KASL clinical practice guidelines for management of chronic hepatitis B

Article information

Clin Mol Hepatol. 2022;28(2):276-331
Publication date (electronic) : 2022 April 1
doi : https://doi.org/10.3350/cmh.2022.0084
The Korean Association for the Study of the Liver (KASL)*
Corresponding author : The Korean Association for the Study of the Liver (KASL) (Committee Chair: Jeong Won Jang) Room A1210 MapoTrapalace, 53 Mapo-daero, Mapo-gu, Seoul 04158, Korea Tel: +82-2-703-0051, Fax: +82-2-703-0071 E-mail: kasl@kams.or.kr or garden@catholic.ac.kr
*Clinical Practice Guidelines Committee of KASL for the Management of Chronic Hepatitis B: Jeong Won Jang (Committee chair, The Catholic University of Korea), Jun Yong Park (Yonsei University College of Medicine), Jung Hyun Kwon (The Catholic University of Korea), Su Jong Yu (Seoul National University College of Medicine), Wonseok Kang (Sungkyunkwan University School of Medicine), Young Eun Chon (CHA University College of Medicine), Tae Hyung Kim (Korea University College of Medicine), Jonggi Choi (University of Ulsan College of Medicine), Jihyun An (Hanyang University College of Medicine)
Editor: Seung Up Kim, Yonsei University College of Medicine, Korea
Received 2022 March 31; Accepted 2022 April 1.

INTRODUCTION

Chronic hepatitis B (CHB) is a major cause of chronic liver diseases in Korea and remains a threat to public health due to its high morbidity and disease-related mortality. Since the latest version of clinical practice guidelines of CHB in 2018 by the Korean Association for the Study of the Liver (KASL), many studies related to hepatitis B virus (HBV) have been published, raising the need to revise the recommendations reflecting the most up-to-date information on management of CHB. The guidelines are intended to provide useful information and medical guidance for clinicians responsible for diagnosing and treating Korean patients with CHB. The current version differs from the published 2018 KASL guidelines, which revised the entire part about CHB, in that it primarily focuses on the 12 major clinical topics that require updated medical information and the latest knowledge. In addition to the natural course of CHB, treatment indication, cessation of antiviral therapy, and management for special populations covered in the previous version of the guidelines, this version will establish new topics on ‘Emerging Markers of HBV Infection’ and ‘New Drugs for Functional Cure,’ which have recently showed advancements for diagnosis and treatment of CHB. Recommendations on parts of CHB not included in this guideline are noted in the 2018 guidelines.

The current practice of managing hepatitis B has become more complex as many targeted and immune therapies are being widely used for systemic treatments of cancer patients with chronic HBV infection. In the updated guidelines, the reported rates of hepatitis B reactivation associated with these new drugs were added, and prophylactic antiviral treatment was recommended accordingly. In transplant settings of the liver and solid organs, the risk of and management against post-transplant hepatitis B recurrence were subclassified according to the status of HBV markers of donors and recipients. In particular, prophylactic management of patients undergoing hematopoietic stem cell transplantation who are at a very high rate of HBV reactivation was separately described. The 2022 guidelines summarized the emerging therapies for a cure of HBV and newly provided hepatocellular carcinoma (HCC) risk models and emerging markers of HBV for clinical utility. In addition, the guidelines updated the feasibility of cessation of antiviral drugs using new HBV markers and management of patients with bone and renal disease, co-infections with other hepatitis virus, and those at the so-called ‘grey zone.’

The Committee for 2022 KASL clinical practice guidelines for CHB, launched in accordance with the initiative of the Board of Directors of the KASL and approved by the council, was composed of nine hepatologists. The Committee searched newly published articles related to hepatitis B from PubMed, MEDLINE (up to 2021), KoreaMed, as well as abstracts and proceedings of international academic conferences and collected necessary data since publication of the 2018 guidelines in order to provide updated recommendations based on the latest data. The levels of evidence in the guidelines were classified by the revised Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) system. The levels of evidence were based on the possibility of change in the estimate of clinical effect by further research and were described as high (A), moderate (B), or low (C). Classification of grades of recommendation were either strong (1) or weak (2), by the GRADE system, according to the level of evidence, generalizability, clinical effect of the research result, and socioeconomic aspects. Each recommendation is combined with the level of relevant evidence (A–C) and corresponding recommendation grade (1, 2) as follows: A1, A2, B1, B2, C1, and C2 (Table 1).

Grading of Recommendations, Assessment, Development, and Evaluation (GRADE)

Expert opinions were solicited in cases of insufficient data to make definitive conclusions. In addition, specialists representing the Korean Society for Transplantation were invited to participate as external consultants, regarding the recommendations on hepatitis B in transplant settings. Each member of the committee was responsible for collecting, analyzing, and preparing the manuscript in their respective fields.

Health care utilization varies depending on race, region, institution, and economic conditions. The presented guidelines can differ from other regional guidelines as it reflected our unique medical conditions and research results. It is intended to provide practical and updated information for management of CHB patients. However, as the guidelines do not represent a standard treatment protocol, clinicians should keep in mind that the best management might vary by individual patient setting. The KASL will continue to update part or all of these guidelines based on publication of new study results. Thus, revision of the guidelines is deemed necessary for promoting the health of Korean CHB patients.

NATURAL HISTORY

CHB is defined as persistence of serum hepatitis B surface antigen (HBsAg) for more than six months. The natural course is divided into five clinical phases: immune-tolerant, hepatitis B e antigen (HBeAg)-positive immune-active, immune-inactive, HBeAg-negative immune-active, and HBsAg loss or resolved HBV infection (Table 2). The duration of these clinical phases can vary, and the sequences of phases might not be continuous in the patient. In addition, there can be grey zone in which the features do not correspond to any specific phase. Therefore, it is often insufficient to determine the clinical stage of infection or to decide antiviral treatment based on a single alanine aminotransferase (ALT) or HBV DNA test (Fig. 1) [1].

Natural course of chronic hepatitis B

Figure 1.

Natural course of chronic hepatitis B (CHB). HBV, hepatitis B virus; HBsAg, hepatitis B surface antigen; anti-HBs, antibody to HBsAg; HBeAg, hepatitis B e antigen; anti-HBe, antibody to HBeAg; ALT, alanine aminotransferase.

Immunological features of CHB during the natural course

CHB, immune-tolerant phase

The immune-tolerant phase is generally related to vertical transmission and is characterized by HBeAg positivity, very high level of serum HBV DNA (generally ≥107 IU/mL), persistently normal level of ALT, and minimal or absence of hepatic necroinflammation [2,3]. In a follow-up of immune-tolerant CHB patients, serum ALT was elevated in 16% of cases, and the follow-up fibrosis stage was not different from the initial stage in those who remained immune-tolerant for 5 years [2]. In another study from Taiwan, 5% of 240 immune-tolerant CHB patients progressed to cirrhosis and did not develop HCC in 10 years of follow-up [4]. In a Korean multi-center study, 1% of 946 immune-tolerant CHB patients progressed to cirrhosis, and 1.7% developed HCC during 10 years of follow-up [5].

However, a small-scale in vitro study suggested that early hepatocarcinogenesis could be progressing even during the immune-tolerant phase, as was evident by a high level of HBV DNA integration and clonal hepatocyte expansion in liver tissues from immune-tolerant CHB patients as with immune-active CHB patients [6]. Recently, several studies reported that development of HCC was not uncommon in the immune-tolerant phase, although the studies included patients with serum HBV DNA <107 IU/mL, serum ALT >upper limit of normal (ULN), or high fibrosis index [7], necessitating further studies to clarify the criteria for discriminating patients in “true” immune-tolerant phase.

The immune-tolerant phase can last for more than three decades in patients infected with HBV genotype C due to late HBeAg seroconversion [8]. Therefore, many female patients infected with this genotype are in the immune-tolerant phase when they are of childbearing age, which can lead to vertical transmission of HBV to a child [9].

HBeAg-positive CHB, immune-active phase

With increasing age, most patients in the immune-tolerant phase experience immune responses to HBV. Such changes are due to increased response of cytotoxic T lymphocytes to hepatitis B core antigen (HBcAg) and HBeAg [10], resulting in destruction of infected hepatocytes. This phase is characterized by HBeAg positivity and fluctuating courses of serum ALT and HBV DNA levels [11,12]. Histological findings reveal moderate-to-severe necroinflammation [13]. There can be various stages of liver fibrosis according to severity of liver injury.

Once HBeAg seroconversion occurs, the natural course of the disease can have one of three clinical features: (1) repeated HBeAg reversion and seroconversion, (2) immune-inactive phase, or (3) HBeAg-negative, immune-active phase of CHB [14,15]. Typically, 10–40% of patients who experience seroconversion revert to an HBeAg-positive state and then experience recurrence of seroconversion at least once with progression of hepatitis activity [16,17]. In particular, reversion frequently occurs in patients with HBV genotype C, and the rate decreases with age [9]. Hepatic decompensation, which occurs in 5% of patients with acute exacerbation, can be fatal [18].

CHB, immune-inactive phase

Most patients who seroconvert during the immune-active phase progress to the immune-inactive phase, which is characterized by HBeAg negativity, antibody to HBeAg (anti-HBe) positivity, persistent normal ALT level, and HBV DNA level below 2,000 IU/mL [19-21]. Typical histological findings in the third phase are mild liver inflammation [19], and various stages of liver fibrosis can reflect previous liver injury [22].

This phase persists for an extended period in most patients but has a relatively good prognosis. However, an estimated 20% of such patients will revert to the HBeAg-negative or HBeAg-positive immune-active phase and can experience recurring periods of reactivation and inactivation throughout their lives, which can lead to cirrhosis or HCC [23,24].

HBeAg-negative CHB, immune-active phase

Approximately 20% of patients who experience HBeAg seroconversion during their immune-active HBeAg-positive phase progress to the immune-active HBeAg-negative phase, with HBV DNA level ≥2,000 IU/mL, increased ALT level, and active necroinflammation of the liver [14]. These patients show HBeAg-negativity because they harbor HBV variants in the precore (PC) or basal core promoter (BCP) regions of HBV DNA, resulting in failure or reduction of HBeAg production [25-28]. The immune-active HBeAg-negative phase is associated with older age and lower rates of prolonged spontaneous disease remission, and most patients in this phase will experience persistent hepatocellular inflammation and progress to hepatic fibrosis and cirrhosis [27,29,30]. Severe fluctuations of HBV DNA and ALT levels can make it difficult to differentiate these patients from those in the immune-inactive phase [31]. Therefore, HBV DNA and ALT levels should be monitored in every 3 months for at least 1 year in case of immune-inactive phase to find out the HBeAg-negative immune-active phase requiring antiviral treatment [32,33].

HBsAg loss phase

During the natural course of CHB, HBsAg loss is a very rare transition that indicates potential cure of HBV infection (Table 3) [34-37]. Complete cure, or “sterilizing cure,” of HBV infection implies seroclearance of HBsAg and HBV DNA as well as complete clearance of intrahepatic covalently closed circular DNA (cccDNA) and/or integrated HBV DNA. However, it is difficult to achieve these goals at present [37]. Accordingly, the realistic goal suggested is a “functional cure,” which refers to seroclearance of HBsAg and HBV DNA regardless of antibody to HBsAg (anti-HBs) [37]. Despite the presence of intrahepatic cccDNA and/or integrated HBV DNA, it is a successful immunological control state of CHB and therefore can be viewed as a concept similar to idealistic functional cure [36]. In certain circumstances, such as immunosuppression, the risk of HBV reactivation persists [38].

Definitions of HBV cure

Patients in the immune-inactive phase subsequently progress to HBsAg loss or clearance phase at a rate of 1–2% annually [31,39,40]. According to Liaw’s prospective data, HBsAg loss occurs in 0.5% of CHB patients per year and 0.8% of asymptomatic chronic HBV carriers per year [35]. Korean patients reportedly experience a relatively low rate of HBsAg loss (0.4% annually) [41]. In a few patients, serum HBV DNA can be detected at a very low titer during this phase [42,43]. HBsAg loss is the state of functional cure and is associated with a reduced risk of cirrhosis. However, significant risk of HCC development remains even after HBsAg loss in male patients and in settings where HBsAg loss has been achieved late (presence of cirrhosis or age ≥50 years) [42,44].

HBsAg seroclearance is also achieved in a few patients on antiviral treatment, and the long-term clinical outcome compared with that of spontaneous HBsAg seroclearance is unclear [45,46]. Two studies showed no difference in HCC incidence between patients who achieved treatment-induced and spontaneous HBsAg seroclearance [47,48], yet another study reported a 5-year cumulative incidence of HCC of 0.9% and 3.9% in spontaneous and treatment-induced HBsAg seroclearance patients, respectively [49].

Grey zone

The natural course of CHB is divided into five phases with regard to clinical indicators such as ALT and HBV DNA – immune-tolerant, HBeAg-positive immune-active, immune-inactive, HBeAg-negative immune-active, and HBsAg loss. However, patients who do not fit into any of the usual clinical phases are considered to be in the “grey zone.” Approximately 30% of CHB patients are in the grey zone [50-52], and the prognosis and treatment of CHB patients in the grey zone are under study [52-57].

The decision whether to initiate antiviral therapy for patients in the grey zone is a clinically challenging question, and it is often difficult to make an accurate decision without the results of a liver biopsy. Treatment of patients in the grey zone is summarized in the ‘Treatment Indication’ session.

Risk factors that influence the natural history and progression of CHB

In the natural course of CHB, the cumulative incidences of cirrhosis and HCC were approximately 8–20%/year and 2–5%/year [58,59]. In Korea, the reported annual and 5-year accumulated incidences of cirrhosis are 5.1% and 23%, respectively, while those for HCC are 0.8% and 3% [60]. The risk factors for CHB progressing to cirrhosis or HCC can be divided into host, viral, and social-environmental factors (Table 4). For host factors, cirrhosis, persistent necroinflammation, old age, male gender, family history of HCC, and co-infection of other hepatitis virus or human immunodeficiency virus (HIV) affect the risk. High level of serum HBV DNA and/or serum HBsAg, HBV genotype C, and specific genotypic mutations are included as viral factors [61-67]. Social-environmental factors for progression to cirrhosis or HCC include alcohol consumption, metabolic syndrome, diabetes, obesity, and smoking [68-70]. In contrast, coffee [71-73], metformin [74], aspirin [75,76], and statins [77-84] exert protective effects against development of HCC.

Factors associated with development of liver cirrhosis and hepatocellular carcinoma in persons with chronic hepatitis B

Multiple prognostic prediction models have been developed to estimate the risk of HCC in CHB patients (Table 5). The REACH-B (risk estimation for HCC in chronic hepatitis B) model, which consists of gender, age, serum ALT, HBeAg, and serum HBV DNA level, has been developed for HCC risk prediction in non-cirrhotic, treatment-naïve CHB patients. REACH-B model has been validated in Hong Kong and Korean cohorts of CHB patients including those with liver cirrhosis. Areas under the receiver operating characteristic curve for HCC prediction at 3 years, 5 years, and 10 years are 0.77–0.81 in those cohort [85]. Modified REACH-B (mREACH-B) model, which substituted serum HBV DNA for the liver stiffness value from the original REACH-B model, showed better outcomes in assessment of 3-year and 5-year HCC prediction in several prospective Korean studies [86,87]. Meanwhile, PAGE-B (platelets, age, gender, and hepatitis B) model, which was developed from Western studies [88], has been validated by several Korean retrospective studies [89,90]. Modified PAGE-B (adding serum albumin) was superior to the original PAGE-B in prediction of 5-year HCC risk in Korean CHB patients [90-92]. Although THRI (Toronto HCC risk index) has shown good predictive ability for HCC in patients with cirrhosis [93], it was not superior to mPAGE-B in Korean CHB patients [91]. Meanwhile, the CAGE-B (cirrhosis and age) and SAGE-B (stiffness and age) models, which were developed from Western studies [94], has been validated by several Korean retrospective studies [95,96]. A recently developed FSAC (fibrosis marker response, sex, age, cirrhosis) model that incorporates on-therapy changes in non-invasive fibrosis markers (fibrosis-4 [FIB-4] or aspartate aminotransferase-to-platelet ratio index [APRI]) has been validated in a separate cohort of Korean CHB patients [97].

HCC risk prediction models

Recently, deep learning or artificial intelligence-assisted prediction models were developed to predict the risk of HCC in patients with CHB on antiviral treatment, and these models demonstrated superior performance in risk stratification compared with previous risk scores [98,99].

Summary

1. The natural course of CHB is divided into five phases – immune-tolerant, HBeAg-positive immune-active, immuneinactive, HBeAg-negative immune-active, and HBsAg loss. Studies on the prognosis and treatment of grey zones that are not classified by clinical indicators, such as ALT and HBV DNA, are in progress.

2. Several models have recently been developed to predict the risk of HCC in CHB patients using clinical parameters, artificial intelligence, and deep-learning technology. There are increasing efforts to apply these models to clinical care in patients receiving antiviral treatment.

EMERGING MARKERS OF HBV INFECTION

HBV, the Dane particle, has a diameter of approximately 44 nm and consists of a double-layered capsid particle enclosing a circular, incomplete double-stranded DNA genome (3.2 kb in length). HBV has been classified into eight genotypes (A to H) by a divergence >8% in the entire genomic sequence. The distribution of HBV genotypes differs between regions and ethnicity. Genotype C, especially the C2 subgenotype, prevails predominantly among chronic HBV carriers in Korea [100,101]. Genotype C HBV infection is an independent risk factor for HCC development in addition to liver cirrhosis [102,103]. Patients infected with HBV genotype C tended to have persistently positive HBeAg or fluctuating HBeAg status.

The structure of HBV consists of an external surface protein and an internal core protein. The antigenicity to each protein is called HBsAg and HBcAg, respectively. There are four known genes encoded by the genome, called C, X, P, and S. Gene S codes for the HBsAg. The core protein (HBcAg) is coded for by gene C, and its start codon is preceded by an upstream in-frame AUG start codon from which the pre-core protein is produced. HBeAg is produced by proteolytic processing of the pre-core protein. HBsAg and HBcAg can be measured in peripheral blood because they circulate in serum. Otherwise, HBcAg is an intracellular antigen that is expressed in infected hepatocytes. It is not detectable in serum with available commercial assays. Antibodies for each antigen, such as anti-HBs, anti-HBc, and anti-HBe, can be detected by serological tests.

In addition to traditional tests above, novel virological markers have been developed to predict prognosis, treatment response, and off-treatment viral suppression in patients infected with HBV (Table 6, Fig. 2) [104].

Role of emerging HBV markers

Figure 2.

Emerging markers for chronic hepatitis B infection. rcDNA, relaxed circular DNA; pgRNA, pregenomic RNA; p22Cr, p22 core-related antigen; HBcAg, hepatitis B core antigen; HBeAg, hepatitis B e antigen; HBsAg, hepatitis B surface antigen; HBcrAg, hepatitis B core-related antigen; dslDNA, double stranded linear DNA; cccDNA, circular covalently closed DNA; HBV, hepatitis B virus.

Serum marker

Quantitation of HBsAg

The HBV encodes the three proteins of the HBsAg, which form the viral envelope, small (SHBsAg), middle (MHBsAg), and large (LHBsAg). These proteins are translated from two HBV subgenomic mRNA transcripts, the preS1 mRNA and the preS2/S mRNA. HBsAg is not only generated by transcription and translation of cccDNA, but also can be generated from HBV DNA episomally integrated into the host genome. HBsAg is assembled with core proteins and DNA polymerases during the viral replication process to form a complete virion (Dane particle). The surface proteins also generate non-infectious excess sub-viral particles, approximately 22 nm in diameter, with either a spherical or a long filamentous form. The quantitation of serum HBsAg is a method of detecting all three forms. There is a positive correlation between levels of serum HBsAg and intrahepatic cccDNA in patients with CHB.

Levels of HBsAg helps to distinguish inactive carriers from CHB patients. In HBeAg-negative patients, one-time measurements of serum HBV DNA <2,000 IU/mL and HBsAg <1,000 IU/mL suggest future inactive carriers [105,106]. In contrast, among HBeAg-negative patients with lower viral load (HBV DNA <2,000 IU/mL), HCC risk is higher in those with a high HBsAg titer (>1,000 IU/mL) than in those with a low HBsAg titer [107]. Levels of HBsAg can predict response during pegylated interferon (peginterferon) therapy in HBeAg-positive patients, possibly providing a guide to stopping treatment earlier [108]. For nucleos(t)ide analogue (NA) treatment, levels of HBsAg has emerged as a valuable tool in identifying patients who will maintain or attain inactive status or develop HBsAg loss if they discontinue long-term NA therapy [109-111].

Hepatitis B core-related antigen (HBcrAg)

HBcrAg is a composite biomarker incorporating several viral antigens expressed from the pre-core/core gene: the HBcAg, HBeAg, and p22 core-related antigen (p22Cr) [112]. Although it is not yet commonly used in Korea, HBcrAg can be easily detected using a chemiluminescent immunoassay kit. Significant positive correlations between serum HBcrAg level and HBV DNA level, as well as amount of intrahepatic cccDNA, have been observed in several Asian and European studies [113]. Serum HBcrAg level varies significantly among the phases of HBV infection. HBcrAg is, therefore, also a good virologic marker to differentiate HBeAg-negative CHB patients (active disease) from HBeAg-negative chronic HBV infection (inactive disease) [114]. Level of HBcrAg can potentially predict the possibility of achieving partial cure in patients on antiviral treatment, as defined by a sustained off-therapy virological control. In the study of 130 Hong Kong patients with undetectable serum HBV DNA during NA, HBcrAg was detectable in 101 (78%) samples [115]. After eight years of NA treatment, 21.3% of patients achieved serum HBcrAg <3 log10 U/mL [116]. Based on these findings, monitoring of HBcrAg and HBsAg quantification is recommended by the Japan Society of Hepatology guidelines to identify patients who can discontinue NA [117]. Furthermore, HBcrAg can be an aid for clinicians in identifying patients with a higher risk of HCC development or post-treatment recurrence [118,119]. Recently, the HBcrAg level has been the most emerging noninvasive predictor reflecting intrahepatic viral replication.

HBV RNA

Serum HBV RNA represents partially reverse-transcribed, encapsidated pregenomic RNA (pgRNA) in virus-like particles. Given that pgRNA is transcribed directly from cccDNA, level of serum HBV RNA can potentially serve as a surrogate marker for transcriptionally active cccDNA [120]. Serum HBV RNA may be an indicator for predicting off-treatment response in patients with suppressed HBV under NA treatment [121,122], although it has limited additional value in differentiating CHB phases [123]. Measurement of HBV RNA could be used to determine cccDNA activity, especially with therapies in development that aim for functional cure [119,124]. Levels of HBV RNA is also associated with increased HCC risk and post-resection recurrence in NA-treated patients [125,126]. Before a widespread clinical application of serum HBV RNA for CHB can be accepted and applied, the methodology for detecting and quantifying serum HBV RNA should be standardized.

Intracellular marker

HBV cccDNA

In the nucleus of hepatocytes, cccDNA is maintained as a stable mini-chromosome and acts as the template for pgRNA transcription and viral protein production [127]. The existence of intrahepatic cccDNA is the molecular basis of disease chronicity and represents a major barrier to HBV eradication. cccDNA is responsible for persistence of the virus in the liver even after HBsAg loss and seroconversion. Occult HBV infection is a state in which HBsAg is not detected in serum, but HBV DNA is present in liver tissue or blood, which is believed to be due to cccDNA remaining in the nucleus. cccDNA also causes hepatitis B reactivation in patients receiving immunosuppressants or chemotherapy [128]. In terms of HBV treatment, current widely used NAs can block the transcription process of RNA production from cccDNA but not remove cccDNA itself. Therefore, cccDNA acts as a major cause of HBV reactivation after stopping the NA, and the development of drugs that can remove cccDNA itself is essential to cure hepatitis B [129]. In order to quantify cccDNA in the liver, invasive liver biopsy is required, making it difficult to use as a marker in clinical settings.

HBV integration into the host genome

HBV can integrate in human DNA and promote hepatocarcinogenesis by insertional mutagenesis, increased genomic instability, or expression of viral oncoproteins such as the protein HBx [130,131]. The application of next-generation sequencing led to characterization of HBV integration events. Recent studies reported that, among patients with primary liver cancer, a higher number of HBV integration events was associated with worse outcome [130,132-134]. In addition, cell-free HBV-integrated tumor DNA was shown to be a circulating biomarker for detecting tumor load in a majority of patients with HBV-related HCC and aided in monitoring residual tumor and recurrence clonality after tumor resection [135,136].

HBV pgRNA

Among HBV mRNAs, 3.5-kb preC and pgRNA originate from HBV cccDNA, and pgRNA is reverse transcribed to enable the synthesis of HBV DNA. Intrahepatic pgRNA is considered a marker of active HBV replication. RNA interference (RNAi) therapeutics such as siRNA and anti-sense molecules targeting pgRNA are being developed. However, the role of intrahepatic pgRNA as a marker for treatment response has recently been replaced by serum HBV RNA measurements.

Summary

1. Serum HBsAg, HBcrAg, and HBV RNA measurements can be valuable indicators in stratifying CHB phases, determining suitability for NA cessation, and predicting off-treatment response.

2. Intracellular markers of HBV such as cccDNA and pgRNA can be effective targets for drug development for HBV cure.

TREATMENT INDICATION

Active HBV replication is associated with increased risk of liver damage, progression of liver disease, and liver-related complications [22]. Antiviral therapy can effectively inhibit replication of the virus [137]. Inhibition of HBV replication by antiviral therapy can improve hepatic inflammation, normalize serum ALT level, improve liver fibrosis, reduce the incidence of HCC, and decrease liver-related death [138].

However, currently available antiviral therapies cannot eradicate or eliminate the virus. Furthermore, the efficacy and side effects of the same drug can vary depending on the clinical situation [137]. Therefore, benefits and risks of antiviral therapy should be carefully evaluated on an individual basis. The following three factors are fundamental components that should be taken into consideration when deciding antiviral therapy: 1) the severity of liver disease, 2) the degree of HBV replication, and 3) the presence of liver injury (Fig. 3). The severity of liver disease can be categorized into chronic hepatitis, compensated cirrhosis, and decompensated cirrhosis according to the degree of liver fibrosis. Severity of liver fibrosis can be evaluated by liver biopsy or non-invasive methods using serum markers (e.g., APRI, FIB-4 index, M2BPGi) [118,139] or transient elastography (TE) using Fibroscan® (Echosense, Paris, France) [140,141]. In general, greater than F2 fibrosis in liver biopsy is considered significant [142], and the diagnostic cut-off for F2 is 7.8 kPa in Fibroscan® in a meta-analysis from Korea [140]. The degree of HBV replication can be assessed by measuring serum HBV DNA level. The presence of liver injury can be estimated using serum ALT level or can be assessed by a liver biopsy. When it is accompanied by inflammation greater than grade A2–A3 in liver biopsy, it is defined as moderate or more severe inflammation [142].

Figure 3.

Algorithm for management of chronic hepatitis B virus infection. HBsAg, hepatitis B surface antigen; HBeAg, hepatitis B e antigen; HBV, hepatitis B virus; AST, aspartate aminotransferase; ALT, alanine aminotransferase; anti-HBe, hepatitis B e antibody. *Serum HBV DNA ≥107 IU/mL. An upper limit of normal (ULN) for ALT of 34 IU/L for males and 30 IU/L for females. Serum HBV DNA ≥20,000 IU/mL for HBeAg-positive patients and HBV DNA ≥2,000 IU/mL for HBeAg-negative patients.

CHB, immune-tolerant phase

The immune-tolerant phase is characterized by HBeAg positivity, very high serum HBV DNA level (usually ≥107 IU/mL), and persistently normal serum ALT level. In this phase, long-term prognosis is excellent without antiviral therapy [143-145]. In a long-term observation of CHB immune-tolerant patients in Korea, the incidence of HCC at 10 years was very low (1.7–2.7%) and not higher than that of patients in the immune-active phase who received antiviral treatment [5,146,147]. To verify the immune-tolerant phase, a liver biopsy is necessary and will show no or mild inflammation without fibrosis. However, liver biopsy is an invasive procedure with potential complications that limit its widespread use and repetitive testing in clinical practice. Hence, in real-life clinical practice, a combination of clinical findings (HBeAg positivity, high serum HBV DNA level, normal ALT level, and no evidence of cirrhosis) is typically used to define the immune-tolerant phase without liver biopsy. However, caution should be exercised considering the results of a recent study suggesting that HCC and liver cirrhosis-related complications occur in a considerable number of such patients during long-term follow-up [7]. In the study, the rate of HCC at 10 years was high (12.7%) compared to another study, although enrolled patients showed HBeAg positivity, HBV DNA (>20,000 IU/mL), normal ALT level, and no evidence of cirrhosis [7].

In recent studies, older age, male, relatively low serum HBV DNA level [7,148], high liver stiffness value [147,149], and normal but high-normal ALT level were associated with HCC development or liver-related complications among patients presumed to be in the immune-tolerant phase by combinations of clinical findings without a liver biopsy [7].

The immune-tolerant phase is usually characterized by little or no necroinflammation without liver fibrosis, but significant fibrosis as seen using non-invasive serum fibrosis markers (e.g., APRI, FIB-4, M2BPGi) or TE (Fibroscan®) suggests that antiviral treatment can be considered. The immune-tolerant phase is usually observed in young adults and is not common in elderly patients as immune reaction against HBV onsets with age. Indeed, elderly patients in the presumed immune-tolerant phase showed higher possibility of significant fibrosis (≥F2) or necroinflammation (≥A2) in liver biopsy [150], and increased risks of HCC development or liver-related complications [7,148]. Therefore, even when all the other clinical findings suggest the immune-tolerant phase, a liver biopsy can be considered to verify the immune-tolerant phase in older adults. An age cut-off for liver biopsy consideration was suggested to be 30–40 years [144,151], but evidence to support this approach is limited. Also, non-invasive serum fibrosis markers (e.g., APRI, FIB-4, M2BPGi) or TE (Fibroscan®) can be used to evaluate the degree of hepatic fibrosis and is helpful in distinguishing patients with genuine immune tolerance. Among CHB patients without clinical cirrhosis, those with a higher liver stiffness value (≥13 kPa) showed a higher rate of HCC than those with a lower value (<13 kPa) [149]. In contrast, the cumulative probability of HCC at 5 years was negligible in the immune-tolerant group stringently defined by a low FIB-4 index (<1.45) [147].

The immune-tolerant phase is also characterized by very high level of HBV DNA, as there is little or minimal immune response to the virus [144,151]. In one study, among patients with HBeAg positivity and normal ALT level, relatively low serum HBV DNA level (<107 IU/mL) was associated with a higher risk of HCC and death compared to those with very high serum HBV DNA level (≥107 IU/mL) [7,148]. Relatively low serum HBV DNA level indicates that the immune response has already begun to suppress the virus. In a recent study about treatment-naïve CHB patients with ALT <2 times the ULN, HBV DNA >106 IU/mL, HBeAg positivity, and HCC risk was highest with baseline HBV DNA level of 106–7 IU/mL compared to HBV DNA level greater than 107 IU/mL [152].

ALT is a good indicator of liver necroinflammation, so patients in the immune-tolerant phase show persistently normal ALT level, as there is no or little liver necroinflammation. Patients with slightly elevated ALT level are more likely to have fibrosis and necroinflammation on a liver biopsy and have a higher risk of complications during follow-up [148,150]. Therefore, ALT at the borderline of or slightly higher than ULN is a sign that a patient is not genuinely in the immune-tolerant phase. However, careful interpretation is needed in defining normal or elevated ALT level. There is controversy about what constitutes healthy, normal ALT level. Elevation of ALT can be caused by obesity and other conditions not related to HBV. Recently, the cut-off level for ALT associated with increased liver-related mortality among Korean chronic HBV-infected patients was reported to be 34 IU/L for men and 30 IU/L for women [153]. Therefore, the present guidelines recommend using these values to define normal ALT level.

The efficacy of currently available antiviral regimens is limited for patients in the immune-tolerant phase. Antiviral treatment using NAs resulted in a poor antiviral response rate and a low HBeAg seroclearance rate [154]. Furthermore, when NA treatment was discontinued for those who started oral NA therapy at the immune-tolerant phase, all patients showed a rebound of serum HBV DNA level above 2,000 IU/mL, 70% showed an elevation of ALT level, and 55% had to re-start NA therapy [155]. Long-term treatment might be necessary, and treatment discontinuation can be difficult. However, in one study from Korea that compared 87 NAtreated immune-tolerant CHB patients to 397 monitored immunetolerant patients as a control group, increased risk of HCC and cirrhosis was observed in the control group despite favorable baseline liver function [156]. In a recent Korean analysis about immune-tolerant phase patients based on several studies, starting antiviral therapy in this phase is cost-effective compared with delaying treatment until the active phase in CHB patients, especially considering increasing HCC risk, decreasing drug costs, and consideration of productivity loss [157]. This finding suggests that some patients who are presumed to be in the immune-tolerant phase will develop complications during follow-up, and that antiviral treatment can decrease this risk. Further studies are needed to identify appropriate antiviral treatment indications in patients in the immune-tolerant phase.

[Recommendations]

1. CHB patients in the immune-tolerant phase, as defined by HBeAg positivity, very high serum HBV DNA level (≥107 IU/mL), persistently normal ALT level, and no inflammation or fibrosis on liver biopsy, should be monitored without antiviral therapy (B1).

2. Evaluation of the degree of liver fibrosis using non-invasive fibrosis tests or liver biopsy among presumed immunetolerant phase patients with normal ALT level is suggested if the patient’s age is ≥30–40 years, serum HBV DNA level is <107 IU/mL, or ALT level is approaching the borderline of ULN, and antiviral therapy can be considered for patients with significant liver fibrosis (B2).

HBeAg-positive and HBeAg-negative CHB, immune-active phase

The immune-active phase is characterized by active replication of HBV and moderate or severe necroinflammation with or without fibrosis. A systematic review and meta-analysis of 15 randomized controlled trials and 44 observational studies showed that antiviral treatment in the immune-active phase reduced the risk of cirrhosis, hepatic decompensation, and HCC [138]. Therefore, patients in the immune-active phase are indicated for antiviral treatment. Nevertheless, careful attention to HCC development is needed as antiviral treatment cannot completely eliminate the risk of HCC [158]. A recent study from Korea reported a marked reduction in liver disease mortality by widespread use of antiviral treatments against HBV but paradoxical increased burden of liver cancer [159].

Active replication of HBV can be confirmed by serum HBV DNA measurement using polymerase chain reaction. Detection of HBV DNA in the serum indicates active replication of the virus. However, the lower limit of detection is different among HBV DNA assays. Moreover, many patients with low-level viremia (serum HBV DNA level <2,000 IU/mL) show normal ALT level, little or no necroinflammation or fibrosis on liver biopsy, and favorable outcomes without antiviral therapy [68]. Hence, not all patients with detectable serum HBV DNA, but patients with serum HBV DNA level ≥2,000–20,000 IU/mL for HBeAg-positive patients and serum HBV DNA level ≥2,000 IU/mL for HBeAg-negative patients are considered for antiviral therapy [56,68,70].

Serum ALT is a convenient indicator of necroinflammation of the liver and can be easily used in clinical practice [160]. Elevation of ALT suggests hepatocellular injury and requires assessment and evaluation. However, the degree of ALT elevation does not always correlate with necroinflammation of the liver and can be affected by body mass index and gender [161,162], alcohol use, drug use, fatty liver, and other causes unrelated to HBV [162,163]. A normal ALT level does not exclude significant liver disease [164]. Hence, the use of ALT as a criterion for treatment initiation requires consideration of the threshold of elevation. If the ALT level is elevated more than ≥2 times the ULN, antiviral treatment for HBV is recommended unless the increase is due to other causes [144,151]. When ALT is elevated above but <2 times the ULN, controversy exists as to whether these patients require antiviral treatment [144,151]. Patients with serum ALT elevated above but <2 times the ULN have increased risk of liver cirrhosis and HCC compared to patients with serum ALT within the normal range [165,166]. Yet, “normal” ALT level varies among studies and by ethnicity [162,167]. The specific ALT levels used in clinical trials to initiate antiviral therapy also differ [168-173]. Therefore, sufficient data are not available to judge whether it is necessary to start antiviral treatment in patients with serum ALT elevated above but <2 times the ULN. In this case, trends in serum ALT and HBV DNA levels should be closely monitored to identify possible causes and to verify whether treatment for such patients should be initiated (Fig. 3). If a patient shows persistently elevated ALT level <2 times the ULN, the degree of fibrosis can be further investigated by non-invasive fibrosis tests or by liver biopsy to verify whether patients require antiviral treatment due to significant fibrosis.

Histological assessment of the liver, liver biopsy, is a cornerstone in the evaluation of hepatic necroinflammation and fibrosis[174]. Findings of moderate to severe necroinflammation or significant fibrosis (≥F2) indicate that antiviral treatment for HBV is needed [137]. However, a liver biopsy is an invasive procedure requiring special resources that limit widespread clinical use. Serum fibrosis biomarkers or TE (Fibroscan®) of the liver are alternatives that can be used to estimate degree of fibrosis [175]. These non-invasive biomarkers for liver fibrosis are less accurate than liver biopsy but can be used to rule in or rule out patients with significant fibrosis. HBeAg, HBV DNA concentration, and ALT level have been traditionally used to determine starting antiviral therapy, and liver biopsy was used in a case of difficult decision for antiviral therapy [176]. Recently, treatment initiation based on liver disease severity as assessed by non-invasive tests (e.g., Fibroscan®) has been suggested [175]. However, more evidence is needed for cut-off levels of non-invasive tests to support treatment initiation.

Among HBeAg-positive CHB patients, spontaneous HBeAg seroconversion has been reported for those experiencing increase of ALT level with HBV DNA elevation. Hence, 3–6 months of observation without antiviral treatment can be considered if spontaneous HBeAg seroconversion is expected [176]. However, biochemical deterioration leading to liver failure is of concern. A prospective cohort study of 90 patients from Korea with HBeAg-positive CHB who were monitored without antiviral therapy showed a very low rate of spontaneous HBeAg seroconversion (1.1%), while there was frequent biochemical deterioration and one case of liver transplantation due to liver failure [177]. Therefore, when expecting HBeAg seroconversion, the risk of acute decompensation leading to liver failure warrants careful attention. Another report from Korea showed that spontaneous HBeAg seroconversion can be expected for patients with non-vertical transmission and low serum HBV DNA level [178].

CHB patients can present with severe acute exacerbation, characterized by elevated HBV DNA level, serum ALT level 5–10 times greater than ULN, jaundice, coagulopathy, ascites, and/or hepatic encephalopathy. They can also be classified as having acute-on-chronic liver failure (ACLF) when they present with symptoms and signs of liver failure [179]. Severe acute exacerbation can occur spontaneously [180], by drug-resistant HBV during antiviral therapy [181], with cessation of antiviral therapy [182], or with anticancer chemotherapy [183]. NA therapy reduces mortality in patients with severe reactivation of CHB presenting as ACLF [184]. Therefore, immediate antiviral treatment is recommended for CHB patients with severe acute exacerbation or ACLF [176]. Some studies have reported a higher mortality rate among entecavir-treated patients than lamivudine-treated patients [185,186], but a meta-analysis of three prospective and eight retrospective studies showed similar effects on the mortality rate between entecavir and lamivudine treatment, with a more favorable long-term outcome in entecavir [179]. However, antiviral treatment cannot fully prevent progression to liver failure, which can lead to mortality in the case of high Model for End-stage Liver Disease (MELD) score, moderate to severe ascites, and/or aggravation of hepatic encephalopathy [187-189], Emergent liver transplantation should be considered and prepared. Steroid or plasma exchange has been suggested in cases of severe acute exacerbation and ACLF, but data are currently limited to a small number of cases [190,191].

Some HBeAg-negative CHB patients show normal or mildly elevated ALT level despite elevated HBV DNA level (>2,000 IU/mL). Some patients progress to the immune-inactive phase spontaneously—especially patients with low HBsAg level and low serum HBV DNA level [192]. In a recent study about patients with HBeAg negativity and replicative HBV DNA ≥2,000 IU/mL, according to ALT level (persistently normal ALT, ALT 1–2 times the ULN, ALT >2 times the ULN), untreated patients with ALT 1–2 times the ULN had higher risks of HCC and death/transplantation than antiviral-treated patients with ALT >2 times the ULN [54]. HBeAg-negative patients are those who have experienced the prior immune-active phase, and there is possibility that various degrees of fibrosis remain in these patients. For those with advanced fibrosis, antiviral treatment can be considered for those with elevated HBV DNA level regardless of ALT level [138,151]. Hence, HBeAg-negative CHB patients showing elevated HBV DNA level (>2,000 IU/mL) but normal or mildly elevated ALT level require careful evaluation of their degree of fibrosis to decide if they should undergo antiviral treatment or monitoring.

[Recommendations]

1. Antiviral therapy is recommended in HBeAg-positive CHB patients with HBV DNA ≥20,000 IU/mL and in HBeAgnegative CHB patients with HBV DNA ≥2,000 IU/mL if serum ALT level is ≥2 times the ULN (A1). In cases where ALT is 1–2 times the ULN, close ALT monitoring or liver biopsy should be considered. Antiviral therapy is recommended if liver biopsy reveals moderate to severe necroinflammation or significant fibrosis (≥F2) (A1), which can be assessed by non-invasive fibrosis tests (B1).

2. In patients with HBeAg-positive or HBeAg-negative CHB, prompt antiviral therapy should be initiated in cases of acute exacerbation, with elevation of ALT ≥5–10 times the ULN and signs of liver failure such as jaundice, prothrombin time prolongation, ascites, or hepatic encephalopathy (A1).

3. In HBeAg-negative CHB patients with HBV DNA ≥2,000 IU/mL and normal ALT level, follow-up can be considered. Otherwise, liver biopsy or non-invasive fibrosis tests can be considered for assessment of the degree of necroinflammation and/or fibrosis to determine whether treatment is needed (B2).

CHB, immune-inactive phase

The immune-inactive phase is characterized by HBeAg-negative, anti-HBe-positive, persistently normal ALT level, and undetectable or low (<2,000 IU/mL) serum HBV DNA level. In this phase, long-term outcome without antiviral treatment is good for those without advanced fibrosis [68]. In contrast, risk of HCC is not low for these patients with high FIB-4 or APRI index suggesting advanced liver fibrosis [52,54,193,194]. The immune-inactive phase is a dynamic phase that can reactivate to an immune-active phase [14]. Hence, patients in the immune-inactive phase require careful assessment of the degree of fibrosis and close monitoring of serum ALT and HBV DNA levels to verify whether they remain in the immune-inactive phase.

HBsAg loss or seroclearance is observed in 1–2% of patients per year in the immune-inactive phase [39,41]. HBsAg seroclearance is considered a surrogate endpoint for a functional cure of CHB. Hence, several studies investigated whether antiviral therapy in the immune-inactive phase can further induce HBsAg seroclearance. In a study about immune-inactive phase patients with HBsAg <1,000 IU/mL, HBsAg loss was achieved in 44.7% of 102 patients treated with peginterferon or peginterferon/adefovir compared to only 2.4% of 42 patients without treatment over 96 weeks. This study suggested that antiviral therapy in immune-inactive phase patients with low HBsAg titer could be considered for HBsAg loss [195]. However, in a randomized prospective controlled study for 48 weeks, when 151 immune-inactive phase patients irrespective of HBsAg titer were randomized into two groups, the treatment group with peginterferon/adefovir or peginterferon/tenofovir (DF or AF) and the no-treatment group, the rate of HBsAg loss was not different between the two groups at 72 weeks (4% vs. 0%, respectively) [196]. This suggests that the benefit of antiviral therapy is limited in immune-inactive phase patients, who have low risk for HCC or liver-related complications during follow-up. The clinical benefit of inducing HBsAg loss by antiviral treatment in the immune-inactive phase, in terms of achieving treatment goals for CHB (improving overall survival or preventing development of HCC) has not yet been demonstrated and requires further investigation.

[Recommendations]

1. Antiviral treatment is not indicated in CHB patients in the immune-inactive phase, determined by serum HBV DNA <2,000 IU/mL, a normal ALT level, and no evidence of advanced liver fibrosis (B1).

2. Antiviral treatment can be considered in CHB patients in the immune-inactive phase with significant liver fibrosis suggested by liver biopsy or non-invasive fibrosis tests, even if the serum HBV DNA is less than 2,000 IU/mL (B2).

Compensated cirrhosis

Liver cirrhosis could be diagnosed by liver biopsy. Since liver biopsy is limited in real practice, clinical diagnosis of cirrhosis is usually assigned when an image study (computed tomography, abdominal ultrasonography, and magnetic resonance imaging) shows nodular liver surface, splenomegaly, or presence of intraabdominal collaterals suggesting portal hypertension or when endoscopy reveals esophageal or gastric varix or clinical symptoms of cirrhosis [197]. In addition to imaging studies, laboratory findings such as serum albumin, bilirubin, prothrombin time, and platelets can be helpful in diagnosis of liver cirrhosis.

Antiviral treatment for compensated cirrhosis patients can decrease the risk of HCC and liver-related complications [138] and improve liver fibrosis [198,199]. Serum ALT level might not be elevated in patients with cirrhosis, and the risk of complication is high even for those with normal ALT level [200]. Hence, cirrhotic patients with active HBV replication require antiviral treatment regardless of ALT level. For cirrhotic patients, the risk of HCC decreases but remains even after achieving a virological response to antiviral therapy [201], requiring HCC surveillance.

For compensated cirrhosis patients, those with elevated HBV DNA level (≥2,000 IU/mL) are indicated for antiviral therapy. For patients with detectable but low-level viremia (<2,000 IU/mL), recent European Association for the Study of the Liver and American Association for the Study of Liver Diseases guidelines recommend antiviral therapy [144,151]. An observational cohort study from Korea reported that 33% of compensated cirrhosis patients with low-level viremia experienced HBV DNA elevation ≥2,000 IU/mL during follow-up, and this was associated with increased risk for HCC [202]. Furthermore, HCC risk was higher for patients who remained at low-level viremia compared to those with undetectable HBV DNA level, antiviral treatment was inversely associated with HCC risk in this group, and antiviral treatment could decrease the risk of reactivation of HBV and have survival benefit in patients with low-level viremia even after HCC development [202,203]. For compensated cirrhosis patients with low-level viremia, prompt antiviral treatment has the advantage of preventing HBV DNA elevation during follow-up and can decrease the risk of complications, as shown in another observational study from Korea. In that study, the cumulative incidence rate of HCC among cirrhotic patients with low-level viremia was 13.9% in 10 years, and they showed higher risk of death and liver-related complications than did treated cirrhotic patients [193,204]. These data support prompt antiviral therapy for compensated cirrhosis with low-level viremia. However, in a recent study from Korea, the incidence rate of HCC of compensated cirrhotic patients was not different between those with HBV DNA <2,000 IU/mL without antiviral treatment and those with complete virological response to antiviral treatment [205]. However, until now, there have not been any randomized controlled trials that can assess the benefits and risks of prompt antiviral therapy for compensated cirrhosis patients showing lowlevel viremia.

[Recommendations]

1. In patients with compensated cirrhosis, antiviral therapy should be initiated regardless of ALT level if serum HBV DNA level is ≥2,000 IU/mL (A1).

2. Antiviral therapy can be initiated in compensated cirrhosis patients regardless of ALT level, even in those with detectable but low-level viremia (<2,000 IU/mL) (B1).

Decompensated cirrhosis

Decompensated cirrhosis includes cases with ascites, variceal bleeding, hepatic encephalopathy, or jaundice [197]. Patients with decompensated cirrhosis might be managed in an institution that can respond appropriately to complications and are candidates for liver transplantation. Antiviral therapy modifies the natural history of decompensated cirrhosis, improves liver function, decreases the need for liver transplantation, and improves survival [206,207]. However, even if antiviral therapy is administered, it takes time to produce a virological response and recover clinically. Some patients with severely impaired liver function do not recover despite antiviral therapy, and liver transplantation should be considered for such cases [208]. Patients with decompensated cirrhosis are prone to liver failure with HBV reactivation, which requires prompt antiviral therapy when serum HBV DNA is detectable, regardless of its serum level. Administration of interferon is contraindicated because it can cause serious side effects including liver failure even with small doses [209].

[Recommendations]

1. In patients with decompensated cirrhosis, NAs should be initiated if serum HBV DNA is detected regardless of ALT level. Liver transplantation should also be considered (A1).

NEW DRUGS FOR A FUNCTIONAL CURE

HBsAg seroclearance is the desired endpoint of functional cure for CHB. However, it cannot be fully achieved with conventional treatment including interferon or NAs [210]. Thus, many new drugs are being developed and investigated for functional cure of CHB, some of which have proceeded to phase 2 trials (Table 7). These drugs can be classified into two types based on mechanism of action: (1) drugs that directly interfere with the HBV life cycle and (2) drugs that strengthen the host immune response to HBV infection (Fig. 4). However, as safe and effective NAs are already available, the success of any newly developed drug will warrant significant reduction in serum HBsAg titer and few side effects.

Novel antiviral agents under development for HBV functional cure

Figure 4.

The mechanisms of novel antiviral agents for the functional cure. NK, natural killer; TLR, toll-like receptor; ICI, immune checkpoint inhibitor; HBV, hepatitis B virus; NTCP, sodium taurocholate co-transporting polypeptide; SVP, subviral particle; HBsAg, hepatitis B surface antigen; HBeAg, hepatitis B e antigen; HBc, hepatitis B core; HBx, hepatitis B virus X protein; cccDNA, covalently closed circular DNA; ASO, antisense oligonucleotide; siRNA, small interfering RNA.

Virus-targeting agents

Virus-targeting agents focus on viral genomes or proteins crucial for the viral life cycle to disrupt its replication and infection. These agents for treatment of CHB decrease titers of cccDNA or HBsAg, and include core protein inhibitors, RNA targeted therapeutics, HBsAg release inhibitors, and gene-editing agents.

Capsid assembly modulator

The HBV core protein aids in capsid assembly and imports pgRNAs and polymerases into the capsid for reverse transcription. Then, enveloped HBV DNA is transmitted to infect other hepatocytes or reenter the nucleus for viral cccDNA replenishment. Thus, inhibition of encapsidation could induce reduction of cccDNA expression [211,212].

NVR 3-778, ABI-H0731, and JNJ-6379 are core protein inhibitors developed as oral drugs. Clinical studies of these drugs have reported a significant reduction in serum HBV DNA but a minor reduction in serum HBsAg titer. Therefore, use of core protein inhibitors alone might be inadequate for functional cure of CHB. Thus, combination therapies with interferon or NAs were tested, but the decrease in serum HBsAg titer remained nonsignificant compared to that in the control group [213-217].

A recent study from Korea demonstrated that ciclopirox, a synthetic antifungal agent, strongly inhibits viral replication by blocking the HBV capsid assembly. When combined with NAs, ciclopirox significantly reduced the serum HBV DNA level and HBsAg titer in in vitro and in vivo studies [218,219]. Preclinical studies for the same are currently underway.

RNA targeted therapeutics

Viral RNA forms the backbone of viral antigens and proteins. RNAi inhibits translation of viral transcripts to prevent its replication and HBsAg production, restore HBV-specific immune response, and potentially lead to a functional cure [220]. Currently, two therapeutics are being used for RNAi-based therapy: antisense oligonucleotides (ASO) and small interfering RNAs (siRNAs). ASOs are 15–20-nucleotide-long single-stranded DNA oligomers, which binds to its complementary site on the target viral RNA to form a DNA-RNA duplex. The viral RNA in the DNA-RNA duplex is then cleaved by ribonuclease H, inhibiting the expression of the corresponding gene. On the other hand, siRNAs are 20–25 nucleotide-long double-stranded RNA molecules. The siRNA guide strand directs the RNA-induced silencing complex (RISC) to the complementary target RNA, which is then cleaved by the Argonaute protein present in the RISC [221,222]. Lipid nanoparticles or N-acetylgalactosamine (GalNac/NAG)-linked particles are used to deliver these agents to hepatocytes [222,223].

The main concerns regarding these agents include the risk of off-target toxicity, the potential toxicity of the delivery vehicle, and the risk of post-treatment reactivation by remaining cccDNA [219]. In addition, ALT flare should be monitored during the course of the treatment [224].

In the ASO, a phase 2 clinical study was conducted for GSK3228836, and both treatment-naïve and previously NA-treated groups each receiving 300 mg six times showed a mean reduction of more than 1.5 log10 IU/mL in serum HBsAg titer [224]. Several siRNA drugs are in phase 2 clinical trials, and interim analyses were presented at an international conference. With NA co-administration, JNJ-3989 induced a decrease in HBsAg greater than 1.0 log10 IU/mL in 98% of the participants and an average decrease of 1.93 log10 IU/mL [225]. Similar reduction in HBsAg titer was shown with AB-729, Vir-2218, and RG6346 [226-228].

HBsAg release inhibitor

HBsAg release inhibitors suppress the assembly and secretion of incomplete HBV particles (subviral particles, SVPs) to reduce serum HBsAg titer and mitigate immune exhaustion in the host, enhancing the HBV-specific immune response. Most of these drugs are nucleic acid polymers, of which REP 2139 and REP 2165 have completed phase 2 clinical trials. After 48 weeks of triple combination therapy with peginterferon and tenofovir disoproxil fumarate (tenofovir DF) in patients with CHB, HBsAg loss was observed in 60% of the patients. Among them, 58.3% maintained HBsAg loss for 48 weeks after discontinuation of REP 2139 or REP 2165 [229]. In addition, both these drugs showed a significant therapeutic effect in the treatment of HBV and hepatitis D virus (HDV) co-infection, supporting the potential of HBsAg release inhibitors for the effective treatment of HDV infection [230].

Immune modulators

One of the viral mechanisms driving CHB pathogenesis is to overwhelm the patient’s immune response. Therefore, various drugs are being developed to overcome this problem and achieve a functional cure for CHB. To this end, toll-like receptor (TLR) agonists, retinoic acid-inducible gene (RIG) agonists, immune checkpoint inhibitors, therapeutic vaccines, genetically engineered T cells, and monoclonal antibodies are being explored.

TLR agonist

TLRs recognize viral and bacterial pathogen-associated molecular patterns (PAMPs) and induce downstream signaling pathways that culminate in the transcriptional activation of interferon-stimulating genes. GS-9688 (selgantolimod) is a TLR-8 agonist, and the interim results of the phase 2 clinical study in combination with NAs were recently presented. Weekly administration of 3 mg GS-9668 for 24 weeks decreased HBsAg titer by 0.5 log10 IU/mL or more in 7% of treatment-naïve patients (vs. 0% in the control group) and demonstrated functional cure in 5% of patients who were virologically stable [231,232].

Immune checkpoint inhibitor

HBV evades the HBV-specific immune response by inducing sustained expression of inhibitory receptors on T cells. Therefore, a functional cure for CHB can be achieved by overcoming the mechanisms underlying immune inhibition using an immune checkpoint inhibitor. A pilot study reported that single administration of nivolumab (anti-PD-1), a widely used anticancer drug, to patients with HBeAg-negative CHB significantly reduced serum HBsAg titer for 24 weeks. This showed the possibility of a functional cure using immune checkpoint inhibitors [233]. Envafolimab, developed as an anti-PD-L1, is undergoing a phase 2 clinical trial as a combination treatment with NAs, and the interim results were recently announced. Serum HBsAg titer decreased by 0.49 log10 IU/mL during a 12-week follow-up after a single dose of 2.5 mg Envafolimab [234].

Therapeutic vaccine

A therapeutic vaccine can enhance the host HBV-specific immune response through exposure to various antigens and proteins of HBV.

NASVAC is a nasal spray vaccine currently undergoing phase 3 clinical trials. During a follow-up period of 138 weeks, 6.3% of the previously NA-treated group and 21.7% of the treatment-naïve group showed a reduction in the serum HBsAg titer [235]. GS-4774, expressing HBs, HBc, and HBx antigens, with tenofovir DF did not show a significant decrease in serum HBsAg titer compared to the control group [236,237].

Combination treatment

Several drugs are being developed for the functional cure of CHB. In many phase 2 clinical trials, NA or peginterferon was used in combination with these drugs rather than being administered alone for effectiveness and safety. Accordingly, combinations of three or more classes of drugs are also being evaluated.

A previous study confirmed the reduction of serum HBsAg upon combined administration of entecavir, peginterferon, and HBV vaccine in patients with CHB who sustained virological response. The HBsAg loss rate of 16.2% at 100 weeks was significant compared to that in the control group [238].

In a recent pilot study, a combination of JNJ-3989, a siRNA agent; JNJ-6379, a capsid assembly modulator; and NAs was administered to the participants. All participants showed a decrease in serum HBsAg titer by 1.0 log10 IU/mL or more in 16 weeks, and the combination treatment was declared a breakthrough treatment [239]. Based on this observation, a phase 2 clinical trial was conducted. The recent interim result showed that the three-agent combination treatment was less effective with more adverse events than the two-agent combination treatment with JNJ-3989 and NAs [240]. Therefore, not all combinations are advantageous in terms of therapeutic and adverse effects, and further research is warranted to determine optimal drug combinations.

Summary

1. New drugs that can achieve a functional cure for CHB are under development. Further research is crucial to understand the usage, efficacy, and adverse effects of each drug alone or in combination.

CESSATION OF TREATMENT AND MONITORING AFTER ANTIVIRAL TREATMENT

Clinical biomarkers for treatment endpoint

The ultimate goal of hepatitis B treatment is to reduce mortality and increase survival by continuously suppressing the proliferation of HBV. This goal can only be achieved by complete eradication of HBV in the liver in the early stages of infection; however, cccDNA persists in the hepatocyte nucleus despite antiviral treatment, so it is difficult to expect complete elimination of HBV. Therefore, it is practically very difficult to determine the end of NA therapy. Alternative biomarkers that are readily measurable and reflect the achievement of treatment goals are needed when considering cessation of NA therapy. In clinical practice, ALT normalization, undetectable HBV DNA, HBeAg loss or seroconversion, and HBsAg loss or seroconversion have been used as treatment endpoints. Recently, studies that can predict sustained off-treatment response and determine cessation of NA therapy through serum HBsAg quantitative test, serum HBcrAg quantitative test, and serum HBV RNA have been introduced [118,124,241-243]. Identification of biomarkers to find the best candidates who can cease NA therapy without clinical relapse and can achieve sustained off-treatment response or HBsAg loss is an unmet clinical need. When stopping NA, the virological relapse of HBV is reported to be around 20–70%, although there are differences depending on the patient’s condition and follow-up period [244-249]. Meanwhile, there are studies that the possibility of a functional cure, which is HBsAg loss, can increase as the immune response increases with ALT flare after cessation of NA therapy. Especially, HBsAg loss is reported to increase progressively with a higher probability in Caucasian patients with HBeAg-negative CHB who cease NA therapy [244,245,250]. Therefore, cessation of treatment should be carefully decided in consideration of safety and expected off-treatment response. Closing monitoring is recommended in all patients who discontinue NA. In particular, in cirrhotic patients, if clinical relapse occurs, there is a risk of acute exacerbation, hepatic decompensation, and death, so special attention is required [144,151,251,252].

The standard treatment duration of peginterferon alfa is 48 weeks [253,254]. However, there have been reports that extended dosing could be more effective in HBeAg-negative CHB [255].

ALT normalization

Normalization of ALT in CHB treatment reflects a decrease in hepatic inflammatory response, mostly associated with undetectable HBV DNA, and reduces clinical deterioration [166]. Normalization of ALT during treatment reflects improvement in cirrhosis and could be considered reflective of treatment goals. However, 14–40% of patients with persistently normal ALT could have significant fibrosis (≥F2), and there is a variety of concurrent liver conditions affecting ALT level, such as non-alcoholic or alcoholic fatty liver [198,256]. As such, ALT normalization alone is insufficient when determining the endpoint of treatment.

Undetectable HBV DNA

HBV DNA level is the strongest indicator of disease progression and long-term outcomes in the natural course of CHB [68,70]. HBV DNA level is associated with histological activity in CHB patients, with low rate of progression to decompensation and high rate of survival in patients with low HBV DNA [257,258]. Antiviral therapy can reduce HBV DNA level, and histological improvement can be achieved in proportion to HBV DNA reduction [198,258-260]. In addition, since it reduces the progression and exacerbation of liver disease and prevents HCC [261,262], HBV DNA is a useful alternative indicator that reflects treatment endpoints. When HBV DNA is not detectable over a long term, and virological response is well maintained, the HBsAg loss rate increases even after cessation of therapy in patients with HBeAg-negative CHB. Therefore, cessation of therapy could be considered in these patients with long-term undetectable HBV DNA [244-246,250]. The lower is the HBV DNA level, the better is the clinical status; however, when the HBV DNA level is 60–2,000 IU/mL, the risk of cirrhosis and HCC is similar to that of patients with undetectable HBV DNA [68,70]. Evidence for HBV DNA level as a surrogate indicator in these patients is lacking. Also, in practice, most patients with undetectable HBV DNA relapsed after cessation of NA therapy [194,244-246]. Hence, undetectable HBV DNA cannot be the sole indicator determining treatment cessation.

HBeAg loss and/or seroconversion

HBeAg seroconversion in patients with HBeAg-positive CHB is accompanied by HBV DNA reduction, ALT normalization, and histological improvement. After HBeAg seroconversion, HBsAg loss increases to 1.15% per year [263,264]. Therefore, HBeAg loss/seroconversion in HBeAg-positive CHB could be an indicator reflecting achievement of the treatment goal. However, after cessation of NA therapy following HBeAg loss/seroconversion, the sustained virological response is reported to be 62.5%, 53.4%, and 51.5% at 1, 2, and 3 years, respectively [265]. Also, in some patients, HBeAg-negative CHB and HBeAg reversion or acute exacerbation accompanied by jaundice can occur in severe cases [266], so there is a limit to the indicator determining treatment cessation with only HBeAg loss/seroconversion. Sustained undetectable HBV DNA after HBeAg loss/seroconversion and the duration of consolidation therapy to maintain ALT normalization are important for maintaining response after cessation of NA therapy [248,267], but studies on the specific period are lacking. Nevertheless, it is recommended to maintain consolidation therapy for at least 12 months after HBeAg loss/seroconversion [144,151,252,265,268].

Quantitative HBsAg level, quantitative HBcrAg level, and HBV RNA

HBsAg is translated from pre-S1 and S2 messenger RNAs that are transcribed from the S gene. HBsAg exists as three protein subtypes (small, middle, and large) that are not differentiated by commercial assays. Additionally, there are two non-infectious subviral particles secreted, in spherical and filamentous forms, with 100-fold to 100,000-fold higher levels than mature virions [104]. The level of HBsAg has been found to be an indicator for off-therapy sustained response in many studies [269-274]. The degree of decline in HBsAg level during peginterferon can predict the efficacy of treatment-induced immune response. In patients whose HBsAg level does not sufficiently decrease in the early phase of peginterferon, additional treatment is unlikely to be successful, so it is used as the stopping rule, reducing unnecessary treatment [269]. Additionally, HBsAg level can be useful to determine if cessation of NA therapy is an option [270-274]. Chen et al. found the HBsAg level at the end-of-treatment (EOT) was the most important predictor for HBsAg loss and sustained response after stopping lamivudine treatment. At EOT, the HBsAg cutoff value of 300 IU/mL could predict 55.6% HBsAg loss in HBeAg-positive patients. In HBeAgnegative patients, the HBsAg cutoff values of 120 and 200 IU/mL could predict 79.2% HBsAg loss and 93.3% post-treatment sustained response, respectively [271]. In a Taiwanese study of patients with EOT HBsAg level <100 IU/mL, the cumulative rates of clinical relapse and sustained virological response were 9.3% and 45.5%, respectively, during a mean 2-year follow-up period after cessation of entecavir treatment [272]. In a large retrospective study of 691 HBeAg-negative patients who had discontinued NAs, during a median follow-up period of 156 (2–614) weeks, virological and clinical relapse rates were reported in 79.2% and 60.6%, respectively. In this study, EOT HBsAg level <100 IU/mL was an independent factor for HBsAg loss [273]. Another study showed EOT HBsAg level to be an independent predictor of virological and clinical relapse in HBeAg-negative patients who had discontinued tenofovir DF treatment, and HBsAg level of 80 IU/mL was the optimal value. The virological and clinical relapse rates at 78 weeks were 19.6% and 15.4%, respectively, in patients who achieved EOT HBsAg ≤80 IU/mL. Also, the cumulative rate of HBsAg loss at 104 weeks were 45.5% in these patients [274]. As such, low EOT HBsAg level (10–200 IU/mL) has been reported to be a good predictor of sustained virological response and HBsAg loss after cessation of NA therapy. Therefore, monitoring of quantitative HBsAg level during NA therapy could be helpful in practice [243,270,273].

HBcrAg contains three products encoded by the PC/core gene, HBcAg, p22cr, and HBeAg, all of which can be measured by serological testing. Serum HBcrAg level is closely associated with intrahepatic total HBV DNA and cccDNA, as well as serum HBV DNA [104,113,118,241]. HBcrAg level is useful for predicting off-treatment sustained response after cessation of NA therapy. Recently, HBcrAg level alone or a combination of HBsAg levels has been proposed [247,275]. SCALE-B score (HBsAg level [S], HBcrAg [C], age [A], ALT [L], and tenofovir [E] used for HBV [B]) including HBsAg and HBcrAg levels at NA discontinuation was suggested for prediction of post-NA relapse and HBsAg loss in Asian CHB patients [276-278]. The scoring criteria for including HBsAg and HBcrAg levels for cessation of NA therapy have been proposed and incorporated into the Japan Society of Hepatology Guidelines for the Management of HBV infection [117].

HBV RNA has been introduced as a novel biomarker for cessation of NA therapy [104,242]. There are also studies that predict off-treatment response with a combination of other indicators, such as HBsAg level or HBcrAg level [119,124,279]. However, the value of this indicator has not yet been evaluated as a standardized test, and it is necessary to explore the role of HBV RNA in predicting outcomes after cessation of NA therapy.

HBsAg loss

HBsAg loss is defined as a sustained loss of HBsAg and HBV DNA from serum, with or without anti-HBs seroconversion [37]. HBsAg loss is considered “functional cure” as the optimal treatment endpoint from a virological and clinical point of view. The prognosis following spontaneous HBsAg loss is excellent, except in patients with cirrhosis or those with concurrent other viral infection [23]. The incidence of HCC is significantly reduced when HBsAg loss occurs before ages 45–50 years [280]. Some patients with HBsAg loss during antiviral therapy showed HBsAg reversion or low but detectable HBV DNA, but most patients maintain HBsAg loss and undetectable HBV DNA, and their incidence of liver complications, including HCC, is significantly lower compared to that of patients without HBsAg loss [46,47,49,281]. Therefore, HBsAg loss is generally regarded as the optimal outcome of antiviral therapy in CHB, reflecting the treatment goal [37], at which point NAs can be discontinued [46,281,282]. Recently, it has been reported that HBsAg reversion and serum HBV DNA detection are rare if antiviral therapy is discontinued after HBsAg loss, defined as testing HBsAg negative on two separate occasions at least 6 months apart [281,282].

Monitoring after antiviral treatment

Although off-treatment response can persist after cessation of NA therapy, clinical relapse can occur in some patients, and there is a risk of acute hepatitis flare, liver decompensation, or fulminant hepatitis. Therefore, regular monitoring of liver function tests, HBeAg, anti-HBe, and HBV DNA is needed to evaluate the durability of the treatment response, relapse, and deterioration in liver function. In particular, if serum HBV DNA increases after cessation of NA therapy, a more intensive monitoring plan should be implemented to determine whether NA should be re-administered [243]. HBsAg level measurement can help monitor HBsAg reduction or loss in patients without HBsAg loss on cessation of NA therapy [104,269,270]. Even in patients in whom HBsAg loss has been achieved, there is the potential risk for reversion of HBsAg or development of HCC [46,49,131,280-282]. Therefore, serum HBsAg and/or anti-HBs should be monitored, and continuous HCC surveillance should be performed.

[Recommendations]

1. Cessation of NA therapy is recommended after serum HBsAg loss in CHB patients (A1).

2. In HBeAg-positive CHB patients, cessation of NA therapy could be considered at least 12 months after HBV DNA is undetectable and serum HBeAg loss or seroconversion has been achieved (B2).

3. Long-term treatment should be considered in patients with liver cirrhosis. Indefinite NA therapy is recommended in patients with decompensated liver cirrhosis (B1).

4. Peginterferon alfa is administered for 48 weeks (A1).

5. With reference to quantitative HBsAg level, cessation of NA therapy could be considered (B1).

6. Biomarker test such as HBcrAg and HBV RNA can be performed when considering cessation of NA therapy (B2).

7. Liver function testing and serum HBV DNA measurement at 1- to 6-month intervals and HBeAg/anti-HBe testing at 3- to 6-month intervals are recommended during the first year after cessation of antiviral treatment. Liver function testing and serum HBV DNA measurement at 3- to 6-month intervals and HBeAg/anti-HBe testing at 6- to 12-month intervals are recommended if treatment response is maintained beyond one year after antiviral therapy (B1).

8. If virological response is maintained after cessation of NA therapy, follow-up HBsAg/anti-HBs testing should be performed to confirm HBsAg loss, maintenance, or reversion (B1).

MANAGEMENT IN SPECIAL CONDITIONS

Patients with HCC

The aims of antiviral treatment in patients with HBV-related HCC are suppression of HBV replication to prevent the progression of hepatic dysfunction, thereby enabling active treatment of HCC, and reduction of HCC recurrence after curative treatment.

HBV treatment in patients undergoing curative treatment for HCC

A recent meta-analysis demonstrated that the pooled rates of HBV reactivation and biochemical reactivation were 20% and 9%, respectively, in patients who underwent surgical resection for HCC without NA treatment [283]. In contrast, patients receiving antiviral prophylaxis had a pooled HBV reactivation rate of 2–4%. In terms of local ablation therapies such as radiofrequency ablation or percutaneous ethanol injection, the HBV reactivation rate in patients who received no prophylactic NA was 5–9%, whereas HBV reactivation was rarely diagnosed in those who received prophylactic NA therapy [283-286]. In HBsAg-positive HCC patients with undetectable serum HBV DNA level at HCC diagnosis, some retrospective studies in Korea and China reported that 22–33% of patients experienced HBV reactivation after surgical resection [287-290]. The risk of HBV reactivation was lower for those receiving NA.

Several studies have reported that antiviral treatment was associated with lower risk of tumor recurrence after curative treatment for HBV-related HCC. A Taiwanese large-scale retrospective study showed that patients who underwent NA treatment with entecavir, lamivudine, telbivudine, etc. showed a significantly lower risk of tumor recurrence or overall death after surgical resection for HCC compared to those not treated with these drugs, although the prevalence of cirrhosis was significantly higher [291]. A Chinese randomized controlled trial (RCT) to evaluate the effect of NA treatment on postoperative prognosis of HBV-HCC showed that antiviral treatment significantly decreased HCC recurrence (hazard ratio [HR], 0.48; 95% confidence interval [CI], 0.32–0.70) and HCC-related death (HR, 0.26; 95% CI, 0.14–0.50) [292]. Even in RCTs including patients with low-level viremia (HBV DNA <2,000 IU/mL), antiviral treatment was associated with longer recurrence-free survival and overall survival [293]. NA therapy was also associated with a decreased risk of HCC recurrence among patients with HBV-related HCC post-RFA (HR, 0.69; 95% CI, 0.50–0.95) [294]. A meta-analysis showed that the antiviral treatment group had significantly lower risk of tumor recurrence (odds ratio [OR], 0.59; 95% CI, 0.35–0.97), liver-related mortality (OR, 0.13; 95% CI, 0.02–0.69), and overall mortality (OR, 0.27; 95% CI, 0.14–0.50) than the no antiviral treatment group [295].

Regarding the choice of oral antivirals, a Korean retrospective study demonstrated that antiviral treatment with high-potency NAs (i.e., entecavir and tenofovir) showed significantly longer recurrence-free survival than both antiviral treatment with low-potency NAs (i.e., lamivudine, clevudine, and telbivudine; HR, 0.47; 95% CI, 0.34–0.65) and no antiviral treatment (HR, 0.39; 95% CI, 0.30–0.51) [296]. However, it is not clear whether tenofovir and entecavir have different effects on HCC recurrence in patients receiving curative hepatectomy for HBV-related HCC. Recent Korean retrospective study reported that tenofovir DF therapy was associated with a significantly lower risk of HCC recurrence after curative resection and better overall patient survival compared with entecavir [297]. In contrast, no difference in the risk of death or liver transplantation was observed in a Taiwanese retrospective cohort with hepatectomy for early-stage HCC, despite tenofovir DF therapy being associated with a lower risk of HCC recurrence than entecavir [298]. The prognoses in terms of recurrence and death after curative treatment of HBV-related HCC were not statistically different between the entecavir and tenofovir DF groups in a multi-center retrospective study in Korea [299].

HBV treatment in patients undergoing locoregional therapy for HCC

After transarterial chemoembolization (TACE), approximately 4–40% of patients with HBV-related HCC developed HBV reactivation [283,300-304]. Post-TACE risks of HBV reactivation, flare-up hepatitis, and liver failure due to HBV reactivation were 2.8%, 2.8%, and 0%, respectively, in the prophylactic lamivudine treatment group and 40.5%, 29.7%, and 8.1% in the control group. There was a significant difference between the two groups [302]. After hepatic artery infusion chemotherapy (HAIC), HBV reactivation was reported in 24–67% of patients, which was relatively higher than that after TACE. This finding can potentially be explained by a larger total amount of cytotoxic chemotherapeutic agents due to shorter treatment intervals than TACE [305-307]. Furthermore, prophylactic antiviral treatment is significantly associated with improved long-term survival among patients undergoing transarterial chemotherapy, including TACE and HAIC (10-year overall survival, 26.5% vs. 12.8%) [203]. The prospective Chinese study involved 98 patients with HBsAg-positive/HBV DNA-negative HCC reported that HBV reactivation occurred in 23.4% of patients in the nonantiviral group (11/47, 23.4%), but only 5.9% of patients in the antiviral group (P<0.05) [308].

In patients who underwent external beam radiation therapy (EBRT) for HCC, reactivation and ALT elevation were reported in 0% and 2.3%, respectively, of the lamivudine-prophylaxis group and 21.8% and 12.5% of the control group. The control group had a significantly higher risk of HBV reactivation [283,309]. A recent Korean retrospective study also showed that the absence of antiviral treatment was an independent risk factor for HBV reactivation in EBRT for HCC (OR, 8.34; 95% CI, 2.53–27.47) [310]. Therefore, preventive antiviral therapy is recommended for patients with HCC who are scheduled to receive EBRT. The combination treatment with TACE and EBRT had twice the risk of HBV reactivation compared to TACE treatment alone [311]. In addition, either TACE or combination treatment of TACE with EBRT can reactivate HBV replication in HBsAg-negative/anti-HBc-positive individuals [312,313]. Patients with prior chronic HBV infection who undergo intensive TACE should be closely monitored, with an alternative approach of antiviral prophylaxis against HBV reactivation.

HBV treatment in patients undergoing systemic therapy/immune checkpoint inhibitors for HCC

HBV reactivation was diagnosed in 6.2% of patients treated with sorafenib based on the data of a recently published meta-analysis [283]. A few retrospective studies showed that underlying HBV DNA >2,000 IU/mL was associated with poor prognosis in sorafenib-treated patients, whereas antiviral prophylaxis was significantly related to the improvement of overall survival [314,315].

Immune checkpoint inhibitors can enhance host immunity and consequently have a lower risk of HBV reactivation. However, immune checkpoints can result in severe acute aggravation of hepatitis since it can upregulate antiviral immunity against HBV. Therefore, suppression of HBV replication with antiviral treatment is necessary before use of immune checkpoint inhibitors [316]. The overall pooled HBV reactivation rate was 7.8% in HCC patients receiving immune checkpoint inhibitors, and most of them did not receive antivirals [283]. A Korean retrospective study including 398 HBV-HCC patients receiving both immune checkpoint inhibitors and antiviral prophylaxis demonstrated that two (0.5%) showed HBV reactivation, both of whom showed poor adherence to antivirals [317]. In addition, retrospective studies reported that antiviral prophylaxis is less likely to reactivate HBV regardless of baseline HBV DNA level [318,319]. Thus, antiviral prophylaxis is recommended in HCC patients receiving immune checkpoint inhibitors.

In atezolizumab and bevacizumab combination treatment or lenvatinib, which have recently been approved as the first-line treatment for liver cancer, further studies are needed due to the lack of data on HBV reactivation.

[Recommendations]

1. In patients with HBV-related HCC, antiviral therapy should be initiated if serum HBV DNA is detected (A1).

2. In patients with HBsAg-positive HCC who undergo anticancer treatment, prophylactic antiviral therapy with NAs should be considered regardless of detectable serum HBV DNA (B1).

3. HBV reactivation should be monitored in patients with past HBV infection undergoing anti-HCC treatment, especially TACE or EBRT (B2).

Patients with renal dysfunction or metabolic bone disease

Long-term administration of adefovir or tenofovir DF in the patients with CHB can result in decreased renal function and bone mineral density. Side effects such as acute or chronic renal failure, hypophosphatemia, and Fanconi syndrome have been reported [320-323]. If patients already have risk factors for renal dysfunction and/or metabolic bone disease, or if worsening kidney function or bone disease is detected during treatment, a change in treatment regimen needs to be considered.

Patients with renal dysfunction or metabolic disease prior to starting treatment

Patients with chronic kidney disease have relatively higher rate of exposure to HBV infection [324]. In Korea, a 4–7% HBsAg-positive rate has been reported among hemodialysis patients [325-329]. When starting NA treatment in patients with chronic kidney disease, the dose must be adjusted according to the creatinine clearance (Table 8). Tenofovir alafenamide fumarate (tenofovir AF) and tenofovir DF are not recommended in patients with creatinine clearance below 15 and 10 mL/min, respectively, without renal replacement. This is also true for besifovir in cases of creatinine clearance below 15 mL/min.

Dose adjustment of NAs for adult patients with altered creatinine clearance

Because NA treatment itself can affect renal function or bone density, it is necessary to select an appropriate drug if there is any risk factor such as long-term steroid use. In addition, patients treated with tenofovir DF should be monitored for bone mineral density during treatment. In a large phase 3 trial comparing tenofovir AF and tenofovir DF over a 96-week treatment period, among those with any risk factor for renal dysfunction (age over 50 years, hypertension, cardiovascular disease, diabetes, or hyperlipidemia), patients treated with tenofovir DF, compared to patients treated with tenofovir AF, had worsening renal function (compared to baseline, median changes in estimated glomerular filtration rate [eGFR] were -5.0 mL/min and -0.3 mL/min, respectively) [330-332]. Additionally, among those with a risk factor for decreasing bone density (female, age over 50 years, Asian, and baseline eGFR <90 mL/min) patients treated with tenofovir DF showed a greater decrease in bone mineral density compared to patients treated with tenofovir AF (-3.29% and 1.23%, respectively) [330-332]. Therefore, it is recommended to avoid the use of tenofovir DF among patients with risk factors for renal dysfunction such as baseline eGFR <60 mL/min, proteinuria, albuminuria (urine albumin: creatinine ratio >30 mg/g), hypophosphatemia (<2.5 mg/dL), uncontrolled diabetes, or hypertension. If patients have a diagnosis of osteopenia or osteoporosis, need to be on chronic steroid treatment, or take other medications that can lower the bone density, other NAs that more weakly affect bone density should be considered over tenofovir DF (Fig. 5) [151].

Figure 5.

Indications for selecting entecavir, tenofovir alafenamide fumarate, or besifovir over tenofovir disoproxil fumarate. AF, alafenamide fumarate. *Dose was adjusted if creatinine clearance <50 mL/min, refer to Table 8. Not indicated if creatinine clearance <15 mL/min without dialysis. Not indicated if creatinine clearance <15 mL/min.

In addition to tenofovir AF, entecavir and besifovir have less of an effect on renal function and bone metabolism. Besifovir, recently approved for use, had been evaluated in a clinical trial for safety in reduction of renal function and bone density. In a multicenter phase 3 trial from Korea, the median decrease from baseline in eGFR was -0.5 mL/min in patients with besifovir and -7.8 mL/min in patients with tenofovir DF. In terms of bone mineral density, patients treated with besifovir showed median decreases from baseline in bone mineral density in spine and hip of 0.33% and 0.44%, respectively, whereas patients treated with tenofovir DF had median decreases of 0.85% and 1.29% [333]. Therefore, besifovir is thought to have a better effect on renal function or bone density compared to tenofovir DF. However, besifovir is not indicated in cases of severely decreased renal dysfunction (eGFR <15 mL/min) due to lack of clinical data.

Patients who developed renal dysfunction or decrease in bone density on treatment with NAs

If patients develop renal dysfunction or decrease in bone density while on NAs, the causative factors must be identified and corrected. Then, the dose must be modified accordingly (Table 8) or a review should be performed for drug change (Fig. 5).

A phase 3 clinical trial compared the safety of tenofovir AF to that of tenofovir AF during 240 weeks. In this trial, patients were given either treatment for 96 weeks each, after which some patients treated with tenofovir DF switched to tenofovir AF, while the remaining switched to tenofovir AF at 144 weeks. Patients who switched from tenofovir DF to tenofovir AF at 96 weeks (tenofovir AF for 48 weeks afterward) showed improved renal function at 144 weeks compared to patients who continued tenofovir DF at 144 weeks (compared to baseline, mean change in eGFR: 4.2 mL/min and -0.9 mL/min, respectively). In terms of bone mineral density in spine and hip, patients who were treated with tenofovir DF showed greater decreases than patients treated with tenofovir AF until week 96. However, after switching to tenofovir AF at week 96 or week 144, bone mineral density recovered to baseline at week 240 in these patients, resulting no significant difference in patients initially treated and then switched and those who maintained tenofovir AF for 240 weeks [334].

In another phase 3 non-inferiority switching trial, patients who switched to tenofovir AF from tenofovir DF had improvements in renal function (compared to baseline, median change 0.94 mL/min vs. -2.74 mL/min, respectively) and bone mineral density (compared to baseline, median change 0.66% vs. -0.51% [hip]; 1.74% vs. -0.11% [spine]) compared to patients who continued tenofovir AF at week 48 [335]. Additionally, in a randomized non-inferiority trial of patients with multidrug-resistant HBV, patients who switched to tenofovir AF from tenofovir DF, compared to patients continuing tenofovir AF, showed a greater increase in median eGFR (compared to baseline, 7.3% vs. 1.9%, respectively) and an improvement in bone mineral density (compared to baseline, 1.8% vs. 0.2%, respectively) [336]. Therefore, it is thought that reduction in renal function and/or bone density while on tenofovir DF can be improved by switching to tenofovir AF.

In a phase 3 trial comparing besifovir with tenofovir D, patients receiving tenofovir DF for 48 weeks showed a decrease in eGFR by -7.8 mL/min. However, renal function as measured by eGFR was recovered to baseline after switching to besifovir and continuing to 144 weeks. In terms of bone mineral density in spine and hip, patients receiving tenofovir DF showed decrease of 1.12% and 0.62%, respectively. After switching to besifovir, bone density was improved and recovered to baseline, showing no significant difference from patients who were initially treated with besifovir. These improvement in renal function and bone density were maintained after 144 weeks of besifovir treatment [337,338]. Therefore, during the NA treatment of CHB, if patients develop renal dysfunction or metabolic bone disease, and/or carry risk factors, appropriate drug change can be an option for overcoming the side effects (Fig. 5).

[Recommendations]

1. Entecavir, tenofovir AF, and besifovir are preferred over tenofovir DF in treatment-naïve CHB patients with or at risk of renal dysfunction or metabolic bone disease (A1).

2. Treatment can be switched to tenofovir AF, besifovir, or entecavir depending on treatment history in patients on tenofovir DF with or at risk of renal dysfunction or metabolic bone disease (A1).

3. NA dose should be adequately adjusted for creatinine clearance (A1).

Patients on immunosuppression or chemotherapy

The progression of CHB is determined by the interaction between the virus and host immune response. Therefore, if the immune response is suppressed by immunosuppressive therapy or anticancer chemotherapy, the risk of reactivation increases [339].

Reactivation of CHB

Reactivation of hepatitis B indicates recurrence of active necrotizing inflammatory disease in patients in the inactive phase of CHB or those who recovered from previous active infection. Such reactivation can be largely divided into two categories, “exacerbation of chronic HBV infection” for those with positive HBsAg and “relapse of past HBV infection” for those with negative HBsAg and positive anti-HBc [340]. In the latter category, patients who remained in an “occult HBV infection” status can show viral replication triggered by immunosuppression, leading to reverse seroconversion or seroreversion, with redetection of HBsAg [341-343]. Exacerbation of chronic HBV infection is defined in those with seropositive HBsAg as an increase of serum HBV DNA by more than 100 times the baseline level. Relapse of past HBV infection is defined as seroconversion of HBsAg-negative to positive or detection of serum HBV DNA from undetectable level to greater than 100 IU/mL. Hepatitis flare due to HBV reactivation is defined as a ≥3-fold increase in serum ALT level, to a total exceeding 100 IU/L [344,345].

Various rates of reactivation have been reported but are about 20–50% in HBsAg-positive patients receiving anti-cancer chemotherapy. For an accurate diagnosis, liver damage related to chemotherapy, tumor metastasis, or hepatitis secondary to other viruses should be excluded. In many cases, patients are asymptomatic but occasionally present with jaundice or are in various stages such as decompensated liver disease or death [343,346-348]. Typical reactivation is noted by detection of serum HBV DNA during immunosuppression or chemotherapy or elevation of serum ALT after stopping the immunosuppressive therapy. If the reactivation occurs during chemotherapy, it can lead to reduction or discontinuation of chemotherapy, adversely affecting treatment success [349-351]. There are risk factors of hepatitis B reactivation related to the virus, the host, and treatment. Virus factors include serum HBV DNA, HBeAg seropositivity, hepatocyte cccDNA, and PC/BCP mutation prior to treatment; host factors include type of malignant tumor, male gender, young age, and high serum ALT level; and treatment factors include the type and intensity of immunosuppressant or chemotherapy regimen, hematopoietic stem-cell transplantaton (HSCT), and/or solid organ transplantation [352]. The type and intensity of chemotherapy regimen related to the risk of hepatitis B reactivation can be classified into three categories: high risk group (reactivation risk of 10% or more), moderate risk group (reactivation risk between 1–10%), and low risk group (reactivation risk less than 1%) (Table 9) [316,353,354].

Risk of hepatitis B reactivation associated with immune-related therapies

Reactivation of hepatitis B during chemotherapy for lymphoma and other hematologic malignancies

During chemotherapy for lymphoma, hepatitis B reactivation is reported to be frequent, with the rate up to 24–67%. This not only implies that the chemotherapy used for lymphoma is strong enough to cause bone marrow suppression, but also that patients with lymphoma have higher rates of seropositive HBsAg than those without lymphoma [347,355-357]. Rituximab, commonly used in combination with steroids for the treatment of lymphoma, is known to increase the risk of reactivation [358,359]. Rituximab therapy increased the risk of hepatitis B reactivation in patients with non-Hodgkin’s lymphoma who had seropositive HBsAg or seronegative HBsAg/seropositive anti-HBc combination (relative risk [RR], 2.14; 95% CI, 1.42–3.22; P<0.001). Even in patients with seronegative HBsAg/seropositive anti-HBc combination, the use of rituximab therapy was associated with higher risk of reactivation (RR, 5.52) [360]. In a retrospective observational study of lymphoma patients treated with rituximab, hepatitis B reactivation rate was 27.8% in patients with seropositive HBsAg, and the risk could be lowered by the prophylactic antiviral treatment (22.9% [32/140] vs. 59.1% [13/22]; P<0.001). However, hepatitis B reactivation developed in more than 20% of patients who received prophylactic antiviral treatment. In a retrospective study, hepatitis B reactivation rate was as low as 2.4% in patients with seronegative HBsAg/seropositive anti-HBc combination [361]. According to a recent meta-analysis, this rate was 17% in prospective studies and 7% in retrospective studies [362]. In addition, in a prospective observational study of lymphoma patients with seronegative HBsAg/seropositive anti-HBc combination who were treated with rituximabCHOP (R-CHOP) chemotherapy, hepatitis B reactivation and subsequent worsening of hepatitis were common (10.4 vs. 6.4 per 100 person-year). In that study, there were cases where hepatitis worsened during periodic monitoring of HBsAg and serum HBV DNA, and immediate antiviral treatment was safe for reactivation of hepatitis B. In particular, seroreversion of HBsAg was the most important indicator of hepatitis B-related liver damage (100% vs. 28%) [363]. There was a significant difference in the reactivation of hepatitis B in patients with and without prophylactic antiviral therapy (13.3% vs. 60%) during treatment with rituximab [364]. Furthermore, prior to receiving chemotherapy (R-CHOP), screening for hepatitis B in all patients, rather than limiting to high-risk groups, resulted in a 10-fold reduction in hepatitis B reactivation rate and economic and survival benefits [365]. Lamivudine was the most commonly used antiviral agent, but the hepatitis B reactivation rate was significantly lower in the entecavir group than in the lamivudine group (6.3% vs. 39.3%; P<0.05). In a recent, prospective, randomized control study, hepatitis B reactivation was reported to be 0% in the group treated with prophylactic tenofovir DF during rituximab treatment but 10.7% (P=0.09) in the group not receiving prophylactic antiviral therapy [366].

With other hematologic malignancies, if patients are receiving high-intensity chemotherapy prior to HSCT, the risk of reactivation is high [366,367]. In particular, in patients with seropositive HBsAg or seronegative HBsAg/seropositive anti-HBc awaiting high intensity chemotherapy prior to HSCT, antiviral therapy with a high barrier to resistance is recommended [368]. During immunosuppressive therapy or chemotherapy for hematologic disorders, for patients with evidence of hepatitis B infection, prophylactic treatment with lamivudine or entecavir can significantly lower the reactivation rate [369,370].

Meanwhile, in a recent Korean study evaluating 93 multiple myeloma patients with seronegative HBsAg who were treated with daratumumab (anti-CD38), hepatitis B reactivation occurred in 6 (6.5%, 6/93). Subgroup analysis for patients with seronegative HBsAg/seropositive anti-HBc combination showed a higher risk of 12.5% (3/24). Furthermore, special attention is needed because one patient eventually died due to liver failure even though all six hepatitis B-reactivated patients were treated with prompt antiviral therapy [371].

Reactivation of hepatitis B during chemotherapy for solid tumors

Reactivation of hepatitis B in patients with solid tumors occurs in 14–21%, while that of breast cancer is higher at about 41– 70%, which is thought to be related to the high doses of treatment agents and the use of anthracycline-based chemotherapy and steroids [351,372,373]. Steroids not only suppress the immune system, but directly stimulate the replication of HBV, increasing the risk of reactivation. Use of prophylactic antiviral agents in most solid tumors, not only breast cancer, but also lung cancer in which immunotherapy is being used as a first-line treatment, has significantly reduced the rates of hepatitis B reactivation and discontinuation of chemotherapy treatment [374,375].

Reactivation of hepatitis B during immune-related treatments for inflammatory bowel disease (IBD) or rheumatoid arthritis (RA)

The reactivation of hepatitis B also might be associated with the use of tumor necrosis factor-alpha (TNFα) inhibitors (infliximab, etanercept, adalimumab, etc.) for the treatment of IBD or RA [375-380]. For TNF α inhibitors used for the treatment of IBD, hepatitis B reactivation occurred in 22.5–39% of patients with seropositive HBsAg and 5% of seronegative HBsAg/seropositive anti-HBc combination [381]. In cases treated with TNFα inhibitors and disease-modifying antirheumatic drugs (DMARDs) for RA treatment, the rate of reactivation of hepatitis B was around 12.3% in patients with seropositive HBsAg [382]. In another study, reactivation was reported in 39% of HBsAg-positive patients and 5% of antiHBc-positive patients; among those given antiviral prophylaxis, the reactivation rate was significantly lower (23% vs. 62%; P=0.003) [383]. In a meta-analysis of patients treated for RA, hepatitis B reactivation occurred in 14.6% of HBsAg-positive patients and was lower at 9.0% in those treated with prophylactic antiviral therapy [384]. However, the reactivation rate tended to decrease in the TNFα inhibitor treatment group (4.4% vs. 15.6%; P=0.05), but no significant difference was observed from the DMARD treatment group (27.1% vs. 22.4%) [384]. This finding might be related with the high heterogeneity of studies included in this metaanalysis, and further studies are needed. In the same study, hepatitis B reactivation occurred in 1.6% of cases where only anti-HBc was positive [384].

Acute exacerbation of hepatitis B during immunotherapy treatment

Recently, immune checkpoint inhibitors such as anti-PD-1 (nivolumab), anti-PD-L1 (atezolizumab), and anti-CTLA4 (ipilimumab) have been used in various carcinomas including liver cancer. According to a recent retrospective study analyzing patients with multiple solid cancers and some lymphomas, the incidence rates of hepatitis B reactivation of the total patients, HBsAg-positive patients, and HBsAg-negative patients were 0.14% (5/3,465), 1.0% (5/511), and 0.0% (0/2,954), respectively. The hepatitis B reactivation rates were 0.4% (2/464) and 6.4% (3/47) in patients with and without antiviral prophylaxis, respectively [317]. Hepatitis B reactivation is rare in immune checkpoint inhibitor chemotherapy, but prophylactic antiviral therapy can be considered in HBsAgpositive patients. Pembrolizumab among various immune checkpoint inhibitors showed a significant correlation with hepatitis B reactivation [385]. In a retrospective study evaluating 89 patients with hematologic malignancies treated with chimeric antigen receptor (CAR)-T cells, all 19 HBsAg-positive patients received prophylactic antiviral therapy with entecavir, but hepatitis B reactivation occurred in one (5.3%). However, hepatitis B reactivation was not observed in patients with seronegative HBsAg/seropositive anti-HBc combination despite only two of 37 patients receiving prophylactic entecavir therapy. Furthermore, no hepatitis B reactivation occurred during prophylactic antiviral therapy in the 33 patients with seronegative HBsAg/seronegative anti-HBc combination. Therefore, prophylactic antiviral therapy might be beneficial to HBsAg-positive patients treated with CAR-T cells, who are thought to have higher than intermediate risk of hepatitis B reactivation [386]. There are concerns about the possibility of acute exacerbation of hepatitis B in relation to these treatments, but there is insufficient data, and further consideration of prophylactic antiviral therapy is needed.

Start and end points of prophylactic antiviral therapy

When the reactivation of hepatitis B occurs, there is a risk of liver failure or even death. Therefore, prevention is most important. Prior to starting an immunosuppressive therapy or chemotherapy, screening for HBsAg and anti-HBc is necessary. If there is no evidence of HBV infection in the past (with negative HBsAg and anti-HBc), HBV vaccination can be considered. In HBsAg-positive cases, regardless of serum HBV DNA level, antiviral prophylaxis is recommended. Instead of waiting for the serum HBV DNA level to rise, administering an antiviral agent at the start of the immunosuppressive therapy or chemotherapy or 7 days prior to the treatment start date is reported to be more effective [377,387-389]. The end point of the prophylactic antiviral treatment should theoretically be continued until the immune system is adequately recovered, but there is lack of sufficient evidence to suggest a specific end point. It has been reported that the risk of HBV reactivation is high when prophylactic lamivudine is discontinued about 3 months after the end of chemotherapy. The risk is especially higher when the serum HBV DNA prior to the treatment is elevated (≥2,000 IU/mL) [390]. Therefore, when HBV is actively replicating before prophylactic antiviral therapy, following the present CHB treatment guidelines for the discontinuation of antiviral agents might prevent virus reactivation after treatment. However, regardless of the serum HBV DNA level prior to the treatment, reactivation is reported more than 6 months after the completion of chemotherapy, so caution is required. Therefore, antiviral prophylaxis should be maintained for at least 6 months after the chemotherapy is completed, and extension should be considered according to the chemotherapy risk. Especially, for patients receiving chemotherapy involving rituximab, it is recommended to extend the antiviral prophylaxis to at least 12 months after the completion of chemotherapy [391-393]. Even after the end of prophylactic antiviral therapy, it is necessary to closely monitor for relapse for more than at least 12 months.

Meanwhile, as described above, hepatitis B reactivation requires attention because it can occur not only when HBsAg is positive, but also when HBsAg is negative and anti-HBc is positive. In particular, under immunosuppressive conditions, patients who were only positive for anti-HBc had a higher risk of hepatitis B reactivation than patients who were positive for both anti-HBc and anti-HBs [316,394]. Therefore, when HBsAg-negative and anti-HBc-positive patients are treated with a rituximab-containing regimen or HSCT for leukemia, it is necessary to consider prophylactic antiviral therapy because the risk of hepatitis B reactivation is greater than 10%. Meanwhile, in a Korean study analyzing the risk of hepatitis B reactivation in patients who received chemotherapy including rituximab for lymphoma, the risk of hepatitis B reactivation was low in patients with resolved HBV (HBsAg-negative and HBcAb-positive) when anti-HBs titer was high (>100 IU/mL). For these patients, meticulous follow-up for ALT and HBV DNA and prompt commencement of antiviral therapy upon hepatitis B reactivation are a possible strategy [395]. For patients receiving intermediate- or low-risk groups of chemotherapy, HBsAg and HBV DNA should be monitored periodically (every 1–3 months) during and after the chemotherapy, and antiviral treatment should be commenced upon hepatitis B reactivation [316].

Treatment medications

Lamivudine is the most widely studied drug for prophylactic antiviral therapy. It is well known to significantly reduce reactivation, liver failure, and death according to randomized controlled trials of lymphoma patients in Hong Kong and Taiwan [356,367,387,396]. However, lamivudine has been reported to be resistant even during prophylaxis. If the treatment duration is expected to be long, it is necessary to select a therapeutic agent with a high barrier to resistance considering the resistance rate [356]. In a retrospective study of lymphoma patients, the incidence of hepatitis and chemotherapy disruption due to HBV reactivation was significantly lower in the entecavir group than in the lamivudine group [397]. In a randomized controlled study comparing 121 patients treated with RCHOP chemotherapy for lymphoma as entecavir-treated (n=61) and lamivudine-treated groups (n=60), the entecavir group performed better in terms of the risk of hepatitis B reactivation (4% vs. 18%; P=0.001) and the rate of discontinuation of chemotherapy due to exacerbation of hepatitis B (1% vs. 11%; P=0.002) [398]. In a meta-analysis, entecavir prophylaxis was shown to prevent reactivation of hepatitis B more effectively than lamivudine prophylaxis [399]. In a retrospective comparative analysis of 419 CHB patients who received chemotherapy for solid cancers and lymphoma in Korea, HBeAg positivity, serum HBV DNA, and cancer types were identified as risk factors related to hepatitis B reactivation, and a risk stratification tool was developed based these factors. This study also confirmed that entecavir had a better preventive effect than lamivudine or telbivudine in the high-risk group [400]. However, since most prophylactic therapy studies were conducted on lymphoma patients, prospective studies evaluating various malignancies including solid cancers on appropriate antiviral agents and treatment duration for each cancer type and anticancer drug are needed. Considering the failure rate and tolerance rate, entecavir and tenofovir are expected to be relatively safe options. In a retrospective study evaluating patients receiving chemotherapy or immunosuppressant treatment for solid cancers, lymphoma, or rheumatic diseases, the tenofovir AF group (n=11) had similar effects compared to the entecavir group (n=66) in terms of HBV DNA reduction effect (-2.83±1.45 log IU/mL vs. -3.05±2.47 log IU/mL; P=0.86), HBV non-detection rate (78.8 vs. 90.9%; P=0.68), and renal function decline rate (-0.62±11.2 mL/min/1.73 m2 vs. -3.67±13.2 mL/min/1.73 m2; P=0.29), respectively [401]. Therefore, tenofovir AF can also be considered as a safe drug with good preventive effects. Such a conclusion regarding besifovir requires additional data. However, considering the characteristics of the drugs, entecavir, tenofovir AF, and besifovir are expected to be relatively safe choices as a prophylactic antiviral treatment for patients at risk or with renal and/or bone diseases.

[Recommendations]

1. If HBV infection is not confirmed, screening for HBsAg and anti-HBc before immunosuppression or chemotherapy is recommended. If either is positive, HBV DNA testing should be performed (A1).

2. If either HBsAg is positive or HBV DNA is detected, prophylactic antiviral therapy should be initiated before or at the start of immunosuppression or chemotherapy (A1). Antiviral agents should be selected based on comprehensive consideration of serum HBV DNA level, the intensity and duration of immunosuppression or chemotherapy, and presence or risk of renal and/or bone diseases. Entecavir or tenofovir (DF or AF) is preferred (B1), and besifovir could be an alternative option (C1).

3. In HBsAg-negative, HBV DNA-undetectable, and anti-HBc-positive patients, serum HBsAg and HBV DNA should be monitored during high-risk immunosuppression/chemotherapy and antiviral therapy started when HBV reactivation occurs (A1). In particular, when a regimen includes rituximab and/ or other B-cell depleting agents, antiviral therapy can be initiated at the start of immunosuppression or chemotherapy (B1).

4. Prophylactic antiviral therapy should be maintained for at least 6 months after the termination of immunosuppression or chemotherapy and for at least 12 months after the termination of therapy if rituximab and/or other B-cell depleting agents were included (B1).

5. Periodic monitoring of serum HBV DNA is recommended during and after prophylactic antiviral therapy (A1).

Hematopoietic stem cell transplantation

Patients with CHB who require HSCT for hematologic malignancies are immunosuppressed for a prolonged period due to the high-dose chemotherapy and hematological diseases itself. This increases the risk of hepatitis B reactivation and leads to a poor prognosis [402,403]. Therefore, all HSCT recipients should be tested for HBsAg, anti-HBc, and anti-HBs before transplantation. Quantitative serum HBV DNA test should be performed for patients who are HBsAg-positive/anti-HBc-positive or HBsAg-negative/anti-HBcpositive, regardless of whether they have anti-HBs or not. Hepatitis B vaccination should be administered and anti-HBs titers should be monitored for all hematologic malignancy patients who are HBsAg-negative/anti-HBs-negative. In small retrospective studies of HBsAg-positive recipients of allogeneic or autologous stem cell transplantation, prophylactic lamivudine treatment for 6–12 months significantly reduced the frequency of hepatitis B reactivation [367,404]. In another study, HBsAg-positive recipients of allogeneic stem cell transplants underwent prophylactic antiviral treatment for up to 6 months after termination of immunosuppressive therapy and were followed for 24 months after transplantation. The cumulative reactivation rate of hepatitis B was significantly higher in patients receiving lamivudine (24%) compared to patients receiving entecavir (2%). Recent meta-analyses have also demonstrated the efficacy of entecavir in preventing hepatitis B reactivation [405,406].

Hepatitis B reactivation is not infrequent in HSCT recipients with seronegative HBsAg/seropositive anti-HBc. In a prospective cohort study in which 62 HBsAg-negative and anti-HBc-positive allogeneic stem cell transplant recipients were followed for 48 weeks, the 2-year cumulative reactivation rate (detectable HBV DNA >10 IU/mL) was 40.8% [407]. In a retrospective study that followed HSCT recipients with past HBV infection in Korea for a median of 78 months, the hepatitis B reactivation rate was 2.6% (3/114). In another retrospective study with median follow-up of 21 months, hepatitis B reactivation occurred in four of 96 patients who received prophylactic antiviral therapy for 7 months and in 8 of 219 patients who did not receive prophylactic antiviral therapy [408,409]. According to observational studies, in HBsAg-negative and anti-HBc-positive allogeneic HSCT recipients, the 5-year cumulative incidence of hepatitis B reactivation is frequent (10.5–43.0%), so it is reasonable to commence prophylactic antiviral agents [410]. Various global guidelines recommend maintaining prophylactic antiviral treatment until 6–18 months after completion of HSCT; however, there is no unified consensus [144,151,368,411-413]. According to a recent study, hepatitis B reactivation was continuously reported up to 5–7 years after HSCT [410,414]. Therefore, it might be safe to maintain prophylactic antiviral therapy for a long time after the completion of HSCT, and further studies on the timing of discontinuation of prophylactic antiviral therapy are needed.

All HBsAg-positive solid organ transplantation and HSCT recipients should receive prophylactic antiviral therapy along with transplantation, and entecavir or tenofovir DF is preferred because long-term treatment is required. Although additional data are needed, given the characteristics of the drugs, entecavir, tenofovir AF, or besifovir could be recommended as a prophylactic antiviral treatment for patients at risk or with renal and/or bone diseases.

[Recommendations]

1. All HSCT recipients should be tested for HBsAg, anti-HBc, and anti-HBs before transplantation (A1).

2. Quantitative serum HBV DNA test should be performed for patients who are HBsAg-positive/anti-HBc-positive or HBsAg-negative/anti-HBc-positive, regardless of whether they have anti-HBs or not (B1).

3. Hepatitis B vaccination should be administered and anti-HBs titers should be monitored for all hematologic malignancy patients who are HBsAg-negative/anti-HBs-negative (C1).

4. All HBsAg-positive or HBV DNA-positive HSCT recipients should receive prophylactic antiviral therapy at the time of transplantation (A1). Antiviral agents should be selected based on comprehensive consideration of serum HBV DNA level, the intensity and duration of immunosuppression or chemotherapy, and presence or risk of renal and/or bone diseases. Entecavir or tenofovir (DF or AF) is preferred for long-term treatment (B1), and besifovir could be an alternative option (C1).

5. HBsAg-negative, HBV DNA undetectable, but anti-HBc-positive HSCT recipients are recommended to start prophylactic antiviral therapy at the time of transplantation (B1).

6. Prophylactic antiviral therapy should be maintained for at least 12 months after HSCT (B1).

Liver transplant patient

Prevention of HBV recurrence after liver transplantation is very important in patients receiving a liver transplantation. The current standard treatment is a combination of NA and hepatitis B immunoglobulin (HBIG), which lowers the posttransplant HBV reinfection rate to less than 5% [151,415]. Entecavir, tenofovir DF, and tenofovir AF are preferred antivirals because of their high potency and low rate of drug resistance [144,416,417]. As switching from other antiviral to tenofovir AF for HBV prophylaxis led to excellent virological response and a trend toward improvement in renal function in several recent studies, tenofovir AF is a reasonable option in posttransplant patients who have or are at risk for renal or bone disease [416,417]. With the advent of NA with high potency, efforts to reduce the duration of HBIG, which is expensive and inconvenient due to the parenteral route of administration, have been made in many transplantation centers [418-425]. In these studies, HBV recurrence rate was low at 0–13% when lifelong highly potent NAs were combined with a short period of HBIG (7 days to 1 year) after transplantation. In addition, HBIG-free prophylaxis using NA alone showed 8-year HBsAg negativity of 92% and 88%, respectively, in two studies, showing that NA monotherapy can effectively prevent posttransplant HBV recurrence [426,427]. NA monotherapy without HBIG has shown to be safe and effective in preventing HBV recurrence and transplant failure and warrants favorable long-term survival rate in liver transplant patients. Therefore, adjusting the period of HBIG use by evaluating the risk of HBV recurrence at the time of transplantation for each patient is recommended. Patients with a low risk of recurrence (HBV DNA negative at liver transplantation) can receive a short course or HBIG-free regimens but need continued monoprophylaxis with a potent NA. Conversely, patients with a high risk of recurrence (HBV DNA positive at liver transplantation, HDV coinfection, or poor adherence to NA therapy) should receive lifelong combination of HBIG and a potent NA [428-430].

Since a HBsAg-negative/anti-HBc-positive donor liver is suggestive of occult HBV infection, it is appropriate to preferentially transplant it to a HBsAg-positive recipient considering the possibility HBV reactivation after liver transplantation. HBsAg-negative patients receiving anti-HBc-positive liver grafts have different HBV reactivation rates from 10–80% depending on recipient’s immunization against HBV [431]. The HBV incidence rate was highest at 47.8% when the recipient was not immune to HBV (anti-HBs-negative, anti-HBc-negative) and lowest at 1.4% when both anti-HBs and anti-HBc were positive [432]. The HBV incidence rate was 13.1% in recipients with anti-HBs-negative and anti-HBc-positive, and 9.7% in recipients who developed immunity through HBV vaccine (anti-HBs-positive, anti-HBc-negative). In HBsAg-negative patients receiving liver transplantation from an anti-HBc-positive donor, administration of prophylactic antiviral drugs significantly reduced the incidence of HBV infection from 47.8% to 12% in recipients who were not immune to hepatitis B (anti-HBs-negative, anti-HBc-negative), from 15.2% to 3.4% in those with anti-HBc-positivity, and from 9.7% to 0% in those who developed immunity through HBV vaccine (anti-HBs-positive, anti-HBc-negative) [431]. Thus, treatment of patients receiving anti-HBc-positive liver grafts should depend on their immunization against HBV (Fig. 6). NAs should be started immediately after surgery and be continued. Further studies are needed to determine whether antiviral therapy is unnecessary in patients who are both anti-HBs and anti-HBc-positive. Lamivudine has been used mostly because of its favorable cost-effectiveness [433,434]; however, drugs with high potency and low rate of resistance such as entecavir, tenofovir DF, and tenofovir AF are expected to be used more in the future.

Figure 6.

Strategies after liver transplantation in patients receiving anti-HBc-positive liver graft. Anti-HBc, antibody to HBcAg; HBsAg, hepatitis B surface antigen; NA, nucleos(t)ide analogue; HBIG, hepatitis B immunoglobulin; anti-HBs, antibody to HBsAg; HBV, hepatitis B virus. *Pooled data from the study by Cholongitas et al [431].

[Recommendations]

1. In HBV-related liver transplant recipients, prophylactic combination of HBIG and a potent NA posttransplantation is recommended for the prevention of HBV recurrence (B1).

- Patients with a low risk of recurrence (HBV DNA negative at liver transplantation) can receive a short course or HBIG-free regimens but need continued monoprophylaxis with a potent NA (B1).

- Patients with a high risk of recurrence (HBV DNA positive at liver transplantation, HDV coinfection, or poor adherence to NA therapy) should receive lifelong combination of HBIG and a potent NA (B1).

2. Considering the risk of renal dysfunction and/or bone diseases after liver transplantation, potent antivirals with low rate of resistance such as entecavir and tenofovir (AF or DF) are preferred antiviral drugs (B1), and besifovir can be considered (C1).

3. HBsAg-negative patients receiving anti-HBc-positive liver grafts show variation in HBV reactivation rate depending on the recipient’s immunization status against HBV (Fig. 6) and should receive NA therapy accordingly (B1).

Non-liver solid organ transplant recipients

All non-liver solid organ transplantation recipients should be evaluated for HBV infection and immunity with HBsAg, anti-HBc, and anti-HBs. Patients who are HBsAg-positive should undergo ALT and HBV DNA measurements and staging with biopsy or non-invasive fibrosis tests to determine whether advanced fibrosis or cirrhosis is present. HBsAg-positive renal transplant recipients are at high risk for persistent viral activity or reactivation and have a significantly higher mortality rate due to liver-related complications such as liver cirrhosis and HCC [435,436]. Recent reports indicate that antiviral therapy significantly decreased the HBV reactivation rate and increased the survival of HBsAg-positive renal transplant recipients [437,438]. Therefore, all HBsAg-positive organ transplant recipients should receive antiviral therapy before or right after liver transplantation. Entecavir and tenofovir (AF or DF) can be considered because of high potency and the low rate of resistance in long-term use, and patients who are at risk for renal dysfunction should receive entecavir and tenofovir AF preferably. With the introduction of effective antiviral therapy, patients with CHB and those with compensated cirrhosis without portal hypertension became able to receive solid organ transplants, and successful cases of kidney transplantation have been reported [439]. However, in patients with advanced or decompensated cirrhosis who are at risk of liver failure, simultaneous transplantation of the liver and other organs, such as kidneys, heart, and lungs, should be considered.

Recurrence of HBV in HBsAg-negative solid organ transplant recipients can be divided into two cases: one with transmission of HBV from a HBsAg-negative and anti-HBc-positive donor and two with reactivation in an anti-HBs-negative and anti-HBc-positive recipient after transplantation. The rate of HBsAg positivity after kidney transplantation caused by HBV transmission from HBsAg-negative and anti-HBc-positive kidney donor was very low at 0% to 3% [440-442]. The HBV reactivation rate in anti-HBs-negative and anti-HBc-positive kidney recipients varied from 0% to 12% depending on the recipient’s immunization against HBV [443-453]. The reactivation rate was significantly higher in recipients with antiHBc-positive and anti-HBs-negative (5.6–12.0%) than in those with anti-HBc-positive and anti-HBs positive (1.1–2.7%) [443,452,453]. Thus, HBsAg-negative, anti-HBc-positive recipients should be monitored for HBsAg positivity after solid organ transplantation, and NA of entecavir, tenofovir DF, or tenofovir AF should be administered in case of HBsAg detection.

[Recommendations]

1. All non-liver solid organ transplantation recipients should be tested for HBsAg, anti-HBc, and anti-HBs before transplantation (A1).

2. All HBsAg-positive or HBV DNA-positive solid organ transplant recipients should start prophylactic antiviral treatment at the time of transplantation (A1). Considering the risk of renal dysfunction and/or bone diseases after transplantation, potent antivirals with low rate of resistance such as entecavir and tenofovir (AF or DF) are preferred (B1), and besifovir can be considered (C1).

3. HBsAg-negative, HBV DNA-undetectable, but anti-HBc-positive solid organ transplant recipients should be monitored for HBV infection after transplantation (B1).

Coinfection with other viruses

Hepatitis C virus (HCV) coinfection

In patients with CHB, the anti-HCV-positivity rate varies from 1.5% to 2.4% in Korea [454,455]. Patients with HBV/HCV coinfection have more severe necroinflammation and fibrosis as well as high risk of cirrhosis and HCC compared to those with a monoinfection [456-459]. Patients with HBV/HCV coinfection should be tested for HBV DNA and HCV RNA before antiviral treatment for each virus. If HCV RNA is detectable, antiviral treatment for HCV (direct acting antiviral [DAA] therapy) should be started. If HBV DNA is detectable, check for ALT, HBV DNA, and presence of liver cirrhosis and treat according to the HBV guidelines, as with HBV monoinfection (Fig. 3).

There is a potential risk of HBV reactivation during DAA therapy or after clearance of HCV. In a meta-analysis, HBV DNA was newly detected or the level increased in 14.1% of patients after administration of DAAs for 4–12 weeks, whereas active hepatitis accompanied by ALT elevation was noted in 12.2% of patients [460]. Therefore, patients with HBV/HCV coinfection should be monitored for the risk of HBV reactivation by monitoring ALT and HBV DNA level during and after DAA therapy. In addition, if HBV is indicated for treatment at the time of starting DAA therapy for HCV, NA therapy for HBV should be started. In patients with a history of cirrhosis or HCC, simultaneous NA therapy for HBV can be considered along with DAA therapy for HCV to reduce the risk of liver failure risk resulting from HBV reactivation [461,462]. Entecavir, tenofovir DF, and tenofovir AF are preferred choices. There was no significant drug-drug interaction between NAs for HBV (entecavir, tenofovir DF, tenofovir AF) and DAAs or HCV. However, when selecting antiviral drugs in patients with HBV/HCV/HIV triple infection, drug-drug interactions should be considered [463,464].

In HBsAg-negative and anti-HBc-positive patients with HCV, HBV reactivation rate (HBsAg positivity) during DAA therapy is very low at 0–0.1% [465-467]. Although HBV reactivation rate during DAA therapy is low, HBsAg and HBV DNA should be tested to check for HBV reactivation in cases of ALT elevation.

[Recommendations]

1. If HCV RNA is detectable in patients with HBV/HCV coinfection, antiviral treatment for HCV should be started (A1).

2. If antiviral treatment for HBV is indicated in HBV/HCV coinfection, antiviral treatment for HBV should be started (Fig. 3) (A1).

3. Patients with HBV/HCV coinfection should be monitored for the risk of HBV reactivation by monitoring ALT and HBV DNA levels during and after DAA therapy (B1).

4. HBsAg-negative and anti-HBc-positive patients with HCV infection are at very low risk of reactivation with HCV-DAA therapy; however, HBsAg and HBV DNA should be tested to check for HBV reactivation in cases of ALT elevation (B1).

HDV coinfection

HDV is a defective RNA virus that does not encode its own envelope proteins, enters the liver through HBsAg-expressing hepatocytes, and replicates the HDV genome to express HDV antigens [468]. Thus, HDV infection occurs when people become infected with both HBV and HDV simultaneously (co-infection) or in CHB patients newly infected with HDV (super-infection). An estimated 12 million people worldwide have experienced HDV infection, and approximately 4.5% of HBV patients are estimated to have HDV [469]. In one Korean study, the HDV coinfection rate was 0.3% in 940 patients with CHB, including 75 patients with HCC [470]. In another study with 194 patients including 64 CHB patients and 130 HCC patients, the HDV infection rate was 3.6% [471]. Compared to HBV monoinfection, HBV/HDV coinfection is associated with higher rates of cirrhosis and HCC [472-474].

HDV infection can be diagnosed by detecting anti-HDV or HDV RNA in the serum or by detecting HDV antigens in liver tissues using immunohistochemistry. In patients with HDV infection, degree of hepatic fibrosis should be evaluated by liver biopsy or non-invasive methods irrelevant to HDV RNA detection. In HBV/HDV coinfected patients having advanced fibrosis or cirrhosis or patients who are indicated for antiviral treatment of CHB, NA therapy should be administered to prevent the progression of liver disease. However, NA therapy is not recommended in cases where HBV treatment is not indicated because NAs do not inhibit HDV replication.

All patients with HDV RNA positivity are eligible for treatment regardless of the degree of liver disease, and the primary endpoint of treatment is the suppression of HDV replication, which is usually accompanied by normalization of ALT level and improvement of necroinflammation on liver biopsy. The current standard treatment for HDV is weekly peginterferon alpha by subcutaneous injection for 48 weeks, and a sustained viral response at 24 weeks has been reported in 23–28% of patients [475,476]. A sustained virological response can be predicted by measuring serum HDV RNA level after 24 weeks of peginterferon alpha therapy [477]. However, relapse is frequent during long-term follow-up, as seen in one study with an average follow-up of 4.3 years where sustained virological response was maintained at only 12% [478]. Combination therapy using NAs and peginterferon alfa did not improve virological response compared to peginterferon alfa monotherapy [479]. In a small study using extended peginterferon alfa therapy for 24 months, 47% of patients achieved sustained virological response during an average follow-up of 19.5 months after treatment, but further studies are needed [480].

Because of the low efficacy of peginterferon alpha, new therapeutic antiviral agents for HDV are under development. Bulevirtide (BLV), a drug in phase III study, is an entry inhibitor for HDV. It is a lipopeptide that competitively acts on sodium taurocholate cotransporting polypeptide (NTCP), an entry receptor shared by HBV and HDV viruses [481]. In 2020, BLV was first approved as an antiviral agent for HDV under conditional authorization by the European Medical Agency for patients with compensated liver disease and positive HDV RNA with or without NAs. BLV 2 mg is administered subcutaneously daily and can be maintained as long as the virologic response persists. According to interim analysis of the ongoing phase 3 study, BLV treatment for 24 weeks resulted in significant decrease in HDV RNA and ALT. Virologic response (undetectable HDV RNA or ≥2 log10 IU/mL decrease from baseline) in patients with no treatment, BLV 2 mg, and BLV 10 mg was 4%, 55%, and 68%, respectively. Combined response (undetectable HDV RNA or ≥2 log10 IU/mL decrease from baseline and normal ALT) rate in patients with no treatment, BLV 2 mg, and BLV 10 mg was 0%, 37%, and 28%, respectively [482].

Another drug in phase III study is lonafarnib, an oral prenylation inhibitor that reduces HDV virus load by blocking prenylation, a process involved in virus assembly, replication, and subsequent hepatocyte infectivity [483]. Combination with ritonavir is known to increase by 4–5-fold in systemic exposure and to decrease gastrointestinal side effects. Virologic responses (undetectable HDV RNA or ≥2 log10 IU/mL decrease from baseline) at the end of treatment were reached in 46% and 89% of patients receiving the all-oral regimen of lonafarnib 50 mg bid + ritonavir, and combination regimen of lonafarnib (25 or 50 mg bid) + ritonavir + peginterferon alpha, respectively [484].

Antiviral agents for HDV under phase II study include peginterferon lambda, REP 2139 (nucleic acid polymers), and JNJ-3989 (RNAi) [485,486].

[Recommendations]

1. In CHB patients with low HBV DNA and elevated ALT levels, HDV screening with anti-HDV test can be considered, unless there is another suspected cause of elevated liver enzyme (C1).

2. In CHB patients with HDV coinfection having advanced fibrosis or cirrhosis, or patients who are indicated for antiviral treatment of CHB, NA therapy should be administered to prevent the progression of liver disease (B1).

3. CHB patients with HDV coinfection are recommended to be treated with peginterferon alfa for at least 1 year (A2), and administration of a new therapeutic agent can be considered according to the clinical course of the patient (C1).

Notes

Authors’ contributions

Jang JW, directed the guideline committee, and outlined and supervised the manuscript writing and editing; Choi J and An J edited the manuscript. All the committee members participated drafting the manuscript; Jang JW (introduction), Kang W (natural history), An J (emerging markers of HBV infection, and patients with HCC), Kwon JH (treatment indication), Kim TH (new drugs for a functional cure), Park JY (cessation of treatment and monitoring after antiviral treatment), Choi J (patients with renal dysfunction or metabolic bone disease and supplementary material), Yu SJ (patients on immunosuppression or chemotherapy, and liver transplant patients), and Chon YE (liver transplant patient, nonliver organ transplant recipients, and coinfection with other viruses) contributed to the manuscript writing.

Conflicts of Interest

Jang JW: Received honoraria from Gilead, Abbvie, Bayer, Roche, Sysmex, Ipsen. Received grants from Sysmex, Gilead, Yuhan, Dong-A ST, Hanmi

Park JY: Received grants from Gilead, Pfizer, MSD, BMS, Roche, Astrazeneca, Aramchol, Novonorodisk, Yuhan, Hanmi

Kwon JH: Received honoraria and grants from Gilead, Abbvie

Yu SJ: Received honoraria and grants from Gilead, Abbvie, BMS, Daewoong, Yuhan, Chongkundang Pharm, Dong-A ST, Samil, Samjin

Kang W: Received honoraria and grants from Gilead, BMS, Abbvie, Eisai, Bayer, Roche, MSD, Yuhan, Dong-A ST, Ildong, Daewoong, Samil, Boryung

Chon YE: Received honoraria from BMS, Dong-A ST, Abbvie, Daewoong

Kim TH: Nothing to declare

Choi J: Received honoraria from Gilead, Abbvie, Daewoong

An J: Received honoraria from Yuhan

Abbreviations

ACLF

acute-on-chronic liver failure

AF

alafenamide fumarate

ALT

alanine aminotransferase

anti-HBe

antibody to HBeAg

anti-HBs

antibody to HBsAg

anti-HBc

antibody to HBcAg

APRI

aspartate aminotransferase-to-platelet ratio index

ASO

antisense oligonucleotides

BCP

basal core promoter

BLV

bulevirtide

CAGE-B

cirrhosis and age

CAR

chimeric antigen receptor

cccDNA

covalently closed circular DNA

CHB

chronic hepatitis B

CI

confidence interval

DAA

direct acting antiviral

DF

disoproxil fumarate

DMARDs

disease-modifying antirheumatic drugs

EBRT

external beam radiation therapy

eGFR

estimated glomerular filtration rate

EOT

end-of-treatment

FIB-4

fibrosis-4

GRADE

Grading of Recommendations, Assessment, Development, and Evaluation

HBcAg

hepatitis B core antigen

HBcrAg

hepatitis B core-related antigen

HBeAg

hepatitis B e antigen

HBIG

hepatitis B immunoglobulin

HBsAg

hepatitis B surface antigen

HBV

hepatitis B virus

HCC

hepatocellular carcinoma

HCV

hepatitis C virus

HDV

hepatitis D virus

HIV

human immunodeficiency virus

HR

hazard ratio

HSCT

hematopoietic stem-cell transplantation

IBD

inflammatory bowel disease

KASL

Korean Association for the Study of the Liver

MELD

Model for End-stage Liver Disease

NA

nucleos(t)ide analogue

NTCP

sodium taurocholate cotransporting polypeptide

OR

odds ratio

p22Cr

p22 corerelated antigen

PAGE-B

platelets, age, gender, and hepatitis B scores

PAMPs

pathogen-associated molecular patterns

PC

precore

pgRNA

pregenomic RNA

R-CHOP

rituximab-CHOP

RA

rheumatoid arthritis

RCT

randomized controlled trial

REACH-B

risk estimation for HCC in chronic hepatitis B

RIG

retinoic acidinducible gene

RISC

RNA-induced silencing complex

RNAi

RNA interference

RR

relative risk

SAGE-B

stiffness and age

siRNA

small interfering RNA

SVPs

subviral particles

TACE

transarterial chemoembolization

TE

transient elastography

THRI

Toronto HCC risk index

TLR

toll-like receptor

TNFα

tumor necrosis factor-alpha

ULN

upper limit of normal

References

1. Terrault NA, Bzowej NH, Chang KM, Hwang JP, Jonas MM, Murad MH, et al. AASLD guidelines for treatment of chronic hepatitis B. Hepatology 2016;63:261–283.
2. Hui CK, Leung N, Yuen ST, Zhang HY, Leung KW, Lu L, et al. Natural history and disease progression in Chinese chronic hepatitis B patients in immune-tolerant phase. Hepatology 2007;46:395–401.
3. Tran TT. Immune tolerant hepatitis B: a clinical dilemma. Gastroenterol Hepatol (N Y) 2011;7:511–516.
4. Chu CM, Hung SJ, Lin J, Tai DI, Liaw YF. Natural history of hepatitis B e antigen to antibody seroconversion in patients with normal serum aminotransferase levels. Am J Med 2004;116:829–834.
5. Lee HA, Lee HW, Kim IH, Park SY, Sinn DH, Yu JH, et al. Extremely low risk of hepatocellular carcinoma development in patients with chronic hepatitis B in immune-tolerant phase. Aliment Pharmacol Ther 2020;52:196–204.
6. Mason WS, Gill US, Litwin S, Zhou Y, Peri S, Pop O, et al. HBV DNA integration and clonal hepatocyte expansion in chronic hepatitis B patients considered immune tolerant. Gastroenterology 2016;151:986–998. e4.
7. Kim GA, Lim YS, Han S, Choi J, Shim JH, Kim KM, et al. High risk of hepatocellular carcinoma and death in patients with immunetolerant-phase chronic hepatitis B. Gut 2018;67:945–952.
8. Chu CJ, Hussain M, Lok AS. Hepatitis B virus genotype B is associated with earlier HBeAg seroconversion compared with hepatitis B virus genotype C. Gastroenterology 2002;122:1756–1762.
9. Livingston SE, Simonetti JP, Bulkow LR, Homan CE, Snowball MM, Cagle HH, et al. Clearance of hepatitis B e antigen in patients with chronic hepatitis B and genotypes A, B, C, D, and F. Gastroenterology 2007;133:1452–1457.
10. Tsai SL, Chen PJ, Lai MY, Yang PM, Sung JL, Huang JH, et al. Acute exacerbations of chronic type B hepatitis are accompanied by increased T cell responses to hepatitis B core and e antigens. Implications for hepatitis B e antigen seroconversion. J Clin Invest 1992;89:87–96.
11. Lee PI, Chang MH, Lee CY, Hsu HY, Chen JS, Chen PJ, et al. Changes of serum hepatitis B virus DNA and aminotransferase levels during the course of chronic hepatitis B virus infection in children. Hepatology 1990;12(4 Pt 1):657–660.
12. Lok AS, Lai CL. Acute exacerbations in Chinese patients with chronic hepatitis B virus (HBV) infection. Incidence, predisposing factors and etiology. J Hepatol 1990;10:29–34.
13. McMahon BJ. The natural history of chronic hepatitis B virus infection. Hepatology 2009;49(5 Suppl):S45–S55.
14. Hsu YS, Chien RN, Yeh CT, Sheen IS, Chiou HY, Chu CM, et al. Long-term outcome after spontaneous HBeAg seroconversion in patients with chronic hepatitis B. Hepatology 2002;35:1522–1527.
15. Bortolotti F, Guido M, Bartolacci S, Cadrobbi P, Crivellaro C, Noventa F, et al. Chronic hepatitis B in children after e antigen seroclearance: final report of a 29-year longitudinal study. Hepatology 2006;43:556–562.
16. Yim HJ, Lok AS. Natural history of chronic hepatitis B virus infection: what we knew in 1981 and what we know in 2005. Hepatology 2006;43(2 Suppl 1):S173–S181.
17. Lok AS, Lai CL, Wu PC, Leung EK, Lam TS. Spontaneous hepatitis B e antigen to antibody seroconversion and reversion in Chinese patients with chronic hepatitis B virus infection. Gastroenterology 1987;92:1839–1843.
18. Sheen IS, Liaw YF, Tai DI, Chu CM. Hepatic decompensation associated with hepatitis B e antigen clearance in chronic type B hepatitis. Gastroenterology 1985;89:732–735.
19. Martinot-Peignoux M, Boyer N, Colombat M, Akremi R, Pham BN, Ollivier S, et al. Serum hepatitis B virus DNA levels and liver histology in inactive HBsAg carriers. J Hepatol 2002;36:543–546.
20. Zacharakis GH, Koskinas J, Kotsiou S, Papoutselis M, Tzara F, Vafeiadis N, et al. Natural history of chronic HBV infection: a cohort study with up to 12 years follow-up in North Greece (part of the Interreg I-II/EC-project). J Med Virol 2005;77:173–179.
21. de Franchis R, Meucci G, Vecchi M, Tatarella M, Colombo M, Del Ninno E, et al. The natural history of asymptomatic hepatitis B surface antigen carriers. Ann Intern Med 1993;118:191–194.
22. Fattovich G. Natural history and prognosis of hepatitis B. Semin Liver Dis 2003;23:47–58.
23. Chen YC, Sheen IS, Chu CM, Liaw YF. Prognosis following spontaneous HBsAg seroclearance in chronic hepatitis B patients with or without concurrent infection. Gastroenterology 2002;123:1084–1089.
24. Lok AS, McMahon BJ. Chronic hepatitis B: update 2009. Hepatology 2009;50:661–662.
25. Funk ML, Rosenberg DM, Lok AS. World-wide epidemiology of HBeAg-negative chronic hepatitis B and associated precore and core promoter variants. J Viral Hepat 2002;9:52–61.
26. Lok AS, Akarca U, Greene S. Mutations in the pre-core region of hepatitis B virus serve to enhance the stability of the secondary structure of the pre-genome encapsidation signal. Proc Natl Acad Sci U S A 1994;91:4077–4081.
27. Okamoto H, Tsuda F, Akahane Y, Sugai Y, Yoshiba M, Moriyama K, et al. Hepatitis B virus with mutations in the core promoter for an e antigen-negative phenotype in carriers with antibody to e antigen. J Virol 1994;68:8102–8110.
28. Tacke F, Gehrke C, Luedde T, Heim A, Manns MP, Trautwein C. Basal core promoter and precore mutations in the hepatitis B virus genome enhance replication efficacy of Lamivudine-resistant mutants. J Virol 2004;78:8524–8535.
29. Croagh CM, Bell SJ, Slavin J, Kong YX, Chen RY, Locarnini S, et al. Increasing hepatitis B viral load is associated with risk of significant liver fibrosis in HBeAg-negative but not HBeAg-positive chronic hepatitis B. Liver Int 2010;30:1115–1122.
30. Yoo BC, Park JW, Kim HJ, Lee DH, Cha YJ, Park SM. Precore and core promoter mutations of hepatitis B virus and hepatitis B e antigen-negative chronic hepatitis B in Korea. J Hepatol 2003;38:98–103.
31. Raimondo G, Allain JP, Brunetto MR, Buendia MA, Chen DS, Colombo M, et al. Statements from the Taormina expert meeting on occult hepatitis B virus infection. J Hepatol 2008;49:652–657.
32. Hoofnagle JH, Doo E, Liang TJ, Fleischer R, Lok AS. Management of hepatitis B: summary of a clinical research workshop. Hepatology 2007;45:1056–1075.
33. Hadziyannis SJ, Vassilopoulos D. Hepatitis B e antigen-negative chronic hepatitis B. Hepatology 2001;34(4 Pt 1):617–624.
34. Wu TT, Hsu HC, Chen DS, Sheu JC, Su IJ, Chen SL, et al. Clearance of hepatitis B surface antigen (HBsAg) after surgical resection of hepatocellular carcinoma. J Hepatol 1987;4:45–51.
35. Liaw YF, Sheen IS, Chen TJ, Chu CM, Pao CC. Incidence, determinants and significance of delayed clearance of serum HBsAg in chronic hepatitis B virus infection: a prospective study. Hepatology 1991;13:627–631.
36. Tout I, Loureiro D, Mansouri A, Soumelis V, Boyer N, Asselah T. Hepatitis B surface antigen seroclearance: immune mechanisms, clinical impact, importance for drug development. J Hepatol 2020;73:409–422.
37. Cornberg M, Lok AS, Terrault NA, Zoulim F, ; 2019 EASL-AASLD HBV Treatment Endpoints Conference Faculty. Guidance for design and endpoints of clinical trials in chronic hepatitis B - report from the 2019 EASL-AASLD HBV Treatment Endpoints Conference‡. J Hepatol 2020;72:539–557.
38. Raimondo G, Locarnini S, Pollicino T, Levrero M, Zoulim F, Lok AS, et al. Update of the statements on biology and clinical impact of occult hepatitis B virus infection. J Hepatol 2019;71:397–408.
39. Chu CM, Liaw YF. HBsAg seroclearance in asymptomatic carriers of high endemic areas: appreciably high rates during a long-term follow-up. Hepatology 2007;45:1187–1192.
40. Liu J, Yang HI, Lee MH, Lu SN, Jen CL, Wang LY, et al. Incidence and determinants of spontaneous hepatitis B surface antigen seroclearance: a community-based follow-up study. Gastroenterology 2010;139:474–482.
41. Nam SW, Jung JJ, Bae SH, Choi JY, Yoon SK, Cho SH, et al. Clinical outcomes of delayed clearance of serum HBsAG in patients with chronic HBV infection. Korean J Intern Med 2007;22:73–76.
42. Kim GA, Lee HC, Kim MJ, Ha Y, Park EJ, An J, et al. Incidence of hepatocellular carcinoma after HBsAg seroclearance in chronic hepatitis B patients: a need for surveillance. J Hepatol 2015;62:1092–1099.
43. Seto WK, Wong DK, Fung J, Huang FY, Liu KS, Lai CL, et al. Linearized hepatitis B surface antigen and hepatitis B core-related antigen in the natural history of chronic hepatitis B. Clin Microbiol Infect 2014;20:1173–1180.
44. Yip TC, Chan HL, Wong VW, Tse YK, Lam KL, Wong GL. Impact of age and gender on risk of hepatocellular carcinoma after hepatitis B surface antigen seroclearance. J Hepatol 2017;67:902–908.
45. Yip TC, Wong GL, Chan HL, Tse YK, Lam KL, Lui GC, et al. HBsAg seroclearance further reduces hepatocellular carcinoma risk after complete viral suppression with nucleos(t)ide analogues. J Hepatol 2019;70:361–370.
46. Kim GA, Lim YS, An J, Lee D, Shim JH, Kim KM, et al. HBsAg seroclearance after nucleoside analogue therapy in patients with chronic hepatitis B: clinical outcomes and durability. Gut 2014;63:1325–1332.
47. Choi J, Yoo S, Lim YS. Comparison of long-term clinical outcomes between spontaneous and therapy-induced HBsAg seroclearance. Hepatology 2021;73:2155–2166.
48. Yip TC, Wong VW, Tse YK, Liang LY, Hui VW, Zhang X, et al. Similarly low risk of hepatocellular carcinoma after either spontaneous or nucleos(t)ide analogue-induced hepatitis B surface antigen loss. Aliment Pharmacol Ther 2021;53:321–331.
49. Park Y, Lee JH, Sinn DH, Park JY, Kim MA, Kim YJ, et al. Risk and risk score performance of hepatocellular carcinoma development in patients with hepatitis B surface antigen seroclearance. Clin Transl Gastroenterol 2021;12:e00290.
50. Di Bisceglie AM, Lombardero M, Teckman J, Roberts L, Janssen HL, Belle SH, et al. Determination of hepatitis B phenotype using biochemical and serological markers. J Viral Hepat 2017;24:320–329.
51. Hsu YN, Pan CQ, Abbasi A, Xia V, Bansal R, Hu KQ. Clinical presentation and disease phases of chronic hepatitis B using conventional versus modified ALT criteria in Asian Americans. Dig Dis Sci 2014;59:865–871.
52. Yao K, Liu J, Wang J, Yan X, Xia J, Yang Y, et al. Distribution and clinical characteristics of patients with chronic hepatitis B virus infection in the grey zone. J Viral Hepat 2021;28:1025–1033.
53. Bonacci M, Lens S, Mariño Z, Londoño MC, Rodríguez-Tajes S, Mas A, et al. Anti-viral therapy can be delayed or avoided in a significant proportion of HBeAg-negative Caucasian patients in the grey zone. Aliment Pharmacol Ther 2018;47:1397–1408.
54. Choi GH, Kim GA, Choi J, Han S, Lim YS. High risk of clinical events in untreated HBeAg-negative chronic hepatitis B patients with high viral load and no significant ALT elevation. Aliment Pharmacol Ther 2019;50:215–226.
55. Hoang JK, Yang HI, Le A, Nguyen NH, Lin D, Vu VD, et al. Lower liver cancer risk with antiviral therapy in chronic hepatitis B patients with normal to minimally elevated ALT and no cirrhosis. Medicine (Baltimore) 2016;95:e4433.
56. Yuen MF, Tanaka Y, Fong DY, Fung J, Wong DK, Yuen JC, et al. Independent risk factors and predictive score for the development of hepatocellular carcinoma in chronic hepatitis B. J Hepatol 2009;50:80–88.
57. Teng W, Chang TT, Yang HI, Peng CY, Su CW, Su TH, et al. Risk scores to predict HCC and the benefits of antiviral therapy for CHB patients in gray zone of treatment guidelines. Hepatol Int 2021;15:1421–1430.
58. Lin SM, Sheen IS, Chien RN, Chu CM, Liaw YF. Long-term beneficial effect of interferon therapy in patients with chronic hepatitis B virus infection. Hepatology 1999;29:971–975.
59. Raffetti E, Fattovich G, Donato F. Incidence of hepatocellular carcinoma in untreated subjects with chronic hepatitis B: a systematic review and meta-analysis. Liver Int 2016;36:1239–1251.
60. Lee KJ, Han KH, Chun JY, Moon YM, Lee SI, Park IS, et al. Natural history of chronic hepatitis type B throughout long-term follow-up. Korean J Gastroenterol 1997;29:343–351.
61. Liu S, Zhang H, Gu C, Yin J, He Y, Xie J, et al. Associations between hepatitis B virus mutations and the risk of hepatocellular carcinoma: a meta-analysis. J Natl Cancer Inst 2009;101:1066–1082.
62. Yang HI, Yeh SH, Chen PJ, Iloeje UH, Jen CL, Su J, et al. Associations between hepatitis B virus genotype and mutants and the risk of hepatocellular carcinoma. J Natl Cancer Inst 2008;100:1134–1143.
63. Tseng TC, Liu CJ, Yang HC, Chen CL, Yang WT, Tsai CS, et al. Higher proportion of viral basal core promoter mutant increases the risk of liver cirrhosis in hepatitis B carriers. Gut 2015;64:292–302.
64. Jang JW, Chun JY, Park YM, Shin SK, Yoo W, Kim SO, et al. Mutational complex genotype of the hepatitis B virus X/precore regions as a novel predictive marker for hepatocellular carcinoma. Cancer Sci 2012;103:296–304.
65. Kim JK, Chang HY, Lee JM, Baatarkhuu O, Yoon YJ, Park JY, et al. Specific mutations in the enhancer II/core promoter/precore regions of hepatitis B virus subgenotype C2 in Korean patients with hepatocellular carcinoma. J Med Virol 2009;81:1002–1008.
66. Kao JH, Chen PJ, Lai MY, Chen DS. Basal core promoter mutations of hepatitis B virus increase the risk of hepatocellular carcinoma in hepatitis B carriers. Gastroenterology 2003;124:327–334.
67. Wei F, Zheng Q, Li M, Wu M. The association between hepatitis B mutants and hepatocellular carcinoma: a meta-analysis. Medicine (Baltimore) 2017;96:e6835.
68. Chen CJ, Yang HI, Su J, Jen CL, You SL, Lu SN, et al. Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level. JAMA 2006;295:65–73.
69. Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 2004;127(5 Suppl 1):S35–S50.
70. Iloeje UH, Yang HI, Su J, Jen CL, You SL, Chen CJ, et al. Predicting cirrhosis risk based on the level of circulating hepatitis B viral load. Gastroenterology 2006;130:678–686.
71. Bravi F, Tavani A, Bosetti C, Boffetta P, La Vecchia C. Coffee and the risk of hepatocellular carcinoma and chronic liver disease: a systematic review and meta-analysis of prospective studies. Eur J Cancer Prev 2017;26:368–377.
72. Kennedy OJ, Roderick P, Buchanan R, Fallowfield JA, Hayes PC, Parkes J. Coffee, including caffeinated and decaffeinated coffee, and the risk of hepatocellular carcinoma: a systematic review and dose-response meta-analysis. BMJ Open 2017;7:e013739.
73. Petrick JL, Freedman ND, Graubard BI, Sahasrabuddhe VV, Lai GY, Alavanja MC, et al. Coffee consumption and risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma by sex: the liver cancer pooling project. Cancer Epidemiol Biomarkers Prev 2015;24:1398–1406.
74. Zhou YY, Zhu GQ, Liu T, Zheng JN, Cheng Z, Zou TT, et al. Systematic review with network meta-analysis: antidiabetic medication and risk of hepatocellular carcinoma. Sci Rep 2016;6:33743.
75. Hwang IC, Chang J, Kim K, Park SM. Aspirin use and risk of hepatocellular carcinoma in a national cohort study of Korean adults. Sci Rep 2018;8:4968.
76. Lee M, Chung GE, Lee JH, Oh S, Nam JY, Chang Y, et al. Antiplatelet therapy and the risk of hepatocellular carcinoma in chronic hepatitis B patients on antiviral treatment. Hepatology 2017;66:1556–1569.
77. Chen CI, Kuan CF, Fang YA, Liu SH, Liu JC, Wu LL, et al. Cancer risk in HBV patients with statin and metformin use: a population-based cohort study. Medicine (Baltimore) 2015;94:e462.
78. Hsiang JC, Wong GL, Tse YK, Wong VW, Yip TC, Chan HL. Statin and the risk of hepatocellular carcinoma and death in a hospitalbased hepatitis B-infected population: a propensity score landmark analysis. J Hepatol 2015;63:1190–1197.
79. Kim G, Jang SY, Nam CM, Kang ES. Statin use and the risk of hepatocellular carcinoma in patients at high risk: a nationwide nested case-control study. J Hepatol 2018;68:476–484.
80. Pradelli D, Soranna D, Scotti L, Zambon A, Catapano A, Mancia G, et al. Statins and primary liver cancer: a meta-analysis of observational studies. Eur J Cancer Prev 2013;22:229–234.
81. Singh S, Singh PP, Singh AG, Murad MH, Sanchez W. Statins are associated with a reduced risk of hepatocellular cancer: a systematic review and meta-analysis. Gastroenterology 2013;144:323–332.
82. Zhou YY, Zhu GQ, Wang Y, Zheng JN, Ruan LY, Cheng Z, et al. Systematic review with network meta-analysis: statins and risk of hepatocellular carcinoma. Oncotarget 2016;7:21753–21762.
83. Goh MJ, Sinn DH, Kim S, Woo SY, Cho H, Kang W, et al. Statin use and the risk of hepatocellular carcinoma in patients with chronic hepatitis B. Hepatology 2020;71:2023–2032.
84. Choi WM, Kim HJ, Jo AJ, Choi SH, Han S, Ko MJ, et al. Association of aspirin and statin use with the risk of liver cancer in chronic hepatitis B: a nationwide population-based study. Liver Int 2021;41:2777–2785.
85. Yang HI, Yuen MF, Chan HL, Han KH, Chen PJ, Kim DY, et al. Risk estimation for hepatocellular carcinoma in chronic hepatitis B (REACH-B): development and validation of a predictive score. Lancet Oncol 2011;12:568–574.
86. Lee HW, Yoo EJ, Kim BK, Kim SU, Park JY, Kim DY, et al. Prediction of development of liver-related events by transient elastography in hepatitis B patients with complete virological response on antiviral therapy. Am J Gastroenterol 2014;109:1241–1249.
87. Jung KS, Kim SU, Song K, Park JY, Kim DY, Ahn SH, et al. Validation of hepatitis B virus-related hepatocellular carcinoma prediction models in the era of antiviral therapy. Hepatology 2015;62:1757–1766.
88. Papatheodoridis G, Dalekos G, Sypsa V, Yurdaydin C, Buti M, Goulis J, et al. PAGE-B predicts the risk of developing hepatocellular carcinoma in Caucasians with chronic hepatitis B on 5-year antiviral therapy. J Hepatol 2016;64:800–806.
89. Kim MN, Hwang SG, Rim KS, Kim BK, Park JY, Kim DY, et al. Validation of PAGE-B model in Asian chronic hepatitis B patients receiving entecavir or tenofovir. Liver Int 2017;37:1788–1795.
90. Chon HY, Lee HA, Suh SJ, Lee JI, Kim BS, Kim IH, et al. Addition of liver stiffness enhances the predictive accuracy of the PAGEB model for hepatitis B-related hepatocellular carcinoma. Aliment Pharmacol Ther 2021;53:919–927.
91. Kim JH, Kim YD, Lee M, Jun BG, Kim TS, Suk KT, et al. Modified PAGE-B score predicts the risk of hepatocellular carcinoma in Asians with chronic hepatitis B on antiviral therapy. J Hepatol 2018;69:1066–1073.
92. Lee HW, Kim SU, Park JY, Kim DY, Ahn SH, Han KH, et al. External validation of the modified PAGE-B score in Asian chronic hepatitis B patients receiving antiviral therapy. Liver Int 2019;39:1624–1630.
93. Sharma SA, Kowgier M, Hansen BE, Brouwer WP, Maan R, Wong D, et al. Toronto HCC risk index: a validated scoring system to predict 10-year risk of HCC in patients with cirrhosis. J Hepatol 2018;68:92–99.
94. Papatheodoridis GV, Sypsa V, Dalekos GN, Yurdaydin C, Van Boemmel F, Buti M, et al. Hepatocellular carcinoma prediction beyond year 5 of oral therapy in a large cohort of Caucasian patients with chronic hepatitis B. J Hepatol 2020;72:1088–1096.
95. Ji JH, Park SY, Son WJ, Shin HJ, Lee H, Lee HW, et al. External validation of CAGE-B and SAGE-B scores for Asian chronic hepatitis B patients with well-controlled viremia by antivirals. J Viral Hepat 2021;28:951–958.
96. Chon HY, Lee JS, Lee HW, Chun HS, Kim BK, Tak WY, et al. Predictive performance of CAGE-B and SAGE-B models in Asian treatment-naive patients who started entecavir for chronic hepatitis B. Clin Gastroenterol Hepatol 2022;20:e794–e807.
97. Nam H, Lee SW, Kwon JH, Lee HL, Yoo SH, Kim HY, et al. Prediction of hepatocellular carcinoma by on-therapy response of noninvasive fibrosis markers in chronic hepatitis B. Am J Gastroenterol 2021;116:1657–1666.
98. Kim HY, Lampertico P, Nam JY, Lee HC, Kim SU, Sinn DH, et al. An artificial intelligence model to predict hepatocellular carcinoma risk in Korean and Caucasian patients with chronic hepatitis B. J Hepatol 2022;76:311–318.
99. Nam JY, Sinn DH, Bae J, Jang ES, Kim JW, Jeong SH. Deep learning model for prediction of hepatocellular carcinoma in patients with HBV-related cirrhosis on antiviral therapy. JHEP Rep 2020;2:100175.
100. Bae SH, Yoon SK, Jang JW, Kim CW, Nam SW, Choi JY, et al. Hepatitis B virus genotype C prevails among chronic carriers of the virus in Korea. J Korean Med Sci 2005;20:816–820.
101. Cho JH, Yoon KH, Lee KE, Park DS, Lee YJ, Moon HB, et al. Distribution of hepatitis B virus genotypes in Korea. Korean J Hepatol 2009;15:140–147.
102. Lin CL, Kao JH. Natural history of acute and chronic hepatitis B: the role of HBV genotypes and mutants. Best Pract Res Clin Gastroenterol 2017;31:249–255.
103. Lin CL, Kao JH. The clinical implications of hepatitis B virus genotype: recent advances. J Gastroenterol Hepatol 2011;26 Suppl 1:123–130.
104. Coffin CS, Zhou K, Terrault NA. New and old biomarkers for diagnosis and management of chronic hepatitis B virus infection. Gastroenterology 2019;156:355–368. e3.
105. Liu J, Yang HI, Lee MH, Jen CL, Batrla-Utermann R, Lu SN, et al. Serum levels of hepatitis B surface antigen and DNA can predict inactive carriers with low risk of disease progression. Hepatology 2016;64:381–389.
106. Park H, Lee JM, Seo JH, Kim HS, Ahn SH, Kim DY, et al. Predictive value of HBsAg quantification for determining the clinical course of genotype C HBeAg-negative carriers. Liver Int 2012;32:796–802.
107. Tseng TC, Liu CJ, Yang HC, Su TH, Wang CC, Chen CL, et al. High levels of hepatitis B surface antigen increase risk of hepatocellular carcinoma in patients with low HBV load. Gastroenterology 2012;142:1140–1149.e3. quiz e13-e14.
108. Sonneveld MJ, Hansen BE, Piratvisuth T, Jia JD, Zeuzem S, Gane E, et al. Response-guided peginterferon therapy in hepatitis B e antigen-positive chronic hepatitis B using serum hepatitis B surface antigen levels. Hepatology 2013;58:872–880.
109. Chang ML, Liaw YF, Hadziyannis SJ. Systematic review: cessation of long-term nucleos(t)ide analogue therapy in patients with hepatitis B e antigen-negative chronic hepatitis B. Aliment Pharmacol Ther 2015;42:243–257.
110. Lee HA, Seo YS, Park SW, Park SJ, Kim TH, Suh SJ, et al. Hepatitis B surface antigen titer is a good indicator of durable viral response after entecavir off-treatment for chronic hepatitis B. Clin Mol Hepatol 2016;22:382–389.
111. Wursthorn K, Jung M, Riva A, Goodman ZD, Lopez P, Bao W, et al. Kinetics of hepatitis B surface antigen decline during 3 years of telbivudine treatment in hepatitis B e antigen-positive patients. Hepatology 2010;52:1611–1620.
112. Park Y, Hong DJ, Shin S, Cho Y, Kim HS. Performance evaluation of new automated hepatitis B viral markers in the clinical laboratory: two quantitative hepatitis B surface antigen assays and an HBV core-related antigen assay. Am J Clin Pathol 2012;137:770–777.
113. Mak LY, Wong DK, Cheung KS, Seto WK, Lai CL, Yuen MF. Review article: hepatitis B core-related antigen (HBcrAg): an emerging marker for chronic hepatitis B virus infection. Aliment Pharmacol Ther 2018;47:43–54.
114. Testoni B, Lebossé F, Scholtes C, Berby F, Miaglia C, Subic M, et al. Serum hepatitis B core-related antigen (HBcrAg) correlates with covalently closed circular DNA transcriptional activity in chronic hepatitis B patients. J Hepatol 2019;70:615–625.
115. Wong DK, Seto WK, Cheung KS, Chong CK, Huang FY, Fung J, et al. Hepatitis B virus core-related antigen as a surrogate marker for covalently closed circular DNA. Liver Int 2017;37:995–1001.
116. Wang ML, Deng R, Chen EQ, Tao CM, Liao J, Zhou TY, et al. Performance of serum HBcrAg in chronic hepatitis B patients with 8-year nucleos(t)ide analogs therapy. Clin Res Hepatol Gastroenterol 2019;43:301–309.
117. Drafting Committee for Hepatitis Management Guidelines, the Japan Society of Hepatology. Japan society of hepatology guidelines for the management of hepatitis B virus infection: 2019 update. Hepatol Res 2020;50:892–923.
118. Inoue T, Tanaka Y. Novel biomarkers for the management of chronic hepatitis B. Clin Mol Hepatol 2020;26:261–279.
119. Carey I, Gersch J, Wang B, Moigboi C, Kuhns M, Cloherty G, et al. Pregenomic HBV RNA and hepatitis B core-related antigen predict outcomes in hepatitis B e antigen-negative chronic hepatitis B patients suppressed on nucleos(t)ide analogue therapy. Hepatology 2020;72:42–57.
120. Wang J, Shen T, Huang X, Kumar GR, Chen X, Zeng Z, et al. Serum hepatitis B virus RNA is encapsidated pregenome RNA that may be associated with persistence of viral infection and rebound. J Hepatol 2016;65:700–710.
121. Giersch K, Allweiss L, Volz T, Dandri M, Lütgehetmann M. Serum HBV pgRNA as a clinical marker for cccDNA activity. J Hepatol 2017;66:460–462.
122. Seto WK, Liu KS, Mak LY, Cloherty G, Wong DK, Gersch J, et al. Role of serum HBV RNA and hepatitis B surface antigen levels in identifying Asian patients with chronic hepatitis B suitable for entecavir cessation. Gut 2021;70:775–783.
123. Ghany MG, King WC, Lisker-Melman M, Lok ASF, Terrault N, Janssen HLA, et al. Comparison of HBV RNA and hepatitis B core related antigen with conventional HBV markers among untreated adults with chronic hepatitis B in north America. Hepatology 2021;74:2395–2409.
124. Kaewdech A, Tangkijvanich P, Sripongpun P, Witeerungrot T, Jandee S, Tanaka Y, et al. Hepatitis B surface antigen, core-related antigen and HBV RNA: predicting clinical relapse after NA therapy discontinuation. Liver Int 2020;40:2961–2971.
125. Liu S, Deng R, Zhou B, Liang X, Liu Z, Peng J, et al. Association of serum hepatitis B virus RNA with hepatocellular carcinoma risk in chronic hepatitis B patients under nucleos(t)ide analogues therapy. J Infect Dis 2021;Dec. :jiab597.
126. Ding WB, Wang MC, Yu J, Huang G, Sun DP, Liu L, et al. HBV/pregenomic RNA increases the stemness and promotes the development of HBV-related HCC through reciprocal regulation with insulin-like growth factor 2 mRNA-binding protein 3. Hepatology 2021;74:1480–1495.
127. Allweiss L, Dandri M. The role of cccDNA in HBV maintenance. Viruses 2017;9:156.
128. Nassal M. HBV cccDNA: viral persistence reservoir and key obstacle for a cure of chronic hepatitis B. Gut 2015;64:1972–1984.
129. Kumar R, Pérez-Del-Pulgar S, Testoni B, Lebossé F, Zoulim F. Clinical relevance of the study of hepatitis B virus covalently closed circular DNA. Liver Int 2016;36 Suppl 1:72–77.
130. Péneau C, Imbeaud S, La Bella T, Hirsch TZ, Caruso S, Calderaro J, et al. Hepatitis B virus integrations promote local and distant oncogenic driver alterations in hepatocellular carcinoma. Gut 2022;71:616–626.
131. Jang JW, Kim JS, Kim HS, Tak KY, Nam H, Sung PS, et al. Persistence of intrahepatic hepatitis B virus DNA integration in patients developing hepatocellular carcinoma after hepatitis B surface antigen seroclearance. Clin Mol Hepatol 2021;27:207–218.
132. Zhao LH, Liu X, Yan HX, Li WY, Zeng X, Yang Y, et al. Genomic and oncogenic preference of HBV integration in hepatocellular carcinoma. Nat Commun 2016;7:12992.
133. Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet 2012;44:765–769.
134. An J, Kim D, Oh B, Oh YJ, Song J, Park N, et al. Comprehensive characterization of viral integrations and genomic aberrations in HBV-infected intrahepatic cholangiocarcinomas. Hepatology 2022;75:997–1011.
135. Mendizabal M, Piñero F, Ridruejo E, Herz Wolff F, Anders M, Reggiardo V, et al. Disease progression in patients with hepatitis C virus infection treated with direct-acting antiviral agents. Clin Gastroenterol Hepatol 2020;18:2554–2563. e3.
136. Zheng B, Liu XL, Fan R, Bai J, Wen H, Du LT, et al. The landscape of cell-free HBV integrations and mutations in cirrhosis and hepatocellular carcinoma patients. Clin Cancer Res 2021;27:3772–3783.
137. Tang LSY, Covert E, Wilson E, Kottilil S. Chronic hepatitis B infection: a review. JAMA 2018;319:1802–1813.
138. Lok AS, McMahon BJ, Brown RS Jr, Wong JB, Ahmed AT, Farah W, et al. Antiviral therapy for chronic hepatitis B viral infection in adults: a systematic review and meta-analysis. Hepatology 2016;63:284–306.
139. Xiao G, Yang J, Yan L. Comparison of diagnostic accuracy of aspartate aminotransferase to platelet ratio index and fibrosis-4 index for detecting liver fibrosis in adult patients with chronic hepatitis B virus infection: a systemic review and meta-analysis. Hepatology 2015;61:292–302.
140. Chon YE, Choi EH, Song KJ, Park JY, Kim DY, Han KH, et al. Performance of transient elastography for the staging of liver fibrosis in patients with chronic hepatitis B: a meta-analysis. PLoS One 2012;7:e44930.
141. Zhu X, Wang LC, Chen EQ, Chen XB, Chen LY, Liu L, et al. Prospective evaluation of FibroScan for the diagnosis of hepatic fibrosis compared with liver biopsy/AST platelet ratio index and FIB-4 in patients with chronic HBV infection. Dig Dis Sci 2011;56:2742–2749.
142. Bedossa P, Poynard T. An algorithm for the grading of activity in chronic hepatitis C. The METAVIR cooperative study group. Hepatology 1996;24:289–293.
143. Andreani T, Serfaty L, Mohand D, Dernaika S, Wendum D, Chazouillères O, et al. Chronic hepatitis B virus carriers in the immunotolerant phase of infection: histologic findings and outcome. Clin Gastroenterol Hepatol 2007;5:636–641.
144. Terrault NA, Lok ASF, McMahon BJ, Chang KM, Hwang JP, Jonas MM, et al. Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology 2018;67:1560–1599.
145. Wong GL. Management of chronic hepatitis B patients in immunetolerant phase: what latest guidelines recommend. Clin Mol Hepatol 2018;24:108–113.
146. Lee HW, Kim SU, Baatarkhuu O, Park JY, Kim DY, Ahn SH, et al. Comparison between chronic hepatitis B patients with untreated immune-tolerant phase vs. those with virological response by antivirals. Sci Rep 2019;9:2508.
147. Jeon MY, Kim BK, Lee JS, Lee HW, Park JY, Kim DY, et al. Negligible risks of hepatocellular carcinoma during biomarker-defined immune-tolerant phase for patients with chronic hepatitis B. Clin Mol Hepatol 2021;27:295–304.
148. Sinn DH, Lee JH, Kim K, Ahn JH, Lee JH, Kim JH, et al. A novel model for predicting hepatocellular carcinoma development in patients with chronic hepatitis B and normal alanine aminotransferase levels. Gut Liver 2017;11:528–534.
149. Kim MN, Kim SU, Kim BK, Park JY, Kim DY, Ahn SH, et al. Increased risk of hepatocellular carcinoma in chronic hepatitis B patients with transient elastography-defined subclinical cirrhosis. Hepatology 2015;61:1851–1859.
150. Park JY, Park YN, Kim DY, Paik YH, Lee KS, Moon BS, et al. High prevalence of significant histology in asymptomatic chronic hepatitis B patients with genotype C and high serum HBV DNA levels. J Viral Hepat 2008;15:615–621.
151. European Association for the Study of the Liver. EASL 2017 clinical practice guidelines on the management of hepatitis B virus infection. J Hepatol 2017;67:370–398.
152. Kim GA, Han S, Choi GH, Choi J, Lim YS. Moderate levels of serum hepatitis B virus DNA are associated with the highest risk of hepatocellular carcinoma in chronic hepatitis B patients. Aliment Pharmacol Ther 2020;51:1169–1179.
153. Shim JJ, Kim JW, Oh CH, Lee YR, Lee JS, Park SY, et al. Serum alanine aminotransferase level and liver-related mortality in patients with chronic hepatitis B: a large national cohort study. Liver Int 2018;38:1751–1759.
154. Chan HL, Chan CK, Hui AJ, Chan S, Poordad F, Chang TT, et al. Effects of tenofovir disoproxil fumarate in hepatitis B e antigenpositive patients with normal levels of alanine aminotransferase and high levels of hepatitis B virus DNA. Gastroenterology 2014;146:1240–1248.
155. Wong VW, Hui AJ, Wong GL, Chan RS, Chim AM, Lo AO, et al. Four-year outcomes after cessation of tenofovir in immune-tolerant chronic hepatitis B patients. J Clin Gastroenterol 2018;52:347–352.
156. Chang Y, Choe WH, Sinn DH, Lee JH, Ahn SH, Lee H, et al. Nucleos(t)ide analogue treatment for patients with hepatitis B virus (HBV) e antigen-positive chronic HBV genotype C infection: a nationwide, multicenter, retrospective study. J Infect Dis 2017;216:1407–1414.
157. Kim HL, Kim GA, Park JA, Kang HR, Lee EK, Lim YS. Cost-effectiveness of antiviral treatment in adult patients with immune-tolerant phase chronic hepatitis B. Gut 2021;70:2172–2182.
158. Cho JY, Paik YH, Sohn W, Cho HC, Gwak GY, Choi MS, et al. Patients with chronic hepatitis B treated with oral antiviral therapy retain a higher risk for HCC compared with patients with inactive stage disease. Gut 2014;63:1943–1950.
159. Choi J, Han S, Kim N, Lim YS. Increasing burden of liver cancer despite extensive use of antiviral agents in a hepatitis B virusendemic population. Hepatology 2017;66:1454–1463.
160. Kwo PY, Cohen SM, Lim JK. ACG clinical guideline: evaluation of abnormal liver chemistries. Am J Gastroenterol 2017;112:18–35.
161. Lee JK, Shim JH, Lee HC, Lee SH, Kim KM, Lim YS, et al. Estimation of the healthy upper limits for serum alanine aminotransferase in Asian populations with normal liver histology. Hepatology 2010;51:1577–1583.
162. Prati D, Taioli E, Zanella A, Della Torre E, Butelli S, Del Vecchio E, et al. Updated definitions of healthy ranges for serum alanine aminotransferase levels. Ann Intern Med 2002;137:1–10.
163. Chung WG, Kim HJ, Choe YG, Seok HS, Chon CW, Cho YK, et al. Clinical impacts of hazardous alcohol use and obesity on the outcome of entecavir therapy in treatment-naïve patients with chronic hepatitis B infection. Clin Mol Hepatol 2012;18:195–202.
164. Chao DT, Lim JK, Ayoub WS, Nguyen LH, Nguyen MH. Systematic review with meta-analysis: the proportion of chronic hepatitis B patients with normal alanine transaminase ≤ 40 IU/L and significant hepatic fibrosis. Aliment Pharmacol Ther 2014;39:349–358.
165. Chen CF, Lee WC, Yang HI, Chang HC, Jen CL, Iloeje UH, et al. Changes in serum levels of HBV DNA and alanine aminotransferase determine risk for hepatocellular carcinoma. Gastroenterology 2011;141:1240–1248. 1248.e1-e2.
166. Yuen MF, Yuan HJ, Wong DK, Yuen JC, Wong WM, Chan AO, et al. Prognostic determinants for chronic hepatitis B in Asians: therapeutic implications. Gut 2005;54:1610–1614.
167. Park HN, Sinn DH, Gwak GY, Kim JE, Rhee SY, Eo SJ, et al. Upper normal threshold of serum alanine aminotransferase in identifying individuals at risk for chronic liver disease. Liver Int 2012;32:937–944.
168. Chang TT, Gish RG, de Man R, Gadano A, Sollano J, Chao YC, et al. A comparison of entecavir and lamivudine for HBeAg-positive chronic hepatitis B. N Engl J Med 2006;354:1001–1010.
169. Lai CL, Shouval D, Lok AS, Chang TT, Cheinquer H, Goodman Z, et al. Entecavir versus lamivudine for patients with HBeAg-negative chronic hepatitis B. N Engl J Med 2006;354:1011–1020.
170. Marcellin P, Heathcote EJ, Buti M, Gane E, de Man RA, Krastev Z, et al. Tenofovir disoproxil fumarate versus adefovir dipivoxil for chronic hepatitis B. N Engl J Med 2008;359:2442–2455.
171. Lai CL, Gane E, Liaw YF, Hsu CW, Thongsawat S, Wang Y, et al. Telbivudine versus lamivudine in patients with chronic hepatitis B. N Engl J Med 2007;357:2576–2588.
172. Hadziyannis SJ, Tassopoulos NC, Heathcote EJ, Chang TT, Kitis G, Rizzetto M, et al. Adefovir dipivoxil for the treatment of hepatitis B e antigen-negative chronic hepatitis B. N Engl J Med 2003;348:800–807.
173. Marcellin P, Chang TT, Lim SG, Tong MJ, Sievert W, Shiffman ML, et al. Adefovir dipivoxil for the treatment of hepatitis B e antigenpositive chronic hepatitis B. N Engl J Med 2003;348:808–816.
174. Rockey DC, Caldwell SH, Goodman ZD, Nelson RC, Smith AD; American Association for the Study of Liver Diseases. Liver biopsy. Hepatology 2009;49:1017–1044.
175. European Association for Study of Liver, ; Asociacion Latinoamericana para el Estudio del Higado. EASL-ALEH clinical practice guidelines: non-invasive tests for evaluation of liver disease severity and prognosis. J Hepatol 2015;63:237–264.
176. Korean Association for the Study of the Liver. KASL clinical practice guidelines: management of chronic hepatitis B. Clin Mol Hepatol 2016;22:18–75.
177. Song BC, Cho YK, Jwa H, Choi EK, Kim HU, Song HJ, et al. Is it necessary to delay antiviral therapy for 3-6 months to anticipate HBeAg seroconversion in patients with HBeAg-positive chronic hepatitis B in endemic areas of HBV genotype C? Clin Mol Hepatol 2014;20:355–360.
178. Kim HS, Kim HJ, Shin WG, Kim KH, Lee JH, Kim HY, et al. Predictive factors for early HBeAg seroconversion in acute exacerbation of patients with HBeAg-positive chronic hepatitis B. Gastroenterology 2009;136:505–512.
179. Huang KW, Tam KW, Luo JC, Kuan YC. Efficacy and safety of lamivudine versus entecavir for treating chronic hepatitis B virus-related acute exacerbation and acute-on-chronic liver failure: a systematic review and meta-analysis. J Clin Gastroenterol 2017;51:539–547.
180. Tsai WL, Sun WC, Cheng JS. Chronic hepatitis B with spontaneous severe acute exacerbation. Int J Mol Sci 2015;16:28126–28145.
181. Chen CH, Lee CM, Lu SN, Wang JH, Tung HD, Hung CH, et al. Comparison of clinical outcome between patients continuing and discontinuing lamivudine therapy after biochemical breakthrough of YMDD mutants. J Hepatol 2004;41:454–461.
182. Kim KH, Sinn DH, Yun WK, Cho HC, Lee YY, Gwak GY, et al. Defining virologic relapse in chronic hepatitis B. Dig Dis Sci 2011;56:2432–2438.
183. Shih CA, Chen WC, Yu HC, Cheng JS, Lai KH, Hsu JT, et al. Risk of severe acute exacerbation of chronic HBV infection cancer patients who underwent chemotherapy and did not receive anti-viral prophylaxis. PLoS One 2015;10:e0132426.
184. Garg H, Sarin SK, Kumar M, Garg V, Sharma BC, Kumar A. Tenofovir improves the outcome in patients with spontaneous reactivation of hepatitis B presenting as acute-on-chronic liver failure. Hepatology 2011;53:774–780.
185. Wong VW, Wong GL, Yiu KK, Chim AM, Chu SH, Chan HY, et al. Entecavir treatment in patients with severe acute exacerbation of chronic hepatitis B. J Hepatol 2011;54:236–242.
186. Tsai WL, Chiang PH, Chan HH, Lin HS, Lai KH, Cheng JS, et al. Early entecavir treatment for chronic hepatitis B with severe acute exacerbation. Antimicrob Agents Chemother 2014;58:1918–1921.
187. Ha JM, Sohn W, Cho JY, Pyo JH, Choi K, Sinn DH, et al. Static and dynamic prognostic factors for hepatitis-B-related acute-on-chronic liver failure. Clin Mol Hepatol 2015;21:232–241.
188. Park JG, Lee YR, Park SY, Lee HJ, Tak WY, Kweon YO, et al. Tenofovir, entecavir, and lamivudine in patients with severe acute exacerbation and hepatic decompensation of chronic hepatitis B. Dig Liver Dis 2018;50:163–167.
189. Chen CH, Lin CL, Hu TH, Hung CH, Tseng PL, Wang JH, et al. Entecavir vs. lamivudine in chronic hepatitis B patients with severe acute exacerbation and hepatic decompensation. J Hepatol 2014;60:1127–1134.
190. Yasui S, Fujiwara K, Nakamura M, Miyamura T, Yonemitsu Y, Mikata R, et al. Virological efficacy of combination therapy with corticosteroid and nucleoside analogue for severe acute exacerbation of chronic hepatitis B. J Viral Hepat 2015;22:94–102.
191. Yue-Meng W, Yang LH, Yang JH, Xu Y, Yang J, Song GB. The effect of plasma exchange on entecavir-treated chronic hepatitis B patients with hepatic de-compensation and acute-on-chronic liver failure. Hepatol Int 2016;10:462–469.
192. Ahn JM, Sinn DH, Gwak GY, Paik YH, Choi MS, Lee JH, et al. Prediction of clinical outcomes in hepatitis B e antigen negative chronic hepatitis B patients with elevated hepatitis B virus DNA levels. PLoS One 2015;10:e0144777.
193. Sinn DH, Kim SE, Kim BK, Kim JH, Choi MS. The risk of hepatocellular carcinoma among chronic hepatitis B virus-infected patients outside current treatment criteria. J Viral Hepat 2019;26:1465–1472.
194. Paik N, Sinn DH, Lee JH, Oh IS, Kim JH, Kang W, et al. Non-invasive tests for liver disease severity and the hepatocellular carcinoma risk in chronic hepatitis B patients with low-level viremia. Liver Int 2018;38:68–75.
195. Cao Z, Liu Y, Ma L, Lu J, Jin Y, Ren S, et al. A potent hepatitis B surface antigen response in subjects with inactive hepatitis B surface antigen carrier treated with pegylated-interferon alpha. Hepatology 2017;66:1058–1066.
196. de Niet A, Jansen L, Stelma F, Willemse SB, Kuiken SD, Weijer S, et al. Peg-interferon plus nucleotide analogue treatment versus no treatment in patients with chronic hepatitis B with a low viral load: a randomised controlled, open-label trial. Lancet Gastroenterol Hepatol 2017;2:576–584.
197. Suk KT, Baik SK, Yoon JH, Cheong JY, Paik YH, Lee CH, et al. Revision and update on clinical practice guideline for liver cirrhosis. Korean J Hepatol 2012;18:1–21.
198. Marcellin P, Gane E, Buti M, Afdhal N, Sievert W, Jacobson IM, et al. Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: a 5-year open-label followup study. Lancet 2013;381:468–475.
199. Chang TT, Liaw YF, Wu SS, Schiff E, Han KH, Lai CL, et al. Longterm entecavir therapy results in the reversal of fibrosis/cirrhosis and continued histological improvement in patients with chronic hepatitis B. Hepatology 2010;52:886–893.
200. Lee J, Sinn DH, Kim JH, Gwak GY, Kim HS, Jung SH, et al. Hepatocellular carcinoma risk of compensated cirrhosis patients with elevated HBV DNA levels according to serum aminotransferase levels. J Korean Med Sci 2015;30:1618–1624.
201. Kim JH, Sinn DH, Kang W, Gwak GY, Paik YH, Choi MS, et al. Lowlevel viremia and the increased risk of hepatocellular carcinoma in patients receiving entecavir treatment. Hepatology 2017;66:335–343.
202. Sinn DH, Lee J, Goo J, Kim K, Gwak GY, Paik YH, et al. Hepatocellular carcinoma risk in chronic hepatitis B virus-infected compensated cirrhosis patients with low viral load. Hepatology 2015;62:694–701.
203. Jang JW, Yoo SH, Nam HC, Jang BH, Sung Sung PS, Lee W, et al. Association of prophylactic anti-hepatitis B virus therapy with improved long-term survival in patients with hepatocellular carcinoma undergoing transarterial therapy. Clin Infect Dis 2020;71:546–555.
204. Cho YY, Lee JH, Chang Y, Nam JY, Cho H, Lee DH, et al. Comparison of overall survival between antiviral-induced viral suppression and inactive phase chronic hepatitis B patients. J Viral Hepat 2018;25:1161–1171.
205. Lee HW, Park SY, Lee YR, Lee H, Lee JS, Kim SU, et al. Episodic detectable viremia does not affect prognosis in untreated compensated cirrhosis with serum hepatitis B virus DNA <2,000 IU/mL. Am J Gastroenterol 2022;117:288–294.
206. Jang JW, Choi JY, Kim YS, Woo HY, Choi SK, Lee CH, et al. Longterm effect of antiviral therapy on disease course after decompensation in patients with hepatitis B virus-related cirrhosis. Hepatology 2015;61:1809–1820.
207. Shim JH, Lee HC, Kim KM, Lim YS, Chung YH, Lee YS, et al. Efficacy of entecavir in treatment-naïve patients with hepatitis B virusrelated decompensated cirrhosis. J Hepatol 2010;52:176–182.
208. Fontana RJ, Hann HW, Perrillo RP, Vierling JM, Wright T, Rakela J, et al. Determinants of early mortality in patients with decompensated chronic hepatitis B treated with antiviral therapy. Gastroenterology 2002;123:719–727.
209. Perrillo R, Tamburro C, Regenstein F, Balart L, Bodenheimer H, Silva M, et al. Low-dose, titratable interferon alfa in decompensated liver disease caused by chronic infection with hepatitis B virus. Gastroenterology 1995;109:908–916.
210. Marcellin P, Ahn SH, Ma X, Caruntu FA, Tak WY, Elkashab M, et al. Combination of tenofovir disoproxil fumarate and peginterferon α-2a increases loss of hepatitis B surface antigen in patients with chronic hepatitis B. Gastroenterology 2016;150:134–144. e10.
211. Lam AM, Espiritu C, Vogel R, Ren S, Lau V, Kelly M, et al. Preclinical characterization of NVR 3-778, a first-in-class capsid assembly modulator against hepatitis B virus. Antimicrob Agents Chemother 2019;63:e01734–18.
212. Huang Q, Cai D, Yan R, Li L, Zong Y, Guo L, et al. Preclinical profile and characterization of the hepatitis B virus core protein inhibitor ABI-H0731. Antimicrob Agents Chemother 2020;64:e01463–20.
213. Yuen MF, Gane EJ, Kim DJ, Weilert F, Yuen Chan HL, Lalezari J, et al. Antiviral activity, safety, and pharmacokinetics of capsid assembly modulator NVR 3-778 in patients with chronic HBV infection. Gastroenterology 2019;156:1392–1403. e7.
214. Yuen MF, Agarwal K, Gane EJ, Schwabe C, Ahn SH, Kim DJ, et al. Safety, pharmacokinetics, and antiviral effects of ABI-H0731, a hepatitis B virus core inhibitor: a randomised, placebo-controlled phase 1 trial. Lancet Gastroenterol Hepatol 2020;5:152–166.
215. Zoulim F, Lenz O, Vandenbossche JJ, Talloen W, Verbinnen T, Moscalu I, et al. JNJ-56136379, an HBV capsid assembly modulator, is well-tolerated and has antiviral activity in a phase 1 study of patients with chronic infection. Gastroenterology 2020;159:521–533.e9.
216. Yuen MF, Agarwal K, Ma X, Nguyen T, Schiff ER, Hann HW, et al. Antiviral activity and safety of the hepatitis B core inhibitor ABIH0731 administered with a nucleos(t)ide reverse transcriptase inhibitor in patients with HBeAg-positive chronic hepatitis B infection in a long-term extension study. J Hepatol 2020;73:S140.
217. Janssen H, Hou J, Asselah T, Chan H, Zoulim F, Tanaka Y, et al. Efficacy and safety results of the phase 2 JNJ-56136379 JADE study in patients with chronic hepatitis B: interim week 24 data. J Hepatol 2020;73:S129–S130.
218. Kang JA, Kim S, Park M, Park HJ, Kim JH, Park S, et al. Ciclopirox inhibits hepatitis B virus secretion by blocking capsid assembly. Nat Commun 2019;10:2184.
219. Kim SW, Yoon JS, Lee M, Cho Y. Toward a complete cure for chronic hepatitis B: novel therapeutic targets for hepatitis B virus. Clin Mol Hepatol 2022;28:17–30.
220. Ko C, Chakraborty A, Chou WM, Hasreiter J, Wettengel JM, Stadler D, et al. Hepatitis B virus genome recycling and de novo secondary infection events maintain stable cccDNA levels. J Hepatol 2018;69:1231–1241.
221. Watts JK, Corey DR. Silencing disease genes in the laboratory and the clinic. J Pathol 2012;226:365–379.
222. Gish RG, Yuen MF, Chan HL, Given BD, Lai CL, Locarnini SA, et al. Synthetic RNAi triggers and their use in chronic hepatitis B therapies with curative intent. Antiviral Res 2015;121:97–108.
223. Hui RW, Mak LY, Seto WK, Yuen MF, et al. RNA interference as a novel treatment strategy for chronic hepatitis B infection. Clin Mol Hepatol 2022;Feb. 17. doi: 10.3350/cmh.2022.0012.
224. Yuen MF, Heo J, Jang JW, Yoon JH, Kweon YO, Park SJ, et al. Safety, tolerability and antiviral activity of the antisense oligonucleotide bepirovirsen in patients with chronic hepatitis B: a phase 2 randomized controlled trial. Nat Med 2021;27:1725–1734.
225. Gane E, Locarnini S, Lim TH, Strasser S, Sievert W, Cheng W, et al. Short interfering RNA JNJ-3989 combination therapy in chronic hepatitis B shows potent reduction of all viral markers but no correlate was identified for HBsAg reduction and baseline factors. J Hepatol 2021;75:S289–S290.
226. Yuen MF, Berliba E, Sukeepaisarnjaroen W, Tangkijvanich P, Leerapun A, Holmes JA, et al. Low HBsAg levels maintained following cessation of the GalNac-siRNA, AB-729, in chronic hepatitis B subjects on nucleos(t)ide analogue therapy. Hepatology 2021;74:1402A–1403A.
227. Yuen MF, Lim YS, Cloutier D, Thanawala V, Shen L, Arizpe A, et al. Preliminary results from a phase 2 study evaluating VIR-2218 alone and in combination with pegylated interferon alfa-2a in participants with chronic hepatitis B infection. Hepatology 2021;74:63A.
228. Yuen MF, Lim TH, Kim W, Tangkijvanich P, Yoon JH, Sievert W, et al. HBV RNAi inhibitor RG6346 in phase 1b-2a trial was safe, welltolerated, and resulted in substantial and durable reductions in serum HBsAg levels. In : American Association for the Study of Liver Diseases the Liver Meeting; 2020 Nov 13-16;
229. Bazinet M, Pântea V, Placinta G, Moscalu I, Cebotarescu V, Cojuhari L, et al. Safety and efficacy of 48 weeks REP 2139 or REP 2165, tenofovir disoproxil, and pegylated interferon alfa-2a in patients with chronic HBV infection naïve to nucleos(t)ide therapy. Gastroenterology 2020;158:2180–2194.
230. Bazinet M, Pântea V, Cebotarescu V, Cojuhari L, Jimbei P, Anderson M, et al. Persistent control of hepatitis B virus and hepatitis delta virus infection following REP 2139-Ca and pegylated interferon therapy in chronic hepatitis B virus/hepatitis delta virus coinfection. Hepatol Commun 2021;5:189–202.
231. Gane E, Dunbar PR, Brooks A, Zhao Y, Tan S, Lau A, et al. Efficacy and safety of 24 weeks treatment with oral TLR8 agonist, selgantolimod, in virally-suppressed adult patients with chronic hepatitis B: a phase 2 study. J Hepatol 2020;73:S52.
232. Janssen HL, Lim YS, Kim HJ, Tseng CH, Coffin CS, Elkashab M, et al. Safety and efficacy of oral TLR8 agonist, selgantolimod, in viremic adult patients with chronic hepatitis B. J Hepatol 2021;75:S757–S758.
233. Gane E, Verdon DJ, Brooks AE, Gaggar A, Nguyen AH, Subramanian GM, et al. Anti-PD-1 blockade with nivolumab with and without therapeutic vaccination for virally suppressed chronic hepatitis B: a pilot study. J Hepatol 2019;71:900–907.
234. Wang G, Qian J, Cui Y, Yan Y, He H, Wu J. A phase IIa trial of subcutaneously administered PD-L1 antibody ASC22 (envafolimab) in patients with chronic hepatitis B. Hepatology 2021;74:62A.
235. Yoshida O, Imai Y, Shiraishi K, Sanada T, Kohara M, Tsukiyama-Kohara K, et al. Long term HBsAg reduction by a nasal administrative therapeutic vaccine containing HBsAg and HBcAg mixed with mucoadhesive CVP (CVP-NASVAC) in patients with chronic HBV infection: the results of 30 months follow up. Hepatology 2021;74:64A.
236. Lok AS, Pan CQ, Han SH, Trinh HN, Fessel WJ, Rodell T, et al. Randomized phase II study of GS-4774 as a therapeutic vaccine in virally suppressed patients with chronic hepatitis B. J Hepatol 2016;65:509–516.
237. Boni C, Janssen HLA, Rossi M, Yoon SK, Vecchi A, Barili V, et al. Combined GS-4774 and tenofovir therapy can improve HBV-specific T-cell responses in patients with chronic hepatitis. Gastroenterology 2019;157:227–241.e7.
238. Lee JH, Lee YB, Cho EJ, Yu SJ, Yoon JH, Kim YJ. Entecavir plus pegylated interferon and sequential hepatitis B virus vaccination increases hepatitis B surface antigen seroclearance: a randomized controlled proof-of-concept study. Clin Infect Dis 2021;73:e3308–e3316.
239. Yuen MF, Locarnini S, Given B, Schluep T, Hamilton J, Biermer M, et al. First clinical experience with RNA interference-based triple combination therapy in chronic hepatitis B: JNJ-3989, JNJ-6379 and a nucleos(t)ide analogue. Hepatology 2019;70:1489A.
240. Yuen MF, Asselah T, Jacobson IM, Brunetto MR, Janssen HL, Takehara T, et al. Efficacy and safety of the siRNA JNJ-3989 and/or the capsid assembly modulator (CAM) JNJ-6379 for the treatment of chronic hepatitis B virus infection (CHB): results from the phase 2B REEF-1 study. Hepatology 2021;74:1390A–1391A.
241. Lee HW, Ahn SH, Chan HL. Hepatitis B core-related antigen: from virology to clinical application. Semin Liver Dis 2021;41:182–190.
242. Liu S, Zhou B, Valdes JD, Sun J, Guo H. Serum hepatitis B virus RNA: a new potential biomarker for chronic hepatitis B virus infection. Hepatology 2019;69:1816–1827.
243. Kao JH, Jeng WJ, Ning Q, Su TH, Tseng TC, Ueno Y, et al. APASL guidance on stopping nucleos(t)ide analogues in chronic hepatitis B patients. Hepatol Int 2021;15:833–851.
244. van Bömmel F, Berg T. Risks and benefits of discontinuation of nucleos(t)ide analogue treatment: a treatment concept for patients With HBeAg-negative chronic hepatitis B. Hepatol Commun 2021;5:1632–1648.
245. Liem KS, Gehring AJ, Feld JJ, Janssen HLA. Challenges with stopping long-term nucleos(t)ide analogue therapy in patients with chronic hepatitis B. Gastroenterology 2020;158:1185–1190.
246. Hall SAL, Vogrin S, Wawryk O, Burns GS, Visvanathan K, Sundararajan V, et al. Discontinuation of nucleot(s)ide analogue therapy in HBeAg-negative chronic hepatitis B: a meta-analysis. Gut 2021;Sep. 7. doi: 10.1136/gutjnl-2020-323979.
247. Jung KS, Park JY, Chon YE, Kim HS, Kang W, Kim BK, et al. Clinical outcomes and predictors for relapse after cessation of oral antiviral treatment in chronic hepatitis B patients. J Gastroenterol 2016;51:830–839.
248. Song DS, Jang JW, Yoo SH, Kwon JH, Nam SW, Bae SH, et al. Improving the prediction of relapse after nucleos(t)ide analogue discontinuation in patients with chronic hepatitis B. Clin Infect Dis 2021;73:e892–e903.
249. Kang SH, Kang K, Eun YJ, Lee YS, Kim TS, Yoo YJ, et al. Antiviral response is not sustained after cessation of lamivudine treatment in chronic hepatitis B patients: a 10-year follow-up study. J Med Virol 2017;89:849–856.
250. Tout I, Lampertico P, Berg T, Asselah T. Perspectives on stopping nucleos(t)ide analogues therapy in patients with chronic hepatitis B. Antiviral Res 2021;185:104992.
251. Yim HJ, Kim JH, Park JY, Yoon EL, Park H, Kwon JH, et al. Comparison of clinical practice guidelines for the management of chronic hepatitis B: when to start, when to change, and when to stop. Clin Mol Hepatol 2020;26:411–429.
252. Sarin SK, Kumar M, Lau GK, Abbas Z, Chan HL, Chen CJ, et al. Asian-Pacific clinical practice guidelines on the management of hepatitis B: a 2015 update. Hepatol Int 2016;10:1–98.
253. Marcellin P, Lau GK, Bonino F, Farci P, Hadziyannis S, Jin R, et al. Peginterferon alfa-2a alone, lamivudine alone, and the two in combination in patients with HBeAg-negative chronic hepatitis B. N Engl J Med 2004;351:1206–1217.
254. Liaw YF, Jia JD, Chan HL, Han KH, Tanwandee T, Chuang WL, et al. Shorter durations and lower doses of peginterferon alfa-2a are associated with inferior hepatitis B e antigen seroconversion rates in hepatitis B virus genotypes B or C. Hepatology 2011;54:1591–1599.
255. Lampertico P, Viganò M, Di Costanzo GG, Sagnelli E, Fasano M, Di Marco V, et al. Randomised study comparing 48 and 96 weeks peginterferon α-2a therapy in genotype D HBeAg-negative chronic hepatitis B. Gut 2013;62:290–298.
256. Chon YE, Jung KS, Ha Y, Kim MN, Lee JH, Hwang SG, et al. High body mass index hinders fibrosis improvement in patients receiving long-term tenofovir therapy in hepatitis B virus-related cirrhosis. J Viral Hepat 2020;27:1119–1126.
257. Yang HC, Shih YF, Liu CJ. Viral factors affecting the clinical outcomes of chronic hepatitis B. J Infect Dis 2017;216(suppl_8):S757–S764.
258. Mommeja-Marin H, Mondou E, Blum MR, Rousseau F. Serum HBV DNA as a marker of efficacy during therapy for chronic HBV infection: analysis and review of the literature. Hepatology 2003;37:1309–1319.
259. Chon YE, Kim SU, Seo YS, Lee HW, Lee HA, Kim MN, et al. Longterm effects of entecavir and tenofovir treatment on the fibrotic burden in patients with chronic hepatitis B. J Gastroenterol Hepatol 2022;37:200–207.
260. Chon YE, Park JY, Myoung SM, Jung KS, Kim BK, Kim SU, et al. Improvement of liver fibrosis after long-term antiviral therapy assessed by fibroscan in chronic hepatitis B patients with advanced fibrosis. Am J Gastroenterol 2017;112:882–891.
261. Lee SW, Choi J, Kim SU, Lim YS. Entecavir versus tenofovir in patients with chronic hepatitis B: enemies or partners in the prevention of hepatocellular carcinoma. Clin Mol Hepatol 2021;27:402–412.
262. Kim WR, Loomba R, Berg T, Aguilar Schall RE, Yee LJ, Dinh PV, et al. Impact of long-term tenofovir disoproxil fumarate on incidence of hepatocellular carcinoma in patients with chronic hepatitis B. Cancer 2015;121:3631–3638.
263. Liaw YF. HBeAg seroconversion as an important end point in the treatment of chronic hepatitis B. Hepatol Int 2009;3:425–433.
264. Xing T, Xu H, Cao L, Ye M. HBeAg seroconversion in HBeAg-positive chronic hepatitis B patients receiving long-term nucleos(t)ide analog treatment: a systematic review and network meta-analysis. PLoS One 2017;12:e0169444.
265. Papatheodoridis G, Vlachogiannakos I, Cholongitas E, Wursthorn K, Thomadakis C, Touloumi G, et al. Discontinuation of oral antivirals in chronic hepatitis B: a systematic review. Hepatology 2016;63:1481–1492.
266. Gara N, Tana MM, Kattapuram M, Auh S, Sullivan L, Fryzek N, et al. Prospective study of withdrawal of antiviral therapy in patients with chronic hepatitis B after prolonged virological response. Hepatol Commun 2021;5:1888–1900.
267. Lee HW, Lee HJ, Hwang JS, Sohn JH, Jang JY, Han KJ, et al. Lamivudine maintenance beyond one year after HBeAg seroconversion is a major factor for sustained virologic response in HBeAg-positive chronic hepatitis B. Hepatology 2010;51:415–421.
268. Chong CH, Lim SG. When can we stop nucleoside analogues in patients with chronic hepatitis B? Liver Int 2017;37 Suppl 1:52–58.
269. Cornberg M, Wong VW, Locarnini S, Brunetto M, Janssen HLA, Chan HL. The role of quantitative hepatitis B surface antigen revisited. J Hepatol 2017;66:398–411.
270. Liu J, Li T, Zhang L, Xu A. The role of hepatitis B surface antigen in nucleos(t)ide analogues cessation among Asian patients with chronic hepatitis B: a systematic review. Hepatology 2019;70:1045–1055.
271. Chen CH, Lu SN, Hung CH, Wang JH, Hu TH, Changchien CS, et al. The role of hepatitis B surface antigen quantification in predicting HBsAg loss and HBV relapse after discontinuation of lamivudine treatment. J Hepatol 2014;61:515–522.
272. Wang CC, Tseng KC, Hsieh TY, Tseng TC, Lin HH, Kao JH. Assessing the durability of entecavir-treated hepatitis B using quantitative HBsAg. Am J Gastroenterol 2016;111:1286–1294.
273. Jeng WJ, Chen YC, Chien RN, Sheen IS, Liaw YF. Incidence and predictors of hepatitis B surface antigen seroclearance after cessation of nucleos(t)ide analogue therapy in hepatitis B e antigen-negative chronic hepatitis B. Hepatology 2018;68:425–434.
274. Chen CH, Hsu YC, Lu SN, Hung CH, Wang JH, Lee CM, et al. The incidence and predictors of HBV relapse after cessation of tenofovir therapy in chronic hepatitis B patients. J Viral Hepat 2018;25:590–597.
275. Huang PY, Wang JH, Hung CH, Lu SN, Hu TH, Chen CH. The role of hepatitis B virus core-related antigen in predicting hepatitis B virus relapse after cessation of entecavir in hepatitis B e antigennegative patients. J Viral Hepat 2021;28:1141–1149.
276. Hsu YC, Nguyen MH, Mo LR, Wu MS, Yang TH, Chen CC, et al. Combining hepatitis B core-related and surface antigens at end of nucleos(t)ide analogue treatment to predict off-therapy relapse risk. Aliment Pharmacol Ther 2019;49:107–115.
277. Sonneveld MJ, Park JY, Kaewdech A, Seto WK, Tanaka Y, Carey I, et al. Prediction of Sustained response after nucleo(s)tide analogue cessation using HBsAg and HBcrAg levels: a multicenter study (CREATE). Clin Gastroenterol Hepatol 2022;20:e784–e793.
278. Sonneveld MJ, Chiu SM, Park JY, Brakenhoff SM, Kaewdech A, Seto WK, et al. Probability of HBsAg loss after nucleo(s)tide analogue withdrawal depends on HBV genotype and viral antigen levels. J Hepatol 2022;Jan. 29. doi: 10.1016/j.jhep.2022.01.007.
279. Fan R, Peng J, Xie Q, Tan D, Xu M, Niu J, et al. Combining hepatitis B virus RNA and hepatitis B core-related antigen: guidance for safely stopping nucleos(t)ide analogues in hepatitis B e antigen-positive patients with chronic hepatitis B. J Infect Dis 2020;222:611–618.
280. Kim JH, Lee YS, Lee HJ, Yoon E, Jung YK, Jong ES, et al. HBsAg seroclearance in chronic hepatitis B: implications for hepatocellular carcinoma. J Clin Gastroenterol 2011;45:64–68.
281. Yip TC, Wong GL, Wong VW, Tse YK, Lui GC, Lam KL, et al. Durability of hepatitis B surface antigen seroclearance in untreated and nucleos(t)ide analogue-treated patients. J Hepatol 2018;68:63–72.
282. Kim MA, Kim SU, Sinn DH, Jang JW, Lim YS, Ahn SH, et al. Discontinuation of nucleos(t)ide analogues is not associated with a higher risk of HBsAg seroreversion after antiviral-induced HBsAg seroclearance: a nationwide multicentre study. Gut 2020;69:2214–2222.
283. Papatheodoridi M, Tampaki M, Lok AS, Papatheodoridis GV. Risk of HBV reactivation during therapies for HCC: a systematic review. Hepatology 2021;Nov. 13. doi: 10.1002/hep.32241.
284. Kubo S, Nishiguchi S, Hamba H, Hirohashi K, Tanaka H, Shuto T, et al. Reactivation of viral replication after liver resection in patients infected with hepatitis B virus. Ann Surg 2001;233:139–145.
285. Dan JQ, Zhang YJ, Huang JT, Chen MS, Gao HJ, Peng ZW, et al. Hepatitis B virus reactivation after radiofrequency ablation or hepatic resection for HBV-related small hepatocellular carcinoma: a retrospective study. Eur J Surg Oncol 2013;39:865–872.
286. Yoshida H, Yoshida H, Goto E, Sato T, Ohki T, Masuzaki R, et al. Safety and efficacy of lamivudine after radiofrequency ablation in patients with hepatitis B virus-related hepatocellular carcinoma. Hepatol Int 2008;2:89–94.
287. Lee JI, Kim JK, Chang HY, Lee JW, Kim JM, Chung HJ, et al. Impact of postoperative hepatitis B virus reactivation in hepatocellular carcinoma patients who formerly had naturally suppressed virus. J Gastroenterol Hepatol 2014;29:1019–1027.
288. Xu M, Zhou Z, Xu R, Zhang H, Lin N, Zhong Y. Antiviral therapy predicts the outcomes following resection of hepatocellular carcinoma in patients negative for HBV DNA: a propensity score matching analysis. World J Surg Oncol 2019;17:45.
289. Sohn W, Paik YH, Cho JY, Ahn JM, Choi GS, Kim JM, et al. Influence of hepatitis B virus reactivation on the recurrence of HBVrelated hepatocellular carcinoma after curative resection in patients with low viral load. J Viral Hepat 2015;22:539–550.
290. Chang JI, Sinn DH, Cho H, Kim S, Kang W, Gwak GY, et al. Clinical outcomes of hepatitis B virus-related hepatocellular carcinoma patients with undetectable serum HBV DNA levels. Dig Dis Sci 2021;Nov. 20. doi: 10.1007/s10620-021-07312-8.
291. Wu CY, Chen YJ, Ho HJ, Hsu YC, Kuo KN, Wu MS, et al. Association between nucleoside analogues and risk of hepatitis B virus–related hepatocellular carcinoma recurrence following liver resection. JAMA 2012;308:1906–1914.
292. Yin J, Li N, Han Y, Xue J, Deng Y, Shi J, et al. Effect of antiviral treatment with nucleotide/nucleoside analogs on postoperative prognosis of hepatitis B virus-related hepatocellular carcinoma: a two-stage longitudinal clinical study. J Clin Oncol 2013;31:3647–3655.
293. Huang G, Li PP, Lau WY, Pan ZY, Zhao LH, Wang ZG, et al. Antiviral therapy reduces hepatocellular carcinoma recurrence in patients with low HBV-DNA levels: a randomized controlled trial. Ann Surg 2018;268:943–954.
294. Lee TY, Lin JT, Zeng YS, Chen YJ, Wu MS, Wu CY. Association between nucleos(t)ide analog and tumor recurrence in hepatitis B virus-related hepatocellular carcinoma after radiofrequency ablation. Hepatology 2016;63:1517–1527.
295. Wong JS, Wong GL, Tsoi KK, Wong VW, Cheung SY, Chong CN, et al. Meta-analysis: the efficacy of anti-viral therapy in prevention of recurrence after curative treatment of chronic hepatitis B-related hepatocellular carcinoma. Aliment Pharmacol Ther 2011;33:1104–1112.
296. Cho H, Ahn H, Lee DH, Lee JH, Jung YJ, Chang Y, et al. Entecavir and tenofovir reduce hepatitis B virus-related hepatocellular carcinoma recurrence more effectively than other antivirals. J Viral Hepat 2018;25:707–717.
297. Choi J, Jo C, Lim YS. Tenofovir versus entecavir on recurrence of hepatitis B virus-related hepatocellular carcinoma after surgical resection. Hepatology 2021;73:661–673.
298. Tsai MC, Wang CC, Lee WC, Liu YW, Lin CC, Hu TH. Tenofovir is superior to entecavir on recurrence in hepatitis B virus-related hepatocellular carcinoma after curative resection. J Gastroenterol Hepatol 2021;36:163.
299. Lee JH, Kim BK, Park SY, Tak WY, Park JY, Kim DY, et al. The efficacies of entecavir and tenofovir in terms of enhancing prognosis after curative treatment of hepatitis B virus-related hepatocellular carcinoma. Eur J Intern Med 2021;89:48–55.
300. Lao XM, Luo G, Ye LT, Luo C, Shi M, Wang D, et al. Effects of antiviral therapy on hepatitis B virus reactivation and liver function after resection or chemoembolization for hepatocellular carcinoma. Liver Int 2013;33:595–604.
301. Lao XM, Wang D, Shi M, Liu G, Li S, Guo R, et al. Changes in hepatitis B virus DNA levels and liver function after transcatheter arterial chemoembolization of hepatocellular carcinoma. Hepatol Res 2011;41:553–563.
302. Jang JW, Choi JY, Bae SH, Yoon SK, Chang UI, Kim CW, et al. A randomized controlled study of preemptive lamivudine in patients receiving transarterial chemo-lipiodolization. Hepatology 2006;43:233–240.
303. Park JW, Park KW, Cho SH, Park HS, Lee WJ, Lee DH, et al. Risk of hepatitis B exacerbation is low after transcatheter arterial chemoembolization therapy for patients with HBV-related hepatocellular carcinoma: report of a prospective study. Am J Gastroenterol 2005;100:2194–2200.
304. Yeo W, Lam KC, Zee B, Chan PS, Mo FK, Ho WM, et al. Hepatitis B reactivation in patients with hepatocellular carcinoma undergoing systemic chemotherapy. Ann Oncol 2004;15:1661–1666.
305. Nagamatsu H, Itano S, Nagaoka S, Akiyoshi J, Matsugaki S, Kurogi J, et al. Prophylactic lamivudine administration prevents exacerbation of liver damage in HBe antigen positive patients with hepatocellular carcinoma undergoing transhepatic arterial infusion chemotherapy. Am J Gastroenterol 2004;99:2369–2375.
306. Tamori A, Nishiguchi S, Tanaka M, Kurooka H, Fujimoto S, Nakamura K, et al. Lamivudine therapy for hepatitis B virus reactivation in a patient receiving intra-arterial chemotherapy for advanced hepatocellular carcinoma. Hepatol Res 2003;26:77–80.
307. Nagamatsu H, Kumashiro R, Itano S, Matsugaki S, Sata M. Investigation of associating factors in exacerbation of liver damage after chemotherapy in patients with HBV-related HCC. Hepatol Res 2003;26:293–301.
308. Wang K, Jiang G, Jia Z, Zhu X, Ni C. Effects of transarterial chemoembolization combined with antiviral therapy on HBV reactivation and liver function in HBV-related hepatocellular carcinoma patients with HBV-DNA negative. Medicine (Baltimore) 2018;97:e10940.
309. Kim JH, Park JW, Kim TH, Koh DW, Lee WJ, Kim CM. Hepatitis B virus reactivation after three-dimensional conformal radiotherapy in patients with hepatitis B virus-related hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 2007;69:813–819.
310. Jun BG, Kim YD, Kim SG, Kim YS, Jeong SW, Jang JY, et al. Hepatitis B virus reactivation after radiotherapy for hepatocellular carcinoma and efficacy of antiviral treatment: a multicenter study. PLoS One 2018;13:e0201316.
311. Jang JW, Kwon JH, You CR, Kim JD, Woo HY, Bae SH, et al. Risk of HBV reactivation according to viral status and treatment intensity in patients with hepatocellular carcinoma. Antivir Ther 2011;16:969–977.
312. Jang JW, Kim YW, Lee SW, Kwon JH, Nam SW, Bae SH, et al. Reactivation of hepatitis B virus in HBsAg-negative patients with hepatocellular carcinoma. PLoS One 2015;10:e0122041.
313. Peng JW, Lin GN, Xiao JJ, Jiang XM. Hepatitis B virus reactivation in hepatocellular carcinoma patients undergoing transcatheter arterial chemoembolization therapy. Asia Pac J Clin Oncol 2012;8:356–361.
314. Yang Y, Wen F, Li J, Zhang P, Yan W, Hao P, et al. A high baseline HBV load and antiviral therapy affect the survival of patients with advanced HBV-related HCC treated with sorafenib. Liver Int 2015;35:2147–2154.
315. Lim S, Han J, Kim GM, Han KH, Choi HJ. Hepatitis B viral load predicts survival in hepatocellular carcinoma patients treated with sorafenib. J Gastroenterol Hepatol 2015;30:1024–1031.
316. Loomba R, Liang TJ. Hepatitis B reactivation associated with immune suppressive and biological modifier therapies: current concepts, management strategies, and future directions. Gastroenterology 2017;152:1297–1309.
317. Yoo S, Lee D, Shim JH, Kim KM, Lim YS, Lee HC, et al. Risk of hepatitis B virus reactivation in patients treated with immunotherapy for anti-cancer treatment. Clin Gastroenterol Hepatol 2022;20:898–907.
318. Sun X, Hu D, Yang Z, Liu Z, Wang J, Chen J, et al. Baseline HBV loads do not affect the prognosis of patients with hepatocellular carcinoma receiving anti-programmed cell death-1 immunotherapy. J Hepatocell Carcinoma 2020;7:337–345.
319. He MK, Peng C, Zhao Y, Liang RB, Lai ZC, Kan A, et al. Comparison of HBV reactivation between patients with high HBV-DNA and low HBV-DNA loads undergoing PD-1 inhibitor and concurrent antiviral prophylaxis. Cancer Immunol Immunother 2021;70:3207–3216.
320. Buti M, Tsai N, Petersen J, Flisiak R, Gurel S, Krastev Z, et al. Seven-year efficacy and safety of treatment with tenofovir disoproxil fumarate for chronic hepatitis B virus infection. Dig Dis Sci 2015;60:1457–1464.
321. Lampertico P, Chan HL, Janssen HL, Strasser SI, Schindler R, Berg T. Review article: long-term safety of nucleoside and nucleotide analogues in HBV-monoinfected patients. Aliment Pharmacol Ther 2016;44:16–34.
322. Lee HW, Park JY, Kim BK, Kim MY, Lee JI, Kim YS, et al. Efficacy of switching from adefovir to tenofovir in chronic hepatitis B patients who exhibit suboptimal responses to adefovir-based combination rescue therapy due to resistance to nucleoside analogues (SATIS study). Clin Mol Hepatol 2016;22:443–449.
323. Maggi P, Montinaro V, Leone A, Fasano M, Volpe A, Bellacosa C, et al. Bone and kidney toxicity induced by nucleotide analogues in patients affected by HBV-related chronic hepatitis: a longitudinal study. J Antimicrob Chemother 2015;70:1150–1154.
324. López-Alcorocho JM, Barril G, Ortiz-Movilla N, Traver JA, Bartolomé J, Sanz P, et al. Prevalence of hepatitis B, hepatitis C, GB virus C/hepatitis G and TT viruses in predialysis and hemodialysis patients. J Med Virol 2001;63:103–107.
325. Gwak GY, Huh W, Lee DH, Min BH, Koh KC, Kim JJ, et al. Occult hepatitis B virus infection in chronic hemodialysis patients in Korea. Hepatogastroenterology 2008;55:1721–1724.
326. Minuk GY, Sun DF, Greenberg R, Zhang M, Hawkins K, Uhanova J, et al. Occult hepatitis B virus infection in a North American adult hemodialysis patient population. Hepatology 2004;40:1072–1077.
327. Burdick RA, Bragg-Gresham JL, Woods JD, Hedderwick SA, Kurokawa K, Combe C, et al. Patterns of hepatitis B prevalence and seroconversion in hemodialysis units from three continents: the DOPPS. Kidney Int 2003;63:2222–2229.
328. Johnson DW, Dent H, Yao Q, Tranaeus A, Huang CC, Han DS, et al. Frequencies of hepatitis B and C infections among haemodialysis and peritoneal dialysis patients in Asia-Pacific countries: analysis of registry data. Nephrol Dial Transplant 2009;24:1598–1603.
329. Finelli L, Miller JT, Tokars JI, Alter MJ, Arduino MJ. National surveillance of dialysis-associated diseases in the United States, 2002. Semin Dial 2005;18:52–61.
330. Agarwal K, Brunetto M, Seto WK, Lim YS, Fung S, Marcellin P, et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol 2018;68:672–681.
331. Buti M, Gane E, Seto WK, Chan HL, Chuang WL, Stepanova T, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate for the treatment of patients with HBeAg-negative chronic hepatitis B virus infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet Gastroenterol Hepatol 2016;1:196–206.
332. Chan HL, Fung S, Seto WK, Chuang WL, Chen CY, Kim HJ, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate for the treatment of HBeAg-positive chronic hepatitis B virus infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet Gastroenterol Hepatol 2016;1:185–195.
333. Ahn SH, Kim W, Jung YK, Yang JM, Jang JY, Kweon YO, et al. Efficacy and safety of besifovir dipivoxil maleate compared With tenofovir disoproxil fumarate in treatment of chronic hepatitis B virus infection. Clin Gastroenterol Hepatol 2019;17:1850–1859. e4.
334. Chan HLY, Buti M, Agarwal K, Marcellin P, Lim YS, Brunetto MR, et al. Maintenance of high levels of viral suppression and improved safety profile of tenofovir alafenamide (TAF) relative to tenofovir disoproxil fumarate (TDF) in chronic hepatitis B patients treated for 5 years in 2 ongoing phase 3 studies. In : American Associatio for the Study of Liver Diseases the Liver Meeting; 2020 Nov 13-16;
335. Lampertico P, Buti M, Fung S, Ahn SH, Chuang WL, Tak WY, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in virologically suppressed patients with chronic hepatitis B: a randomised, double-blind, phase 3, multicentre non-inferiority study. Lancet Gastroenterol Hepatol 2020;5:441–453.
336. Byun KS, Choi J, Kim JH, Lee YS, Lee HC, Kim YJ, et al. Tenofovir alafenamide for drug-resistant hepatitis B: a randomized trial for switching from tenofovir disoproxil fumarate. Clin Gastroenterol Hepatol 2022;20:427–437. e5.
337. Yim HJ, Kim W, Ahn SH, Yang JM, Jang JY, Kweon YO, et al. Besifovir dipivoxil maleate 144-week treatment of chronic hepatitis B: an open-label extensional study of a phase 3 trial. Am J Gastroenterol 2020;115:1217–1225.
338. Song DS, Kim W, Ahn SH, Yim HJ, Jang JY, Kweon YO, et al. Continuing besifovir dipivoxil maleate versus switching from tenofovir disoproxil fumarate for treatment of chronic hepatitis B: results of 192-week phase 3 trial. Clin Mol Hepatol 2021;27:346–359.
339. Gupta S, Govindarajan S, Fong TL, Redeker AG. Spontaneous reactivation in chronic hepatitis B: patterns and natural history. J Clin Gastroenterol 1990;12:562–568.
340. Lok AS, McMahon BJ, ; Practice Guidelines Committee, American Association for the Study of Liver Diseases (AASLD). Chronic hepatitis B: update of recommendations. Hepatology 2004;39:857–861.
341. Tanaka Y, Esumi M, Shikata T. Persistence of hepatitis B virus DNA after serological clearance of hepatitis B virus. Liver 1990;10:6–10.
342. Koo YX, Tan DS, Tan IB, Tao M, Chow WC, Lim ST. Hepatitis B virus reactivation and role of antiviral prophylaxis in lymphoma patients with past hepatitis B virus infection who are receiving chemoimmunotherapy. Cancer 2010;116:115–121.
343. Yeo W, Johnson PJ. Diagnosis, prevention and management of hepatitis B virus reactivation during anticancer therapy. Hepatology 2006;43:209–220.
344. Hwang JP, Lok AS. Management of patients with hepatitis B who require immunosuppressive therapy. Nat Rev Gastroenterol Hepatol 2014;11:209–219.
345. Bessone F, Dirchwolf M. Management of hepatitis B reactivation in immunosuppressed patients: an update on current recommendations. World J Hepatol 2016;8:385–394.
346. Lok AS, Liang RH, Chiu EK, Wong KL, Chan TK, Todd D. Reactivation of hepatitis B virus replication in patients receiving cytotoxic therapy. Report of a prospective study. Gastroenterology 1991;100:182–188.
347. Yeo W, Chan PK, Zhong S, Ho WM, Steinberg JL, Tam JS, et al. Frequency of hepatitis B virus reactivation in cancer patients undergoing cytotoxic chemotherapy: a prospective study of 626 patients with identification of risk factors. J Med Virol 2000;62:299–307.
348. Loomba R, Rowley A, Wesley R, Liang TJ, Hoofnagle JH, Pucino F, et al. Systematic review: the effect of preventive lamivudine on hepatitis B reactivation during chemotherapy. Ann Intern Med 2008;148:519–528.
349. Kwak LW, Halpern J, Olshen RA, Horning SJ. Prognostic significance of actual dose intensity in diffuse large-cell lymphoma: results of a tree-structured survival analysis. J Clin Oncol 1990;8:963–977.
350. Bonadonna G, Valagussa P, Moliterni A, Zambetti M, Brambilla C. Adjuvant cyclophosphamide, methotrexate, and fluorouracil in node-positive breast cancer: the results of 20 years of follow-up. N Engl J Med 1995;332:901–906.
351. Yeo W, Chan PK, Hui P, Ho WM, Lam KC, Kwan WH, et al. Hepatitis B virus reactivation in breast cancer patients receiving cytotoxic chemotherapy: a prospective study. J Med Virol 2003;70:553–561.
352. Jang JW. Hepatitis B virus reactivation in patients with hepatocellular carcinoma undergoing anti-cancer therapy. World J Gastroenterol 2014;20:7675–7685.
353. Perrillo RP, Gish R, Falck-Ytter YT. American Gastroenterological Association Institute technical review on prevention and treatment of hepatitis B virus reactivation during immunosuppressive drug therapy. Gastroenterology 2015;148:221–244.e3.
354. Reddy KR, Beavers KL, Hammond SP, Lim JK, Falck-Ytter YT; American Gastroenterological Association Institute. American Gastroenterological Association Institute guideline on the prevention and treatment of hepatitis B virus reactivation during immunosuppressive drug therapy. Gastroenterology 2015;148:215–219. quiz e16-e17.
355. Takai S, Tsurumi H, Ando K, Kasahara S, Sawada M, Yamada T, et al. Prevalence of hepatitis B and C virus infection in haematological malignancies and liver injury following chemotherapy. Eur J Haematol 2005;74:158–165.
356. Hsu C, Hsiung CA, Su IJ, Hwang WS, Wang MC, Lin SF, et al. A revisit of prophylactic lamivudine for chemotherapy-associated hepatitis B reactivation in non-Hodgkin’s lymphoma: a randomized trial. Hepatology 2008;47:844–853.
357. Engels EA, Cho ER, Jee SH. Hepatitis B virus infection and risk of non-Hodgkin lymphoma in South Korea: a cohort study. Lancet Oncol 2010;11:827–834.
358. Cheng AL, Hsiung CA, Su IJ, Chen PJ, Chang MC, Tsao CJ, et al. Steroid-free chemotherapy decreases risk of hepatitis B virus (HBV) reactivation in HBV-carriers with lymphoma. Hepatology 2003;37:1320–1328.
359. Yeo W, Chan TC, Leung NW, Lam WY, Mo FK, Chu MT, et al. Hepatitis B virus reactivation in lymphoma patients with prior resolved hepatitis B undergoing anticancer therapy with or without rituximab. J Clin Oncol 2009;27:605–611.
360. Dong HJ, Ni LN, Sheng GF, Song HL, Xu JZ, Ling Y. Risk of hepatitis B virus (HBV) reactivation in non-Hodgkin lymphoma patients receiving rituximab-chemotherapy: a meta-analysis. J Clin Virol 2013;57:209–214.
361. Kim SJ, Hsu C, Song YQ, Tay K, Hong XN, Cao J, et al. Hepatitis B virus reactivation in B-cell lymphoma patients treated with rituximab: analysis from the Asia Lymphoma Study Group. Eur J Cancer 2013;49:3486–3496.
362. Tang Z, Li X, Wu S, Liu Y, Qiao Y, Xu D, et al. Risk of hepatitis B reactivation in HBsAg-negative/HBcAb-positive patients with undetectable serum HBV DNA after treatment with rituximab for lymphoma: a meta-analysis. Hepatol Int 2017;11:429–433.
363. Hsu C, Tsou HH, Lin SJ, Wang MC, Yao M, Hwang WL, et al. Chemotherapy-induced hepatitis B reactivation in lymphoma patients with resolved HBV infection: a prospective study. Hepatology 2014;59:2092–2100.
364. Chen XQ, Peng JW, Lin GN, Li M, Xia ZJ. The effect of prophylactic lamivudine on hepatitis B virus reactivation in HBsAg-positive patients with diffuse large B-cell lymphoma undergoing prolonged rituximab therapy. Med Oncol 2012;29:1237–1241.
365. Zurawska U, Hicks LK, Woo G, Bell CM, Krahn M, Chan KK, et al. Hepatitis B virus screening before chemotherapy for lymphoma: a cost-effectiveness analysis. J Clin Oncol 2012;30:3167–3173.
366. Buti M, Manzano ML, Morillas RM, García-Retortillo M, Martín L, Prieto M, et al. Randomized prospective study evaluating tenofovir disoproxil fumarate prophylaxis against hepatitis B virus reactivation in anti-HBc-positive patients with rituximab-based regimens to treat hematologic malignancies: the Preblin study. PLoS One 2017;12:e0184550.
367. Lau GK, He ML, Fong DY, Bartholomeusz A, Au WY, Lie AK, et al. Preemptive use of lamivudine reduces hepatitis B exacerbation after allogeneic hematopoietic cell transplantation. Hepatology 2002;36:702–709.
368. Sarmati L, Andreoni M, Antonelli G, Arcese W, Bruno R, Coppola N, et al. Recommendations for screening, monitoring, prevention, prophylaxis and therapy of hepatitis B virus reactivation in patients with haematologic malignancies and patients who underwent haematologic stem cell transplantation-a position paper. Clin Microbiol Infect 2017;23:935–940.
369. Chen CY, Tien FM, Cheng A, Huang SY, Chou WC, Yao M, et al. Hepatitis B reactivation among 1962 patients with hematological malignancy in Taiwan. BMC Gastroenterol 2018;18:6.
370. Chen FW, Coyle L, Jones BE, Pattullo V. Entecavir versus lamivudine for hepatitis B prophylaxis in patients with haematological disease. Liver Int 2013;33:1203–1210.
371. Lee SK, Sung PS, Park SS, Min CK, Nam H, Jang JW, et al. Reactivation of resolved hepatitis B after daratumumab for multiple myeloma. Clin Infect Dis 2021;73:e1372–e1375.
372. Kim S, Kim HY, Lee S, Kim SW, Sohn S, Kim K, et al. Hepatitis B virus X protein induces perinuclear mitochondrial clustering in microtubule- and dynein-dependent manners. J Virol 2007;81:1714–1726.
373. Dai MS, Wu PF, Shyu RY, Lu JJ, Chao TY. Hepatitis B virus reactivation in breast cancer patients undergoing cytotoxic chemotherapy and the role of preemptive lamivudine administration. Liver Int 2004;24:540–546.
374. Wu YT, Li X, Liu ZL, Xu Z, Dai W, Zhang K, et al. Hepatitis B virus reactivation and antiviral prophylaxis during lung cancer chemotherapy: a systematic review and meta-analysis. PLoS One 2017;12:e0179680.
375. Xu Z, Dai W, Wu YT, Arshad B, Li X, Wu H, et al. Prophylactic effect of lamivudine on chemotherapy-induced hepatitis B virus reactivation in patients with solid tumour: a meta-analysis. Eur J Cancer Care (Engl) 2018;27:e12799.
376. El-Sayed MH, Mohamed MM, Karim A, Maina AM, Oliveri F, Brunetto MR, et al. Severe liver disease is caused by HBV rather than HCV in children with hematological malignancies. Hematol J 2003;4:321–327.
377. Yeo W, Chan PK, Ho WM, Zee B, Lam KC, Lei KI, et al. Lamivudine for the prevention of hepatitis B virus reactivation in hepatitis B santigen seropositive cancer patients undergoing cytotoxic chemotherapy. J Clin Oncol 2004;22:927–934.
378. Chung SJ, Kim JK, Park MC, Park YB, Lee SK. Reactivation of hepatitis B viral infection in inactive HBsAg carriers following anti-tumor necrosis factor-alpha therapy. J Rheumatol 2009;36:2416–2420.
379. Esteve M, Saro C, González-Huix F, Suarez F, Forné M, Viver JM. Chronic hepatitis B reactivation following infliximab therapy in Crohn’s disease patients: need for primary prophylaxis. Gut 2004;53:1363–1365.
380. Kim YJ, Bae SC, Sung YK, Kim TH, Jun JB, Yoo DH, et al. Possible reactivation of potential hepatitis B virus occult infection by tumor necrosis factor-alpha blocker in the treatment of rheumatic diseases. J Rheumatol 2010;37:346–350.
381. Alameel T, Al Sulais E. Risk of HBV reactivation among IBD patients with occult hepatitis B virus infection. Clin Gastroenterol Hepatol 2021;19:621–622.
382. Lee YH, Bae SC, Song GG. Hepatitis B virus reactivation in HBsAgpositive patients with rheumatic diseases undergoing anti-tumor necrosis factor therapy or DMARDs. Int J Rheum Dis 2013;16:527–531.
383. Pérez-Alvarez R, Díaz-Lagares C, García-Hernández F, Lopez-Roses L, Brito-Zerón P, Pérez-de-Lis M, et al. Hepatitis B virus (HBV) reactivation in patients receiving tumor necrosis factor (TNF)-targeted therapy: analysis of 257 cases. Medicine (Baltimore) 2011;90:359–371.
384. Lin TC, Yoshida K, Tedeschi SK, de Abreu MM, Hashemi N, Solomon DH. Risk of hepatitis B virus reactivation in patients with inflammatory arthritis receiving disease-modifying antirheumatic drugs: a systematic review and meta-analysis. Arthritis Care Res (Hoboken) 2018;70:724–731.
385. Burns EA, Muhsen IN, Anand K, Xu J, Umoru G, Arain AN, et al. Hepatitis B virus reactivation in cancer patients treated with immune checkpoint inhibitors. J Immunother 2021;44:132–139.
386. Wang N, Hu X, Cao W, Li C, Xiao Y, Cao Y, et al. Efficacy and safety of CAR19/22 T-cell cocktail therapy in patients with refractory/ relapsed B-cell malignancies. Blood 2020;135:17–27.
387. Lau GK, Yiu HH, Fong DY, Cheng HC, Au WY, Lai LS, et al. Early is superior to deferred preemptive lamivudine therapy for hepatitis B patients undergoing chemotherapy. Gastroenterology 2003;125:1742–1749.
388. Saab S, Dong MH, Joseph TA, Tong MJ. Hepatitis B prophylaxis in patients undergoing chemotherapy for lymphoma: a decision analysis model. Hepatology 2007;46:1049–1056.
389. Viganò M, Serra G, Casella G, Grossi G, Lampertico P. Reactivation of hepatitis B virus during targeted therapies for cancer and immune-mediated disorders. Expert Opin Biol Ther 2016;16:917–926.
390. Hui CK, Cheung WW, Au WY, Lie AK, Zhang HY, Yueng YH, et al. Hepatitis B reactivation after withdrawal of pre-emptive lamivudine in patients with haematological malignancy on completion of cytotoxic chemotherapy. Gut 2005;54:1597–1603.
391. Cerva C, Colagrossi L, Maffongelli G, Salpini R, Di Carlo D, Malagnino V, et al. Persistent risk of HBV reactivation despite extensive lamivudine prophylaxis in haematopoietic stem cell transplant recipients who are anti-HBc-positive or HBV-negative recipients with an anti-HBc-positive donor. Clin Microbiol Infect 2016;22:946.e1–946.e8.
392. Liu WP, Wang XP, Zheng W, Ping LY, Zhang C, Wang GQ, et al. Hepatitis B virus reactivation after withdrawal of prophylactic antiviral therapy in patients with diffuse large B cell lymphoma. Leuk Lymphoma 2016;57:1355–1362.
393. Nakaya A, Fujita S, Satake A, Nakanishi T, Azuma Y, Tsubokura Y, et al. Delayed HBV reactivation in rituximab-containing chemotherapy: how long should we continue anti-virus prophylaxis or monitoring HBV-DNA? Leuk Res 2016;50:46–49.
394. Paul S, Dickstein A, Saxena A, Terrin N, Viveiros K, Balk EM, et al. Role of surface antibody in hepatitis B reactivation in patients with resolved infection and hematologic malignancy: a meta-analysis. Hepatology 2017;66:379–388.
395. Cho Y, Yu SJ, Cho EJ, Lee JH, Kim TM, Heo DS, et al. High titers of anti-HBs prevent rituximab-related viral reactivation in resolved hepatitis B patient with non-Hodgkin’s lymphoma. J Med Virol 2016;88:1010–1017.
396. Rossi G, Pelizzari A, Motta M, Puoti M. Primary prophylaxis with lamivudine of hepatitis B virus reactivation in chronic HbsAg carriers with lymphoid malignancies treated with chemotherapy. Br J Haematol 2001;115:58–62.
397. Li HR, Huang JJ, Guo HQ, Zhang X, Xie Y, Zhu HL, et al. Comparison of entecavir and lamivudine in preventing hepatitis B reactivation in lymphoma patients during chemotherapy. J Viral Hepat 2011;18:877–883.
398. Huang H, Li X, Zhu J, Ye S, Zhang H, Wang W, et al. Entecavir vs lamivudine for prevention of hepatitis B virus reactivation among patients with untreated diffuse large B-cell lymphoma receiving R-CHOP chemotherapy: a randomized clinical trial. JAMA 2014;312:2521–2530.
399. Yu S, Luo H, Pan M, Luis AP, Xiong Z, Shuai P, et al. Comparison of entecavir and lamivudine in preventing HBV reactivation in lymphoma patients undergoing chemotherapy: a meta-analysis. Int J Clin Pharm 2016;38:1035–1043.
400. Kim HY, Yoo JJ, Oh S, Yu SJ, Kim YJ, Yoon JH, et al. Scoring system for risk stratification of viral reactivation during prophylactic antiviral treatment in Korean patients with hepatitis B undergoing anticancer chemotherapy: a multicenter study. J Med Virol 2018;90:1593–1603.
401. Inada K, Kaneko S, Kurosaki M, Yamashita K, Kirino S, Osawa L, et al. Tenofovir alafenamide for prevention and treatment of hepatitis B virus reactivation and de novo hepatitis. JGH Open 2021;5:1085–1091.
402. Lau GK, Leung YH, Fong DY, Au WY, Kwong YL, Lie A, et al. High hepatitis B virus (HBV) DNA viral load as the most important risk factor for HBV reactivation in patients positive for HBV surface antigen undergoing autologous hematopoietic cell transplantation. Blood 2002;99:2324–2330.
403. Li J, Liu J, Huang B, Zheng D, Chen M, Zhou Z, et al. Hepatitis B virus infection status is an independent risk factor for multiple myeloma patients after autologous hematopoietic stem cell transplantation. Tumour Biol 2013;34:1723–1728.
404. Huang H, Cai Q, Lin T, Lin X, Liu Y, Gao Y, et al. Lamivudine for the prevention of hepatitis B virus reactivation after high-dose chemotherapy and autologous hematopoietic stem cell transplantation for patients with advanced or relapsed non-Hodgkin’s lymphoma single institution experience. Expert Opin Pharmacother 2009;10:2399–2406.
405. Shang J, Wang H, Sun J, Fan Z, Huang F, Zhang Y, et al. A comparison of lamivudine vs entecavir for prophylaxis of hepatitis B virus reactivation in allogeneic hematopoietic stem cell transplantation recipients: a single-institutional experience. Bone Marrow Transplant 2016;51:581–586.
406. Siyahian A, Malik SU, Mushtaq A, Howe CL, Majeed A, Zangeneh T, et al. Prophylaxis for hepatitis B virus reactivation after allogeneic stem cell transplantation in the era of drug resistance and newer antivirals: a systematic review and meta-analysis. Biol Blood Marrow Transplant 2018;24:1483–1489.
407. Seto WK, Chan TS, Hwang YY, Wong DK, Fung J, Liu KS, et al. Hepatitis B reactivation in occult viral carriers undergoing hematopoietic stem cell transplantation: a prospective study. Hepatology 2017;65:1451–1461.
408. Park S, Kim K, Kim DH, Jang JH, Kim SJ, Kim WS, et al. Changes of hepatitis B virus serologic status after allogeneic hematopoietic stem cell transplantation and impact of donor immunity on hepatitis B virus. Biol Blood Marrow Transplant 2011;17:1630–1637.
409. Yoo JJ, Cho EJ, Cho YY, Lee M, Lee DH, Cho Y, et al. Efficacy of antiviral prophylaxis in HBsAg-negative, anti-HBc positive patients undergoing hematopoietic stem cell transplantation. Liver Int 2015;35:2530–2536.
410. Liu YC, Hsu CM, Hsiao SY, Hsiao HH. Hepatitis B virus infection in patients receiving allogeneic hematopoietic stem cell transplantation. J Pers Med 2021;11:1108.
411. Hwang JP, Somerfield MR, Alston-Johnson DE, Cryer DR, Feld JJ, Kramer BS, et al. Hepatitis B virus screening for patients with cancer before therapy: American Society of Clinical Oncology provisional clinical opinion update. J Clin Oncol 2015;33:2212–2220.
412. Hwang JP, Feld JJ, Hammond SP, Wang SH, Alston-Johnson DE, Cryer DR, et al. Hepatitis B virus screening and management for patients with cancer prior to therapy: ASCO provisional clinical opinion update. J Clin Oncol 2020;38:3698–3715.
413. Lau G, Yu ML, Wong G, Thompson A, Ghazinian H, Hou JL, et al. APASL clinical practice guideline on hepatitis B reactivation related to the use of immunosuppressive therapy. Hepatol Int 2021;15:1031–1048.
414. Lee HL, Jang JW, Han JW, Lee SW, Bae SH, Choi JY, et al. Early hepatitis B surface antigen seroclearance following antiviral treatment in patients with reactivation of resolved hepatitis B. Dig Dis Sci 2019;64:2992–3000.
415. European Association for the Study of the Liver. EASL clinical practice guidelines: liver transplantation. J Hepatol 2016;64:433–485.
416. Sripongpun P, Mannalithara A, Kwo PY, Kim WR. Potential benefits of switching liver transplant recipients to tenofovir alafenamide prophylaxis. Clin Gastroenterol Hepatol 2020;18:747–749.
417. Rashidi-Alavijeh J, Straub K, Achterfeld A, Wedemeyer H, Willuweit K, Herzer K. Safety and efficacy of tenofovir alafenamide in liver transplant recipients: a single center experience. Transpl Infect Dis 2021;23:e13522.
418. Wesdorp DJ, Knoester M, Braat AE, Coenraad MJ, Vossen AC, Claas EC, et al. Nucleoside plus nucleotide analogs and cessation of hepatitis B immunoglobulin after liver transplantation in chronic hepatitis B is safe and effective. J Clin Virol 2013;58:67–73.
419. Teperman LW, Poordad F, Bzowej N, Martin P, Pungpapong S, Schiano T, et al. Randomized trial of emtricitabine/tenofovir disoproxil fumarate after hepatitis B immunoglobulin withdrawal after liver transplantation. Liver Transpl 2013;19:594–601.
420. Stravitz RT, Shiffman ML, Kimmel M, Puri P, Luketic VA, Sterling RK, et al. Substitution of tenofovir/emtricitabine for hepatitis B immune globulin prevents recurrence of hepatitis B after liver transplantation. Liver Int 2012;32:1138–1145.
421. Degertekin B, Han SH, Keeffe EB, Schiff ER, Luketic VA, Brown RS Jr, et al. Impact of virologic breakthrough and HBIG regimen on hepatitis B recurrence after liver transplantation. Am J Transplant 2010;10:1823–1833.
422. Cholongitas E, Goulis I, Antoniadis N, Fouzas I, Imvrios G, Papanikolaou V, et al. New nucleos(t)ide analogue monoprophylaxis after cessation of hepatitis B immunoglobulin is effective against hepatitis B recurrence. Transpl Int 2014;27:1022–1028.
423. Radhakrishnan K, Chi A, Quan DJ, Roberts JP, Terrault NA. Short course of postoperative hepatitis B immunoglobulin plus antivirals prevents reinfection of liver transplant recipients. Transplantation 2017;101:2079–2082.
424. Manini MA, Whitehouse G, Bruce M, Passerini M, Lim TY, Carey I, et al. Entecavir or tenofovir monotherapy prevents HBV recurrence in liver transplant recipients: a 5-year follow-up study after hepatitis B immunoglobulin withdrawal. Dig Liver Dis 2018;50:944–953.
425. Tanaka T, Benmousa A, Marquez M, Therapondos G, Renner EL, Lilly LB. The long-term efficacy of nucleos(t)ide analog plus a year of low-dose HBIG to prevent HBV recurrence post-liver transplantation. Clin Transplant 2012;26:E561–E569.
426. Fung J, Wong T, Chok K, Chan A, Cheung TT, Dai JW, et al. Longterm outcomes of entecavir monotherapy for chronic hepatitis B after liver transplantation: results up to 8 years. Hepatology 2017;66:1036–1044.
427. Fung J, Chan SC, Cheung C, Yuen MF, Chok KS, Sharr W, et al. Oral nucleoside/nucleotide analogs without hepatitis B immune globulin after liver transplantation for hepatitis B. Am J Gastroenterol 2013;108:942–948.
428. Wang P, Tam N, Wang H, Zheng H, Chen P, Wu L, et al. Is hepatitis B immunoglobulin necessary in prophylaxis of hepatitis B recurrence after liver transplantation? A meta-analysis. PLoS One 2014;9:e104480.
429. Miyaaki H, Tamada Y, Hayashi K, Taura N, Miuma S, Shibata H, et al. Recurrent hepatitis B and D virus infection in a liver transplant recipient. Transplant Proc 2017;49:175–177.
430. Mederacke I, Filmann N, Yurdaydin C, Bremer B, Puls F, Zacher BJ, et al. Rapid early HDV RNA decline in the peripheral blood but prolonged intrahepatic hepatitis delta antigen persistence after liver transplantation. J Hepatol 2012;56:115–122.
431. Cholongitas E, Papatheodoridis GV, Burroughs AK. Liver grafts from anti-hepatitis B core positive donors: a systematic review. J Hepatol 2010;52:272–279.
432. Skagen CL, Jou JH, Said A. Risk of de novo hepatitis in liver recipients from hepatitis-B core antibody-positive grafts - a systematic analysis. Clin Transplant 2011;25:E243–E249.
433. Perrillo R. Hepatitis B virus prevention strategies for antibody to hepatitis B core antigen-positive liver donation: a survey of North American, European, and Asian-Pacific transplant programs. Liver Transpl 2009;15:223–232.
434. Huprikar S, Danziger-Isakov L, Ahn J, Naugler S, Blumberg E, Avery RK, et al. Solid organ transplantation from hepatitis B virus-positive donors: consensus guidelines for recipient management. Am J Transplant 2015;15:1162–1172.
435. Lee J, Cho JH, Lee JS, Ahn DW, Kim CD, Ahn C, et al. Pretransplant hepatitis B viral infection increases risk of death after kidney transplantation: a multicenter cohort study in Korea. Medicine (Baltimore) 2016;95:e3671.
436. Thongprayoon C, Kaewput W, Sharma K, Wijarnpreecha K, Leeaphorn N, Ungprasert P, et al. Outcomes of kidney transplantation in patients with hepatitis B virus infection: a systematic review and meta-analysis. World J Hepatol 2018;10:337–346.
437. Yap DY, Yung S, Tang CS, Seto WK, Ma MK, Mok MM, et al. Entecavir treatment in kidney transplant recipients infected with hepatitis B. Clin Transplant 2014;28:1010–1015.
438. Hu TH, Tsai MC, Chien YS, Chen YT, Chen TC, Lin MT, et al. A novel experience of antiviral therapy for chronic hepatitis B in renal transplant recipients. Antivir Ther 2012;17:745–753.
439. Nho KW, Kim YH, Han DJ, Park SK, Kim SB. Kidney transplantation alone in end-stage renal disease patients with hepatitis B liver cirrhosis: a single-center experience. Transplantation 2015;99:133–138.
440. Veroux M, Puliatti C, Gagliano M, Cappello D, Macarone M, Vizcarra D, et al. Use of hepatitis B core antibody-positive donor kidneys in hepatitis B surface antibody-positive and -negative recipients. Transplant Proc 2005;37:2574–2575.
441. Wachs ME, Amend WJ, Ascher NL, Bretan PN, Emond J, Lake JR, et al. The risk of transmission of hepatitis B from HBsAg(-), HBcAb(+), HBIgM(-) organ donors. Transplantation 1995;59:230–234.
442. Fong TL, Bunnapradist S, Jordan SC, Cho YW. Impact of hepatitis B core antibody status on outcomes of cadaveric renal transplantation: analysis of united network of organ sharing database between 1994 and 1999. Transplantation 2002;73:85–89.
443. Jeon JW, Kim SM, Cho H, Baek CH, Kim H, Shin S, et al. Presence of hepatitis B surface antibody in addition to hepatitis B core antibody confers protection against hepatitis B virus infection in hepatitis B surface antigen-negative patients undergoing kidney transplantation. Transplantation 2018;102:1717–1723.
444. Lentine KL, Kasiske BL, Levey AS, Adams PL, Alberú J, Bakr MA, et al. KDIGO clinical practice guideline on the evaluation and care of living kidney donors. Transplantation 2017;101(8S Suppl 1):S1–S109.
445. Duhart BT Jr, Honaker MR, Shokouh-Amiri MH, Riely CA, Vera SR, Taylor SL, et al. Retrospective evaluation of the risk of hepatitis B virus reactivation after transplantation. Transpl Infect Dis 2003;5:126–131.
446. Blanpain C, Knoop C, Delforge ML, Antoine M, Peny MO, Liesnard C, et al. Reactivation of hepatitis B after transplantation in patients with pre-existing anti-hepatitis B surface antigen antibodies: report on three cases and review of the literature. Transplantation 1998;66:883–886.
447. Knöll A, Pietrzyk M, Loss M, Goetz WA, Jilg W. Solid-organ transplantation in HBsAg-negative patients with antibodies to HBV core antigen: low risk of HBV reactivation. Transplantation 2005;79:1631–1633.
448. Berger A, Preiser W, Kachel HG, Stürmer M, Doerr HW. HBV reactivation after kidney transplantation. J Clin Virol 2005;32:162–165.
449. Kanaan N, Kabamba B, Maréchal C, Pirson Y, Beguin C, Goffin E, et al. Significant rate of hepatitis B reactivation following kidney transplantation in patients with resolved infection. J Clin Virol 2012;55:233–238.
450. Bae E, Park CH, Ki CS, Kim SJ, Huh W, Oh HY, et al. Prevalence and clinical significance of occult hepatitis B virus infection among renal transplant recipients in Korea. Scand J Infect Dis 2012;44:788–792.
451. Nishimura K, Kishikawa H, Yoshida Y, Ueda N, Nakazawa S, Yamanaka K, et al. Clinical and virologic courses of hepatitis B surface antigen-negative and hepatitis B core or hepatitis B surface antibody-positive renal transplant recipients. Transplant Proc 2013;45:1600–1602.
452. Lee J, Park JY, Huh KH, Kim BS, Kim MS, Kim SI, et al. Rituximab and hepatitis B reactivation in HBsAg-negative/ anti-HBc-positive kidney transplant recipients. Nephrol Dial Transplant 2017;32:722–729.
453. Chen GD, Gu JL, Qiu J, Chen LZ. Outcomes and risk factors for hepatitis B virus (HBV) reactivation after kidney transplantation in occult HBV carriers. Transpl Infect Dis 2013;15:300–305.
454. Kim YJ, Lee JW, Kim YS, Jeong SH, Kim YS, Yim HJ, et al. Clinical features and treatment efficacy of peginterferon alfa plus ribavirin in chronic hepatitis C patients coinfected with hepatitis B virus. Korean J Hepatol 2011;17:199–205.
455. Choi MS, Sinn DH, Kim SA, Lee YS, Choi W, Paik SW. The clinical and laboratory characteristics of patients with chronic hepatitis B using current or past antiviral therapy in Korea: a multi-center, nation-wide, cross-sectional epidemiologic study. Gut Liver 2012;6:241–248.
456. Sagnelli E, Pasquale G, Coppola N, Scarano F, Marrocco C, Scolastico C, et al. Influence of chronic coinfection with hepatitis B and C virus on liver histology. Infection 2004;32:144–148.
457. Lee LP, Dai CY, Chuang WL, Chang WY, Hou NJ, Hsieh MY, et al. Comparison of liver histopathology between chronic hepatitis C patients and chronic hepatitis B and C-coinfected patients. J Gastroenterol Hepatol 2007;22:515–517.
458. Yang WT, Wu LW, Tseng TC, Chen CL, Yang HC, Su TH, et al. Hepatitis B surface antigen loss and hepatocellular carcinoma development in patients with dual hepatitis B and C infection. Medicine (Baltimore) 2016;95:e2995.
459. Pol S, Haour G, Fontaine H, Dorival C, Petrov-Sanchez V, Bourliere M, et al. The negative impact of HBV/HCV coinfection on cirrhosis and its consequences. Aliment Pharmacol Ther 2017;46:1054–1060.
460. Chen G, Wang C, Chen J, Ji D, Wang Y, Wu V, et al. Hepatitis B reactivation in hepatitis B and C coinfected patients treated with antiviral agents: a systematic review and meta-analysis. Hepatology 2017;66:13–26.
461. Yeh ML, Huang CF, Huang CI, Holmes JA, Hsieh MH, Tsai YS, et al. Hepatitis B-related outcomes following direct-acting antiviral therapy in Taiwanese patients with chronic HBV/HCV co-infection. J Hepatol 2020;73:62–71.
462. Calvaruso V, Ferraro D, Licata A, Bavetta MG, Petta S, Bronte F, et al. HBV reactivation in patients with HCV/HBV cirrhosis on treatment with direct-acting antivirals. J Viral Hepat 2018;25:72–79.
463. Bunnell KL, Vibhakar S, Glowacki RC, Gallagher MA, Osei AM, Huhn G. Nephrotoxicity associated with concomitant use of ledipasvir-sofosbuvir and tenofovir in a patient with hepatitis C virus and human immunodeficiency virus coinfection. Pharmacotherapy 2016;36:e148–e153.
464. Solas C, Bregigeon S, Faucher-Zaegel O, Quaranta S, Obry-Roguet V, Tamalet C, et al. Ledipasvir and tenofovir drug interaction in human immunodeficiency virus-hepatitis C virus coinfected patients: impact on tenofovir trough concentrations and renal safety. Br J Clin Pharmacol 2018;84:404–409.
465. Mücke VT, Mücke MM, Peiffer KH, Weiler N, Welzel TM, Sarrazin C, et al. No evidence of hepatitis B virus reactivation in patients with resolved infection treated with direct-acting antivirals for hepatitis C in a large real-world cohort. Aliment Pharmacol Ther 2017;46:432–439.
466. Tamori A, Abiru S, Enomoto H, Kioka K, Korenaga M, Tani J, et al. Low incidence of hepatitis B virus reactivation and subsequent hepatitis in patients with chronic hepatitis C receiving direct-acting antiviral therapy. J Viral Hepat 2018;25:608–611.
467. Sulkowski MS, Chuang WL, Kao JH, Yang JC, Gao B, Brainard DM, et al. No evidence of reactivation of hepatitis B virus among patients treated with ledipasvir-sofosbuvir for hepatitis C virus infection. Clin Infect Dis 2016;63:1202–1204.
468. Urban S, Neumann-Haefelin C, Lampertico P. Hepatitis D virus in 2021: virology, immunology and new treatment approaches for a difficult-to-treat disease. Gut 2021;70:1782–1794.
469. Stockdale AJ, Kreuels B, Henrion MYR, Giorgi E, Kyomuhangi I, de Martel C, et al. The global prevalence of hepatitis D virus infection: systematic review and meta-analysis. J Hepatol 2020;73:523–532.
470. Kim HS, Kim SJ, Park HW, Shin WG, Kim KH, Lee JH, et al. Prevalence and clinical significance of hepatitis D virus coinfection in patients with chronic hepatitis B in Korea. J Med Virol 2011;83:1172–1177.
471. Jeong SH, Kim JM, Ahn HJ, Park MJ, Paik KH, Choi W, et al. The prevalence and clinical characteristics of hepatitis-delta infection in Korea. Korean J Hepatol 2005;11:43–50.
472. Alfaiate D, Clément S, Gomes D, Goossens N, Negro F. Chronic hepatitis D and hepatocellular carcinoma: a systematic review and meta-analysis of observational studies. J Hepatol 2020;73:533–539.
473. Bahcecioglu IH, Aygun C, Gozel N, Poyrazoglu OK, Bulut Y, Yalniz M. Prevalence of hepatitis delta virus (HDV) infection in chronic hepatitis B patients in eastern Turkey: still a serious problem to consider. J Viral Hepat 2011;18:518–524.
474. Fattovich G, Giustina G, Christensen E, Pantalena M, Zagni I, Realdi G, et al. Influence of hepatitis delta virus infection on morbidity and mortality in compensated cirrhosis type B. The European concerted action on viral hepatitis (Eurohep). Gut 2000;46:420–426.
475. Wedemeyer H, Yurdaydìn C, Dalekos GN, Erhardt A, Çakaloğlu Y, Değertekin H, et al. Peginterferon plus adefovir versus either drug alone for hepatitis delta. N Engl J Med 2011;364:322–331.
476. Abbas Z, Memon MS, Mithani H, Jafri W, Hamid S. Treatment of chronic hepatitis D patients with pegylated interferon: a real-world experience. Antivir Ther 2014;19:463–468.
477. Keskin O, Wedemeyer H, Tüzün A, Zachou K, Deda X, Dalekos GN, et al. Association between level of hepatitis D virus RNA at week 24 of pegylated interferon therapy and outcome. Clin Gastroenterol Hepatol 2015;13:2342–2349. e1-e2.
478. Heidrich B, Yurdaydın C, Kabaçam G, Ratsch BA, Zachou K, Bremer B, et al. Late HDV RNA relapse after peginterferon alpha-based therapy of chronic hepatitis delta. Hepatology 2014;60:87–97.
479. Triantos C, Kalafateli M, Nikolopoulou V, Burroughs A. Metaanalysis: antiviral treatment for hepatitis D. Aliment Pharmacol Ther 2012;35:663–673.
480. Karaca C, Soyer OM, Baran B, Ormeci AC, Gokturk S, Aydin E, et al. Efficacy of pegylated interferon-α treatment for 24 months in chronic delta hepatitis and predictors of response. Antivir Ther 2013;18:561–566.
481. Bogomolov P, Alexandrov A, Voronkova N, Macievich M, Kokina K, Petrachenkova M, et al. Treatment of chronic hepatitis D with the entry inhibitor myrcludex B: first results of a phase Ib/IIa study. J Hepatol 2016;65:490–498.
482. Wedemeyer H, Aleman S, Andreone P, Blank A, Brunetto M, Bogomolov P, et al. Bulevirtide monotherapy at low and high dose in patients with chronic hepatitis delta: 24 weeks interim data of the phase 3 MYR301 study. J Hepatol 2021;75:S294–S295.
483. Dhillon S. Lonafarnib: first approval. Drugs 2021;81:283–289.
484. Yurdaydin C, Keskin O, Yurdcu E, Çalişkan A, Önem S, Karakaya F, et al. A phase 2 dose-finding study of lonafarnib and ritonavir with or without interferon alpha for chronic delta hepatitis. Hepatology 2021;Dec. 3. doi: 10.1002/hep.32259.
485. Bazinet M, Pântea V, Cebotarescu V, Cojuhari L, Jimbei P, Albrecht J, et al. Safety and efficacy of REP 2139 and pegylated interferon alfa-2a for treatment-naive patients with chronic hepatitis B virus and hepatitis D virus co-infection (REP 301 and REP 301-LTF): a non-randomised, open-label, phase 2 trial. Lancet Gastroenterol Hepatol 2017;2:877–889.
486. Sandmann L, Cornberg M. Experimental drugs for the treatment of hepatitis D. J Exp Pharmacol 2021;13:461–468.
487. Wong VW, Chan SL, Mo F, Chan TC, Loong HH, Wong GL, et al. Clinical scoring system to predict hepatocellular carcinoma in chronic hepatitis B carriers. J Clin Oncol 2010;28:1660–1665.
488. Wong GL, Chan HL, Wong CK, Leung C, Chan CY, Ho PP, et al. Liver stiffness-based optimization of hepatocellular carcinoma risk score in patients with chronic hepatitis B. J Hepatol 2014;60:339–345.

Article information Continued

Figure 1.

Natural course of chronic hepatitis B (CHB). HBV, hepatitis B virus; HBsAg, hepatitis B surface antigen; anti-HBs, antibody to HBsAg; HBeAg, hepatitis B e antigen; anti-HBe, antibody to HBeAg; ALT, alanine aminotransferase.

Figure 2.

Emerging markers for chronic hepatitis B infection. rcDNA, relaxed circular DNA; pgRNA, pregenomic RNA; p22Cr, p22 core-related antigen; HBcAg, hepatitis B core antigen; HBeAg, hepatitis B e antigen; HBsAg, hepatitis B surface antigen; HBcrAg, hepatitis B core-related antigen; dslDNA, double stranded linear DNA; cccDNA, circular covalently closed DNA; HBV, hepatitis B virus.

Figure 3.

Algorithm for management of chronic hepatitis B virus infection. HBsAg, hepatitis B surface antigen; HBeAg, hepatitis B e antigen; HBV, hepatitis B virus; AST, aspartate aminotransferase; ALT, alanine aminotransferase; anti-HBe, hepatitis B e antibody. *Serum HBV DNA ≥107 IU/mL. An upper limit of normal (ULN) for ALT of 34 IU/L for males and 30 IU/L for females. Serum HBV DNA ≥20,000 IU/mL for HBeAg-positive patients and HBV DNA ≥2,000 IU/mL for HBeAg-negative patients.

Figure 4.

The mechanisms of novel antiviral agents for the functional cure. NK, natural killer; TLR, toll-like receptor; ICI, immune checkpoint inhibitor; HBV, hepatitis B virus; NTCP, sodium taurocholate co-transporting polypeptide; SVP, subviral particle; HBsAg, hepatitis B surface antigen; HBeAg, hepatitis B e antigen; HBc, hepatitis B core; HBx, hepatitis B virus X protein; cccDNA, covalently closed circular DNA; ASO, antisense oligonucleotide; siRNA, small interfering RNA.

Figure 5.

Indications for selecting entecavir, tenofovir alafenamide fumarate, or besifovir over tenofovir disoproxil fumarate. AF, alafenamide fumarate. *Dose was adjusted if creatinine clearance <50 mL/min, refer to Table 8. Not indicated if creatinine clearance <15 mL/min without dialysis. Not indicated if creatinine clearance <15 mL/min.

Figure 6.

Strategies after liver transplantation in patients receiving anti-HBc-positive liver graft. Anti-HBc, antibody to HBcAg; HBsAg, hepatitis B surface antigen; NA, nucleos(t)ide analogue; HBIG, hepatitis B immunoglobulin; anti-HBs, antibody to HBsAg; HBV, hepatitis B virus. *Pooled data from the study by Cholongitas et al [431].

Table 1.

Grading of Recommendations, Assessment, Development, and Evaluation (GRADE)

Criteria
Quality of evidence
 High (A) Further research is unlikely to change confidence in the estimate of the clinical effect.
 Moderate (B) Further research might change confidence in the estimate of the clinical effect.
 Low (C) Further research is very likely to impact confidence in the estimate of the clinical effect.
Strength of recommendations
 Strong (1) Factors influencing the strength of the recommendation were quality of the evidence, presumed patient-important outcomes, and cost.
 Weak (2) Variability in preferences and values or high uncertainty. Recommendation is made with less certainty, higher cost, or resource consumption.

Of the quality levels of evidence, we excluded “very low quality (D)” for convenience, which was originally included in the GRADE system and indicates that any estimate of effect is very uncertain.

Table 2.

Natural course of chronic hepatitis B

Phases* Serologic marker ALT HBV DNA Histologic activity
CHB, immune-tolerant phase HBeAg (+) Persistently normal Very high level of viral replication (HBV DNA level ≥10,000,000 IU/mL) None/minimal
Anti-HBe (-)
HBeAg-positive CHB, immune-active phase HBeAg (+) Elevated (persistently or intermittently) High level of viral replication (HBV DNA level ≥20,000 IU/mL) Moderate/severe
Can develop anti-HBe
CHB, immune-inactive phase HBeAg (-) Persistently normal Low or undetectable HBV DNA (HBV DNA level <2,000 IU/mL) Minimal
Anti-HBe (+)
HBeAg-negative CHB, immune-active phase HBeAg (-) Elevated (persistently or intermittently) Moderate to high level of HBV replication (HBV DNA level ≥2,000 IU/mL) Moderate/severe
Anti-HBe (+/-)
HBsAg loss phase (resolved CHB) HBsAg (-) Normal Not detected -
Anti-HBc (+)
Anti-HBs (+/-)

ALT, alanine aminotransferase; HBV, hepatitis B virus; CHB, chronic hepatitis B; HBeAg, hepatitis B e antigen; anti-HBe, antibody to HBeAg; HBsAg, hepatitis B surface antigen; anti-HBs, antibody to HBsAg; anti-HBc, antibody to HBcAg.

*

There can be areas of grey zone among the various phases of natural courses.

Fibrosis stage can progress during the natural course but might vary according to degree of accumulation of liver injury.

Table 3.

Definitions of HBV cure

Sterilizing cure Idealistic functional cure Realistic functional cure
Clinical scenario Never infected Recovery after acute HBV Chronic HBV with HBsAg loss
HBsAg Negative Negative Negative
Anti-HBs Negative/positive Positive Negative/positive
HBeAg Negative Negative Negative
Serum HBV DNA Not detected Not detected Not detected
Hepatic cccDNA and/or integrated HBV DNA Not detected Detected* Detected
Liver disease None None Inactive, fibrosis regress over time
Risk of HCC Not increased Not increased Declines with time

HBV, hepatitis B virus; HBsAg, hepatitis B surface antigen; anti-HBs, antibody to HBsAg; HBeAg, hepatitis B e antigen; cccDNA, covalently closed circular DNA; HCC, hepatocellular carcinoma.

*

Insufficient evidence.

Table 4.

Factors associated with development of liver cirrhosis and hepatocellular carcinoma in persons with chronic hepatitis B

Risk factor Host Viral Miscellaneous
HCC and liver cirrhosis • Older age (>40 years) • High serum HBV DNA (>2,000 IU/mL) • Alcohol
• Persistent ALT elevation • High serum HBsAg • Metabolic syndrome
• Male • Genotype C • Diabetes
• Concurrent infection (HCV, HDV, HIV) • Delayed HBeAg seroconversion • Obesity
• Basal core promotor mutation
HCC • Presence of cirrhosis • Aflatoxin
• Family history of HCC • Smoking

HCC, hepatocellular carcinoma; ALT, alanine aminotransferase; HCV, hepatitis C virus; HDV, hepatitis D virus; HIV, human immunodeficiency virus; HBsAg, hepatitis B surface antigen; HBeAg, hepatitis B e antigen.

Table 5.

HCC risk prediction models

Prediction model Patients Antiviral therapy Variables Cutoff Cumulative incidence of HCC NPV (%)
GAG-HCC [56] 820 Asian (Hong Kong) Treatment-naïve Age, gender, HBV DNA, cirrhosis Low (≤100) - 99% at 10 years
High (>100)
CU-HCC4 [87] 1,005 Asian (Hong Kong) Treatment-naïve Age, HBV DNA, cirrhosis, albumin, bilirubin Low (<5) Low (2.2% at 10 years) 97.8% at 10 years
Intermediate (5–19) Intermediate (14.5% at 10 years)
High (>19) High (29.4% at 10 years)
REACH-B [85] 3,584 Asian (Taiwan) Treatment-naïve Age strata, gender, HBV DNA, ALT, HBeAg Low (0–5) Low (0.5% at 10 years) 98% at 10 years
Intermediate (6–11) Intermediate (8.4% at 10 years)
High (12–17) High (81.6% at 10 years)
LSM-HCC4 [88] 1,035 Asian (Hong Kong) 38% on antiviral therapy Age, HBV DNA, albumin, LSM Low (0–10) Low (0.6% at 5 years) 99.4% at 5 years
High (11–30) High (8.8% at 5 years)
THRI [93] 2,079 (396 HBV) Western (Canada) Regardless of antiviral therapy Age, gender, platelet, etiology Low (<120) Low (2.7% at 10 years) -
Intermediate (120–240) Intermediate (9.8% at 10 years)
High (>240) High (32.1% at 10 years)
mREACH-B [86] 192 Asian (Korea) ETV Age, gender, ALT, HBeAg, LSM Low (<10) Low (5.6% at 5 years) 96.8% at 5 years
High (≥10) High (20.6% at 5 years)
PAGE-B [88] 1,325 Western (Europe) ETV or TDF Age, gender, platelet Low (0–9) Low (0% at 5 years) 100% at 5 years
Intermediate (10–17) Intermediate (3% at 5 years)
High (≥18) High (17% at 5 years)
mPAGE-B [91] 2,001 Asian (Korea) ETV or TDF Age, gender, platelet, albumin Low (0–8) Low (0.7% at 5 years) -
Intermediate (9–12) Intermediate (5.1% at 5 years)
High (≥13) High (18.4% at 5 years)
CAGE-B [94] 1,427 Western (Europe) ETV or TDF Age, cirrhosis, LSM Low (0–5) Low (0% at 12 years) 100% at 12 years
Intermediate (6–10) Intermediate (1.8% at 12 years)
High (11–16) High (15.4% at 12 years)
SAGE-B [94] 1,427 Western (Europe) ETV or TDF Age, LSM Low (0–5) Low (0% at 12 years) 100% at 12 years
Intermediate (6–10) Intermediate (4.0% at 12 years)
High (11–15) High (13.8% at 12 years)
FSAC [97] 5,147 Asian (Korea) ETV or TDF Age, gender, cirrhosis, noninvasive fibrosis marker (FIB-4, APRI) response Low (≤2) Low (0.4% at 10 years) 99.0% at 10 years
Intermediate (3–8) Intermediate (7.5% at 10 years)
High (≥9) High (36.3% at 10 years)
mPAGELS-B [90] 2,184 Asian (Korea) ETV or TDF Age, gender, platelet, LSM Low (<12) Low (0.5% at 5 years) -
Intermediate (12–24) Intermediate (4.3% at 5 years)
High (≥24) High (18.1% at 5 years)

HCC, hepatocellular carcinoma; NPV, negative predictive value; HBV, hepatitis B virus; ALT, alanine aminotransferase; HBeAg, hepatitis B e antigen; LSM, liver stiffness measurement; FIB-4, fibrosis-4; APRI, aspartate aminotransferase-to-platelet ratio index.

Table 6.

Role of emerging HBV markers

Category Potential role HBV marker
Natural history HBeAg seroconversion Quantification of HBsAg, HBcrAg
Diagnostic tool for differentiating disease states Quantification of HBsAg, HBcrAg
cccDNA activity Amounts of intrahepatic cccDNA and cccDNA activities Quantification of HBsAg, HBcrAg, HBV RNA
Endpoint for testing therapeutic agents that target cccDNA Quantification of HBsAg, HBV RNA, HBcrAg
HBV treatment Predictors of successful withdrawal of therapy Quantification of HBsAg, HBcrAg, HBV RNA, cccDNA
Risk of reactivation during therapy or after therapy withdrawal Quantification of HBsAg, HBcrAg, HBV RNA, cccDNA
HCC occurrence/recurrence Evaluation of HCC occurrence HBcrAg, HBV RNA, HBV integration
HCC recurrence HBcrAg, HBV RNA, HBV integration
HBV reactivation HBV reactivation by immune-related therapy HBcrAg, cccDNA

HBV, hepatitis B virus; HBeAg, hepatitis B e antigen; HBsAg, hepatitis B surface antigen; HBcrAg, hepatitis B core-related antigen; cccDNA, covalently closed circular DNA; HCC, hepatocellular carcinoma.

Table 7.

Novel antiviral agents under development for HBV functional cure

Drug class Treatment regimen Efficacy
Route Company Phase
Mean reduction of log10 IU/mL Proportion of subjects
Capsid assembly modulator ABI-H0731 (Vebicorvir)+Nas [216] ΔlogHBsAg >0.5: 40% in TN PO Assembly Biosciences, USA II/III
JNJ-6379+TDF/ETV [217] 0.4 in TN & HBeAg (+) (250 mg) PO J&J with Arrowhead, USA II
ASO GSK3228836 [224] 1.56 in TN (300 mg) ΔlogHBsAg>1: 50% in TN, 60% in VS (300 mg) SC Ionis with GSK, USA II
1.99 in VS (300 mg) HBsAg loss: 17% in TN, 40% in VS (300 mg)
siRNA JNJ-3989+TDF/ETV [225] 1.93 ΔlogHBsAg>1: 98% SC J&J with Arrowhead, USA II
ΔlogHBsAg>2: 30%
AB-729+NAs [226] 2.03 in TN (90 mg q 8 w) HBsAg <1: 14% in TN, SC Arbutus Biopharma, USA II
2.16 in VS (90 mg q 8 w) 17% in VS (90 mg q 8 w)
Vir2218±pegIFN [227] 2.3 in VS (with pegIFN) HBsAg loss: 8.1% (with pegIFN) SC Vir Biotech, USA II
RG6346 [228] 1.0 in TN ΔlogHBsAg >1: 92% in VS SC Roche, Switzerland, with Dicerna II
1.7 in VS
HBsAg release inhibitor REP 2139 or REP 2165+pegIFN+TDF [229] 4.2 in VS HBsAg loss: 60% IV Replicor, Canada II
ΔlogHBsAg >1: 87.5%
TLR-8 agonist GS-9688 (selgantolimod)+NAs [231,232] ΔlogHBsAg >0.5: 7.4% in TN PO Gilead Sciences, USA II
ΔlogHBsAg >1: 2.6% in VS
HBsAg loss: 5% in VS
ICI ASC22 (envafolimab)+TDF/ETV [234] 0.49 in VS (2.5 mg/kg) IV Ascletis Pharma, China II
Therapeutic vaccine NASVAC±NAs [235] 0.579 in TN HBsAg loss: 21.7% in TN, 6.3% in VS Nasal CIGB, Cuba III
0.216 in VS
GS-4774+TDF [236,237] 0.135 in TN (40 YU) SC Gobelmmune with Gilead, USA II
0.166 in VS (40 YU)
Combination ETV+pegIFNα2+HBV vaccine [238] 0.57 (100 w, sVIP) HBsAg loss: 16.2% in sVIP III
JNJ-3989±JNJ-6379+ETV/TDF/TAF [240] 2.6 (JNJ-3989 200 mg) HBsAg <10: 29.7% (JNJ3989 200 mg) J&J with Arrowhead, USA II
1.8 (JNJ-3989 100 mg+JNJ-6379 250 mg) 12.8% (JNJ-3989 100 mg+JNJ-6379 250 mg)

HBV, hepatitis B virus; NAs, nucleos(t)ide analogues; TDF, tenofovir disoproxil fumarate; ETV, entecavir; TN, treatment-naïve; PO, per oral; HBeAg, hepatitis B e antigen; Δ, reduction after treatment; HBsAg, hepatitis B surface antigen; ASO, antisense oligonucleotide; VS, virologically stable; siRNA, small interfering RNA; IFN, interferon; SC, subcutaneous injection; w, weeks; IV, intravenous injection; TLR, toll-like receptor; ICI, immune checkpoint inhibitor; YU, yeast units; pegIFN, pegylated interferon; TAF, tenofovir alafenamide; sVIP, sequential HBV vaccination on an intensified schedule.

Table 8.

Dose adjustment of NAs for adult patients with altered creatinine clearance

Creatinine clearance (mL/min)* Recommended dose
Nucleoside analogues
Lamivudine
≥50 100 mg q 24 hours
30–49 100 mg first dose, then 50 mg q 24 hours
15–29 100 mg first dose, then 25 mg q 24 hours
5–14 35 mg first dose, then 15 mg q 24 hours
<5 35 mg first dose, then 10 mg q 24 hours
Telbivudine
≥50 600 mg q 24 hours
30–49 600 mg q 48 hours
<30 (not requiring dialysis) 600 mg q 72 hours
End-stage renal disease 600 mg q 96 hours
Entecavir NA naïve Lamivudine refractory/resistant
≥50 0.5 mg q 24 hours 1 mg q 24 hours
30–49 0.25 mg q 24 hours or 0.5 mg q 48 hours 0.5 mg q 24 hours or 1 mg q 48 hours
10–29 0.15 mg q 24 hours or 0.5 mg q 72 hours 0.3 mg q 24 hours or 1 mg q 72 hours
<10 or hemodialysis or continuous ambulatory peritoneal dialysis 0.05 mg q 24 hours or 0.5 mg q 7 days 0.1 mg q 24 hours or 1 mg q 7 days
Nucleotide analogues
Adefovir
≥50 10 mg q 24 hours
20–49 10 mg q 48 hours
10–19 10 mg q 72 hours
<10 Not indicated
Hemodialysis 10 mg q 7 days following dialysis
Besifovir
≥50 150 mg q 24 hours
30–49 150 mg q 48 hours
15–29 150 mg q 96 hours
<15 Not indicated
Tenofovir disoproxil fumarate
≥50 300 mg q 24 hours
30–49 300 mg q 48 hours
10–29 300 mg q 72–96 hours
<10 with dialysis 300 mg q 7 days or after a total of approximately 12 hours of dialysis
<10 without dialysis Not indicated
Tenofovir alafenamide fumarate
≥15 25 mg q 24 hours
<15 with dialysis§ 25 mg q 24 hours
<15 without dialysis Not indicated

NA, nucleos(t)ide analogue.

*

Calculated using ideal (lean) body weight.

Administered after hemodialysis.

Generally once a weekly, assuming three hemodialysis sessions of approximately 4 hours duration per week. Administered following completion of dialysis.

§

Evidence is insufficient.

Table 9.

Risk of hepatitis B reactivation associated with immune-related therapies

Risk of reactivation Immune-related therapies
HBsAg-positive
High risk (≥10%) B-cell depleting agents (rituximab, ofatumumab, natalizumab, alemtuzumab, ibritumomab, and obinutuzumab)
Human immunoglobulin G1 monoclonal antibody targeting CD38-expressing cells (daratumumab)*
High-dose corticosteroids (prednisone ≥20 mg/day, ≥4-week)
Anthracycline derivatives (doxorubicin, daunorubicin, and epirubicin)
Potent TNFα inhibitors (infliximab, adalimumab, certolizumab, and golimumab)
Local therapy for HCC (TACE)
Chimeric antigen receptor (CAR) T cell therapy*
Moderate risk (1–10%) Cytotoxic systemic chemotherapies other than anthracycline derivatives
Moderate-dose corticosteroids (prednisone 10–20 mg/day, ≥4-week)
Less potent TNFα inhibitors (etanercept)
Cytokine-based therapies (abatacept, ustekinumab, mogamulizumab, natalizumab, vedolizumab, secukinumab*, tofacitinib*, baricitinib*, and guselkumab*)
Immunophilin inhibitors (cyclosporine)
mTOR inhibitors (everolimus*, temsirolimus*)
Tyrosine-kinase inhibitors (osimertinib, imatinib, nilotinib, gefitinib, dasatinib*, erlotinib, afatinib, ibrutinib*, idelalisib*, palbociclib*, and ribociclib*)
Proteasome inhibitors (bortezomib)
Histone deacetylase inhibitors
Immune checkpoint inhibitors (ICIs) (pembrolizumab, nivolumab, atezolizumab, durvalumab)
Low risk (<1%) Antimetabolites, azathioprine, 6-mercaptopurine, methotrexate, mycophenolate mofetil, leflunomide, hydroxychloroquine, hydroxyurea*, immunomodulatory drugs (thalidomide, lenalidomide, and pomalidomide)*
Low-dose corticosteroids (prednisone <10 mg/day)
Intra-articular steroid injections (extremely low risk)
HBsAg-negative/anti-HBc-positive
High risk (≥10%) B-cell depleting agents (rituximab, ofatumumab, natalizumab, alemtuzumab, ibritumomab, and obinutuzumab)
Moderate risk (1–10%) High-dose corticosteroids (prednisone ≥20 mg/day, ≥4-week)
Anthracycline derivatives (doxorubicin, daunorubicin, and epirubicin)
Potent TNFα inhibitors (infliximab, adalimumab, certolizumab, and golimumab)
Cytotoxic systemic chemotherapies other than anthracycline derivatives
Cytokine-based therapies (abatacept, ustekinumab, mogamulizumab, natalizumab, vedolizumab, secukinumab*, tofacitinib*, baricitinib*, and guselkumab*)
Immunophilin inhibitors (cyclosporine)
mTOR inhibitors (everolimus*, temsirolimus*)
Tyrosine-kinase inhibitors (osimertinib, imatinib, nilotinib, gefitinib, dasatinib*, erlotinib, afatinib, ibrutinib*, idelalisib*, palbociclib*, and ribociclib*)
Proteasome inhibitors (bortezomib)
Human immunoglobulin G1 monoclonal antibody targeting CD38-expressing cells (daratumumab)*
Histone deacetylase inhibitors
Low risk (<1%) Moderate-dose (prednisone 10–20 mg/day) or low-dose (prednisone <10 mg/day) corticosteroids
Antimetabolites, azathioprine, 6-mercaptopurine, methotrexate, mycophenolate mofetil, leflunomide, hydroxychloroquine, hydroxyurea*, immunomodulatory drugs (thalidomide, lenalidomide, and pomalidomide)*
ICIs (pembrolizumab, nivolumab, atezolizumab, durvalumab)
CAR T cell therapy*

Modified from Loomba et al. [316]

HBsAg, hepatitis B surface antigen; TNFα, tumor necrosis factor-alpha; HCC, hepatocellular carcinoma; TACE, transarterial chemoembolization; mTOR, mammalian target of rapamycin; HBc, hepatitis B core.

*

For some immune-related therapies, the evidence for risk of hepatitis B virus reactivation is insufficient.