Clin Mol Hepatol > Volume 31(3); 2025 > Article
Lee, Yang, Tsai, Huang, Chen, Hung, Chen, Tai, Cheng, Kuo, Tseng, Mo, Lo, Huang, Lin, Lee, Bair, Chang, Lin, Wang, Hsieh, Yang, Peng, Yang, Chong, Huang, Lin, Chu, Tsai, Kao, Liu, Chuang, Lee, Yu, and on behalf of TACR investigators: Direct-acting antiviral therapy for patients with hepatitis C virus-related hepatocellular carcinoma: A nationwide cohort study

ABSTRACT

Background/Aims

The survival benefit of direct-acting antiviral (DAA) therapy for hepatitis C virus (HCV) infection in patients with hepatocellular carcinoma (HCC), particularly in Barcelona Clinic Liver Cancer (BCLC) stages B/C, remains largely uncertain. We aimed to explore the impact of DAA therapy on overall survival (OS) in HCC patients using a nationwide cohort study.

Methods

We utilized the nationwide Taiwan Association for the Study of the Liver (TASL) HCV Registry (TACR) database to include all adults receiving a DAA therapy for HCV, excluding those with other viral infections, liver transplantation, non-HCC malignancies, and terminal-staged HCC. We respectively analyzed the adjusted odds ratio (aOR) for sustained virological response (SVR) and adjusted hazard ratio (aHR) for OS.

Results

Between December 2013 and December 2020, 2,205 (9.3%) patients with HCC and 21,569 (90.7%) patients without HCC were include. The SVR rates were 96.6% in the HCC group and 98.8% in the non-HCC group (P<0.001), with HCC being an independent risk factor affecting SVR (aOR 0.41; 95% CI 0.31–0.54; P<0.001). In the whole patient cohort, SVR was independently associated with improved OS (aHR 0.46; 95% CI 0.35–0.60; P<0.001). Among patients with baseline HCC, SVR remained an independent factor related to OS (aHR 0.41; 95% CI 0.28–0.59; P<0.001). The impact of SVR on OS persisted significantly across BCLC stages 0/A and stages B/C.

Conclusions

High SVR rates among HCC patients underscore the importance of DAA therapy in enhancing OS, reaffirming its efficacy across various HCC stages.

Graphical Abstract

INTRODUCTION

Hepatitis C virus (HCV) infection is one of the major causes of hepatocellular carcinoma (HCC), with the outcomes of individuals diagnosed with HCC depending on both tumor burden and the degree of liver dysfunction [1]. To date, direct-acting antiviral agents (DAAs) are recommended for HCV infection due to their high rates of eradication and low rates of adverse events. Prior studies have reported the benefits of DAA therapy in reducing incident HCC and mortality among HCV-infected patients [2-4]. Additionally, DAAs are also known to improve survival outcomes, even in HCV-infected patients with a history of curative treatment for early HCC [5,6]. However, the survival benefit of DAA therapy in patients with HCC, particularly in Barcelona Clinic Liver Cancer (BCLC) stages B/C, remains largely uncertain.
In the era of DAA therapy, HCV eradication has become faster and safer, with HCV eradication therapy no longer being a difficult task for HCC patients. A high rate of sustained virological response (SVR) among HCC patients has been reported, though it seems to be lower when compared to patients without HCC. However, the use of DAA in patients with unresectable HCC remains controversial. In our previous hospital-based cohort study which enrolled 97 patients with HCC at BCLC stage B or C, a better survival outcome was disclosed in patients undergoing SVR to DAA therapy [7]. However, considering the limited sample size and potential selection bias, the benefits of DAA therapy in HCC patients need to be further confirmed through a largescale multicenter study.
The Taiwan Association for the Study of the Liver (TASL) HCV Registry (TACR) is a nationwide registry program that establishes and manages the database and biobank of HCV patients receiving DAA therapy in Taiwan. The demographic and virological data of all patients from each participating site have been completely collected and feature well-characterized demographic and virological characteristics, as detailed in previous publications [8-15]. Its nationwide scale is particularly suitable for overcoming the limitations in studies involving HCC patients. The aim of this study was to explore the survival benefit of DAA therapy in patients with HCV-related HCC using a nationwide cohort study.

MATERIALS AND METHODS

Study design

This was an observational, nationwide, cohort study of HCV-infected patients receiving DAA therapy, with or without HCC, through application of TACR data. The study has been approved by the Institutional Review Board of each study site (IRB No. SF19088B), while conforming to the guidelines of the International Conference on Harmonization for Good Clinical Practice. All patients provided written informed consent.

The insurance reimbursement criteria & the consensus for hepatitis C management in Taiwan

According to the reimbursement criteria of Taiwan National Health Insurance, DAA therapy is reimbursed for any patient with a detectable HCV viral load, regardless of underlying conditions, including HCC [16]. Therefore, DAA therapy can be initiated even in patients with active, untreated HCC prior to HCC treatment. However, for patients eligible for curative HCC treatments, the Taiwan consensus on the management of hepatitis C recommends prioritizing HCC treatment before initiating DAA therapy [17]. Conversely, given the accumulating evidence demonstrating high SVR rates in patients with active HCC treated with highly potent, pangenotypic DAA regimens [18,19], it is recommended that DAA therapy be initiated immediately for patients with incurable HCC, irrespective of HCC treatment.
As shown in Supplementary Table 1, the selection of DAAs must conform to their specific indications, including HCV genotypes and/or nonstructural 5A resistance-associated substitutions. In addition, due to the underlying comorbidities commonly observed in HCV carriers, a high prevalence of comedications has been reported [20,21]. To mitigate potential harmful effects caused by drug-drug interactions (DDIs), the Taiwan consensus on the management of hepatitis C recommends a thorough assessment of DDIs before initiating DAA therapy, including interactions with medications used for HCC treatment [17].

Study population and data collection

The TACR is a nationwide registry program, including patients who have received DAA therapy for HCV in Taiwan. By the end of 2020, 48 study sites, including 21 medical centers, 22 regional hospitals and five primary clinics, were participating in the registry [10]. As shown in Figure 1, we screened all patients registered in the TACR during the period from 25 December, 2013 to 31 December, 2020. The inclusion criteria were as follows: (1) aged 20 years or more, (2) detectable HCV RNA, (3) received DAA‐containing regimes for at least one dosage of any DAA, and (4) had virological response available. The exclusion criteria included: (1) hepatitis B virus (HBV) or HIV coinfection, (2) liver transplantation, (3) cancers other than HCC, or (4) terminal-stage HCC prior to DAA treatment initiation.
As outlined in our previously published studies [8-15], we retrieved comprehensive information from the TACR for analysis, including patient demographic characteristics (e.g., age, gender, body mass index [BMI], smoking and alcoholism), underlying comorbidities (e.g., liver cirrhosis, HCC, diabetes mellitus [DM], hypertension [HTN], cerebral vascular accident [CVA], coronary artery disease [CAD] and chronic kidney disease [CKD]), laboratory data (e.g., alanine aminotransferase [ALT], bilirubin, albumin, and HCV virological characteristics (HCV genotype, HCV RNA level, and virological responses to antivirals). Liver cirrhosis was defined by liver histology, transient elastography or the presence of clinical radiological evidence of cirrhosis. The presentation of liver cirrhosis and Child-Turcotte-Pugh (CTP) scores were recorded. Hepatic decompensation was defined as CTP scores ≥7. HCC was diagnosed by histological or typical image findings based on the guidelines of the American Association for the Study of Liver Diseases [22].
The DAA regimens used in this study were recorded, and pangenotypic regimens included glecaprevir/pibrentasvir, sofosbuvir/velpatasvir, and sofosbuvir/velpatasvir/voxilaprevir. Adherence to the DAA treatment of the enrolled subjects was also recorded and analyzed.

Study outcomes and linked databases

The primary endpoint of this study was overall survival (OS) of the study subjects. We linked the data taken from TACR with the National Health Insurance Research Database, where the dates of patient death could be retrieved [8]. Patients were followed up from the date of accessing SVR, i.e., the 12th week after completing DAA therapy, until the occurrence of death or the end of this study period (31 December, 2020), in which OS was defined.

Statistical analysis

Data was expressed as mean±standard deviation for continuous variables, and case number (percentage of the total number) for categorical variables. As appropriate, the Student’s t-test was used to compare the data of continuous variables, while chi-square or Fisher’s exact tests were used to compare those of categorical variables. The multivariable logistic regression model was applied to determine the independent factors related to SVR, in which the adjusted odds ratio (aOR) with a 95% confidence interval (CI) was presented. The multivariable Cox regression model was applied to determine the independent factors related to OS, in which the adjusted hazard ratio (aHR) with a 95% CI was presented. The Kaplan–Meier method was used to estimate the cumulative incidences of patient mortality, while the log-rank test was used to compare the incidence differences seen in different study groups. A P-value below 0.05 was defined as being statistically significant. All statistical analyses were performed using the SAS Enterprise Guide.

RESULTS

Patient characteristics

Figure 1 demonstrates the patient selection process, in which 31,894 patients were enrolled in the TACR cohort during the period from December 2013 to December 2020. After excluding patients as the exclusion criteria, 23,774 individuals were eventually enrolled for final analysis, with 2,205 cases (9.3%) and 21,569 (90.7%) having underlying HCC or not, respectively. As shown in Table 1, when compared to patients in the non-HCC group, patients in the HCC group were significantly older (70.5 vs. 61.6 years) and male-predominant (49.8% vs. 45.2%), with higher percentages in underlying comorbidities such as DM, HTN, CAD and CKD. However, BMI was not significantly different between the two study groups. Not surprisingly, the liver related parameters were more severe in the HCC group, including ALT, bilirubin and CTP scores, with the patient proportion of liver cirrhosis being significantly higher (63.7% vs. 25.7%). The mean HCV RNA was 5.9±1.0 log IU/mL, while HCV genotype 1 was significantly higher. The adherence to DAA treatment was better in those without HCC (99.6%). The proportion of BCLC stage 0, A, B and C was 22.2%, 52.3%, 16.5% and 9.0%, respectively.

Analysis of SVR in the whole population

The SVR rate of DAA therapy in the whole patient population was 98.6%, with it being seen as significantly lower in the HCC group when compared to that of the non-HCC group (96.6% vs. 98.8%; P<0.001). However, the SVR rate remained higher than 95% among patients with HCC.
As shown in Table 2, in the analysis for SVR-related factors, female gender (aOR 1.42; 95% CI 1.14–1.77; P=0.002), HCV genotype 1 (aOR 2.20; 95% CI 1.74–2.77; P<0.001), pangenotypic DAA regimen (aOR 2.14; 95% CI 1.63–2.81; P<0.001), DAA adherence >80% (aOR 31.51; 95% CI 20.23–49.09; P<0.001), liver cirrhosis (CTP score 5-6: aOR 0.76; 95% CI 0.60–0.97; P=0.028 & CTP score ≥7: aOR 0.47; 95% CI 0.26–0.85; P=0.012) and coexisting HCC (aOR 0.41; 95% CI 0.31–0.54; P<0.001) were all independent factors related to SVR.

Analysis of OS in the whole patient population

As shown in Table 3, in analysis of the prognostic factors related to OS, age >65 years (aHR 1.79; 95% CI 1.55–2.07; P<0.001), DM (aHR 1.51; 95% CI 1.37–1.81; P<0.001), CVA (aHR 1.45; 95% CI 1.10–1.91; P=0.009), CKD (aHR 2.22; 95% CI 1.93–2.54; P<0.001), liver cirrhosis (CTP score 5-6: aHR 1.59; 95% CI 1.37–1.83; P<0.001 & CTP score ≥7: aHR 5.49; 95% CI 4.35–6.92; P<0.001) and coexisting HCC (aHR 3.99, 95% CI 3.48-4.58; P<0.001) were all independent factors related to a decreased OS rate. In contrast, female gender (aHR 0.61; 95% CI 0.53–0.69; P<0.001) and SVR to DAA therapy (aHR 0.46; 95% CI 0.35–0.60; P<0.001) were independent factors related to an increased OS rate.
As shown in Figure 2, the 3-year cumulative incidence of OS was significantly higher in patients with SVR, when compared to that seen in patients without SVR (94.0%, 95% CI 93.5–94.4 vs. 78.5%, 95% CI 71.8–83.8; P<0.001). After adjusting for the factors of age, gender, DM, HTN, CVA, CAD, CKD, HCC and liver cirrhosis, SVR was associated with improved OS (aHR 0.46; 95% CI 0.35–0.60; P<0.001), when compared to non-SVR.

Analysis of OS among patients with baseline HCC

As shown in Table 4, DM (aHR 1.32; 95% CI 1.06–1.64; P=0.012), CKD (aHR 1.56; 95% CI 1.21–1.90; P<0.001), liver cirrhosis (CTP score 5-6: aHR 1.27; 95% CI 1.00–1.62; P=0.048 & CTP score ≥7: aHR 3.21; 95% CI 2.25–4.60; P<0.001), and HCC in BCLC stage B or C (aHR 1.78; 95% CI 1.44–2.21; P<0.001) were all independent factors related to a decreased OS. In contrast, female gender (aHR 0.74; 95% CI 0.61–0.93; P=0.005) and SVR (aHR 0.41; 95% CI 0.28–0.59; P<0.001) were independent factors related to an increased OS.
As shown in Figure 3, the 3-year cumulative incidence of OS was significantly higher in patients with SVR, when compared to that seen in patients without SVR (79.2%, 95% CI 77.2–81.2 vs. 48.3%, 95% CI 34.3–61.0; P<0.001). After adjusting for gender, DM, CKD, liver cirrhosis and BCLC stage, SVR was associated with improved OS (aHR 0.41; 95% CI 0.28–0.59; P<0.001), when compared to non-SVR.
We further analyzed the impact of SVR on different HCC stages. As shown in Figure 4, the 3-year cumulative incidence of OS was the highest among BCLC stage 0/A patients with SVR, but was lowest among BCLC stage B/C patients without SVR (P<0.001). After adjusting for gender, DM, CKD, and liver cirrhosis, SVR was respectively associated with improved OS in BCLC stage 0/A patients (aHR 0.47; 95% CI 0.29–0.77; P=0.002) and BCLC stage B/C patients (aHR 0.34; 95% CI 0.19–0.59; P<0.001), when compared to non-SVR.

DISCUSSION

This study is so far the first nationwide and largest cohort study investigating the survival benefits of DAA therapy in HCC patients. We report that although HCC may be a factor related to a reduced SVR rate in DAA therapy, the SVR rate among HCC patients remains very high. A lower SVR rate should not be a reason to withhold DAA therapy for HCC patients. Importantly, HCV eradication, i.e., SVR, through DAA therapy, is associated with improved OS in patients no matter whether they have HCC or not. Moreover, not only in early-stage HCC, i.e., BCLC stage 0/A, but also in BCLC stage B/C HCC, SVR is associated with improved OS, with DAA therapy being helpful for patients diagnosed with any-stage HCC who do not have a short-life expectancy.
In previous small-scale studies, although the SVR rates of DAA therapy in HCC patients were varied, ranging from 79% to 98%, HCC was commonly found to be a factor related to a lower SVR rate [23,24]. However, due to the rapid progress being seen in DAA development in recent years, the SVR rate in patients with genotype 1 to genotype 6 has become very high, nearly 100% in the per-protocol analysis of previous clinical trials using pangenotypic DAAs [25,26]. The high SVR rate among HCC patients in this study may be due to a large proportion of patients receiving HCV pangenotypic genotype-oriented DAA therapies, which have been reimbursed by the National Health Insurance program of Taiwan [27]. Although the SVR rate in HCC patients is expected to be lower when compared to non-HCC patients, the small difference in the rate may be omitted. This is because the SVR-related survival benefits in HCC patients should be certainly cost-effective.
Liver function is one of the most important prognostic factors in patients with HCC during each tumor stage [28], however most HCC patients suffer from underlying advanced liver fibrosis, significant portal HTN, and even liver decompensation [29]. Moreover, the HCC treatments, including curative treatments, locoregional treatments, and systemic therapies, may further impair liver function. However, impaired liver function may not only result in liver decompensation and death, but it may also delay HCC treatments [29]. Because achieving SVR through DAA therapy can improve liver function, it is reasonable to believe that pa-tient survival can be prolonged after curative or even palliative HCC treatments. However, the process of liver function improvement is not dramatically changed, and DAA therapy may not be beneficial in time for HCC patients with a short life expectancy, such as those in the terminal stage of HCC [30]. As demonstrated in this study, the survival benefit of SVR became more and more significant during the follow-up period.
In the past, although SVR achieved through interferon therapy could reduce the HCC recurrence rate and improve patient survival rates in those with early-stage HCC receiving curative treatments [31], patients with unresectable HCC or severe liver cirrhosis were often excluded from interferon therapy due to its high side effect possibility, long treatment period, and low SVR rate [32]. However, in the current era of DAA therapy, HCV eradication has become faster, safer and easier, even in those patients experiencing advanced liver diseases or severe underlying disorders [33-35]. Therefore, DAA therapy can be safely used in HCC patients. Although determining the optimal timing for HCV treatment intervention following HCC development is a clinical issue, particularly for patients with untreated or active HCC, the high SVR rates observed among HCC patients across various stages in this study may provide indirect evidence addressing this issue. The timing of DAA therapy and HCC treatment is no longer as conflicting as it was in the era of interferon-based therapy. In patients with early-stage HCC following curative treatments, DAA therapy can reduce the HCC recurrence rate and further improve OS. For example, in a prospective multiple-center study performed in Italy, DAA therapy significantly reduced as high as a 61% risk of death, a 30% risk of HCC recurrence and a 68% risk of hepatic decompensation, after a mean follow-up period of 21.4 months [5]. Moreover, in previous single center-based studies, DAA therapy was considered to be helpful in improving OS in patients diagnosed with BCLC stage B/C HCC [7,24]. This nationwide study further provides strong evidence that DAA therapy should become an essential component nowadays in the management of HCV-infected HCC patients across various HCC stages.
Several limitations should be addressed in this study. First, although selection bias should have been minimized in this nationwide study, all study subjects used in this study were Taiwanese. More large-scale validation studies from other countries or populations should be welcome. Second, some patients receiving DAA therapy had been excluded in the final analysis, such as those with HBV or HIV coinfection; therefore, further investigations for these special populations should be encouraged in the future. Third, a DAA-untreated control group was lacking. Conducting a study to establish a DAA-untreated control group should not be considered an ethical procedure. Fortunately, we were able to include patients who did not achieve SVR as the control group in this large-scale study. Fourth, due to the limited resources available for this study, including the constrained study timeframe, further analyses focusing on specific subpopulations, such as HCC patients receiving immunotherapy, could not be performed. Nevertheless, the positive findings of our study provide a strong foundation for future investigations.
In conclusion, this nationwide cohort study provides strong evidence that DAA therapy can achieve high SVR rates, even in HCC patients. Achieving SVR can significantly improve the survival of HCC patients, across various HCC stages. DAA therapy is highly recommended for all HCC patients who do not have a short life expectancy.

FOOTNOTES

Authors’ contribution
Study conception and design: S.W.L., S.S.Y., T.Y.L., M. L.Y. Data acquisition, analysis and interpretation: S.W.L., S.S.Y., P.C.T., C.F.H., C.Y.C., C.H.H., C.H.C., C.M.T., P.N.C., H.T.K., K.C.T., L.R.M., C.C.L., Y.H.H., H.C.L., P.L.L., M.J.B., T.S.C., C.Y.L., S.J.W., T.Y.H., T.H.Y., C.Y.P., C.C.Y., L.W.C., C.W.H., C.W.L., C.H.C., M.C.T., J.H.K., C.J.L., W.L.C., T.Y.L., M.L.Y. Manuscript drafting: S.W.L, S.S.Y., P.C.T., T.Y.L. Critical revision for important intellectual content: S.W.L, S.S.Y., P.C.T., T.Y.L., M.L.Y. All authors contributed to the revision of the final manuscript.
Acknowledgements
The authors would like to thank both the Taiwan Association for the Study of Liver (TASL) and the Taiwan Association for the Study of the Liver Foundation (TASLF) for grant support, and the TASL HCV Registry (TACR) study group for data collection. We also wish to thank the Center for Medical Informatics and Statistics of Kaohsiung Medical University for providing administrative and funding support. All linkage databases were supported from the Health and Welfare Data Science Center, Taiwan.
This work was supported in part by the Ministry of Health and Welfare (MOHW111-TDU-B-221-114007, MOHW112-TDU-B-221-124007, MOHW113-TDU-B-221-134007), Nat ional Science and Technology Counci l (NSTC 112-2314-B-075A-010-MY3), Taichung Veterans General Hospital (TCVGH-1123301C, TCVGH-1133301C, VTA112-V1-3-3, VTA113-V1-1-2), Kaohsiung Medical University (KMU-NSTC 112-2635-B-037-001-MY2, KMU-MOST 111-2314-B-037-069-MY2, MOHW113-TDU-B-221-134007), Kaohsiung Medical University Hospital (KMUH112-2R09, KMUH111-1R04, KMUH-DK(A)113002), all in Taiwan. The funders had no role in the design or conduct of the study, the collection, analysis and interpretation of the data, nor the preparation, review or approval of the manuscript.
Conflicts of Interest
TY Lee: research support from Gilead, Merck and Roche diagnostics; consultant of BMS, Gilead, and Astra- Zeneca and speaker of Abbvie, BMS, Eisai, Gilead, Roche and AstraZeneca.
ML Yu: research support from Abbvie, Abbott Diagnostic, BMS, Gilead, Merck and Roche diagnostics; consultant of Abbvie, Abbott Diagnostic, BMS, Gilead, Roche and Roche diagnostics and speaker of Abbvie, BMS, Eisai, Gilead, Roche and Roche diagnostics. All other authors declare that they do not have any relevant conflict of interest.

SUPPLEMENTAL MATERIAL

Supplementary material is available at Clinical and Molecular Hepatology website (http://www.e-cmh.org).
Supplementary Table 1.
The virological limitations of DAA regimens reimbursed in Taiwan National Health Insurance
cmh-2024-1015-Supplementary-Table-1.pdf

Figure 1.
Flowchart of patient selection. BCLC, Barcelona Clinic Liver Cancer; DAA, direct-acting antiviral; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; HIV, human immunodeficiency virus; N, numbers; SVR, sustained virological response.

cmh-2024-1015f1.jpg
Figure 2.
Kaplan–Meier analysis of overall survival in patients with or without SVR. All HRs (95% CIs) and P-values were calculated by the Cox subdistribution hazards method. *Adjusted for age, gender, diabetes mellitus, hypertension, cerebral vascular accident, coronary heart disease, chronic kidney disease, hepatocellular carcinoma, and liver cirrhosis. CI, confidence interval; HR, hazard ratio; N, numbers; OS, overall survival; SVR, sustained virological response.

cmh-2024-1015f2.jpg
Figure 3.
Kaplan–Meier analysis of overall survival in HCC patients with or without SVR. All HRs (95% CIs) and P-values were calculated by the Cox subdistribution hazards method. *Adjusted for gender, diabetes mellitus, chronic kidney disease, liver cirrhosis, and BCLC stage. BCLC, Barcelona Clinic Liver Cancer; CI, confidence interval; HCC, hepatocellular carcinoma; HR, hazard ratio; N, numbers; OS, overall survival; SVR, sustained virological response.

cmh-2024-1015f3.jpg
Figure 4.
Kaplan–Meier analysis of overall survival in HCC patients with or without SVR (BCLC stage 0-A or B-C). All HRs (95% CIs) and P-values were calculated by the Cox subdistribution hazards method. *Adjusted for gender, diabetes mellitus, chronic kidney disease, and liver cirrhosis. BCLC, Barcelona Clinic Liver Cancer; CI, confidence interval; HCC, hepatocellular carcinoma; HR, hazard ratio; N, numbers; OS, overall survival; SVR, sustained virological response.

cmh-2024-1015f4.jpg

cmh-2024-1015f5.jpg
Table 1.
The baseline characteristics of all (HCC & non-HCC) patients receiving a DAA therapy
Variables Total Non-HCC HCC P-value
n=23,774 (100) n=21,569 (90.7) n=2,205 (9.3)
Age (yr) 62.4±12.4 61.6±12.4 70.5±9.1 <0.001
 >65 10,096 (42.5) 8,503 (39.4) 1,593 (72.2) <0.001
Male 10,845 (45.6) 9,748 (45.2) 1,097 (49.8) <0.001
BMI (kg/m2) 24.6±3.5 24.7±3.5 24.6±3.5 0.725
 ≥27 4,564 (19.2) 4,127 (19.1) 437 (19.8) 0.437
DM 4,780 (20.1) 4,156 (19.3) 624 (28.3) <0.001
HTN 7,645 (32.2) 6,669 (30.9) 976 (44.3) <0.001
CVA 670 (2.8) 598 (2.8) 72 (3.3) 0.183
CAD 2,259 (9.5) 1,989 (9.2) 270 (12.2) <0.001
CKD 3,788 (15.9) 3,294 (15.3) 494 (22.4) <0.001
ALT (IU/L) 75.1±78.4 74.5±79.6 81.2±65.4 <0.001
 ≥80 7,172 (30.2) 6,377 (29.6) 795 (36.1) <0.001
Total bilirubin (mg/dL) 0.8±0.5 0.8±0.5 1.0±0.6 <0.001
Albumin (g/dL) 4.2±0.4 4.2±0.4 3.8±0.5 <0.001
Liver cirrhosis 6,956 (29.3) 5,551 (25.7) 1,405 (63.7) <0.001
 CTP score 5–6 6,533 (27.5) 5,258 (24.4) 1,275 (57.8)
 CTP score ≥7 423 (1.8) 293 (1.4) 130 (5.9)
HCV viral load (log IU/mL) 5.9±1.0 5.9±1.0 5.7±0.9 <0.001
HCV genotype
 1 12,263 (51.7) 10,849 (50.4) 1,414 (64.2) <0.001
 2 9,319 (39.3) 8,650 (40.1) 669 (30.4)
 Mixed 290 (1.2) 255 (1.2) 35 (1.6)
 Other 1,727 (7.3) 1,657 (7.7) 70 (3.2)
 Unclassified 139 (0.6) 126 (0.6) 13 (0.6)
DAA regimen
 Pangenotypic* 8,811 (37.1) 8,388 (38.9) 423 (19.2) <0.001
 Non-pangenotypic 14,963 (62.9) 13,181 (61.1) 1,782 (80.8)
DAA adherence >80% 23,662 (99.5) 21,482 (99.6) 2,180 (98.9) <0.001
BCLC stage
 0 - - 432 (22.2)
 A - - 1,017 (52.3)
 B - - 322 (16.5)
 C - - 175 (9.0)

Values are presented as number (%) or mean±standard deviation.

ALT, alanine aminotransferase; BCLC, Barcelona Clinic Liver Cancer; BMI, body mass index; CAD, coronary artery disease; CKD, chronic kidney disease; CTP, Child-Turcotte-Pugh; CVA, cerebral vascular accident; DAA, direct-acting antiviral therapy; DM, diabetes mellitus; HCC, hepatocellular carcinoma; HTN, hypertension; N, numbers.

* Pangenotypic DAA included glecaprevir/pibrentasvir, sofosbuvir/velpatasvir, and sofosbuvir/velpatasvir/voxilaprevir.

Table 2.
Logistic regression model for risk factors of SVR in all (HCC & non-HCC) patients receiving a DAA therapy
Variables N SVR N (%) Crude
Adjusted
OR (95% CI) P-value OR (95% CI) P-value
Age (yr) ≤65 13,678 13,492 (98.6) 1
>65 10,096 9,942 (98.5) 0.89 (0.72–1.10) 0.288
Gender Male 10,845 10,667 (98.4) 1 1
Female 12,929 12,767 (98.7) 1.32 (1.06–1.63) 0.012 1.42 (1.14–1.77) 0.002
BMI (kg/m2) ≤27 19,210 18,941 (98.6) 1
>27 4,564 4,493 (98.4) 0.90 (0.69–1.17) 0.427
DM No 18,994 18,742 (98.7) 1 1
Yes 4,780 4,692 (98.2) 0.72 (0.56–0.92) 0.008 0.88 (0.68–1.14) 0.344
HTN No 16,129 15,901 (98.6) 1
Yes 7,645 7,533 (98.5) 0.96 (0.77–1.21) 0.754
CVA No 23,104 22,772 (98.6) 1
Yes 670 662 (98.8) 1.21 (0.60–2.44) 0.602
CAD No 21,515 21,217 (98.6) 1
Yes 2,259 2,217 (98.1) 0.74 (0.54–1.03) 0.072
CKD No 19,986 19,698 (98.6) 1
Yes 3,788 3,736 (98.6) 1.05 (0.78–1.41) 0.746
HCV viral load ≤8×105 21,438 21,138 (98.6) 1
>8×105 2,336 2,296 (98.3) 0.82 (0.58–1.14) 0.227
HCV genotype Non-1 11,475 11,278 (98.3) 1 1
1 12,263 12,122 (98.9) 1.50 (1.21–1.87) <0.001 2.20 (1.74–2.77) <0.001
DAA regimen Non-pangenotypic 14,963 14,702 (98.3) 1 1
Pangenotypic* 8,811 8,732 (99.1) 1.96 (1.52–2.53) <0.001 2.14 (1.63–2.81) <0.001
DAA adherence ≤80% 112 79 (70.5) 1 1
>80% 23,662 23,355 (98.7) 31.80 (20.86–48.47) <0.001 31.51 (20.23–49.09) <0.001
ALT (IU/L) ≤80 16,602 16,373 (98.6) 1
>80 7,172 7,061 (98.5) 0.89 (0.71–1.12) 0.316
Liver cirrhosis No 16,818 16,624 (98.9) 1 1
CTP score 5–6 6,533 6,400 (98.0) 0.56 (0.45–0.70) <0.001 0.76 (0.60–0.97) 0.028
CTP score ≥7 423 410 (96.9) 0.37 (0.21–0.65) <0.001 0.47 (0.26–0.85) 0.012
HCC No 21,569 21,305 (98.8) 1 1
Yes 2,205 2,129 (96.6) 0.35 (0.27–0.45) <0.001 0.41 (0.31–0.54) <0.001

ALT, alanine aminotransferase; BMI, body mass index; CAD, coronary artery disease; CI, confidence interval; CKD, chronic kidney disease; CTP, Child-Turcotte-Pugh; CVA, cerebral vascular accident; DAA, direct-acting antiviral therapy; DM, diabetes mellitus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; HTN, hypertension; N, numbers; OR, adds ratio; SVR, sustained virological response.

* Pangenotypic DAA included glecaprevir/pibrentasvir, sofosbuvir/velpatasvir, and sofosbuvir/velpatasvir/voxilaprevir.

Table 3.
Cox subdistribution hazards model for risk factors of overall survival in all (HCC & non-HCC) patients receiving a DAA therapy
Variables N Death N (%) Crude
Adjusted
HR (95% CI) P-value HR (95% CI) P-value
Total 23,774 954 (4.0) - - - -
Age (yr) ≤65 13,678 299 (2.2) 1 1
>65 10,096 655 (6.5) 2.60 (2.27–2.98) <0.001 1.79 (1.55–2.07) <0.001
Gender Male 10,845 522 (4.8) 1 1
Female 12,929 432 (3.3) 0.62 (0.54–0.70) <0.001 0.61 (0.53–0.69) <0.001
BMI (kg/m2) ≤27 19,210 773 (4.0) 1 - -
>27 4,564 181 (4.0) 0.92 (0.78–1.08) 0.296 - -
DM No 18,994 601 (3.2) 1 1
Yes 4,780 353 (7.4) 2.16 (1.89–2.46) <0.001 1.51 (1.37–1.81) <0.001
HTN No 16,129 521 (3.2) 1
Yes 7,645 433 (5.7) 1.63 (1.44–1.85) <0.001 0.92 (0.80–1.06) 0.244
CVA No 23,104 898 (3.9) 1 1
Yes 670 56 (8.4) 2.21 (1.68–2.89) <0.001 1.45 (1.10–1.91) 0.009
CAD No 21,515 808 (3.8) 1 1
Yes 2,259 146 (6.5) 1.73 (1.45–2.06) <0.001 1.17 (0.98–1.41) 0.088
CKD No 19,986 616 (3.1) 1 1
Yes 3,788 338 (8.9) 3.18 (2.79–3.63) <0.001 2.22 (1.93–2.54) <0.001
Liver cirrhosis No 16,818 348 (2.1) 1 1
CTP score 5–6 6,533 509 (7.8) 2.37 (2.06–2.72) <0.001 1.59 (1.37–1.83) <0.001
CTP score ≥7 423 97 (22.9) 9.45 (7.55–11.84) <0.001 5.49 (4.35–6.92) <0.001
HCC No 21,569 527 (2.4) 1 1
Yes 2,205 427 (19.4) 6.09 (5.35–6.92) <0.001 3.99 (3.48–4.58) <0.001
SVR No 340 51 (15.0) 1 1
Yes 23,434 903 (3.9) 0.29 (0.22–0.39) <0.001 0.46 (0.35–0.60) <0.001

BMI, body mass index; CAD, coronary artery disease; CI, confidence interval; CKD, chronic kidney disease; CTP, Child-Turcotte-Pugh; CVA, cerebral vascular accident; DAA, direct-acting antiviral therapy; DM, diabetes mellitus; HCC, hepatocellular carcinoma; HR, hazard ratio; HTN, hypertension; N, numbers; SVR, sustained virological response.

Table 4.
Cox subdistribution hazards model for risk factors of overall survival in HCC patients receiving a DAA therapy
Variables N Death N (%) Crude
Adjusted
HR (95% CI) P-value HR (95% CI) P-value
Total 2,205 427 (19.4) - - - -
Age (yr) ≤65 612 104 (17.0) 1 - -
>65 1,593 323 (20.3) 1.22 (0.98–1.53) 0.075 - -
Gender Male 1,097 234 (21.3) 1 1
Female 1,108 193 (17.4) 0.76 (0.62–0.91) 0.004 0.74 (0.61–0.93) 0.005
BMI (kg/m2) ≤27 1,768 350 (19.8) 1 - -
>27 437 77 (17.6) 0.89 (0.70–1.14) 0.355 - -
DM No 1,581 287 (18.2) 1 1
Yes 624 140 (22.4) 1.29 (1.05–1.58) 0.014 1.32 (1.06–1.64) 0.012
HTN No 1,229 234 (19.0) 1 - -
Yes 976 193 (19.8) 1.06 (0.88–1.28) 0.560 - -
CVA No 2,133 407 (19.1) 1 - -
Yes 72 20 (27.8) 1.51 (0.96–2.36) 0.074 - -
CAD No 1,935 369 (19.1) 1 - -
Yes 270 58 (21.5) 1.24 (0.94–1.64) 0.125 - -
CKD No 1,711 304 (17.8) 1 1
Yes 494 123 (24.9) 1.62 (1.31–2.00) <0.001 1.56 (1.21–1.90) <0.001
Liver cirrhosis No 800 110 (13.8) 1 1
CTP score 5–6 1,275 268 (21.0) 1.29 (1.04–1.62) 0.023 1.27 (1.00–1.62) 0.048
CTP score ≥7 130 49 (37.7) 3.14 (2.24–4.39) <0.001 3.21 (2.25–4.60) <0.001
BCLC stage B/C 1,449 250 (17.3) 1 1
0/A 497 129 (26.0) 2.02 (1.47–2.78) <0.001 1.78 (1.44–2.21) <0.001
SVR No 76 34 (44.7) 1 1
Yes 2,129 393 (18.5) 0.37 (0.26–0.53) <0.001 0.41 (0.28–0.59) <0.001

BCLC, Barcelona Clinic Liver Cancer; BMI, body mass index; CAD, coronary artery disease; CI, confidence interval; CKD, chronic kidney disease; CTP, Child-Turcotte-Pugh; CVA, cerebral vascular accident; DAA, direct-acting antiviral therapy; DM, diabetes mellitus; HCC, hepatocellular carcinoma; HR, hazard ratio; HTN, hypertension; N, numbers; SVR, sustained virological response.

Abbreviations

aHR
adjusted hazard ratio
ALT
alanine aminotransferase
AST
aspartate aminotransferase
aOR
adjusted odds ratio
BCLC
Barcelona Clinic Liver Cancer
BMI
body mass index
CAD
coronary artery disease
CHC
chronic hepatitis C
CI
confidence interval
CKD
chronic kidney disease
CTP
Child-Turcotte-Pugh
CVA
cerebral vascular accident
DAA
direct-acting antiviral
DDI
drug-drug interaction
DM
diabetes mellitus
eGFR
estimated glomerular filtration rate
FIB-4
fibrosis-4 index
HCC
hepatocellular carcinoma
HCV
hepatitis C virus
HTN
hypertension
LC
liver cirrhosis
OR
odds ratio
OS
overall survival
SVR
sustained virological response

REFERENCES

1. Reig M, Forner A, Rimola J, Ferrer-Fàbrega J, Burrel M, Garcia-Criado Á, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol 2022;76:681-693.
crossref pmid pmc
2. Ioannou GN, Green PK, Berry K. HCV eradication induced by direct-acting antiviral agents reduces the risk of hepatocellular carcinoma. J Hepatol 2017;68:25-32.
crossref pmid pmc
3. Calvaruso V, Cabibbo G, Cacciola I, Petta S, Madonia S, Bellia A, et al.; Rete Sicilia Selezione Terapia–HCV (RESIST-HCV). Incidence of hepatocellular carcinoma in patients with HCVassociated cirrhosis treated with direct-acting antiviral agents. Gastroenterology 2018;155:411-421.e4.
pmid
4. Singal AG, Lim JK, Kanwal F. AGA clinical practice update on interaction between oral direct-acting antivirals for chronic hepatitis C infection and hepatocellular carcinoma: expert review. Gastroenterology 2019;156:2149-2157.
crossref pmid pmc
5. Cabibbo G, Celsa C, Calvaruso V, Petta S, Cacciola I, Cannavò MR, et al.; Rete Sicilia Selezione Terapia – HCV (RESISTHCV) and Italian Liver Cancer (ITA.LI.CA.) Group. Direct-acting antivirals after successful treatment of early hepatocellular carcinoma improve survival in HCV-cirrhotic patients. J Hepatol 2019;71:265-273.
crossref pmid
6. Ohki T, Sato K, Kondo M, Goto E, Sato T, Kondo Y, et al. Effectiveness of direct acting antiviral agents for hepatitis C virus related recurrent hepatocellular carcinoma patients who had multiple courses of recurrence. J Viral Hepat 2021;28:1597-1603.
crossref pmid pdf
7. Lee SW, Chen LS, Yang SS, Huang YH, Lee TY. Direct-acting antiviral therapy for hepatitis C virus in patients with BCLC stage B/C hepatocellular carcinoma. Viruses 2022;14:2316.
crossref pmid pmc
8. Tsai PC, Kuo HT, Hung CH, Tseng KC, Lai HC, Peng CY, et al.; T-COACH Study Group. Metformin reduces hepatocellular carcinoma incidence after successful antiviral therapy in patients with diabetes and chronic hepatitis C in Taiwan. J Hepatol 2023;78:281-292.
pmid
9. Chang TS, Huang CF, Kuo HT, Lo CC, Huang CW, Chong LW, et al. Effectiveness and safety of 8-week glecaprevir/pibrentasvir in HCV treatment-naïve patients with compensated cirrhosis: real-world experience from Taiwan nationwide HCV registry. Hepatol Int 2023;17:550-561.
pmid
10. Chen CY, Huang CF, Cheng PN, Tseng KC, Lo CC, Kuo HT, et al. Factors associated with treatment failure of direct-acting antivirals for chronic hepatitis C: a real-world nationwide hepatitis C virus registry programme in Taiwan. Liver Int 2021;41:1265-1277.
crossref pmid pmc pdf
11. Huang CF, Tseng KC, Cheng PN, Hung CH, Lo CC, Peng CY, et al. Impact of Sofosbuvir-based direct-acting antivirals on renal function in chronic hepatitis C patients with impaired renal function: a large cohort study from the nationwide HCV registry program (TACR). Clin Gastroenterol Hepatol 2022;20:1151-1162.e6.
crossref pmid
12. Chen YC, Chang TS, Chen CH, Cheng PN, Lo CC, Mo LR, et al. Factors associated with significant platelet count improvement in thrombocytopenic chronic hepatitis C patients receiving direct-acting antivirals. Viruses 2022;14:333.
pmid pmc
13. Hong CM, Lin YY, Liu CJ, Lai YY, Yeh SH, Yang HC, et al. Drug resistance profile and clinical features for hepatitis C patients experiencing DAA failure in Taiwan. Viruses 2021;13:2294.
crossref pmid pmc
14. Cheng PN, Mo LR, Chen CT, Chen CY, Huang CF, Kuo HT, et al.; TACR investigators. Sofosbuvir/velpatasvir for hepatitis C virus infection: real-world effectiveness and safety from a nationwide registry in Taiwan. Infect Dis Ther 2022;11:485-500.
pmid
15. Yeh ML, Lin CL, Lin CY, Lee PL, Chong LW, Hung CH, et al.; TACR investigators. Ledipasvir/sofosbuvir for HCV genotype 1, 2, 4-6 infection: real-world evidence from a nationwide registry in Taiwan. J Formos Med Assoc 2022;121:1567-1578.
pmid
16. Chien RN, Lu SN, Pwu RF, Wu GH, Yang WW, Liu CL. Taiwan accelerates its efforts to eliminate hepatitis C. Glob Health Med 2021;3:293-300.
crossref pmid pmc
17. Yu ML, Chen PJ, Dai CY, Hu TH, Huang CF, Huang YH, et al. 2020 Taiwan consensus statement on the management of hepatitis C: part (II) special populations. J Formos Med Assoc 2020;119:1135-1157.
crossref pmid
18. Fukumoto T, Minami T, Moriyama M, Yamada T, Wake T, Kinoshita MN, et al. Improved prognosis of hepatitis C-related hepatocellular carcinoma in the era of direct-acting antivirals. Hepatol Commun 2022;6:2496-2512.
crossref pmid pmc pdf
19. Baumert TF, Berg T, Lim JK, Nelson DR. Status of direct-acting antiviral therapy for hepatitis C virus infection and remaining challenges. Gastroenterology 2019;156:431-445.
crossref pmid
20. Hsu PY, Wei YJ, Lee JJ, Niu SW, Huang JC, Hsu CT, et al. Comedications and potential drug-drug interactions with directacting antivirals in hepatitis C patients on hemodialysis. Clin Mol Hepatol 2021;27:186-196.
crossref pmid pdf
21. Hui VW, Au CL, Lam ASM, Yip TC, Tse YK, Lai JC, et al. Drugdrug interactions between direct-acting antivirals and co-medications: a territory-wide cohort study. Hepatol Int 2022;16:1318-1329.
crossref pmid pdf
22. Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology 2018;67:358-380.
crossref pmid pdf
23. Ji F, Yeo YH, Wei MT, Ogawa E, Enomoto M, Lee DH, et al. Sustained virologic response to direct-acting antiviral therapy in patients with chronic hepatitis C and hepatocellular carcinoma: a systematic review and meta-analysis. J Hepatol 2019;71:473-485.
pmid
24. Prenner SB, VanWagner LB, Flamm SL, Salem R, Lewandowski RJ, Kulik L. Hepatocellular carcinoma decreases the chance of successful hepatitis C virus therapy with directacting antivirals. J Hepatol 2017;66:1173-1181.
crossref pmid pmc
25. Zoratti MJ, Siddiqua A, Morassut RE, Zeraatkar D, Chou R, van Holten J, et al. Pangenotypic direct acting antivirals for the treatment of chronic hepatitis C virus infection: a systematic literature review and meta-analysis. EClinicalMedicine 2020;18:100237.
crossref pmid pmc
26. Brown RS Jr, Buti M, Rodrigues L, Chulanov V, Chuang WL, Aguilar H, et al. Glecaprevir/pibrentasvir for 8 weeks in treatment-naïve patients with chronic HCV genotypes 1-6 and compensated cirrhosis: the EXPEDITION-8 trial. J Hepatol 2020;72:441-449.
crossref pmid
27. Feld JJ, Jacobson IM, Hézode C, Asselah T, Ruane PJ, Gruener N, et al.; ASTRAL-1 Investigators. Sofosbuvir and velpatasvir for HCV genotype 1, 2, 4, 5, and 6 infection. N Engl J Med 2015;373:2599-2607.
crossref pmid
28. Bruix J, Reig M, Sherman M. Evidence-based diagnosis, staging, and treatment of patients with hepatocellular carcinoma. Gastroenterology 2016;150:835-853.
crossref pmid
29. Vogel A, Meyer T, Sapisochin G, Salem R, Saborowski A. Hepatocellular carcinoma. Lancet 2022;400:1345-1362.
crossref pmid
30. Reig M, Cabibbo G. Antiviral therapy in the palliative setting of HCC (BCLC-B and -C). J Hepatol 2021;74:1225-1233.
crossref pmid
31. Yoshida H, Tateishi R, Arakawa Y, Sata M, Fujiyama S, Nishiguchi S, et al. Benefit of interferon therapy in hepatocellular carcinoma prevention for individual patients with chronic hepatitis C. Gut 2004;53:425-430.
crossref pmid pmc
32. Chen LS, Lee SW, Yang SS, Tsai HJ, Lee TY. Advances in the era of direct-acting antivirals for hepatitis C in patients with unresectable hepatocellular carcinoma. Dig Dis 2022;40:616-624.
crossref pmid pdf
33. Backus LI, Belperio PS, Shahoumian TA, Mole LA. Impact of sustained virologic response with direct-acting antiviral treatment on mortality in patients with advanced liver disease. Hepatology 2019;69:487-497.
crossref pmid pdf
34. Herzer K, Gerken G, Kroy D, Tacke F, Plewe J, Eurich D, et al. Impact of direct-acting antiviral therapy on the need for liver transplantation related to hepatitis C in Germany. J Hepatol 2018;69:982-984.
crossref pmid
35. Lai PC, Chen CH, Jeng LB, Yu TM, Tsai SF, Wu MJ, et al. Grazoprevir/elbasvir treatment in liver or kidney transplant recipients with genotype 1b hepatitis C virus infection. Antimicrob Agents Chemother 2022;66:e0200321.
crossref pmid pdf

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 3826
TOTAL : 2926018
Close layer