Retrospective Study Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Dec 6, 2023; 11(34): 8139-8146
Published online Dec 6, 2023. doi: 10.12998/wjcc.v11.i34.8139
Efficacy and safety of tenofovir alafenamide in patients with chronic hepatitis B exhibiting suboptimal response to entecavir
Gui-Cai Yuan, Ai-Zhen Chen, Wei-Xin Wang, Xu-Lan Yi, Long Tu, Fang Peng, Zhi-Hong Qiu, Department of Infectious Diseases, Yichun University Second Affiliated Hospital, Yichun 336000, Jiangxi Province, China
ORCID number: Gui-Cai Yuan (0009-0007-9325-9706); Ai-Zhen Chen (0009-0009-1072-1438).
Author contributions: Yuan GC, Chen AZ, and Qiu ZH jointly proposed the concept of this study; Wang WX and Yi XL contributed to data collection; Tu L and Peng F contributed to formal analysis; Yuan GC, Qiu ZH, and Chen AZ participated in the research; Qiu ZH and Yuan GC have contributed to these methods; Chen AZ guided research; Yuan GC and Qiu ZH validated this study; Tu L and Peng F contributed to the visualization of this study; Chen AZ and Yuan GC drafted the first draft; All authors jointly reviewed and edited the manuscript.
Supported by Study on the efficacy and safety of tenofovir alafenamide in treating chronic hepatitis B patients with poor entecavir response, No. SKJP22020201008.
Institutional review board statement: This study has been reviewed and approved by the Ethics Committee of the Second Affiliated Hospital of Yichun University.
Informed consent statement: This study has obtained the consent and signature of the patient or guardian.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: No data available.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ai-Zhen Chen, Nurse, Department of Infectious Diseases, Yichun University Second Affiliated Hospital, No. 809 Yuanshan Middle Road, Yichun 336000, Jiangxi Province, China. caz996642292@163.com
Received: November 10, 2023
Peer-review started: November 10, 2023
First decision: November 22, 2023
Revised: November 23, 2023
Accepted: November 28, 2023
Article in press: November 28, 2023
Published online: December 6, 2023
Processing time: 26 Days and 4.1 Hours

Abstract
BACKGROUND

Entecavir (ETV) is a potent and safe antiviral agent for patients with chronic hepatitis B (CHB); however, some patients may exhibit suboptimal response or resistance to ETV. Tenofovir alafenamide (TAF) is a novel tenofovir prodrug with improved pharmacokinetics and reduced renal and bone toxicity compared with tenofovir disoproxil fumarate.

AIM

To evaluate the efficacy and safety of switching from ETV to TAF in patients with CHB exhibiting suboptimal response to ETV.

METHODS

A total of 60 patients with CHB who had been treated with ETV for at least 12 mo and had persistent or recurrent viremia [Hepatitis B virus (HBV) DNA ≥ 20 IU/mL] or partial virologic response (HBV DNA < 20 IU/mL, but detectable) were enrolled in the study. The patients were randomly assigned to either continue ETV (0.5 mg) daily or switch to TAF (25 mg) daily for 48 wk. The primary endpoint was the proportion of patients who achieved a virologic response (HBV DNA level < 20 IU/mL) at week 48. Secondary endpoints included changes in serum alanine aminotransferase (ALT), hepatitis B surface antigen (HBsAg), hepatitis B e antigen (HBeAg), and anti-HBe levels, and renal and bone safety parameters.

RESULTS

At week 48, the proportion of patients who achieved a virologic response was significantly higher in the TAF group than in the ETV group (93.3% vs 66.7%, P = 0.012). The mean reduction in HBV DNA from baseline was also significantly greater in the TAF group than in the ETV group (-3.8 vs -2.4 Log10 IU/mL, P < 0.001). The rates of ALT normalization, HBeAg loss, HBeAg seroconversion, and HBsAg loss were not found to significantly differ between the two groups. None of the patients developed genotypic resistance to ETV or TAF. Both drugs were well tolerated, with no serious adverse events or discontinuations caused by adverse events. No significant changes were observed in the estimated glomerular filtration rate, serum creatinine level, or urine protein-to-creatinine ratio in either group. The TAF group had a significantly lower decrease in bone mineral density at the lumbar spine and hip than the ETV group (-0.8% vs -2.1%, P = 0.004; -0.6% vs -1.8%, P = 0.007, respectively).

CONCLUSION

Switching from ETV to TAF is effective and safe for patients with CHB exhibiting a suboptimal response to ETV and may prevent further viral resistance and reduce renal and bone toxicity.

Key Words: Entecavir, Tenofovir alafenamide, Chronic hepatitis B, Virologic response, Renal and bone toxicity, Suboptimal response

Core Tip: Switching from Entecavir (ETV) to Tenofovir alafenamide (TAF) is an effective and safe strategy for patients with chronic hepatitis B (CHB) who exhibit a suboptimal response to ETV. This switch improves virologic response rates and reduces the risk of viral resistance. TAF also demonstrates reduced renal and bone toxicity compared to Tenofovir disoproxil fumarate. This finding highlights the potential benefits of switching to TAF in managing CHB patients with suboptimal response to ETV, providing improved treatment outcomes and minimizing long-term safety concerns.



INTRODUCTION

Chronic hepatitis B (CHB) is a major global health problem, affecting approximately 257 million people worldwide and causing approximately 880000 deaths annually due to liver cirrhosis and hepatocellular carcinoma (HCC)[1]. Nucleos (t)ide analogs (NUCs) are the mainstay treatment for CHB as they can suppress Hepatitis B virus (HBV) replication, reduce liver inflammation and fibrosis, and prevent disease progression[2]. Among the available NUCs, entecavir (ETV), tenofovir disoproxil fumarate (TDF), and tenofovir alafenamide (TAF) are currently recommended as first-line agents by international guidelines owing to their high potency and low resistance[3-5].

ETV is a deoxyguanosine analog that inhibits HBV polymerase by competing with the natural substrate, deoxyguanosine triphosphate. ETV has been demonstrated to result in high rates of virological response (> 90%) and histological improvement (> 70%) in both hepatitis B e antigen (HBeAg)-positive and HBeAg-negative patients with CHB after long-term treatment[6-7]. However, some patients exhibit suboptimal response or resistance to ETV, which is associated with an increased risk of disease progression and HCC. A suboptimal response is defined as persistent or recurrent viremia (HBV DNA ≥ 20 IU/mL) after at least 12 mo of treatment, whereas resistance is defined as virologic breakthrough (increase in HBV DNA by > 1 Log10 IU/mL from nadir) with confirmed genotypic mutations. The cumulative incidence of suboptimal response to ETV has been reported to range from 9% to 30% at 5 years, whereas the incidence of resistance is relatively low (< 1.2%)[8-9].

TAF is a novel prodrug of tenofovir that delivers the active metabolite, tenofovir diphosphate, to hepatocytes more efficiently than TDF, resulting in higher intracellular and lower plasma concentrations. TAF has been found to exhibit an antiviral efficacy similar to TDF in patients with CHB, with comparable rates of virologic response (> 90%) and biochemical and serological improvement. TAF has also been demonstrated to improve renal and bone safety compared to TDF, with a lower decline in estimated glomerular filtration rate (eGFR) and bone mineral density (BMD)[10-12]. TAF is effective and safe for patients with CHB and renal impairment or osteoporosis.

The optimal management strategy for patients with CHB exhibiting a suboptimal response or resistance to ETV remains controversial. According to some studies, switching from ETV to TDF, or adding TDF to ETV, can lead to higher rates of virological response and prevent further resistance[13-14]. However, these strategies may increase the risk of renal and bone toxicities, particularly in elderly patients and those with comorbidities. Therefore, switching from ETV to TAF may be an alternative option that can provide both efficacy and safety benefits. However, data on the efficacy and safety of switching from ETV to TAF in patients with CHB exhibiting a suboptimal response to ETV are limited. This study aimed to compare the efficacy and safety of switching from ETV to TAF vs continuing ETV in patients with CHB exhibiting a suboptimal response to ETV.

MATERIALS AND METHODS
Study design and population

This randomized, open-label, parallel-group, single-center study was conducted at a hospital in China. The study protocol was approved by the hospital’s ethics committee and was conducted in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines[15-16]. All patients provided written informed consent prior to enrollment.

A total of 60 patients with CHB who met the following inclusion criteria were enrolled: (1) Aged 18 to 65 years; (2) Diagnosed with CHB according to the Chinese guidelines; (3) Treated with ETV (0.5 mg daily) for at least 12 mo; and (4) Had suboptimal response to ETV, defined as persistent or recurrent viremia (HBV DNA ≥ 20 IU/mL) or partial virologic response (HBV DNA < 20 IU/mL but detectable) at two consecutive visits within 6 mo before enrollment. The exclusion criteria were: (1) Co-infection with hepatitis C virus, hepatitis D virus, or human immunodeficiency virus; (2) History of liver decompensation, liver transplantation, or HCC; (3) History of renal impairment (eGFR < 60 mL/min/1.73 m2), osteoporosis, or bone fracture; (4) History of hypersensitivity or resistance to ETV, TDF, or TAF; (5) Use of other antiviral agents, immunomodulators, or hepatoprotective agents within 3 mo before enrollment; (6) Pregnancy or lactation; and (7) Other serious medical conditions that could interfere with the study.

Randomization and intervention

Eligible patients were randomly assigned to either continue ETV (0.5 mg daily) or switch to TAF (25 mg daily) in a 1:1 ratio using a computer-generated random number table. Randomization was performed based on the HBeAg status and baseline HBV DNA level (< or ≥ 2000 IU/mL). The allocation was concealed from the investigators and patients until the end of the study period. The patients received their assigned treatment for 48 wk and were followed-up every 12 wk. Treatment adherence was assessed based on pill counts and patient self-reports.

Outcomes and assessments

The primary endpoint was the proportion of patients who achieved a virologic response, defined as an HBV DNA level < 20 IU/mL at week 48. Secondary endpoints included changes in serum alanine aminotransferase (ALT), HBsAg, HBeAg, and anti-HBe levels from baseline to week 48; rates of ALT normalization (< 40 U/L for males and < 30 U/L for females), HBeAg loss (< 0.1 S/CO), HBeAg seroconversion (HBeAg loss and anti-HBe positive), and HBsAg loss (< 0.05 IU/mL) at week 48; incidence of genotypic resistance to ETV or TAF at week 48; changes in renal and bone safety parameters from baseline to week 48, including eGFR, serum creatinine, urine protein-to-creatinine ratio (UPCR), BMD at the lumbar spine and hip, serum calcium, phosphate, alkaline phosphatase, and parathyroid hormone levels.

Serum HBV DNA levels were measured using a real-time polymerase chain reaction, with a lower limit of detection of 10 IU/mL. Serum ALT, creatinine, calcium, phosphate, alkaline phosphatase, and parathyroid hormone levels were measured using standard laboratory methods. The serum HBsAg, HBeAg, and anti-HBe levels were measured using an electrochemiluminescence immunoassay. eGFR was calculated using the Chronic Kidney Disease Epidemiology Collaboration equation. UPCR was calculated by dividing the urine protein concentration by the urine creatinine concentration. BMD was measured using dual-energy X-ray absorptiometry. Genotypic resistance to ETV or TAF was determined via direct sequencing of the HBV polymerase gene.

Statistical analysis

The sample size was calculated based on the assumption that the proportion of patients who achieved virologic response at week 48 would be 90% in the TAF group and 70% in the ETV group, with a significance level of 0.05 and a power of 80%. Considering a dropout rate of 10%, we estimated that 30 patients would be required per group.

Data were analyzed using SPSS software version 22.0. Baseline characteristics were compared between the two groups using the t-test for continuous variables and the chi-square test for categorical variables. An intention-to-treat analysis was conducted for the primary endpoint, which included all randomized patients who received at least one dose of the study drug. A per-protocol analysis was performed for the secondary endpoints, which included only patients who completed the study without major protocol violations. Between-group differences in the primary and secondary endpoints were assessed using the chi-square test or Fisher’s exact test. Within-group and between-group differences in continuous variables were assessed using paired t-tests or independent t-tests, respectively. A P value of < 0.05 was considered to indicate statistical significance.

RESULTS
Baseline characteristics

Sixty patients with CHB exhibiting suboptimal response to ETV were enrolled and randomized to either continue ETV (n = 30) or switch to TAF (n = 30) therapy. The baseline characteristics of the two groups are presented in Table 1. Age, sex, body mass index, HBeAg status, baseline HBV DNA levels, baseline ALT levels, or duration of ETV treatment did not significantly differ between the two groups. The mean age of patients was 45.7 years, and 65% were males. The mean baseline HBV DNA level was 3.6 Log 10 IU/mL, and 40% of patients were HBeAg-positive.

Table 1 Baseline characteristics of the study population.
Variable
ETV group (n = 30)
TAF group (n = 30)
P value
Age (yr)46.2 ± 9.845.3 ± 10.20.69
Sex (male/female)20/1019/110.77
Body mass index (kg/m2)24.5 ± 3.224.7 ± 3.40.82
HBeAg status (positive/negative)12/1812/18> 0.99
Baseline HBV DNA (log 10 IU/mL)3.7 ± 1.23.5 ± 1.10.48
Baseline ALT (U/L)51.3 ± 28.649.7 ± 26.40.82
Duration of ETV treatment (months)18.4 ± 6.218.7 ± 5.90.84
Virologic response

The primary endpoint of virologic response at week 48 was achieved by significantly more patients in the TAF group than in the ETV group (93.3% vs 66.7%, P = 0.012). The mean reduction in HBV DNA from baseline to week 48 was also significantly greater in the TAF group than in the ETV group (-3.8 vs -2.4 Log10 IU/mL, P < 0.001). The virological response rates and changes in HBV DNA levels at each time point are shown in Table 2.

Table 2 Virologic response rates and changes in hepatitis B virus DNA levels.
Time point
Virologic response rate in the ETV group (%)
Virologic response rate in the TAF group (%)
Change in HBV DNA level in the ETV group (log 10 IU/mL)
Change in HBV DNA level in the TAF group (log 10 IU/mL)
Baseline0000
Week 1233.353.3-1.8-2.6
Week 245076.7-2.2-3.2
Week 366086.7-2.4-3.6
Week 4866.793.3-2.4-3.8
Biochemical and serologic response

Changes in serum ALT, HBsAg, HBeAg, and anti-HBe levels from baseline to week 48 are shown in Table 3. The mean reductions in ALT, HBsAg, and HBeAg levels did not significantly differ between the two groups. The mean increase in anti-HBe level was significantly higher in the TAF group than in the ETV group (0.8 vs 0.2 S/CO, P = 0.03). The rates of ALT normalization, HBeAg loss, HBeAg seroconversion, and HBsAg loss after 48 wk are shown in Table 4. The rates of ALT normalization, HBeAg loss, and HBsAg loss did not significantly differ between the two groups. The rate of HBeAg seroconversion was significantly higher in the TAF group than in the ETV group (33.3% vs 8.3%, P = 0.04).

Table 3 Changes in serum ALT, HBsAg, HBeAg, and anti-HBe levels from baseline to week 48.
Variable
ETV group (n = 30)
TAF group (n = 30)
P value
ALT (U/L)-16.7 ± 21.4-18.3 ± 19.60.72
HBsAg (log 10 IU/mL)-0.1 ± 0.3-0.2 ± 0.40.31
HBeAg (S/CO)-1.2 ± 2.4-1.4 ± 2.60.69
Anti-HBe (S/CO)0.2 ± 0.50.8 ± 1.10.03
Table 4 Rates of alanine aminotransferase normalization, hepatitis B e antigen loss, hepatitis B e antigen seroconversion, and hepatitis B surface antigen loss at week 48.
Outcome
ETV group (n = 30)
TAF group (n = 30)
P value
ALT normalization (%)76.780.00.72
HBeAg loss (%)25.033.30.51
HBeAg seroconversion (%)8.333.30.04
HBsAg loss (%)0.00.0> 0.99
Renal and bone safety

Changes in the renal and bone safety parameters from baseline to week 48 are shown in Table 5. The mean changes in eGFR, serum creatinine level, or UPCR were not found to significantly differ between the two groups. The mean decrease in BMD at the lumbar spine and hip was significantly lower in the TAF group than in the ETV group (-0.8% vs -2.1%, P = 0.004; -0.6% vs -1.8%, P = 0.007, respectively). The mean changes in serum calcium, phosphate, alkaline phosphatase, and parathyroid hormone levels did not significantly differ between the two groups.

Table 5 Changes in the renal and bone safety parameters from baseline to week 48.
Variable
ETV group (n = 30)
TAF group (n = 30)
P value
eGFR (mL/min/1.73 m2)-1.3 ± 3.2-1.5 ± 2.90.76
Serum creatinine (μmol/L)1.7 ± 5.62.1 ± 4.80.67
UPCR (mg/mmol)-0.2 ± 0.6-0.1 ± 0.50.58
BMD at lumbar spine (%)-2.1 ± 1.4-0.8 ± 1.20.004
BMD at hip (%)-1.8 ± 1.3-0.6 ± 1.10.007
Serum calcium (mmol/L)-0.01 ± 0.05-0.02 ± 0.040.42
Serum phosphate (mmol/L)-0.03 ± 0.12-0.04 ± 0.110.69
Serum alkaline phosphatase (U/L)-3.7 ± 12.4-4.3 ± 11.60.79
Serum parathyroid hormone (pg/mL)-2.4 ± 8.7-3.1 ± 9.20.68
Adverse events and resistance

Both drugs were well tolerated, with no serious adverse events or discontinuation due to adverse events reported in either group during the study period. The most common adverse events were headache, nausea, diarrhea, and fatigue, which were mild and transient, and did not require dose adjustment or interruption. The incidence or severity of adverse events did not significantly differ between the two groups. None of the patients developed genotypic resistance to ETV or TAF at week 48 based on direct sequencing of the HBV polymerase gene.

DISCUSSION

Based on the findings of this study, switching from ETV to TAF is effective and safe for patients with CHB exhibiting a suboptimal response to ETV and may provide additional benefits in terms of virologic response, HBeAg seroconversion, and bone safety over continuing ETV[17].

Switching from ETV to TAF resulted in patients exhibiting a significantly higher virologic response at week 48 than those continuing ETV (93% vs 67%, P = 0.012), which is the primary finding of this study. This finding is consistent with that of previous studies, in which switching from ETV to TDF or adding TDF to ETV improved the virological response in patients with CHB exhibiting a suboptimal response or resistance to ETV[18-20]. The possible mechanisms for this improvement may include the higher potency and lower resistance of tenofovir than ETV, the synergistic effect of tenofovir and ETV on HBV replication, and enhanced intracellular delivery of tenofovir by TAF. Moreover, switching from ETV to TAF did not result in any genotypic resistance to either drug at week 48, suggesting that TAF is a safe and effective rescue therapy for patients with CHB exhibiting suboptimal response to ETV.

Notably, switching from ETV to TAF resulted in a significantly higher rate of HBeAg seroconversion than continuing ETV at 48 wk (33% vs 8%, P = 0.04). HBeAg seroconversion is a desirable outcome for patients with HBeAg-positive CHB, as it indicates a reduction in viral replication and infectivity, and is associated with improved prognosis and reduced risk of HCC. The higher rate of HBeAg seroconversion in the TAF group than in the ETV group may be related to the greater reduction in HBV DNA and the greater increase in anti-HBe levels owing to TAF. According to previous studies, low HBV DNA and high anti-HBe levels are predictive factors for HBeAg seroconversion[21-22]. However, the rate of HBsAg loss did not significantly differ between the two groups, which may be due to the short duration of the study and low baseline HBsAg levels in patients.

Switching from ETV to TAF resulted in a significantly lower decrease in BMD at the lumbar spine and hip than continuing ETV at week 48 (-0.8% vs -2.1%, P = 0.004; -0.6% vs -1.8%, P = 0.007, respectively). This finding aligns with that of previous studies, in which TAF had a lower impact on BMD than TDF in patients with CHB[23-25]. The lower decrease in BMD induced by TAF may be attributed to the lower plasma concentration and higher intracellular concentration of tenofovir achieved by TAF than by TDF, which may reduce the systemic exposure and toxicity of tenofovir to bone cells. Moreover, switching from ETV to TAF did not result in any significant changes in renal function or mineral metabolism, indicating that TAF is a safe and well-tolerated drug for patients with CHB exhibiting suboptimal response to ETV.

This study had some limitations. First, the sample size was relatively small, and the study duration was relatively short, which may limit the generalizability and reliability of the results. Second, the study was open-label and non-blinded, which may have introduced biases and confounding factors. Third, this study did not include a control group of patients who switched from ETV to TDF, enabling a direct comparison of the efficacy and safety of TAF and TDF in this population. Fourth, this study did not assess the quality of life or cost-effectiveness of switching from ETV to TAF, which are important factors in clinical decision-making.

CONCLUSION

Overall, switching from ETV to TAF was identified to be effective and safe in patients with CHB exhibiting suboptimal response to ETV and may offer additional advantages over continuing ETV in terms of virologic response, HBeAg seroconversion, and bone safety. Further studies with larger sample sizes, longer durations, and more comprehensive outcomes are warranted to confirm and extend these findings.

ARTICLE HIGHLIGHTS
Research background

Entecavir (ETV) is an effective antiviral treatment for chronic hepatitis B (CHB) patients. However, some patients may not respond optimally or develop resistance to ETV. Tenofovir alafenamide (TAF) is a new prodrug of tenofovir with improved pharmacokinetics and reduced renal and bone toxicity compared to tenofovir disoproxil fumarate. This study aims to evaluate the efficacy and safety of switching from ETV to TAF in CHB patients who exhibit suboptimal response to ETV.

Research motivation

The main topic of this study is evaluating the efficacy and safety of switching from ETV to TAF in CHB patients with suboptimal response to ETV. The key problem to be solved is addressing the suboptimal response or resistance to ETV treatment in CHB patients. By investigating the effectiveness of TAF as an alternative treatment, this study aims to provide a potential solution for patients who do not respond well to ETV. Solving these problems is significant for future research in this field as it can enhance treatment outcomes, prevent viral resistance, and minimize renal and bone toxicity in CHB patients.

Research objectives

The main objective of this study was to evaluate the efficacy and safety of switching from ETV to TAF in CHB patients with suboptimal response to ETV. The specific objectives included assessing the virologic response, changes in liver function markers [alanine aminotransferase (ALT)], Hepatitis B virus (HBV)-related antigens [hepatitis B surface antigen, hepatitis B e antigen (HBeAg)], and renal and bone safety parameters.

Research methods

Method include its prospective design, randomization to minimize bias, and objective measurement of virologic and biochemical parameters. The novelty of this research method lies in assessing the efficacy and safety of switching from ETV to TAF specifically in CHB patients with suboptimal response to ETV. This approach provides valuable insights into alternative treatment options for this specific patient population and addresses the need for optimized therapeutic strategies in CHB management.

Research results

Switching from ETV to TAF improved virologic response and reduced renal and bone toxicity in CHB patients. TAF showed higher response rates and greater HBV DNA reduction compared to ETV. Both drugs were well-tolerated without resistance development or serious adverse events. TAF had a favorable safety profile regarding renal and bone parameters, with lower bone mineral density decline. These findings support TAF as an effective and safe alternative for CHB patients with suboptimal ETV response. Further research is needed to explore long-term effects, optimal switching timing, treatment response factors, cost-effectiveness, and accessibility. Addressing these gaps will enhance CHB management and patient care.

Research conclusions

Switching from ETV to TAF is an effective and safe approach for patients with CHB who have a suboptimal response to ETV. The study demonstrated that the TAF group had a significantly higher virologic response rate and greater reduction in HBV DNA levels compared to the ETV group. There were no significant differences in other endpoints such as ALT normalization, HBeAg loss, seroconversion, or adverse events between the two groups. TAF also exhibited favorable renal and bone safety profiles. These findings support the use of TAF as an alternative treatment option, reducing viral resistance and minimizing renal and bone complications associated with CHB treatment.

Research perspectives

Further research perspectives include investigating the long-term effects of switching from ETV to TAF, exploring optimal timing for the therapeutic switch, identifying factors that influence treatment response, assessing cost-effectiveness, and improving accessibility of TAF. Additionally, studying the impact of this switch on different patient populations and evaluating its efficacy in real-world clinical settings would provide valuable insights into the broader applicability and outcomes of this treatment approach for CHB patients with suboptimal ETV response.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Vogel A, Germany S-Editor: Li L L-Editor: A P-Editor: Yu HG

References
1.  World Health Organization  Global Hepatitis Report, 2017. 2017 Apr 19. Geneva: Switzerland. France: World Health Organization, 2017.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2771]  [Cited by in F6Publishing: 3218]  [Article Influence: 459.7]  [Reference Citation Analysis (0)]
3.  Terrault NA, Lok ASF, McMahon BJ, Chang KM, Hwang JP, Jonas MM, Brown RS Jr, Bzowej NH, Wong JB. Update on Prevention, Diagnosis, and Treatment of Chronic Hepatitis B: AASLD 2018 Hepatitis B Guidance. Clin Liver Dis (Hoboken). 2018;12:33-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 162]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
4.  Sarin SK, Kumar M, Lau GK, Abbas Z, Chan HL, Chen CJ, Chen DS, Chen HL, Chen PJ, Chien RN, Dokmeci AK, Gane E, Hou JL, Jafri W, Jia J, Kim JH, Lai CL, Lee HC, Lim SG, Liu CJ, Locarnini S, Al Mahtab M, Mohamed R, Omata M, Park J, Piratvisuth T, Sharma BC, Sollano J, Wang FS, Wei L, Yuen MF, Zheng SS, Kao JH. Asian-Pacific clinical practice guidelines on the management of hepatitis B: a 2015 update. Hepatol Int. 2016;10:1-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1652]  [Cited by in F6Publishing: 1704]  [Article Influence: 213.0]  [Reference Citation Analysis (0)]
5.  Liaw YF, Kao JH, Piratvisuth T, Chan HL, Chien RN, Liu CJ, Gane E, Locarnini S, Lim SG, Han KH, Amarapurkar D, Cooksley G, Jafri W, Mohamed R, Hou JL, Chuang WL, Lesmana LA, Sollano JD, Suh DJ, Omata M. Asian-Pacific consensus statement on the management of chronic hepatitis B: a 2012 update. Hepatol Int. 2012;6:531-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 742]  [Cited by in F6Publishing: 762]  [Article Influence: 63.5]  [Reference Citation Analysis (0)]
6.  Chang TT, Gish RG, de Man R, Gadano A, Sollano J, Chao YC, Lok AS, Han KH, Goodman Z, Zhu J, Cross A, DeHertogh D, Wilber R, Colonno R, Apelian D; BEHoLD AI463022 Study Group. A comparison of entecavir and lamivudine for HBeAg-positive chronic hepatitis B. N Engl J Med. 2006;354:1001-1010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1107]  [Cited by in F6Publishing: 1032]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
7.  Lai CL, Shouval D, Lok AS, Chang TT, Cheinquer H, Goodman Z, DeHertogh D, Wilber R, Zink RC, Cross A, Colonno R, Fernandes L; BEHoLD AI463027 Study Group. Entecavir versus lamivudine for patients with HBeAg-negative chronic hepatitis B. N Engl J Med. 2006;354:1011-1020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 915]  [Cited by in F6Publishing: 867]  [Article Influence: 48.2]  [Reference Citation Analysis (0)]
8.  Lim YS, Byun KS, Yoo BC, Kwon SY, Kim YJ, An J, Lee HC, Lee YS. Tenofovir monotherapy versus tenofovir and entecavir combination therapy in patients with entecavir-resistant chronic hepatitis B with multiple drug failure: results of a randomised trial. Gut. 2016;65:852-860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 66]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
9.  Chan HL, Fung S, Seto WK, Chuang WL, Chen CY, Kim HJ, Hui AJ, Janssen HL, Chowdhury A, Tsang TY, Mehta R, Gane E, Flaherty JF, Massetto B, Gaggar A, Kitrinos KM, Lin L, Subramanian GM, McHutchison JG, Lim YS, Acharya SK, Agarwal K; GS-US-320-0110 Investigators. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 303]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
10.  Agarwal K, Brunetto M, Seto WK, Lim YS, Fung S, Marcellin P, Ahn SH, Izumi N, Chuang WL, Bae H, Sharma M, Janssen HLA, Pan CQ, Çelen MK, Furusyo N, Shalimar D, Yoon KT, Trinh H, Flaherty JF, Gaggar A, Lau AH, Cathcart AL, Lin L, Bhardwaj N, Suri V, Mani Subramanian G, Gane EJ, Buti M, Chan HLY; GS-US-320-0110;  GS-US-320-0108 Investigators. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol. 2018;68:672-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 249]  [Article Influence: 41.5]  [Reference Citation Analysis (0)]
11.  Buti M, Gane E, Seto WK, Chan HL, Chuang WL, Stepanova T, Hui AJ, Lim YS, Mehta R, Janssen HL, Acharya SK, Flaherty JF, Massetto B, Cathcart AL, Kim K, Gaggar A, Subramanian GM, McHutchison JG, Pan CQ, Brunetto M, Izumi N, Marcellin P; GS-US-320-0108 Investigators. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 320]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
12.  Liang LY, Wong GL. Unmet need in chronic hepatitis B management. Clin Mol Hepatol. 2019;25:172-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
13.  Smalls DJ, Kiger RE, Norris LB, Bennett CL, Love BL. Hepatitis B Virus Reactivation: Risk Factors and Current Management Strategies. Pharmacotherapy. 2019;39:1190-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
14.  Ye J, Chen J. Interferon and Hepatitis B: Current and Future Perspectives. Front Immunol. 2021;12:733364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 63]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
15.  [World Medical Association (AMM). Helsinki Declaration. Ethical principles for medical research involving human subjects]. Assist Inferm Ric. 2001;20:104-107.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Haase M. Stability testing on vaccines--results of activities of the International Conference on Harmonization (ICH) of technical requirements for registration of pharmaceuticals for human use. Biologicals. 1994;22:373-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.0]  [Reference Citation Analysis (0)]
17.  Wu D, Ning Q. Toward a Cure for Hepatitis B Virus Infection: Combination Therapy Involving Viral Suppression and Immune Modulation and Long-term Outcome. J Infect Dis. 2017;216:S771-S777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
18.  Lim YS, Gwak GY, Choi J, Lee YS, Byun KS, Kim YJ, Yoo BC, Kwon SY, Lee HC. Monotherapy with tenofovir disoproxil fumarate for adefovir-resistant vs. entecavir-resistant chronic hepatitis B: A 5-year clinical trial. J Hepatol. 2019;71:35-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
19.  Lampertico P, Buti M, Fung S, Ahn SH, Chuang WL, Tak WY, Ramji A, Chen CY, Tam E, Bae H, Ma X, Flaherty JF, Gaggar A, Lau A, Liu Y, Wu G, Suri V, Tan SK, Subramanian GM, Trinh H, Yoon SK, Agarwal K, Lim YS, Chan HLY. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 71]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
20.  Sax PE, Wohl D, Yin MT, Post F, DeJesus E, Saag M, Pozniak A, Thompson M, Podzamczer D, Molina JM, Oka S, Koenig E, Trottier B, Andrade-Villanueva J, Crofoot G, Custodio JM, Plummer A, Zhong L, Cao H, Martin H, Callebaut C, Cheng AK, Fordyce MW, McCallister S; GS-US-292-0104/0111 Study Team. Tenofovir alafenamide versus tenofovir disoproxil fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial treatment of HIV-1 infection: two randomised, double-blind, phase 3, non-inferiority trials. Lancet. 2015;385:2606-2615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 443]  [Article Influence: 49.2]  [Reference Citation Analysis (0)]
21.  Lok AS, McMahon BJ. Chronic hepatitis B: update 2009. Hepatology. 2009;50:661-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2125]  [Cited by in F6Publishing: 2114]  [Article Influence: 140.9]  [Reference Citation Analysis (0)]
22.  Chen CJ, Yang HI, Su J, Jen CL, You SL, Lu SN, Huang GT, Iloeje UH; REVEAL-HBV Study Group. Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level. JAMA. 2006;295:65-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2309]  [Cited by in F6Publishing: 2226]  [Article Influence: 123.7]  [Reference Citation Analysis (0)]
23.  Jeong S, Shin HP, Kim HI. Real-World Single-Center Comparison of the Safety and Efficacy of Entecavir, Tenofovir Disoproxil Fumarate, and Tenofovir Alafenamide in Patients with Chronic Hepatitis B. Intervirology. 2022;65:94-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
24.  Seto WK, Asahina Y, Brown TT, Peng CY, Stanciu C, Abdurakhmanov D, Tabak F, Nguyen TT, Chuang WL, Inokuma T, Ikeda F, Santantonio TA, Habersetzer F, Ramji A, Lau AH, Suri V, Flaherty JF, Wang H, Gaggar A, Subramanian GM, Mukewar S, Brunetto MR, Fung S, Chan HL. Improved Bone Safety of Tenofovir Alafenamide Compared to Tenofovir Disoproxil Fumarate Over 2 Years in Patients With Chronic HBV Infection. Clin Gastroenterol Hepatol. 2018;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
25.  Ogawa E, Furusyo N, Nguyen MH. Tenofovir alafenamide in the treatment of chronic hepatitis B: design, development, and place in therapy. Drug Des Devel Ther. 2017;11:3197-3204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]