Next Article in Journal
Development of a Cell Culture Model for Inducible SARS-CoV-2 Replication
Previous Article in Journal
Determination and Characterization of Novel Papillomavirus and Parvovirus Associated with Mass Mortality of Chinese Tongue Sole (Cynoglossus semilaevis) in China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Adaptive Immune Response in Hepatitis B Virus-Associated Hepatocellular Carcinoma Is Characterized by Dysfunctional and Exhausted HBV-Specific T Cells

by
Malene Broholm
1,*,
Anne-Sofie Mathiasen
1,
Ása Didriksen Apol
1 and
Nina Weis
1,2
1
Department of Infectious Disease, Copenhagen University Hospital, 2650 Hvidovre, Denmark
2
Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2300 Copenhagen, Denmark
*
Author to whom correspondence should be addressed.
Viruses 2024, 16(5), 707; https://doi.org/10.3390/v16050707
Submission received: 18 March 2024 / Revised: 25 April 2024 / Accepted: 26 April 2024 / Published: 29 April 2024
(This article belongs to the Section Human Virology and Viral Diseases)

Abstract

:
This systematic review investigates the immunosuppressive environment in HBV-associated hepatocellular carcinoma (HCC), characterized by dysfunctional and exhausted HBV-specific T cells alongside an increased infiltration of HBV-specific CD4+ T cells, particularly regulatory T cells (Tregs). Heightened expression of checkpoint inhibitors, notably PD-1, is linked with disease progression and recurrence, indicating its potential as both a prognostic indicator and a target for immunotherapy. Nevertheless, using PD-1 inhibitors has shown limited effectiveness. In a future perspective, understanding the intricate interplay between innate and adaptive immune responses holds promise for pinpointing predictive biomarkers and crafting novel treatment approaches for HBV-associated HCC.

1. Introduction

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths globally [1,2], with 55–80% of the cases linked to chronic hepatitis B virus (CHB) [3], which affects approximately 296 million individuals worldwide [4]. The natural history of CHB varies widely among patients, with some entering a phase of low viral replication and limited inflammation, while others may have persistent inflammation leading to tumorigenesis. Although the precise mechanisms determining disease outcomes remain unclear, there is growing evidence indicating that the prognosis of CHB relies on the interplay between the virus and the host immune response. The adaptive immune response is crucial for viral clearance, but the destruction of virus-infected cells also induces inflammation and liver damage.
During viral infections, CD4+ T cells primarily differentiate into T helper (Th1) effector cells, which induce the activation of cytotoxic CD8+ T cells, and into T follicular (Tfh) helper cells which activate B cells [5,6]. Additionally, CD4+ T cells can differentiate into regulatory T cells (Tregs), a specialized population that suppresses the activation, proliferation, and effector functions of multiple immune cells, including T cells, B cells, natural killer cells and dendritic cells [7]. This regulatory function is essential for maintaining self-tolerance. However, an expanded population of Tregs can inhibit the antiviral immune response, leading to persistent inflammation, and even suppress anti-cancer immunity and protective immune surveillance [8].
During acute HBV infection, the HBV-specific CD8+ T-cell response is usually strong and polyclonal, characterized by the production of pro-inflammatory cytokines such as INF-γ and TNF-α, and cytotoxic molecules such as granzyme and perforin, which are essential for controlling HBV infection [9]. Although levels of HBV-specific CD8+ T cells usually decline and become undetectable during the acute phase [10], these cells persistently remain detectable in CHB, albeit usually at low levels [9]. Previous research has suggested that the persistence of HBV-specific CD8+ T cells may contribute to HBV-induced carcinogenesis by maintaining inflammation and accelerating hepatocyte turnover, thereby promoting fibrosis and cirrhosis [10,11,12,13,14].
Moreover, in CHB, the adaptive immune response is characterized by a dysfunctional and exhausted T-cell response marked by the expression of multiple inhibitory receptors, including programmed cell death-1 (PD-1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), and T-cell immunoreceptor with Ig and ITIM domains (TIGIT). This induces an immunosuppressive microenvironment leading to persistent inflammation and ultimately contributes to tumorigenesis [8,15,16,17,18]. Characterization of the immune response and complex interactions within the tumor microenvironment is essential for understanding the mechanisms of disease progression and for the future development of new targeted therapies. This review aims to explore the role of the adaptive immune response in HBV-associated HCC and to discuss the mechanisms involved in the immunosuppressive tumor microenvironment.

2. Materials and Methods

2.1. Search Strategy

This systematic review was conducted and reported according to the Preferred Reporting Items for Systematic Reviews (PRISMA) 2020 statement [19]. The PRISMA 2020 replaces the PRISMA 2009 guideline with an expanded 27-item checklist. The study was registered at the international prospective register of systematic reviews (PROSPERO) as recommended by the Cochrane Collaboration (registration ID: CRD42023493591). A search strategy was developed and the PubMed/Medline, Cochrane Library, and Embase databases were systematically searched on 1 December 2023 for original studies investigating T-cell activity in patients with HBV-related HCC. We used the following search string: hepatitis B virus OR HBV AND hepatocellular carcinoma OR carcinoma OR cancer OR tumor OR HCC AND T cell OR CD4 OR CD8 OR Treg OR regulatory T cell.
We also manually searched reference lists of relevant articles for additional studies. Assessment of abstracts and inclusion of studies was performed independently by two reviewers (M.B. and A.S.M). Any differences were settled through discussion.

2.2. Eligibility Criteria

All study types were eligible for inclusion except for reviews and meta-analyses. It was not obligatory for the studies to include control groups.
Inclusion criteria were as follows:
  • Age of study population ≥ 18 years;
  • Studies in English language;
  • Only studies including humans;
  • Studies including patients with HBV-associated HCC;
Exclusion criteria were as follows:
  • Pediatric patients < 18 years of age;
  • Studies only performed on animals;
  • Reviews or meta-analyses;
  • Studies on individuals with HBV infection without a group of individuals with HBV-related HCC;
  • Studies including CHB patients coinfected with other hepatitis viruses and/or human immunodeficiency virus (HIV).

2.3. Comparison Groups

We divided comparison groups into three groups: (1) a group of CHB patients, which were defined as chronic HBV infection with or without liver cirrhosis, but without HCC; (2) a group of patients with non-HBV HCC involving patients with HCC with HCV infection and/or non-viral etiologies; (3) the healthy comparison group was defined as patients without hepatitis infection, HCC, alcohol abuse, or other liver diseases.

3. Results

Our search strategy resulted in 1411 studies of which 52 were included in this systematic review, collectively involving a total of 6931 participants. The study participants were categorized into four groups of patients as follows: (1) HBV-associated HCC (n = 3660), (2) CHB without HCC (n = 740), (3) non-HBV HCC (n = 1627), and (4) healthy comparison (n = 650). Among the included studies, 47 conducted a comparative analysis. Studies were clinical translational studies, with 36 studies focusing on local biomarkers in formalin-fixed paraffin-embedded (FFPE) or fresh–frozen tissue samples, and 38 studies investigating systemic biomarkers in peripheral blood mononuclear cells (PBMCs). Furthermore, 21 studies conducted combined analysis of local and systemic biomarkers (Table 1). The study selection process is shown in Figure 1.

3.1. Patient Characteristics

The included patients were chronically infected with HBV, defined as persistence of HBsAg in serum > 6 months. In 22 studies, a control group of CHB patients (n = 740) was included, 18 studies included a group of patients with non-HBV HCC (n = 1627), and 26 studies included a healthy comparison group without HBV infection. The study populations were dominated by male patients, and several studies also included subgroups based on different clinical stages of CHB and/or HCC.

3.1.1. HBV-Specific CD8+ T Cells

A total of 36 studies analyzed HBV-specific CD8+T cells in patients with HBV-associated HCC, of which 24 studies included circulating CD8+ T cells in PBMCs, and 26 studies analyzed infiltrating CD8+ T cells in liver tissue samples.

3.1.2. Phenotypes of Circulating CD8+ T Cells in Patients with HBV-Related HCC

Total levels of global circulating CD8+ T cells were significantly higher in patients with HBV-associated HCC compared with healthy comparisons [41,54], and levels of terminally differentiated effector CD8+ T cells were higher in HBV-associated HCC [21] with the majority being effector CD8+ T cells and memory CD8+ T cells [21,34]. The HBV-specific CD8+ T cells were characterized by exhausted phenotypes with high expression levels of inhibitory markers. In contrast to the comparison group, patients with HBV-associated HCC had markedly higher levels of exhaustion markers, including PD-1 [33,38,61,65], TIGIT [21,38,39], and CTLA-4 expression [39].
In most of the studies, circulating HBV-specific CD8+ T-cell levels were similar between CHB patients and patients with HBV-associated HCC [21,32,41,59,61], whereas one study showed lower levels in HBV-associated HCC [50]. Interestingly, HBV-specific CD8+ T cells showed stronger cytotoxicity in CHB patients [32] and a more exhausted functional phenotype in patients with HBV-associated HCC [67]. Additionally, increased levels of HLA-DR were seen in HBV-associated HCC which indicates highly activated CD8+ T cells [67].
Studies reported significantly higher expression of exhaustion markers, such as PD-1 [38,59,61,65,67], TIGIT [38], and T-cell immunoglobin and mucin domain 3 (TIM-3) [59] in patients with HBV-associated HCC compared with CHB. A study reported that PD-1 expression especially was seen in central memory and effector memory CD8+ T cells [38]. Furthermore, a study found elevated expression of lymphocyte activation gene 3 (LAG-3), CTLA-4, and TIM-3 on PD-1+CD8+ T cells in patients with HBV-associated HCC compared with CHB [59]. Expression of INF-γ decreased in HBV-associated HCC [59] and HBV-specific CD8+ T cells showed impaired capacity for TNF-α secretion [67] in comparison with CHB patients.
Total levels of global circulating CD8+ T cells were elevated in HBV-associated HCC compared with non-HBV HCC [30], and a study reported lower levels of INF-γ, TNF-α, and granzyme B secretion in patients with HBV-associated HCC compared with non-HBV HCC [37], indicating impaired effector function of CD8+ T cells.

3.1.3. Prognostic Value of PD-1 Expression on Circulating CD8+ T Cells

Expression of PD-1 on CD8+ T cells was highly correlated with disease progression [38,61,65] and the association was shown to be stronger in CD8+ T cells than in CD4+ T cells [38]. In addition, high levels of PD-1 were related to higher recurrence rates of HCC [38]. Finally, PD-1 levels were positively correlated with HBV DNA, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) [61].

3.1.4. Tumor-Infiltrating HBV-Specific CD8+ T-Cell Phenotypes in HBV-Associated HCC

The infiltration of CD8+ T cells was markedly lower in the tumor tissue of HBV-associated HCC compared with healthy comparisons or paired non-tumor tissue [40,69,71]. Furthermore, one study reported significantly elevated levels of PD-1 expression in HBV-associated HCC compared with the normal comparison group [65], and a second study reported increased expression of LAG-3 followed by decreasing levels of INF-γ, which reduces the effector function of CD8+ T cells [63]. In addition, a study demonstrated low levels of granzyme A, granzyme B, and perforin in patients with HBV-associated HCC compared with healthy comparisons [69].
The CD8+ T-cell infiltration in HBV-associated HCC was compared within liver tissue from CHB patients in two studies, which reported higher levels of PD-1 on CD8+ T cells in HBV-associated HCC in one study [65], while no difference was found in the second study [29]. Additionally, high expression of TIGIT and CTLA-4 was comparable in the two groups [29].
When comparing tumor tissue from HBV-associated HCC with non-HBV HCC, studies reported higher levels of CD8+ T cells in HBV-associated HCC [47,71]. Tumor-infiltrating CD8+ T cells in HBV-associated HCC were characterized by marked clonal expansion and an immunosuppressive and exhausted tumor microenvironment. Studies demonstrated higher levels of PD-1 [22,24,30,37,47,71], TIGIT [22], CTLA-4 [20,37,71], TIM-3 [30,37], LAG-3 [22], and TOX [22,30] in HBV-associated HCC compared with non-HBV HCC.

3.1.5. Impaired Effector CD8+ T Cell Function in HBV-Associated HCC

The HBV-specific CD8+ T cells are crucial in controlling HBV infection and tumor-infiltrating CD8+ T cells in HBV-associated HCC were positively correlated with survival [30,31,47]. The CD8+ T cells were primarily effector cells and memory cells, and one study reported that effector cells in HBV-associated HCC were characterized by increased levels of CTLA-4, ICOS, and TOX expression [20]. In addition, PD-1 and TIM-3 levels proved to be correlated with poor prognosis [47,65], and PD-1 levels increased with severity of liver fibrosis [27]. Cytotoxic CD8+ T cells in HBV-associated HCC secreted lower levels of INF-γ, TNFα, and granzyme B, compared with non-HBV HCC [37,47]. However, a study reported that inhibition of TIM-3 and PD-1 restored CD8+ T cell function with increasing INF-γ and TNF-α levels, indicating a potential therapeutic target [42]. Finally, a study found decreasing INF-γ levels with increasing LAG-3 expression, suggesting that LAG-3 reduces the effector function of CD8+ T cells [63].

3.2. HBV-Specific CD4+ T Cells

A total of 35 studies investigated CD4+ T cells in HBV-associated HCC, of which 25 studies included circulating CD4+ T cells in PBMCs and 24 studies included infiltrating CD4+ T cells in liver tissue samples.

3.2.1. Circulating CD4+ T Cells in HBV-HCC

A study reported decreased levels of total global circulating CD4+ T cells in patients with HBV-associated HCC (n = 715) compared with healthy controls (n = 100) [54]. In addition, a study showed lower levels of naïve CD4+ T cells compared with a healthy comparison group [21], whereas another study reported increased levels of cytotoxic CD4+ T cells, characterized by granzyme A and B expression [53]. Furthermore, studies found increased levels of PD-1 [33,61] and TIM-3 [21] expression compared with healthy comparisons.
In comparison with CHB patients, total levels of global circulating CD4+ T cells [50] and naïve CD4+ T cells [21] were lower in HBV-associated HCC. The studies found varying results regarding exhaustion markers on circulating CD4+ T cells in patients with CHB compared with HBV-associated HCC. A study found increased expression of TIGIT and TIM-3 in HBV-associated HCC [21]. Expression of PD-1 levels was analyzed in one study and reported similar PD-1 expression between the two groups [61]. It was demonstrated that PD-1 expression on circulating CD4+ T cells was correlated with HBV DNA and ALT levels [61], whereas a second study only found a correlation between PD-1 expression and HBV DNA in CHB patients, but not in patients with HBV-associated HCC [33]. In addition, a study showed elevated HLA-DR in HBV-associated HCC compared with CHB indicating a higher T-cell activation [50].
Only a single study compared total levels of circulating CD4+ T cells in patients with HBV-associated HCC (n = 22) and non-HBV HCC (n = 17) and found no difference [60]. A second study demonstrated that levels of CD4+ memory T cells were an independent predictor for survival in patients with HBV-associated HCC [23].

3.2.2. Tumor-Infiltrating CD4+ T Cells

Levels of infiltrating CD4 T cells in HBV-associated HCC compared with healthy controls were investigated in two studies finding elevated total CD4+ T cells [68], but decreased cytotoxic CD4+ T cells in HBV-associated HCC [53].
The total levels of infiltrating CD4+ T cells in CHB were not reported across the included studies, and a single study found that total CD4+ T-cell levels were higher in HBV-associated HCC compared with non-HBV HCC [71]. Furthermore, exhausted states of CD4+ T cells were markedly higher in HBV-associated HCC than non-HBV HCC, with increased CTLA-4 and PD-1 expression levels [20,71].
A study including 1328 patients showed that CD4+ T-cell subsets were more enriched in HBV-associated HCC tissue than CD8+ T cells [40]. Among CD4+ T-cell populations, Tregs were the predominant subset and highly enriched in the tumor microenvironment of HBV-associated HCC [25,71], with the highest accessibility at the forkhead box P3 (FOXP3) loci [25].

3.2.3. Circulating Regulatory T Cells in HBV-Associated HCC

Circulating Tregs were primarily characterized as CD4+CD25+ T cells and studies found significantly elevated levels in HBV-associated HCC compared with healthy controls [20,36,41,48,58,66,69,70]. In addition, studies found markedly elevated expression levels of FOXP3 [20,41,66,69,70], PD-1 [21], and TIGIT [21].
Levels of Tregs in HBV-associated HCC compared with CHB showed varying results, as two studies found no difference [41,66] and one study found elevated levels in HBV-associated HCC [69]. In comparison with non-HBV HCC, levels of circulating Tregs were significantly increased in HBV-associated HCC [25,37,60] and expression levels of FOXP3 [25,37], CTLA-4, LAG-3, and PD-1 [37] increased in patients with HBV-associated HCC.

3.2.4. Tumor-Infiltrating Regulatory T Cells in HBV-Associated HCC

Levels of infiltrating Tregs in HBV-associated HCC compared with control tissue were analyzed in three studies and in all cases markedly increased levels of Tregs were demonstrated in HBV-related HCC tumor tissue [36,69,70]. Increased levels of Tregs were also found in HBV-associated HCC in comparison with liver tissue from patients with CHB without HCC [60,69].
In three studies, higher levels of Tregs were documented in HBV-associated HCC compared with HBV-non-associated HCC [26,37,60]. Analyses of Tregs in the HBV-associated HCC tumor microenvironment showed a more immunosuppressive and effective status [20], with increased expression levels of FOXP3 [20,26,37], PD-1 [20,25,37], CTLA-4 [20,25], ICOS [20], and LAG-3 [37] compared with non-HBV HCC.

3.2.5. Role of Regulatory T Cells HBV-Associated HCC

It was demonstrated that levels of circulating Tregs increased with advancing stages of HBV-associated HCC, thus being a predicter for prognosis [69]. A study isolated circulating Tregs from patients with HBV-associated HCC to analyze the function of this cell subset and found that Tregs significantly inhibit proliferation [66]. Additionally, a study showed that Tregs in HBV-associated HCC were more suppressive than in non-HBV HCC, through high IL-10 and TGF-β secretion [60]. Moreover, Tregs suppressed CD8+ T-cell proliferation through the inhibition of INF-γ, TNF-α, and reduced HLA-DR [69]. Another interesting finding was that the CD4+ T and CD8+ T-cell interactions were increasingly replaced with rising Treg level in HBV-associated HCC. Spatial proteomics demonstrated increasing cell–cell connections between CD4+ T cells and Tregs in the tumor microenvironment, and distances between these cells became significantly shorter in the mid tumor regions compared to non-tumor regions [24]. Furthermore, spatial proteomics visualized that PD-1+CD8+ T cells also connected with Tregs [24]. Finally, a study demonstrated that the Treg/CD8+ ratio was strongly associated with prognosis [20].

3.2.6. Role of T Helper 17 Cells

Circulating T helper 17 (Th17) cells were analyzed in three studies. One study found higher levels of circulating Th17 cells in HBV-associated HCC compared with CHB [62]; however, a second study found no difference among the two groups [28]. Additionally, a third study found the Th17/Treg ratio to be an independent risk factor of HCC development [52], and that levels of Th17 were associated with degree of liver damage. An interesting finding was that levels of PD-1 increased in patients who progressed from CHB to HCC [62].
Infiltrating Th17 cells were investigated in two studies. The first study showed lower levels of infiltrating Th17 cells in HBV-associated HCC compared with a healthy comparisons group and CHB patients [28]. The second study found that infiltrating Th17 secreted increasing levels of IL-17 along with severity of liver disease and that IL-17 promoted liver fibrosis and tumorigenesis [36].

3.2.7. Role of Follicular Helper T Cells

Studies have found that levels of follicular helper T cells in HBV-associated HCC were similar compared with the CHB group and healthy comparison group [44,55,57]. However, higher expression levels of PD-1 have been shown [55] along with decreased levels of IL-21 secretion, suggesting poorer viability [57].

4. Discussion

This systematic review revealed that HBV-associated HCC is marked by an immunosuppressive tumor microenvironment, characterized by dysfunctional and exhausted T-cell populations. Circulating CD8+ T-cell levels were elevated in HBV-associated HCC patients compared with non-HBV HCC and healthy comparisons [30,41,54], mainly composed of terminally differentiated effector and memory CD8+ T cells [21,34]. Conversely, infiltrating CD8+ T-cell levels in the tumor microenvironment were lower in patients with HBV-associated HCC compared with liver tissue from non-HBV HCC patients and healthy comparisons [40,47,69,71]. Tumor-infiltrating CD8+ T cells in HBV-associated HCC were highly activated but dysfunctional, showing impaired cytotoxicity and exhaustion, with significantly increased expression of immune checkpoint molecules, such as PD-1, CTLA-4, LAG-3, and TIGIT. On the contrary, tumor-infiltrating HBV-specific CD4+ T cells were significantly elevated compared with all control groups (non-HBV HCC, CHB, and HC) [40,53,68,71], with Tregs being the predominant CD4+ T-cell subset [25,71]. The CD4+ T-cell populations, including Tregs, also exhibited high expression levels of immune checkpoint molecules, predominantly PD-1 and CTLA-4. Additionally, Tregs displayed high expression of the transcription factor FOXP3 [25], which plays a suppressive role in the immune system [72]. Interestingly, levels of circulating and infiltrating HBV-specific CD8+ T cells were largely similar in HBV-associated HCC compared with CHB, whereas infiltrating CD4+ T cells and Tregs were significantly elevated in HBV-associated HCC, indicating that CD4+ T-cell populations may be a dominant factor in the progression of CHB to HBV-associated HCC. A limited number of studies have investigated the role of T helper 17 cells and follicular helper T cells and found that T helper 17 cells may be associated with poor prognosis. However, no conclusions should be made based on these findings and further studies are needed to elucidate the role of these cell subsets.
The cytotoxic effects of CD8+ T cells are crucial in controlling viral HBV infection and cancer [16]. Decreasing levels of CD8+ T cells in CHB, leading towards dysfunction and exhaustion, are fundamental mechanisms in disease progression. Hepatitis B surface antigen (HBsAg) and hepatitis B core-related antigen (HBcrAg) are considered crucial factors in HBV specific immune responses and thought to be a hallmark in T-cell dysfunction. However, recent evidence has shown that the phenotypical and functional profiles of CD8+ T cells were unaffected by HBsAg levels [73]. Conversely lower HBcrAg levels correlated with higher HBV-specific CD4+ T-cell responses, indicating that HBcrAg may be a more significant viral biomarker [73]. These findings are important for the development of novel immune-based therapies. The increased enrichment of CD4+ T cells and their differentiation into Tregs have dual effects. Firstly, CD4+ T cells activate CD8+ T cells to a lesser extent. Secondly, Tregs inhibit the proliferation of CD8+ T cells. Furthermore, the expression of immune checkpoint molecules contributes to immunosuppression in the tumor microenvironment.
This systematic review highlights PD-1 expression’s crucial role in the development of HCC. Elevated PD-1 expression in circulating CD8+ and CD4+ T cells strongly correlates with disease progression and higher recurrence rates in HBV-HCC patients. PD-1 levels are notably high on exhausted CD8+ T cells in the tumor microenvironment, indicating immune dysfunction and tumor evasion mechanisms (Figure 2). Moreover, PD-1 expression correlates positively with HBV DNA levels, ALT, and AST, which is an interesting finding as HBV DNA integration in infected hepatocytes is a major driver in HCC development [74]. A study demonstrated that the blocking of TIM-3 and PD-1 restored CD8+ T-cell effector functions [42], suggesting their potential as therapeutic targets for immune checkpoint blockade strategies. The PD-1/PD-L1 axis leads to negative feedback of the immune response by blocking the T-cell receptor. Tumor cells expresses PD-L1 to avoid immunosurveillance. Immune escape is a fundamental Hallmark of cancer [75,76] and the development of PD-1 inhibitors has gained a fundamental role in the treatment of several cancers. However, the response rate to the checkpoint inhibitor nivolumab (PD-1 monoclonal antibody) for HCC is 15–20% [77], and a randomized controlled multicenter trial found no difference in treatment with nivolumab compared with sorafenib (recommended first-line chemotherapeutic drug for HCC) [78]. Monotherapy with PD-1 inhibition has thus far demonstrated questionable efficacy [79], and should be used in a personalized approach. Studies have demonstrated a higher response rate of PD-L1 inhibition in patients with low levels of HBsAg and HBcrAg [73].
The tumor microenvironment in HCC is multifactorial and complex involving HBV DNA integration, chronic inflammation, and a dysfunctional adaptive immune response, and a multi-target treatment strategy may be a potential approach in the future. Efficient preventive and curative treatment for HBV-associated HCC is lacking, and the mechanisms driving the transition towards exhausted T lymphocytes and carcinogenesis is not fully elucidated. Another important aspect is the interplay between the innate and adaptive immune responses, which is also an important driver in the persistent inflammation in CHB. Tumor-promoting inflammation is a fundamental hallmark of cancer [75,76,80,81] and it may be of great importance to characterize the inflammatory phenotypes in CHB and HBV-associated HCC to fully understand the mechanisms, and in a future perspective, identify predictive biomarkers and ultimately develop efficient treatment strategies.

Strengths and Limitations

This systematic review included studies with general similarity regarding eligibility criteria for included study participants. Conditions other than HBV and CHB that may affect the liver and/or immune system led to exclusion across all studies, which is a strength in this review. Additionally, this review included a relatively high number of studies which collectively included 6931 participants and translational analysis was performed on biological samples with similar preparation, including PBMCs, FFPE, or fresh–frozen liver samples. Although all studies used validated platforms and demonstrated consistent results overall, a limitation of this review is the variety of methods employed to analyze T-cell activity. Differences in proteomic panels, gene expression platforms, and proliferation assays could contribute to certain discrepancies observed.

5. Conclusions

This systematic review highlights the immunosuppressive tumor microenvironment in HBV-associated HCC, characterized by dysfunctional and exhausted HBV-specific T cells and increased infiltration of HBV-specific CD4+ T cells, particularly Tregs. The elevated expression of checkpoint inhibitors, notably PD-1, correlates with disease progression and recurrence, suggesting its potential as a prognostic marker and therapeutic target. However, monotherapy with PD-1 inhibitors has demonstrated limited efficacy. Moving forward, a characterization of the complex interplay between the innate and adaptive immune responses holds promise for the identification of predictive biomarkers and development of new treatment strategies in HBV-associated HCC.

Author Contributions

Conceptualization, M.B. and N.W.; methodology, M.B.; Screening and inclusion, M.B. and A.-S.M.; formal analysis, M.B.; writing—original draft preparation, M.B.; writing—review and editing, M.B., A.-S.M., Á.D.A. and N.W.; visualization, Á.D.A.; supervision, N.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brown, Z.J.; Tsilimigras, D.I.; Ruff, S.M.; Mohseni, A.; Kamel, I.R.; Cloyd, J.M.; Pawlik, T.M. Management of Hepatocellular Carcinoma: A Review. JAMA Surg. 2023, 158, 410–420. [Google Scholar] [CrossRef]
  2. McGlynn, K.A.; Petrick, J.L.; El-Serag, H.B. Epidemiology of Hepatocellular Carcinoma. Hepatology 2021, 73 (Suppl. S1), 4–13. [Google Scholar] [CrossRef]
  3. Graber-Stiehl, I. The silent epidemic killing more people than HIV, malaria or TB. Nature 2018, 564, 24–26. [Google Scholar] [CrossRef]
  4. World Health Statistics. 2021. Available online: http://www.who.int/data/gho/publications/world-health-statistics (accessed on 1 December 2023).
  5. Chen, Y.; Tian, Z. HBV-Induced Immune Imbalance in the Development of HCC. Front. Immunol. 2019, 10, 2048. [Google Scholar] [CrossRef]
  6. Buschow, S.I.; Jansen, D.T.S.L. CD4+ T Cells in Chronic Hepatitis B and T Cell-Directed Immunotherapy. Cells 2021, 10, 1114. [Google Scholar] [CrossRef]
  7. Jung, M.K.; Shin, E.C. Regulatory T Cells in Hepatitis B and C Virus Infections. Immune Netw. 2016, 16, 330–336. [Google Scholar] [CrossRef]
  8. Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 116. [Google Scholar] [CrossRef]
  9. Cho, H.J.; Cheong, J.Y. Role of Immune Cells in Patients with Hepatitis B Virus-Related Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 8011. [Google Scholar] [CrossRef]
  10. Phillips, S.; Chokshi, S.; Riva, A.; Evans, A.; Williams, R.; Naoumov, N.V. CD8(+) T cell control of hepatitis B virus replication: Direct comparison between cytolytic and noncytolytic functions. J. Immunol. 2010, 184, 287–295. [Google Scholar] [CrossRef]
  11. Xie, Y.; Hepatitis, B. Virus-Associated Hepatocellular Carcinoma. Adv. Exp. Med. Biol. 2017, 1018, 11–21. [Google Scholar]
  12. Ringehan, M.; McKeating, J.A.; Protzer, U. Viral hepatitis and liver cancer. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2017, 372, 20160274. [Google Scholar] [CrossRef]
  13. Nakamoto, Y.; Guidotti, L.G.; Kuhlen, C.V.; Fowler, P.; Chisari, F.V. Immune pathogenesis of hepatocellular carcinoma. J. Exp. Med. 1998, 188, 341–350. [Google Scholar] [CrossRef]
  14. Hao, X.; Chen, Y.; Bai, L.; Wei, H.; Sun, R.; Tian, Z. HBsAg-specific CD8+ T cells as an indispensable trigger to induce murine hepatocellular carcinoma. Cell Mol. Immunol. 2021, 18, 128–137. [Google Scholar] [CrossRef]
  15. Khanam, A.; Chua, J.V.; Kottilil, S. Immunopathology of Chronic Hepatitis B Infection: Role of Innate and Adaptive Immune Response in Disease Progression. Int. J. Mol. Sci. 2021, 22, 5497. [Google Scholar] [CrossRef]
  16. Iannacone, M.; Guidotti, L.G. Immunobiology and pathogenesis of hepatitis B virus infection. Nat. Rev. Immunol. 2022, 22, 19–32. [Google Scholar] [CrossRef]
  17. Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef]
  18. Apol, Á.D.; Winckelmann, A.A.; Duus, R.B.; Bukh, J.; Weis, N. The Role of CTLA-4 in T Cell Exhaustion in Chronic Hepatitis B Virus Infection. Viruses 2023, 15, 1141. [Google Scholar] [CrossRef]
  19. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.B.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  20. You, M.; Gao, Y.; Fu, J.; Xie, R.; Zhu, Z.; Hong, Z.; Meng, L.; Du, S.; Liu, J.; Wang, F.-S.; et al. Epigenetic regulation of HBV-specific tumor-infiltrating T cells in HBV-related HCC. Hepatology 2023, 78, 943–958. [Google Scholar] [CrossRef]
  21. Sun, R.; Li, J.; Lin, X.; Yang, Y.; Liu, B.; Lan, T.; Xiao, S.; Deng, A.; Yin, Z.; Xu, Y.; et al. Peripheral immune characteristics of hepatitis B virus-related hepatocellular carcinoma. Front. Immunol. 2023, 14, 1079495. [Google Scholar] [CrossRef]
  22. Liu, L.; Liu, J.; Li, P.; Luo, J.; Qin, R.; Peng, Q.; Li, B.; Wei, X.; Wang, T.; Shi, H.; et al. Single-cell analysis reveals HBV-specific PD-1+CD8+ TRM cells in tumor borders are associated with HBV-related hepatic damage and fibrosis in HCC patients. J. Exp. Clin. Cancer Res. 2023, 42, 152. [Google Scholar] [CrossRef]
  23. Chang, Y.-S.; Tu, S.-J.; Chen, H.-D.; Hsu, M.-H.; Chen, Y.-C.; Chao, D.-S.; Chung, C.-C.; Chou, Y.-P.; Chang, C.-M.; Lee, Y.-T.; et al. Integrated genomic analyses of hepatocellular carcinoma. Hepatol. Int. 2023, 17, 97–111. [Google Scholar] [CrossRef]
  24. Li, M.; Wang, L.; Cong, L.; Wong, C.C.; Zhang, X.; Chen, H.; Zeng, T.; Li, B.; Jia, X.; Huo, J.; et al. Spatial proteomics of immune microenvironment in nonalcoholic steatohepatitis-associated hepatocellular carcinoma. Hepatology 2023, 79, 560–574. [Google Scholar] [CrossRef]
  25. Gao, Y.; You, M.; Fu, J.; Tian, M.; Zhong, X.; Du, C.; Zhu, Z.; Liu, J.; Markowitz, G.J.; Wang, F.-S.; et al. Intratumoral stem-like CCR4+ regulatory T cells orchestrate the immunosuppressive microenvironment in HCC associated with hepatitis B. J. Hepatol. 2022, 76, 148–159. [Google Scholar] [CrossRef]
  26. Lu, L.-C.; Deantonio, C.; Palu, C.C.; Lee, Y.-H.; Mitchell, L.S.; Cowan, M.; Corser, M.; Sherry, L.; Cheng, A.-L.; Quaratino, S.; et al. ICOS-Positive Regulatory T Cells in Hepatocellular Carcinoma: The Perspective from Digital Pathology Analysis. Oncology 2022, 100, 419–428. [Google Scholar] [CrossRef]
  27. Li, Y.; Zhao, J.-F.; Zhang, J.; Zhan, G.-H.; Li, Y.-K.; Huang, J.-T.; Huang, X.; Xiang, B.-D. Inflammation and Fibrosis in Patients with Non-Cirrhotic Hepatitis B Virus-Associated Hepatocellular Carcinoma: Impact on Prognosis after Hepatectomy and Mechanisms Involved. Curr. Oncol. 2022, 30, 196–218. [Google Scholar] [CrossRef]
  28. Zhang, M.; Zhao, H.; Gao, H. Interleukin-24 Limits Tumor-Infiltrating T Helper 17 Cell Response in Patients with Hepatitis B Virus-Related Hepatocellular Carcinoma. Viral Immunol. 2022, 35, 212–222. [Google Scholar] [CrossRef]
  29. Ho, D.W.-H.; Tsui, Y.-M.; Chan, L.-K.; Sze, K.M.-F.; Zhang, X.; Cheu, J.W.-S.; Chiu, Y.-T.; Lee, J.M.-F.; Chan, A.C.-Y.; Cheung, E.T.-Y.; et al. Single-cell RNA sequencing shows the immunosuppressive landscape and tumor heterogeneity of HBV-associated hepatocellular carcinoma. Nat. Commun. 2021, 12, 3684. [Google Scholar] [CrossRef]
  30. Cheng, Y.; Gunasegaran, B.; Singh, H.D.; Dutertre, C.-A.; Loh, C.Y.; Lim, J.Q.; Crawford, J.C.; Lee, H.K.; Zhang, X.; Lee, B.; et al. Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma. Immunity 2021, 54, 1825–1840.e7. [Google Scholar] [CrossRef]
  31. Xin, H.; Liang, D.; Zhang, M.; Ren, D.; Chen, H.; Zhang, H.; Li, S.; Ding, G.; Zhang, C.; Ding, Z.; et al. The CD68+ macrophages to CD8+ T-cell ratio is associated with clinical outcomes in hepatitis B virus (HBV)-related hepatocellular carcinoma. HPB 2021, 23, 1061–1071. [Google Scholar] [CrossRef]
  32. Zhang, Q.; Yang, L.; Liu, S.; Zhang, M.; Jin, Z. Interleukin-35 Suppresses Interleukin-9-Secreting CD4+ T Cell Activity in Patients With Hepatitis B-Related Hepatocellular Carcinoma. Front. Immunol. 2021, 12, 645835. [Google Scholar] [CrossRef]
  33. Liu, W.; Zheng, X.; Wang, J.; He, Q.; Li, J.; Zhang, Z.; Liu, H. MicroRNA-138 Regulates T-Cell Function by Targeting PD-1 in Patients with Hepatitis B Virus-Related Liver Diseases. Lab. Med. 2021, 52, 439–451. [Google Scholar] [CrossRef]
  34. Zhao, L.; Jin, Y.; Yang, C.; Li, C. HBV-specific CD8 T cells present higher TNF-α expression but lower cytotoxicity in hepatocellular carcinoma. Clin. Exp. Immunol. 2020, 201, 289–296. [Google Scholar] [CrossRef]
  35. Li, Z.Q.; Wang, H.Y.; Zeng, Q.L.; Yan, J.Y.; Hu, Y.S.; Li, H.; Yu, Z.J. p65/miR-23a/CCL22 axis regulated regulatory T cells recruitment in hepatitis B virus positive hepatocellular carcinoma. Cancer Med. 2020, 9, 711–723. [Google Scholar] [CrossRef]
  36. Zhang, H.; Yan, X.; Yang, C.; Zhan, Q.; Fu, Y.; Luo, H.; Luo, H. Intrahepatic T helper 17 cells recruited by hepatitis B virus X antigen-activated hepatic stellate cells exacerbate the progression of chronic hepatitis B virus infection. J. Viral Hepat. 2020, 27, 1138–1149. [Google Scholar] [CrossRef]
  37. Lim, C.J.; Lee, Y.H.; Pan, L.; Lai, L.; Chua, C.; Wasser, M.; Lim, T.K.H.; Yeong, J.; Toh, H.C.; Lee, S.Y.; et al. Multidimensional analyses reveal distinct immune microenvironment in hepatitis B virus-related hepatocellular carcinoma. Gut 2019, 68, 916–927. [Google Scholar] [CrossRef]
  38. Liu, X.; Li, M.; Wang, X.; Dang, Z.; Jiang, Y.; Wang, X.; Kong, Y.; Yang, Z. PD-1+ TIGIT+ CD8+ T cells are associated with pathogenesis and progression of patients with hepatitis B virus-related hepatocellular carcinoma. Cancer Immunol. Immunother. 2019, 68, 2041–2054. [Google Scholar] [CrossRef]
  39. Wang, Y.-G.; Zheng, D.-H.; Shi, M.; Xu, X.-M. T cell dysfunction in chronic hepatitis B infection and liver cancer: Evidence from transcriptome analysis. J. Med. Genet. 2019, 56, 22–28. [Google Scholar] [CrossRef]
  40. Hsiao, Y.-W.; Chiu, L.-T.; Chen, C.-H.; Shih, W.-L.; Lu, T.-P. Tumor-Infiltrating Leukocyte Composition and Prognostic Power in Hepatitis B- and Hepatitis C-Related Hepatocellular Carcinomas. Genes 2019, 10, 630. [Google Scholar] [CrossRef]
  41. Shen, X.H.; Xu, P.; Yu, X.; Song, H.F.; Chen, H.; Zhang, X.G.; Wu, M.; Wang, X. Discrepant Clinical Significance of CD28+CD8- and CD4+CD25high Regulatory T Cells During the Progression of Hepatitis B Virus Infection. Viral Immunol. 2018, 31, 548–558. [Google Scholar] [CrossRef]
  42. Liu, F.; Zeng, G.; Zhou, S.; He, X.; Sun, N.; Zhu, X.; Hu, A. Blocking Tim-3 or/and PD-1 reverses dysfunction of tumor-infiltrating lymphocytes in HBV-related hepatocellular carcinoma. Bull. Cancer 2018, 105, 493–501. [Google Scholar] [CrossRef] [PubMed]
  43. Ou, X.; Guan, J.; Chen, J.; Ying, J.; Liu, X.; Tian, P.; Liu, J.; Nie, L.; Zhao, Y.; Yu, G. LAP+CD4+ T cells are elevated among the peripheral blood mononuclear cells and tumor tissue of patients with hepatocellular carcinoma. Exp. Ther. Med. 2018, 16, 788–796. [Google Scholar] [CrossRef]
  44. Wu, X.; Su, Z.; Cai, B.; Yan, L.; Li, Y.; Feng, W.; Wang, L. Increased Circulating Follicular Regulatory T-Like Cells May Play a Critical Role in Chronic Hepatitis B Virus Infection and Disease Progression. Viral Immunol. 2018, 31, 379–388. [Google Scholar] [CrossRef] [PubMed]
  45. Li, Z.; Chen, G.; Cai, Z.; Dong, X.; Qiu, L.; Xu, H.; Zeng, Y.; Liu, X.; Liu, J. Genomic and transcriptional Profiling of tumor infiltrated CD8+ T cells revealed functional heterogeneity of antitumor immunity in hepatocellular carcinoma. Oncoimmunology 2018, 8, e1538436. [Google Scholar] [CrossRef] [PubMed]
  46. Meng, F.; Zhen, S.; Song, B. HBV-specific CD4+ cytotoxic T cells in hepatocellular carcinoma are less cytolytic toward tumor cells and suppress CD8+ T cell-mediated antitumor immunity. APMIS 2017, 25, 743–751. [Google Scholar] [CrossRef]
  47. Huang, C.-Y.; Wang, Y.; Luo, G.-Y.; Han, F.; Li, Y.-Q.; Zhou, Z.-G.; Xu, G.-L. Relationship Between PD-L1 Expression and CD8+ T-cell Immune Responses in Hepatocellular Carcinoma. J. Immunother. 2017, 40, 323–333. [Google Scholar] [CrossRef] [PubMed]
  48. Lan, Y.-T.; Fan, X.-P.; Fan, Y.-C.; Zhao, J.; Wang, K. Change in the Treg/Th17 cell imbalance in hepatocellular carcinoma patients and its clinical value. Medicine 2017, 96, e7704. [Google Scholar] [CrossRef] [PubMed]
  49. Jiang, H.; Li, L.; Hang, J.; Sun, Z.; Rong, Y.; Jin, Y. CXCR5+ CD8+ T Cells Indirectly Offer B Cell Help and Are Inversely Correlated with Viral Load in Chronic Hepatitis B Infection. DNA Cell Biol. 2017, 36, 321–327. [Google Scholar] [CrossRef] [PubMed]
  50. Liu, X.; He, L.; Han, J.; Wang, L.; Li, M.; Jiang, Y.; Wang, X.; Yang, Z. Association of neutrophil-lymphocyte ratio and T lymphocytes with the pathogenesis and progression of HBV-associated primary liver cancer. PLoS ONE 2017, 12, e0170605. [Google Scholar] [CrossRef]
  51. Li, X.; Su, Y.; Hua, X.; Xie, C.; Liu, J.; Huang, Y.; Zhou, L.; Zhang, M.; Li, X.; Gao, Z. Levels of hepatic Th17 cells and regulatory T cells upregulated by hepatic stellate cells in advanced HBV-related liver fibrosis. J. Transl. Med. 2017, 15, 75. [Google Scholar] [CrossRef]
  52. Li, K.; Liu, H.; Guo, T. Th17/Treg imbalance is an indicator of liver cirrhosis process and a risk factor for HCC occurrence in HBV patients. Clin. Res. Hepatol. Gastroenterol. 2017, 41, 399–407. [Google Scholar] [CrossRef] [PubMed]
  53. Xue, H.; Lin, F.; Tan, H.; Zhu, Z.-Q.; Zhang, Z.-Y.; Zhao, L. Overrepresentation of IL-10-Expressing B Cells Suppresses Cytotoxic CD4+ T Cell Activity in HBV-Induced Hepatocellular Carcinoma. PLoS ONE 2016, 11, e0154815. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, H.-Z.; Deng, W.; Li, J.-L.; Tang, Y.-M.; Zhang, L.-T.; Cui, Y.; Liang, X.-Q. Peripheral blood lymphocyte subset levels differ in patients with hepatocellular carcinoma. Oncotarget 2016, 7, 77558–77564. [Google Scholar] [CrossRef] [PubMed]
  55. Follicular Helper T Cell Exhaustion Induced by PD-L1 Expression in Hepatocellular Carcinoma Results in Impaired Cytokine Expression and B Cell Help, and Is Associated with Advanced Tumor Stages—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/27508013/ (accessed on 12 February 2024).
  56. Jia, Y.; Zeng, Z.; Li, Y.; Li, Z.; Jin, L.; Zhang, Z.; Wang, L.; Wang, F.-S. Impaired function of CD4+ T follicular helper (Tfh) cells associated with hepatocellular carcinoma progression. PLoS ONE 2015, 10, e0117458. [Google Scholar] [CrossRef] [PubMed]
  57. Duan, Z.; Gao, J.; Zhang, L.; Liang, H.; Huang, X.; Xu, Q.; Zhang, Y.; Shen, T.; Lu, F. Phenotype and function of CXCR5+CD45RA-CD4+ T cells were altered in HBV-related hepatocellular carcinoma and elevated serum CXCL13 predicted better prognosis. Oncotarget 2015, 6, 44239–44253. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, H.R.; Li, W.M. Treg-specific demethylated region activity in isolated regulatory t lymphocytes is a surrogate for disease severity in hepatocellular carcinoma. IUBMB Life 2015, 67, 355–360. [Google Scholar] [CrossRef] [PubMed]
  59. Dinney, C.M.; Zhao, L.-D.; Conrad, C.D.; Duker, J.M.; Karas, R.O.; Hu, Z.; Hamilton, M.A.; Gillis, T.R.; Parker, T.M.; Fan, B.; et al. Regulation of HBV-specific CD8(+) T cell-mediated inflammation is diversified in different clinical presentations of HBV infection. J. Microbiol. 2015, 53, 718–724. [Google Scholar] [CrossRef] [PubMed]
  60. Sharma, S.; Khosla, R.; David, P.; Rastogi, A.; Vyas, A.; Singh, D.; Bhardwaj, A.; Sahney, A.; Maiwall, R.; Sarin, S.K.; et al. CD4+CD25+CD127(low) Regulatory T Cells Play Predominant Anti-Tumor Suppressive Role in Hepatitis B Virus-Associated Hepatocellular Carcinoma. Front. Immunol. 2015, 6, 49. [Google Scholar] [CrossRef] [PubMed]
  61. Xu, P.; Chen, Y.-J.; Chen, H.; Zhu, X.-Y.; Song, H.-F.; Cao, L.-J.; Wang, X.-F. The expression of programmed death-1 in circulating CD4+ and CD8+ T cells during hepatitis B virus infection progression and its correlation with clinical baseline characteristics. Gut Liver 2014, 8, 186–195. [Google Scholar] [CrossRef]
  62. Chen, J.; Wang, L.; Fu, Y.; Li, Y.; Bai, Y.; Luo, L.; Liao, Y. The co-inhibitory pathway and cellular immune imbalance in the progress of HBV infection. Hepatol. Int. 2014, 8, 55–63. [Google Scholar] [CrossRef]
  63. Li, F.-J.; Zhang, Y.; Jin, G.-X.; Yao, L.; Wu, D.-Q. Expression of LAG-3 is coincident with the impaired effector function of HBV-specific CD8(+) T cell in HCC patients. Immunol. Lett. 2013, 150, 116–122. [Google Scholar] [CrossRef] [PubMed]
  64. Li, H.; Wu, K.; Tao, K.; Chen, L.; Zheng, Q.; Lu, X.; Liu, J.; Shi, L.; Liu, C.; Wang, G.; et al. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology 2012, 56, 1342–1351. [Google Scholar] [CrossRef] [PubMed]
  65. Shi, F.; Shi, M.; Zeng, Z.; Qi, R.Z.; Liu, Z.W.; Zhang, J.Y.; Yang, Y.; Tien, P.; Wang, F. PD-1 and PD-L1 upregulation promotes CD8(+) T-cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients. Int. J. Cancer 2011, 128, 887–896. [Google Scholar] [CrossRef]
  66. Zhang, H.H.; Mei, M.H.; Fei, R.; Liu, F.; Wang, J.H.; Liao, W.J.; Qin, L.L.; Wei, L.; Chen, H.S. Regulatory T cells in chronic hepatitis B patients affect the immunopathogenesis of hepatocellular carcinoma by suppressing the anti-tumour immune responses. J. Viral Hepat. 2010, 17 (Suppl. S1), 34–43. [Google Scholar] [CrossRef]
  67. Gehring, A.J.; Ho, Z.Z.; Tan, A.T.; Aung, M.O.; Lee, K.H.; Tan, K.C.; Lim, S.G.; Bertoletti, A. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology 2009, 137, 682–690. [Google Scholar] [CrossRef] [PubMed]
  68. Gao, Y.W.; Chen, Y.X.; Wang, Z.M.; Jin, J.; Li, X.Y.; Du Zhou, L.; Huo, Z.; Zhou, J.H.; Chen, W. Increased expression of cyclooxygenase-2 and increased infiltration of regulatory T cells in tumors of patients with hepatocellular carcinoma. Digestion 2009, 79, 169–176. [Google Scholar] [CrossRef] [PubMed]
  69. Fu, J.; Xu, D.; Liu, Z.; Shi, M.; Zhao, P.; Fu, B.; Zhang, Z.; Yang, H.; Zhang, H.; Zhou, C.; et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 2007, 132, 2328–2339. [Google Scholar] [CrossRef] [PubMed]
  70. Ormandy, L.A.; Hillemann, T.; Wedemeyer, H.; Manns, M.P.; Greten, T.F.; Korangy, F. Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res. 2005, 65, 2457–2464. [Google Scholar] [CrossRef] [PubMed]
  71. Lu, Y.; Yang, A.; Quan, C.; Pan, Y.; Zhang, H.; Li, Y.; Gao, C.; Lu, H.; Wang, X.; Cao, P.; et al. A single-cell atlas of the multicellular ecosystem of primary and metastatic hepatocellular carcinoma. Nat. Commun. 2022, 13, 4594. [Google Scholar] [CrossRef]
  72. Kim, C.H. FOXP3 and its role in the immune system. Adv. Exp. Med. Biol. 2009, 665, 17–29. [Google Scholar]
  73. Aliabadi, E.; Urbanek-Quaing, M.; Maasoumy, B.; Bremer, B.; Grasshoff, M.; Li, Y.; Niehaus, C.E.; Wedemeyer, H.; Kraft, A.R.M.; Cornberg, M. Impact of HBsAg and HBcrAg levels on phenotype and function of HBV-specific T cells in patients with chronic hepatitis B virus infection. Gut 2022, 71, 2300–2312. [Google Scholar] [CrossRef] [PubMed]
  74. Yeh, S.-H.; Li, C.-L.; Lin, Y.-Y.; Ho, M.-C.; Wang, Y.-C.; Tseng, S.-T.; Chen, P.-J. Hepatitis B Virus DNA Integration Drives Carcinogenesis and Provides a New Biomarker for HBV-related HCC. Cell Mol. Gastroenterol. Hepatol. 2023, 15, 921–929. [Google Scholar] [CrossRef] [PubMed]
  75. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  76. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  77. El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.-Y.; Choo, S.-P.; Trojan, J.; Welling, T.H., 3rd; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef] [PubMed]
  78. Yau, T.; Park, J.-W.; Finn, R.S.; Cheng, A.-L.; Mathurin, P.; Edeline, J.; Kudo, M.; Harding, J.J.; Merle, P.; Rosmorduc, O.; et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022, 23, 77–90. [Google Scholar] [CrossRef]
  79. D’Alessio, A.; Rimassa, L.; Cortellini, A.; Pinato, D.J. PD-1 Blockade for Hepatocellular Carcinoma: Current Research and Future Prospects. J. Hepatocell. Carcinoma 2021, 8, 887–897. [Google Scholar] [CrossRef] [PubMed]
  80. Elinav, E.; Nowarski, R.; Thaiss, C.A.; Hu, B.; Jin, C.; Flavell, R.A. Inflammation-induced cancer: Crosstalk between tumours, immune cells and microorganisms. Nat. Rev. Cancer 2013, 13, 759–771. [Google Scholar] [CrossRef]
  81. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
Figure 1. PRISMA 2020 flow diagram of the study selection process. Reason 1: studies without inclusion of human individuals ≥ 18 years of age; Reason 2: not eligible outcome; Reason 3: study without original data; Reason 4: not reported in English.
Figure 1. PRISMA 2020 flow diagram of the study selection process. Reason 1: studies without inclusion of human individuals ≥ 18 years of age; Reason 2: not eligible outcome; Reason 3: study without original data; Reason 4: not reported in English.
Viruses 16 00707 g001
Figure 2. Illustration of the role of PD-1 in T-cell inactivation. PD-1 is a receptor found in T cells, which regulates immune responses by binding to its ligands, PD-L1 or PD-L2, on other cells. This interaction triggers inhibitory signals that dampen T-cell activation and effector function. This mechanism helps maintain immune tolerance but can also be exploited by pathogens and cancer cells to evade immune surveillance. Blocking the PD-1/PD-L1 or PD-L2 interaction with immune checkpoint inhibitors enhances anti-tumor immune responses, making it a potential target in cancer immunotherapy.
Figure 2. Illustration of the role of PD-1 in T-cell inactivation. PD-1 is a receptor found in T cells, which regulates immune responses by binding to its ligands, PD-L1 or PD-L2, on other cells. This interaction triggers inhibitory signals that dampen T-cell activation and effector function. This mechanism helps maintain immune tolerance but can also be exploited by pathogens and cancer cells to evade immune surveillance. Blocking the PD-1/PD-L1 or PD-L2 interaction with immune checkpoint inhibitors enhances anti-tumor immune responses, making it a potential target in cancer immunotherapy.
Viruses 16 00707 g002
Table 1. Overview of 52 studies included in the review.
Table 1. Overview of 52 studies included in the review.
AuthorPublication Year Patients, Total (n)HBV HCC CHBNon-HBV HCCHC Tissue SamplesBlood Samples
You et al. [20]202358430114YesYes
Sun et al. [21]20231062631049NoYes
Liu et al. [22]20232516090YesNo
Chang et al. [23]2023147147000NoYes
Li et al. [24]20233460226YesNo
Gao et al. [25]2022147070YesNo
Lu et al. [26]2022142870550YesNo
Li et al. [27]2022293293000YesYes
Zhang et al. [28]2022864227017YesYes
Ho et al. [29] 20218292200YesNo
Cheng et al. [30]202146300160YesYes
Xin et al. [31]2021220220000YesNo
Zhang et al. [32]2021602227011YesYes
Liu et al. [33]20211523178043NoYes
Zhao et al. [34]202038190019YesYes
Li et al. [35]202060300300YesNo
Zhang et al. [36]2020924921022YesNo
Lim et al. [37]201924113501060YesYes
Liu et al. [38]201920412247035No Yes
Wang et al. [39]20199330 3YesYes
Hsiao et al. [40]20191328313010150YesNo
Shen et al. [41]2018792434021NoYes
Liu et al. [42]20189090000YesYes
Ou et al. [43]201888305800YesYes
Wu et al. [44]2018851847020YesYes
Li et al. [45]201887010YesNo
Meng er al. [46]20171111000YesYes
Huang et al. [47]20174113620490Yes No
Lan et al. [48]201793510042No Yes
Jiang et al. [49]2017421414014No Yes
Liu et al. [50]2017160738700No Yes
Li et al. [51]20173232000YesYes
Li et al. [52]2017296 *0000NoYes
Xue et al. [53]201628150013YesYes
Liu et al. [54]20168155740141100NoYes
Zhou et al. [55]2016442012012YesYes
Jia et al. [56]20158585000YesYes
Duan et al. [57]201533210011No Yes
Liu et al. [58]2015601501530YesYes
Dinney et al. [59]201545153000No Yes
Sharma et al. [60]2015491710220YesYes
Xu et al. [61]2014881652020No Yes
Chen et al. [62]201494306400No Yes
Li et al. [63]201389600029YesYes
Li et al. [64]2012150990510 Yes No
Shi et al. [65]20111025620026YesYes
Zhang et al. [66]201089491,5025YesYes
Gehring et al. [67]200930102000No Yes
Gao et al. [68]200950400010YesNo
Fu et al. [69]200719112321047YesYes
Ormandy et al. [70]20051051706721No Yes
Total (n) 6931366074016276503638
HBV HCC = hepatitis B virus-associated hepatocellular carcinoma; CBH = chronic hepatitis B; non-HBV HCC = hepatocellular carcinoma with other/additional etiologies than hepatitis B virus; HC = healthy comparison group without CHB. * No patients were diagnosed with HCC at inclusion; however, patients were followed to evaluate risk of HCC development.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Broholm, M.; Mathiasen, A.-S.; Apol, Á.D.; Weis, N. The Adaptive Immune Response in Hepatitis B Virus-Associated Hepatocellular Carcinoma Is Characterized by Dysfunctional and Exhausted HBV-Specific T Cells. Viruses 2024, 16, 707. https://doi.org/10.3390/v16050707

AMA Style

Broholm M, Mathiasen A-S, Apol ÁD, Weis N. The Adaptive Immune Response in Hepatitis B Virus-Associated Hepatocellular Carcinoma Is Characterized by Dysfunctional and Exhausted HBV-Specific T Cells. Viruses. 2024; 16(5):707. https://doi.org/10.3390/v16050707

Chicago/Turabian Style

Broholm, Malene, Anne-Sofie Mathiasen, Ása Didriksen Apol, and Nina Weis. 2024. "The Adaptive Immune Response in Hepatitis B Virus-Associated Hepatocellular Carcinoma Is Characterized by Dysfunctional and Exhausted HBV-Specific T Cells" Viruses 16, no. 5: 707. https://doi.org/10.3390/v16050707

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop