J: Association between aspartate Aminotransferase-to-Platelet ratio index and hepatocellular carcinoma risk in patients with chronic hepatitis: A meta-analysis of cohort study. Dis Markers. 2019:20468252019. View Article : Google Scholar : PubMed/NCBI

28 

Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR and Winchester DP: The Eighth edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J Clin. 67:93–99. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Okuda K, Ohtsuki T, Obata H, Tomimatsu M, Okazaki N, Hasegawa H, Nakajima Y and Ohnishi K: Natural history of hepatocellular carcinoma and prognosis in relation to treatment. Study of 850 patients. Cancer. 56:918–928. 1985. View Article : Google Scholar : PubMed/NCBI

30 

Llovet JM, Bru C and Bruix J: Prognosis of hepatocellular carcinoma: The BCLC staging classification. Semin Liver Dis. 19:329–338. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Kudo M, Chung H, Haji S, Osaki Y, Oka H, Seki T, Kasugai H, Sasaki Y and Matsunaga T: Validation of a new prognostic staging system for hepatocellular carcinoma: The JIS score compared with the CLIP score. Hepatology. 40:1396–1405. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Llovet JM and Bruix J: Prospective validation of the Cancer of the Liver Italian Program (CLIP) score: A new prognostic system for patients with cirrhosis and hepatocellular carcinoma. Hepatology. 32:679–680. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Kim JY, Sinn DH, Gwak GY, Choi GS, Saleh AM, Joh JW, Cho SK, Shin SW, Carriere KC, Ahn JH, et al: Transarterial chemoembolization versus resection for intermediate-stage (BCLC B) hepatocellular carcinoma. Clin Mol Hepatol. 22:250–258. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Selby LK, Tay RX, Woon WW, Low JK, Bei W, Shelat VG, Pang TC and Junnarkar SP: Validity of the Barcelona clinic liver cancer and Hong Kong liver cancer staging systems for hepatocellular carcinoma in Singapore. J Hepatobiliary Pancreat Sci. 24:143–152. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Wai CT, Greenson JK, Fontana RJ, Kalbfleisch JD, Marrero JA, Conjeevaram HS and Lok AS: A simple noninvasive index can predict both significant fibrosis and cirrhosis in patients with chronic hepatitis C. Hepatology. 38:518–526. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Zhu GQ, Wang K, Wang B, Zhou YJ, Yang Y, Chen EB, Zhou ZJ, Zhou SL, Shi YH, Zhou J and Dai Z: Aspartate aminotransferase-to-platelet ratio index predicts prognosis of hepatocellular carcinoma after postoperative adjuvant transarterial chemoembolization. Cancer Manag Res. 11:63–79. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Zhang Y, Wan G, Li H, Gao L, Liu N, Gao P, Liu Y, Gao X and Duan X: A prediction nomogram for hepatitis B virus-associated hepatocellular carcinoma. Scand J Gastroenterol. 1–8. 2023.doi: 10.1080/00365521.2023.2252546 (Epub ahead of print). View Article : Google Scholar

38 

Tang T, Qiu JL, Li GW, Huang MP, Li Y, Li YJ and Gu SZ: Aspartate aminotransferase-to-platelet ratio predicts response to transarterial chemoembolisation and prognosis in hepatocellular carcinoma patients. Clin Radiol. 73:259–265. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Zhang X, Svn Z, Liv M, Liu M, Zhang Y and Sun Q: Assessment of prognostic value of aspartate Aminotransferase-to-Platelet ratio index in patients with hepatocellular carcinoma: Meta-analysis of 28 cohort studies. Front Med (Lausanne). 8:7562102021. View Article : Google Scholar : PubMed/NCBI

40 

Hung HH, Su CW, Lai CR, Chau GY, Chan CC, Huang YH, Huo TI, Lee PC, Kao WY, Lee SD and Wu JC: Fibrosis and AST to platelet ratio index predict post-operative prognosis for solitary small hepatitis B-related hepatocellular carcinoma. Hepatol Int. 4:691–699. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Zhang X, Xin Y, Yang Y, Chen Y, Cao XJ, Wang Y, Fan Q, Zhou X and Li X: Aspartate Aminotransferase-to-Platelet ratio index for predicting late recurrence of hepatocellular carcinoma after radiofrequency ablation. Int J Hyperthermia. 39:437–445. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Gorelick C, Andikyan V, Mack M, Lee YC and Abulafia O: Prognostic significance of preoperative thrombocytosis in patients with endometrial carcinoma in an inner-city population. Int J Gynecol Cancer. 19:1384–1389. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Lin MS, Huang JX, Zhu J and Shen HZ: Elevation of platelet count in patients with colorectal cancer predicts tendency to metastases and poor prognosis. Hepatogastroenterology. 59:1687–1690. 2012.PubMed/NCBI

44 

Gao J, Zhang HY and Xia YF: Increased platelet count is an indicator of metastasis in patients with nasopharyngeal carcinoma. Tumour Biol. 34:39–45. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Kim YJ, Borsig L, Varki NM and Varki A: P-selectin deficiency attenuates tumor growth and metastasis. Proc Natl Acad Sci USA. 95:9325–9330. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Pinedo HM, Verheul HM, D'Amato RJ and Folkman J: Involvement of platelets in tumour angiogenesis? Lancet. 352:1775–1777. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Jain S, Harris J and Ware J: Platelets: Linking hemostasis and cancer. Arterioscler Thromb Vasc Biol. 30:2362–2367. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Nieswandt B, Hafner M, Echtenacher B and Mannel DN: Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 59:1295–1300. 1999.PubMed/NCBI

49 

Maini MK and Schurich A: Platelets harness the immune response to drive liver cancer. Proc Natl Acad Sci USA. 109:12840–12841. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Witjes CD, Ijzermans JN, van der Eijk AA, Hansen BE, Verhoef C and de Man RA: Quantitative HBV DNA and AST are strong predictors for survival after HCC detection in chronic HBV patients. Neth J Med. 69:508–513. 2011.PubMed/NCBI

51 

Kamimoto Y, Horiuchi S, Tanase S and Morino Y: Plasma clearance of intravenously injected aspartate aminotransferase isozymes: Evidence for preferential uptake by sinusoidal liver cells. Hepatology. 5:367–375. 1985. View Article : Google Scholar : PubMed/NCBI

52 

Yugawa K, Maeda T, Nagata S, Sakai A, Edagawa M, Omine T, Kometani T, Yamaguchi S, Konishi K and Hashimoto K: A novel combined prognostic nutritional index and aspartate aminotransferase-to-platelet ratio index-based score can predict the survival of patients with hepatocellular carcinoma who undergo hepatic resection. Surg Today. 52:1096–1108. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Kao W, Chiou Y, Hung H, Chou Y, Su C, Wu J, Huo T, Huang Y, Lin H and Lee S: Risk factors for long-term prognosis in hepatocellular carcinoma after radiofrequency ablation therapy. Eur J Gastroenterol Hepatol. 23:12017.

54 

Persico M, Aglitti A, Aghemo A, Rendina M, Lleo A, Ciancio A, Di Marco V, Lampertico P, Brunetto MR, Zuin M, et al: High efficacy of direct-acting anti-viral agents in hepatitis C virus-infected cirrhotic patients with successfully treated hepatocellular carcinoma. Aliment Pharmacol Ther. 47:1705–1712. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2024
Volume 27 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shu Q, Zhang N, Han J, Yan X, Sha B, Zhao L, Yi Y and Zhang Y: Novel predictive nomograms based on aspartate aminotransferase‑to‑platelet ratio index for hepatocellular carcinoma with post‑operative adjuvant transarterial chemoembolization. Oncol Lett 27: 3, 2024
APA
Shu, Q., Zhang, N., Han, J., Yan, X., Sha, B., Zhao, L. ... Zhang, Y. (2024). Novel predictive nomograms based on aspartate aminotransferase‑to‑platelet ratio index for hepatocellular carcinoma with post‑operative adjuvant transarterial chemoembolization. Oncology Letters, 27, 3. https://doi.org/10.3892/ol.2023.14137
MLA
Shu, Q., Zhang, N., Han, J., Yan, X., Sha, B., Zhao, L., Yi, Y., Zhang, Y."Novel predictive nomograms based on aspartate aminotransferase‑to‑platelet ratio index for hepatocellular carcinoma with post‑operative adjuvant transarterial chemoembolization". Oncology Letters 27.1 (2024): 3.
Chicago
Shu, Q., Zhang, N., Han, J., Yan, X., Sha, B., Zhao, L., Yi, Y., Zhang, Y."Novel predictive nomograms based on aspartate aminotransferase‑to‑platelet ratio index for hepatocellular carcinoma with post‑operative adjuvant transarterial chemoembolization". Oncology Letters 27, no. 1 (2024): 3. https://doi.org/10.3892/ol.2023.14137