Skip to main content

REVIEW article

Front. Oncol., 06 September 2021
Sec. Cancer Genetics
This article is part of the Research Topic Predictive Biomarkers of Immune-checkpoint Inhibitors Immunotherapies in Hepatocellular Carcinomas and Gastric Cancers View all 12 articles

Biomarkers and Future Perspectives for Hepatocellular Carcinoma Immunotherapy

  • 1Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
  • 2College of Life Sciences, Wuhan University, Wuhan, China

Hepatocellular cancer is the sixth most frequently diagnosed malignant disease worldwide, and was responsible for tens of millions of deaths in 2020; however, treatment options for patients with advanced hepatocellular carcinoma remain limited. Immunotherapy has undergone rapid development over recent years, especially in the field of immune checkpoint inhibitors (ICIs). These drugs aim to activate and enhance antitumor immunity and represent a new prospect for the treatment of patients with advanced cancer. Nevertheless, only a small proportion of liver cancer patients currently benefit from ICI-based treatment, highlighting the need to better understand how ICIs and tumors interact, as well as identify predictive biomarkers for immunotherapeutic responses. In this review, we highlight clinical trials and basic research in hepatocellular carcinoma, with a particular focus on predictive biomarkers for the therapeutic efficacy of ICIs. Predictive biomarkers for immune-related adverse events are also discussed.

Introduction

Hepatocellular carcinoma (HCC) accounts for over 80% of all primary liver malignancies, while liver cancer ranked as the sixth most frequently diagnosed cancer in 2020, resulting in 83 million deaths (1). Despite these statistics, HCC treatment remains a major healthcare challenge globally. Additionally, because symptoms and physical characteristics of HCC are not easily detected, 80% of patients diagnosed with HCC miss out on curative treatment (2).

HCC typically develops in a background of underlying inflammatory liver disease, especially that associated with hepatitis B (HBV) or hepatitis C virus (HCV) infection (3), while nonalcoholic fatty liver disease (NAFLD) is rapidly becoming a key etiological factor for HCC in many Western countries (4, 5). Current treatment modalities for patients with nonadvanced HCC include resection, transplantation, ablation, or chemoembolization, while patients with advanced HCC receive systemic treatment (6). However, progress in the development of treatments for advanced HCC has been limited, partly due to the complex and heterogeneous etiology of this disease. Additionally, the most common driver mutations (TERT promoter, CTNNB1, TP53, and ARID1A mutations) have not yet been shown to be suitable therapeutic targets (7). Although first-line multikinase inhibitors (sorafenib and lenvatinib) can prolong the survival of patients with advanced HCC (810), and multitarget tyrosine inhibitors (e.g., regorafenib and cabozantinib) and vascular endothelial growth factor (VEGF) receptor inhibitors (e.g., ramucirumab) can provide benefit for patients who previously tolerated sorafenib (11), most cases of HCC show tolerance or become refractory to these agents during the clinical course of the disease (12). Accordingly, the median overall survival (OS) for patients treated with these agents remains under 15 months.

Cancer immunotherapy has undergone rapid development in recent years, especially in the field of immune checkpoint inhibitors (ICI). Immune checkpoint-related molecules, such as programmed cell death-1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), T-cell immunoglobulin, mucin domain-3 (TIM-3), and lymphocyte activating-3 (LAG-3), are important components of the negative feedback regulatory mechanism that serves to suppress excessive immune responses. They are constitutively upregulated in various tumors, generating T-cell exhaustion or anergy, and thereby helping tumors evade immune surveillance (13). The rationale behind utilizing ICIs is to restore and enhance antitumor immunity by relieving the immunosuppressive effects of immune checkpoint-related molecules. The development of anti-PD-1, anti-programmed cell death-ligand 1 (PD-L1), and anti-CTLA-4 monoclonal antibodies has advanced the treatment for advanced cancer, resulting in numerous attempts to apply ICIs for the treatment of multiple advanced solid malignancies, including HCC.

Based on encouraging results from the CheckMate 040 and KEYNOTE-224 clinical trials, the United States Federal Drug Administration (FDA) has granted accelerated approval for the PD-1 inhibitors nivolumab and pembrolizumab as second-line treatments for advanced HCC. The CheckMate 040 phase I/II trial obtained objective response rates (ORRs) of 15% (dose-escalation phase) and 20% (dose-expansion phase) in patients treated with nivolumab (14). Meanwhile, the KEYNOTE-224 phase II trial reported an ORR of 17% for pembrolizumab monotherapy for HCC patients previously treated with sorafenib (15). Disappointingly, however, both the CheckMate 459 and KEYNOTE-240 phase III trials, which evaluated nivolumab versus sorafenib and pembrolizumab versus best supportive care, respectively, failed to meet their predetermined primary endpoints of OS (16, 17). Overall, ICI monotherapy has shown limited efficacy in HCC, benefiting only a limited subgroup of patients. More results of clinical trials for ICIs in HCC are summarized in Table 1.

TABLE 1
www.frontiersin.org

Table 1 Clinical trials of immune-checkpoint inhibitors in hepatocellular carcinoma.

In contrast, the results from the IMbrave150 phase III trial have been encouraging (22). This trial enrolled 501 patients with advanced HCC who had not previously received systemic treatment, two-thirds of whom received atezolizumab (anti-PD-L1 blockade) plus bevacizumab (VEGF monoclonal antibody), while the others received sorafenib monotherapy. The median progression-free survival (PFS) was 6.8 months in the combination arm and 4.3 months in the sorafenib group. In addition, OS at 12 months was 67.2% with atezolizumab + bevacizumab and 54.6% with sorafenib. The two subgroups displayed similar toxic effects, with an incidence of 56.5% for grade 3 or 4 adverse events in the combination arm and 55.1% in the sorafenib arm. Of note, these data are momentous, as they identify the first therapy to improve OS and PFS beyond the standard of care sorafenib in treatment-naïve patients (23). After more than a decade of stagnation in the treatment of advanced HCC, these therapeutic strategies changed the status quo and have entered into clinical practice. The mechanisms underlying the effects of ICI/anti-VEGF-agent combination treatment have also been elucidated, and have been reviewed in detail (24, 25).

Even though the prospect for ICIs seems to be excellent, numerous difficulties remain to be resolved. Pivotal among these is the low response rate in patients treated with ICI monotherapy, with treatment benefiting only 15%–20% of patients with advanced HCC (14, 15). The incidence of immune-related adverse events (irAEs) is another important concern. A better understanding of the dialog between ICIs and tumors is essential, as is the identification of predictive biomarkers for treatment response and toxicity. In this review, we focus on clinical trials and basic research in HCC, with particular emphasis on predictive biomarkers for the therapeutic efficacy of ICIs. Predictive biomarkers for irAEs are also discussed.

Potential Predictive Biomarkers for ICI-Based Treatment

Because immunotherapy for HCC is still in its infancy, studies relating to predictive biomarkers for ICI treatment response are scarce. Nevertheless, several valuable data on potential biomarkers have emerged in recent years, including genetic and protein markers, immune-related cells, and host-related factors (Figures 1, 2), which are described below.

FIGURE 1
www.frontiersin.org

Figure 1 The classification of biomarkers. The biomarkers introduced in this review can be divided into protein markers, gene markers, immune-related cells, and host-related cells. Moreover, according to their localization in vivo, they can be further subdivided into intratumoral and extratumoral biomarkers as they are detected using different methods.

FIGURE 2
www.frontiersin.org

Figure 2 A schematic representation of biomarkers for predicting treatment responses to immune checkpoint inhibitors (ICIs) in vivo and their interactions with each other. Both the genetic characteristics and the expression of PD-L1 in tumor cells, as well as the density and diversity of tumor-infiltrating CD8+ T cells, have the potential to predict the efficacy of ICI treatment in hepatocellular carcinoma (HCC). Other tumor-infiltrating lymphocytes in the tumor microenvironment affect T-cell activity by secreting cytokines (such as IFN-γ and TGF-β), thereby affecting the efficacy of ICI treatment. The gut microbiota also affects the responses to ICI treatment by influencing the activity of tumor-infiltrating lymphocytes (TILs), while circulating biomarkers in peripheral blood can also be used as predictors of immunotherapeutic efficacy.

PD-L1 Expression

PD-L1 is dynamically and widely expressed on the surface of tumor cells, antigen-presenting cells, and other immune cells. PD-L1 expression is reported to be generally low in HCC (~10% of tumor cells) (26) and is associated with recurrence and shorter OS (27). In addition, high PD-L1 expression in inflammatory cells within the tumor microenvironment (TME) correlates with high serum alpha-fetoprotein (AFP) levels, macrovascular invasion, and poor differentiation, resulting in increased tumor aggressiveness (28). PD-L1 was the first proposed predictive biomarker for responses to ICIs, and the relationship between PD-L1 expression and ICI treatment response has been extensively explored in HCC-related clinical trials.

Several results of clinical trials in which PD-L1 was evaluated as a predictive biomarker for ICI responses have been published (the data are summarized in detail in Table 2). In the CheckMate 459 phase III trial, although nivolumab did not elicit prominent improvements in OS as a first-line treatment for HCC, PD-L1-positive tumors nevertheless showed a better response to nivolumab compared to sorafenib (17). This agreed with the results of the KEYNOTE-224 trial, which showed that PD-L1 expression was associated with the response to pembrolizumab, even though the results were not statistically significant (15). However, the predictive value of PD-L1 remains unclear. Disappointing response rates were consistent across all patients in both the CheckMate 040 and NCT02658019 trials, regardless of PD-L1 expression levels (14, 18, 21). Even so, further analysis of patients in the CheckMate 040 trial showed that tumor PD-L1 expression was associated with improved OS, although objective responses could still be observed in PD-L1-negative patients treated with nivolumab. These observations highlight that PD-L1 expression alone may not serve as an adequate biomarker for responses to ICI treatment (29). Although these trials provide valuable clinical data that allow the evaluation of the predictive effect of PD-L1 expression, they have their limitations. First, these are retrospective analyses with small sample sizes. Secondly, there are limitations associated with the detection of PD-L1 expression (discussed in detail later), as well as inconsistencies in sample sources; although they are all collected at baseline, some samples are fresh and some are archival.

TABLE 2
www.frontiersin.org

Table 2 Predictive biomarkers in clinical trials of immune-checkpoint inhibitors in hepatocellular carcinoma.

The contradictory results of these clinical trials may be partly due to limitations in the detection of PD-L1 levels, including a lack of standard methods for evaluating PD-L1 expression and its spatial and temporal heterogeneity, as well as the absence of standard thresholds that allow the determination of “overexpression” (30, 31). The expression of PD-L1 is measured using immunohistochemistry on formalin-fixed paraffin-embedded (FFPA) sections; however, two methods are used for defining PD-L1-positive expression, namely, the ratio of PD-L1-positive tumor cells—the so-called tumor proportion score (TPS)—and the ratio of PD-L1-stained tumor and immune cells, the so-called combined positive score (CPS). The KEYNOTE-224 phase II trial evaluated PD-L1 using both scoring methods, with the CPS turning out to be a more applicable biomarker (15). In addition, the predictive value of PD-L1 expression may be underestimated as it is commonly evaluated at a single time point, even though PD-L1 expression is dynamic and inducible (32, 33). PD-L1 is heavily glycosylated and such modifications significantly affect the detection performance and therapeutic efficacy of PD-L1 antibodies (34, 35). A method was developed to resolve this issue that involved removing the glycan moieties from cell-surface antigens via enzymatic digestion, which boosted the positivity rate of PD-L1 detection in tumor samples (35).

Tumor Mutational Burden/Microsatellite Instability

Neoantigens, arising as a consequence of tumor-specific mutations, are hypothesized to generate robust immune responses (36). The tumor mutational burden (TMB) refers to the number of nonsynonymous mutations found in the genome of a single tumor, including alterations in DNA damage response genes and those encoding the DNA polymerase epsilon (POLE) and delta (POLD) catalytic subunits, and has been assessed for its potential as a biomarker in multiple tumor types (37, 38). It is thought that tumors with a greater TMB can produce a greater number of neoantigens. One cross-cancer study reported that tumors with a high TMB are positively correlated with responses to anti-PD-1/PD-L1 therapy, and a greater TMB is also associated with higher PD-L1 expression in tumor cells (39). To examine this association more broadly, another study analyzed the clinical and genomic data of thousands of patients with advanced cancer and identified an association between a higher TMB and improved survival linked to ICI treatment in most of the cancer types assessed (40). Several studies have shown that a higher TMB is associated with immune microenvironment diversification and worse prognosis in HCC patients (41, 42). However, its putative role as a predictive biomarker has not been reported in HCC patients treated with either nivolumab or pembrolizumab, and whether the TMB has value as a predictive marker for ICI efficacy remains unclear.

A study evaluated the frequency of genomic biomarkers of ICI response in 755 HCC patients and found that the median TMB was four mutations/Mb, with only six tumors (0.8%) found to be TMB-high (43). Furthermore, in a small case series (N=17), the TMB showed no correlation with ICI response. As in the case of PD-L1 expression, however, TMB determination lacks standardized thresholds and there is variability in quantification methods (44). Thus, the clinical value of TMB should be interpreted with caution. Recently, Wong et al. evaluated the TMB in 29 HCC patients through targeted next-generation sequencing (tNGS) on both fresh and archival samples. The authors reported that, while fresh HCC samples were better sources of tumor DNA (45), the low median TMB values observed may limit the usefulness of the TMB as a predictor of response to immunotherapy in HCC.

DNA mismatch repair (MMR) represents a key mechanism for the maintenance of genomic integrity and stability. A deficiency in MMR activity results in a hypermutator phenotype known as microsatellite instability (MSI) (46). High mutation rates in somatic cells are believed to amplify the neoantigen load, leading to lymphocyte activation and enhanced cancer susceptibility to immunotherapy (47, 48). A study assessed the efficacy of PD-1 blockade with pembrolizumab monotherapy in 86 patients with advanced MMR-deficient cancers across 12 different cancer types. Based on ORRs and complete response (CR) rates of 53% and 21%, respectively, the results of this study support that MSI may serve as a predictor of the clinical response of solid tumors to PD-1 blockade (49). Although MSI is considered useful as an agnostic histological indicator for the selection of responders to ICI therapy, there is nonetheless a lack of data for HCC patients. Ang et al. assessed 542 HCC specimens for MSI and found that only one (0.2%) was MSI-high and TMB-high (43), while several other studies have reported that the prevalence of MSI in HCC is also generally low (50, 51). These findings highlight that MSI may not be an ideal biomarker to predict responses to ICIs in HCC patients.

The low frequency of high TMB and high MSI limits their exploration as predictive biomarkers in HCC, while the number of cases included in existing studies is also relatively low (44). However, given that MSI has shown good predictability in immunotherapy for other types of cancers, research efforts should continue to focus on exploring its predictive potential in HCC, including combining data from multiple clinical trials to obtain a sufficiently large sample size that would allow the evaluation of its role in HCC-targeted immunotherapy.

Tumor-Infiltrating Lymphocytes

Because T-cell infiltration within the TME is a prerequisite for immune checkpoint blocking (52), baseline intratumoral T-cell density and phenotype have been extensively studied and closely connected with ICI responses in melanoma and other tumors (53, 54). A retrospective biomarker analysis undertaken in the CheckMate 040 trial indicated that, although not statistically significant, increased numbers of CD3+ or CD8+ tumor-infiltrating T cells were correlated with a trend towards improved OS in nivolumab-treated HCC patients (29). This study further indicated that an association existed between an increased frequency of CD3+ T cells and the best overall response. A different study reported that an increase in CD8+ T cells in six-week tumor biopsies was connected with clinical benefits for HCC patients who received tremelimumab plus ablation combination therapy (55). Furthermore, Kaseb et al. found that the clinical response of HCC patients who received perioperative immunotherapy (nivolumab plus ipilimumab) followed by surgical resection was correlated with an increase in CD8+ T-cell infiltration, and, specifically, with two effector T-cell clusters (CD3+CD8+CD45RO+Eomes+ and CD3+CD8+CD45RO+Eomes+CD57+CD38low) (56).

The connection between TILs and ICI treatment response has also been investigated in other immune cell types. Ng et al. analyzed 49 HCC samples from patients treated with ICIs and reported that patients with a high intratumoral CD38+CD68+ macrophage density had a better median OS compared with those with low CD38+CD68+ macrophage density (34.43 vs. 9.66 months) (57), likely because CD38hi macrophages produce greater amounts of interferon-gamma (IFN-γ) and related cytokines. Notably, some of these 49 patients had received combination immunotherapy. The influence of immune cell infiltration on the effectiveness of ICIs has also been studied in multifocal HCC, with the results showing that small nodules are more sensitive to anti-PD-1 therapy than large nodules, while small tumors exhibit greater immune cell infiltration and an upregulated interferon signature compared with large tumors (58).

Deep single-cell RNA sequencing was performed on 5,063 T cells isolated from peripheral blood, tumor tissue, and adjacent normal tissues of six HCC patients (59). The results demonstrated that specific subsets, such as exhausted CD8+ T cells and regulatory T cells (Tregs), were preferentially enriched in HCC. The authors further identified that increased expression of layilin (encoded by LAYN) was associated with tumor-infiltrating Tregs and activated CD8+ T cells. While these findings need to be validated in a larger cohort, they will undoubtedly open a new avenue for further research into the potential of utilizing CD8+ T-cell infiltration as a biomarker in HCC.

Although it is important to focus on lymphocytes in the TME, applying them as biomarkers in the clinic remains a distant prospect. How to select lymphocytes with specific characteristics, how to define the positive criteria are among the many unanswered questions. Joint evaluation of the number of multiple immune cells and PD-L1 expression to develop TME scores may be one of the answers.

Specific Gene Alterations

Over recent years, molecular profiling associated with the advent of NGS has provided information on actionable targets and identified specific gene alterations associated with responses to ICIs (60).

Mutations in the CTNNB1 gene, which lead to the activation of the WNT/β-catenin signaling pathway, are characteristic of immune-excluded HCC types (cold tumors) and are associated with significantly lower enrichment scores for several immune signatures (61, 62). Harding et al. employed NGS to determine which type of patient with advanced HCC might benefit from systemic treatments (63). For a subgroup of 31 patients treated with diverse ICIs, activating alterations in the WNT/β-catenin pathway were associated with lower disease control rates (DCRs) (0% vs. 53%), shorter median PFS (2.0 vs. 7.4 months), and shorter median OS (9.1 vs. 15.2 months) compared with those in WNT wild-type HCC. Additionally, β-catenin-driven tumors were reported to be resistant to PD-1 therapy in a mouse model of HCC, while the expression of chemokine (C-C motif) ligand 5 (CCL5) could restore the β-catenin-associated loss of immune surveillance (64).

In addition, TP53 gene alterations were the most frequently identified mutations in HCC patients and were mutually exclusive with CTNNB1 mutations (65). Studies have shown that TP53 alterations are strongly related to the immune microenvironment in HCC, with less CD8+ T cell infiltration and more FOXP3+ Treg infiltration, resulting in the downregulation of the immune response (6668). TP53 dysfunction was shown to be linked to chromosomal instability (defined as high broad copy-number alteration loads) and immune-excluded traits in HCC (69). Conversely, Yang et al. reported that HCC patients carrying TP53 neoantigens showed higher cytotoxic lymphocyte infiltration and longer OS (70).

Recent studies have indicated that immune-related long noncoding RNAs (lncRNAs) may predict ICI treatment responses in HCC. Peng et al. reported that the host gene of lncRNA MIR155 was strongly positively correlated with the expression of CTLA-4 and PD-L1 in HCC tissues, and showed predictive value for the curative effect of ICI therapy (71). Additionally, Zhang et al. identified an immune-related lncRNA signature that correlated with worse survival and was an independent prognostic biomarker for HCC patients (72). That this signature was associated with immune cell infiltration and ICI treatment-related molecules (including PD-L1, PD-L2, and IDO1) suggests that it may have the potential to measure the response to ICI immunotherapy. Several studies have reported on the ability of immune-related lncRNAs to predict prognosis in HCC patients (7376). However, prospective validation in HCC patients who received ICI treatment is still lacking. MicroRNAs and circular RNAs have also been reported to reshape the immune microenvironment in HCC (77, 78). Mo et al. identified 5-methylcytosine (5mC)-associated molecular subtypes in HCC and found that they were associated with responses to immunotherapy (79).

Several studies have indicated that cancer cell-intrinsic epigenetic alterations are associated with carcinogenesis and tumor progression (8083), as well as with changes in the TME, such as infiltration of tumor-associated lymphocytes and expression of immune checkpoint molecules (84, 85), indicative of their potential as predictors of immunotherapy. Furthermore, given that DNA methylation can be measured in liquid biopsies, epigenetic biomarkers may provide additional advantages, such as low patient invasiveness (86). In patients with melanoma receiving anti-PD-1 immunotherapy, the hypermethylation of the T-cell costimulatory receptor TNFRSF9 was reported to be correlated with poor PFS and treatment response (87). In a multicenter study, Duruisseaux et al. established an epigenomic profile based on a microarray DNA methylation signature (EPIMMUNE) in a discovery set of tumor samples from patients with advanced nonsmall cell lung cancer (NSCLC) who had received anti-PD-1 therapy (88). The authors found that the unmethylated status of the T-cell differentiation factor forkhead box P1 (FOXP1) was associated with improved PFS and OS, and, therefore, possessed good predictive value for the efficacy of anti-PD-1 treatment. Another study identified a broad DNA methylation signature in peripheral blood mononuclear cells and T cells of HCC patients that differed from that of non-HCC patients (89). Additionally, differences in immune infiltrates related to the methylation levels of cell division cycle-associated (CDCA) family genes in HCC were reported to have potential as predictive biomarkers for responses to immunotherapy (90). Notably, Llopiz et al. reported that an epigenetic drug/ICI combination exerted synergistic antitumor effects in a murine model of HCC (91), thereby providing further evidence that DNA methylation signatures may be related to the efficacy of immunotherapy, and also have the potential to serve as biomarkers of combination therapy.

Combined, these observations indicate that the presence of specific gene alternations, especially those related to CTNNB1, TP53, noncoding RNAs, and methylation, may influence the response to ICI treatment through interacting with the immune microenvironment. Although these studies are still preliminary, such alterations could represent novel biomarkers for HCC patient selection, and patient exclusion in particular.

Immune-Related Gene Signatures

Comprehensive analyses of tumor transcriptomic profiling data have recently been conducted to characterize the responsiveness of the immune microenvironment to ICI treatment. Ayers et al. identified an 18-gene T-cell-inflamed expression profile and a 6-gene IFN-γ signature that could predict responses to pembrolizumab therapy across multiple solid tumors (92). Based on gene expression profiles, Sia et al. classified 25% of HCCs as an “immune-specific class” characterized by high expression levels of PD-L1 and markers of cytolytic activity. This class further comprised two subtypes, namely, an active immune subtype, characterized by significant enrichment of T cells and IFN signatures, and an exhausted immune subtype, associated with a T-cell exhaustion signature and immunosuppressive components (TGF-β and M2 macrophages) (62).

The previously-mentioned retrospective analysis of the findings of the CheckMate 040 trial also included the evaluation of the predictive value of a four-gene inflammatory signature (CD274 [PD-L1], CD8A, LAG3, and STAT1). This signature was found to be associated with improved response and OS related to nivolumab therapy, both in the dose-escalation and dose-expansion phases (29), and may be indicative of IFN-γ/STAT1-dependent CD8+ T-cell expansion, LAG-3-dependent T-cell exhaustion, and/or an immune-suppressed TME with high PD-L1 expression. Importantly, however, although the differences in the above-mentioned clinical trial data were reported to be statistically significant, this gene signature does not adequately distinguish between responders and nonresponders. Recently, a five-gene immune-related signature (LDHA, PPAT, BFSP1, NR0B1, and PFKFB4) was identified and used to establish a prognostic model for responses to HCC treatment that could stratify patients who were sensitive to immunotherapy (93). Other studies using different gene combination strategies have also reported their potential to predict ICI responses by reflecting the characteristics of the immune microenvironment (94, 95). Nevertheless, these signatures need further testing for clinical application.

Immune-related gene signatures are associated with the same obstacles as TILs, namely, how to select the best gene combinations and positive cut-off values. A feasible strategy involves undertaking a wide-ranging bioinformatics analysis of the existing databases to identify possible gene combinations and then verify them through basic research and clinical trials.

Biomarkers in Peripheral Blood

The continuous availability of tumor samples from ICI-treated patients is crucial for biomarker research; however, this is difficult to achieve owing to the invasive nature of biopsies. In contrast, circulating biomarkers can be easily collected and repeatedly measured after treatment, rendering them more convenient for use in the clinic.

Studies have shown that TGF-β plays a central role in immune suppression within the TME and tumor immune evasion (96). In HCC, the potent immune inhibitory function of Tregs is a major obstacle to generating an effective antitumor response, and Treg activation is modulated by the TGF-β pathway (97). In addition, TGF-β enables tumor evasion from host immune responses in part through enhancing SMAD3-mediated PD-1 expression on TILs (98). The TGF-β signaling pathway is activated at the transcriptional level in most HCCs (99, 100). A strong association was identified between the TGF-β signature and the exhausted immune signature in HCC (62, 99). In a phase II trial, several representative circulating biomarkers were analyzed in 29 patients with unresectable HCC treated with pembrolizumab, with the results showing that baseline plasma TGF-β levels of <200 pg/mL were an effective predictor of better OS and PFS (21). The therapeutic co-administration of TGF-β-blocking and anti-PD-L1 antibodies was reported to facilitate T-cell penetration into the center of tumors and elicit strong antitumor immunity (101). Clinical trials of the TGF-β receptor I inhibitor galunisertib have been conducted, and have reported median OS durations of 16.8 months in patients with advanced HCC with baseline AFP <1.5 × ULN (102). Briefly, TGF-β may serve as a negative predictive biomarker for ICI therapy given that HCCs with strong TGF-β and exhausted immune signatures may be resistant to PD-1 blockade, while HCC patients with an activated TGF-β signature are expected to benefit from a combination of ICI and TGF-β inhibitor therapy (99). Thus, it seems likely that drugs targeting both TGF-β and PD-1/PD-L1, such as bintrafusp alfa, will play a role in the treatment of HCC in the future.

Several soluble immune checkpoint-related proteins were recently shown to have promising predictive value in various cancer types. Chen et al. reported that metastatic melanomas released exosomes carrying PD-L1 that suppressed CD8+ T-cell function and supported tumor growth. Additionally, the authors reported that the magnitude of the increase in the levels of circulating exosomal PD-L1 during the early stages of treatment could discriminate clinical responders from nonresponders (103). One preclinical study using mouse models indicated that the removal of exosomal PD-L1 inhibits tumor growth, even in models resistant to PD-L1 blockade (104).

Soluble PD-L1 (sPD-L1) has been reported to be correlated with responses to immunotherapy in patients with NSCLC (105). Additionally, several studies have focused on the prognostic value of sPD-L1 and identified high serum sPD-L1 levels as an independent predictive factor for poor outcomes in HCC patients (106108). Whether an association exists between sPD-L1 and intratumoral PD-L1 expression levels remains unclear, with contradictory results having been reported (106, 108). Moreover, there is a lack of data on the ability of exosomal PD-L1 or sPD-L1 to predict clinical outcomes in HCC patients following ICI therapy, which warrants further study.

Circulating immune cells in peripheral blood have been extensively evaluated as predictive biomarkers (105, 109). Agdashian et al. tested the combination of anti-CTLA-4 treatment (tremelimumab) with locoregional therapy in HCC patients and found that the frequency of CD4+PD-1+ cells among peripheral blood mononuclear cells at baseline was higher in patients responding to therapy than in nonresponding patients (110). In addition, low baseline peripheral B cell PD-1 positivity and constant posttreatment monocyte PD-L1 positivity were observed to be associated with disease control in 16 HCC patients treated with nivolumab (111). Dharmapuri et al. evaluated the relationship between the neutrophil–lymphocyte (NLR) and platelet–lymphocyte (PLR) ratios and survival outcomes in HCC patients treated with nivolumab and reported that patients who achieved a partial or CR had significantly lower posttreatment NLRs and PLRs (112). The predictive value of the NLR and the PLR was also indicated in the CheckMate 040 trial (29).

Necrotic or apoptotic tumor cells release DNA carrying tumor-related genetic and epigenetic alterations into the bloodstream, thereby helping to overcome the limitations related to sample availability. Circulating tumor DNA (ctDNA) can reportedly predict tumor responses to ICI therapy in several types of tumors; specifically, it can be used to distinguish pseudoprogression from true progression (113115). However, a study analyzing the mutational landscape of advanced HCC using ctDNA reported that WNT pathway-related mutations were not associated with clinical outcomes after ICI therapy (65), highlighting that further investigation is needed to determine whether ctDNA can indeed serve as a predictive biomarker in HCC. Meanwhile, although the blood TMB was reported to have good predictive value in some types of cancer (116, 117), evidence is lacking for HCC. Winograd et al. sought to detect PD-L1-expressing circulating tumor cells (CTCs) in HCC patients and found that there was a strong association between the presence of PD-L1+ CTCs and favorable treatment responses to PD-1 blockade (118).

High AFP levels are considered to be a prognostic marker for poor clinical outcomes in HCC patients. Recently, a posttreatment decline in serum AFP levels was reported to be associated with higher ICI treatment efficacy in advanced HCC (119, 120). In addition, Spahn et al. reported that baseline levels of AFP of <400 µg/L at the start of ICI treatment were associated with higher rates of CR or partial response (PR) as best responses (121). However, the results of the checkmate040 clinical trial showed that although baseline AFP <400 µg/L was associated with longer OS compared with AFP ≥400 µg/L, the ORR and DCR were similar regardless of baseline AFP levels (29). Given that the AFP level is closely related to the baseline characteristics of patients, its predictive effect should be interpreted with caution.

Biomarkers in peripheral blood have the great advantage of low invasiveness, while a large number of studies have also shown their potential as predictors of immunotherapeutic efficacy. However, supporting data from large-sample clinical trials are still lacking. In addition, there is currently no evidence that the predictive accuracy of blood samples is better than that of tissue samples for any given biomarker. Nevertheless, these studies provide insights for future research into biomarkers for HCC immunotherapy.

Gut Microbiota

Increasing evidence has indicated that the gut microbiota plays a crucial role in the development and regulation of innate and adaptive immunity, while several studies have described its value in predicting the efficacy of ICIs. For instance, analysis of baseline gut microbiota composition of fecal samples from patients with melanoma or NSCLC before immunotherapy treatment has indicated that commensal microbial composition is associated with an ICI response (122, 123).

A meta-analysis undertaken on 2,424 samples through 16S RNA gene sequencing and machine based-learning indicated that among several major dysbiosis-related diseases, liver cirrhosis is the condition where changes in the gut microbiome most accurately predict the presence of disease (124). Given that there is an anatomical connection between the liver and the gut, and that HCC occurs in the context of chronic liver inflammation concomitant with a defective intestinal barrier and increased hepatic exposure to bacterial products, it seems likely that a relationship exists between the gut microbiota and responses to ICI therapy. Indeed, increasing evidence points towards a key role of the bacterial microbiome in promoting the development of HCC (125). In the context of chronic inflammation, intestinal bacterial translocation is detected by Toll-like receptor (TLR) 4 present on resident liver cells through its ligand lipopolysaccharide, which leads to the upregulation of the expression of the hepatomitogen epiregulin and, consequently, the promotion of hepatocarcinogenesis (126). Microbiota-derived metabolites can also affect the development of HCC. For instance, the gut microbial metabolite deoxycholic acid acts in concert with lipoteichoic acid to enhance the tumor-promoting phenotype of hepatic stellate cells and promote the expression of COX2 through TLR2, resulting in the suppression of antitumor immunity (127). Additionally, gut microbial-dependent bile acid metabolism modulates liver tumor growth by regulating the hepatic expression of CXCL16, a mediator of natural killer T (NKT) cell recruitment (128). Furthermore, Arpaia et al. found that microbe-derived short-chain fatty acids facilitate extrathymic Treg generation (129). Studies have also shown that the gut microbiota may play an important role in regulating ICI treatment responses (130, 131). For instance, fecal microbiota transplantation from cancer patients who responded to ICIs into sterile or antibiotic-treated mice improved the response to anti-PD-1 therapy (122, 123). Moreover, Zheng and colleagues recently reported the dynamic variation in the composition of the gut microbiome during anti-PD-1 immunotherapy in HCC by metagenomic sequencing (132). They observed that fecal samples from patients responding to immunotherapy showed higher taxonomic richness and greater gene counts compared with those of nonresponders; microbial composition remained relatively stable in the responder group, whereas in nonresponders, Proteobacteria abundance markedly increased from week 3 and became predominant at week 12. In addition, antibiotic administration at the initiation of ICI treatment was reportedly associated with worse outcomes, indicative of the influence of the gut microbiota on HCC treatment (121). A clinical trial (NCT03785210) to evaluate the effects of combined antibiotic (vancomycin) and ICI therapies is currently underway.

However, in the aforementioned studies, there was no overlap in gut microbiota associated with responses, which may be due to differences in etiology, geographic location, nutritional intake, and techniques used to analyze the samples. Moreover, Rosshart et al. found that the gut microbiome of laboratory mice differs significantly from that of closely related species in the wild (133), implying that caution is needed when generalizing these research results. The gut microbiota can be influenced by many environmental, dietary, and lifestyle factors, all of which can potentially affect the immune system and, consequentially, regulate the response to ICIs (134). Given these complications, the application of gut microbiota as a biomarker for clinical use remains a distant possibility.

Others

The T-cell receptor (TCR) is composed of multiple antigen-specific peptide chains. Recent studies have used high-throughput sequencing for the in-depth elucidation of the composition and distribution of the TCR. There have been several attempts to use the TCR as a predictive biomarker for ICI responses. A more clonal TCR repertoire or oligoclonal TIL expansion is associated with a better response to PD-1 blockade in melanoma patients (135, 136). In addition, baseline TCR diversity in peripheral blood has been associated with clinical outcomes following ipilimumab treatment in metastatic melanoma (137). Despite the lack of data regarding the suitability of employing TCR as a biomarker in HCC, its predictive potential nevertheless warrants further investigation given the novel TCR analysis approaches proposed (138, 139). Lin et al. found that the combination of TCR repertoires and TNM stage could serve as an efficient prognostic indicator in patients with HBV-associated HCC (140). Additionally, Han et al. identified several specific TRBVTRBJ combinations that could distinguish the TCR repertoires of HCC patients from those of healthy adults and thus have the potential to serve as novel biomarkers (141).

Pfister et al. conducted a meta-analysis that incorporated more than 1,600 patients with advanced HCC in three randomized phase III clinical trials and reported that PD-L1 or PD-1 inhibitors did not improve survival in patients with nonviral HCC, particularly NAFLD (142). This was likely due to the progressive accumulation of exhausted, unconventionally activated CD8+PD-1+ T cells, which contributed to inducing NAFLD/HCC rather than carrying out or enhancing immune surveillance. However, in the CheckMate 040 phase I/II trial, which enrolled HCC patients with or without HBV or HCV infection, responses to nivolumab were observed irrespective of HCC etiology (14).

Epithelial-to-mesenchymal transition (EMT) has been implicated as a resistance mechanism that helps to promote the immune evasion of cancer cells (143). High expression of PD­L1 in HCC patients is reported to be associated with an EMT phenotype and be a predictor of poor survival (41). The correlation between PD-L1 expression and EMT presents a theoretical foundation to investigate EMT as a negative biomarker for ICI responses.

Matrix metalloproteinase 9 (MMP-9) secreted by tumor-associated macrophages was recently reported to be a potential predictor of immune characteristics and immunotherapeutic responses in HCC (144, 145). One study measured ADAM9 mRNA levels in blood samples derived from patients with advanced HCC. Among four patients treated with nivolumab therapy, two who exhibited a clinical response also showed significant decreases in serum ADAM9 mRNA levels, whereas the two who displayed no response to nivolumab did not. Although the sample size was small, the results of this study nevertheless suggested that ADAM9 mRNA might serve as a predictive biomarker for clinical responses (146).

In addition, Qayyum et al. undertook an interesting prospective study that included 15 patients with advanced HCC treated with pembrolizumab and found that the changes in HCC stiffness as measured by magnetic resonance elastography (MRE) at 6 weeks was significantly associated with OS (147). This was the first proposed imaging-based predictor of immunotherapy outcome in HCC and opens up new avenues for predictor selection.

Potential Biomarkers for irAEs

Between 15% and 25% of ICI-treated HCC patients undergo grade 3/4 treatment for immune-related adverse events, including fatigue, pruritus, rash, diarrhea, and increases in aspartate aminotransferase and alanine aminotransferase levels (14, 15). Although ICI treatment-related adverse events are manageable and less frequent than those seen with chemotherapy, it is still necessary to identify biomarkers that can predict irAEs to alleviate unnecessary suffering in patients.

Most studies have focused on identifying biomarkers for predicting the efficacy of immunotherapy, while relatively few studies have investigated biomarkers relating to irAEs. Moreover, a large proportion of research findings come from patients with melanoma (148). Nevertheless, these studies can serve as a reference for identifying biomarkers that can predict irAEs in HCC. Baseline serum IL-6 and IL-17 levels were significantly associated with an increased risk of severe toxicity in patients treated with ipilimumab (149, 150), with IL-17 being related to severe diarrhea/colitis. A retrospective review involving 167 adult patients with solid tumors indicated that increased baseline lymphocyte counts are associated with a greater risk for irAEs in patients treated with nivolumab or pembrolizumab (151). The detection of autoantibodies has been suggested to predict the development of irAEs related to the autoantibodies, and two studies evaluating antithyroid antibodies and diabetes-related autoantibodies have been reported (152, 153). Baseline gut microbiota enriched in Faecalibacterium spp. and other members of the Firmicutes is associated with a more frequent occurrence of ipilimumab-induced colitis (154).

Rogado et al. reported that ICI treatment was markedly more beneficial for patients with advanced cancer presenting with irAEs than for those without irAEs (ORR: 82.5% vs. 16.6%; PFS: 10 vs. 3 months) (155). Cutaneous or early irAEs are associated with improved survival in melanoma patients treated with nivolumab (156, 157). Future studies should address this association to explore the underlying biological mechanisms related to ICI efficacy, while how to balance the incidence of irAEs and the immunotherapeutic response also merits serious consideration.

Conclusion

In HCC, although several studies have been conducted to identify predictive biomarkers that would allow the stratification of patients who could benefit from ICI treatment, few have been prospectively validated and none have resulted in the rewriting of the current clinical guidelines or entered into clinical practice. Here, we summarized the progress of immunotherapy for HCC over recent years, with a particular emphasis on predictive biomarkers. However, as HCC-related immunotherapy is still in its infancy, basic research and clinical trials exploring the predictive efficacy of immunotherapy biomarkers are still limited, and it is not yet possible to determine which biomarker(s) can effectively predict the efficacy of immunotherapy. It is particularly noteworthy that biomarkers represent continuums and undergo dynamic changes in a population of patients. Accordingly, it is pivotal to obtain samples from patients both before and during treatment to evaluate these dynamic changes and properly determine the predictive value of the assessed biomarkers, while adequate consideration should be given to their application in clinical practice. Furthermore, we only reviewed the biomarkers that predict responses to immune monotherapy, especially anti-PD-L1 and anti-PD-1 agents. However, given the success of the IMbrave150 phase III clinical trial, it is clear that antivascular therapy combined with immunotherapy has great potential in patients with advanced HCC, and combination therapy may be the direction of cancer treatment in the future. But while combination therapy can prolong the OS of HCC patients, it also complicates the patient selection process. The development of research techniques such as NGS, single-cell RNA sequencing, and artificial intelligence should allow for a more comprehensive understanding of the various components of the TME and their interactions, and potential biomarkers could be widely screened on a genomic scale to identify the predictors of treatment efficacy.

Author Contributions

PZ conceived of the topic for this review. All authors contributed to the article and approved the submitted version.

Funding

This work was funded by the National Natural Science Foundation of China (No. 81974483).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

The authors would like to acknowledge Yuhao Hu for assistance with referencing.

References

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: Cancer J Clin (2021) 71(3):209–49. doi: 10.3322/caac.21660

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Lin S, Hoffmann K, Schemmer P. Treatment of Hepatocellular Carcinoma: A Systematic Review. Liver Cancer (2012) 1(3-4):144–58. doi: 10.1159/000343828

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Park JW, Chen M, Colombo M, Roberts LR, Schwartz M, Chen PJ, et al. Global Patterns of Hepatocellular Carcinoma Management From Diagnosis to Death: The BRIDGE Study. Liver International: Off J Int Assoc Study Liver (2015) 35(9):2155–66. doi: 10.1111/liv.12818

CrossRef Full Text | Google Scholar

4. Personeni N, Rimassa L. Hepatocellular Carcinoma: A Global Disease in Need of Individualized Treatment Strategies. J Oncol Pract (2017) 13(6):368–9. doi: 10.1200/jop.2017.024604

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Zhang X. NAFLD Related-HCC: The Relationship With Metabolic Disorders. Adv Exp Med Biol (2018) 1061:55–62. doi: 10.1007/978-981-10-8684-7_5

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Forner A, Reig M, Bruix J. Hepatocellular Carcinoma. Lancet (London England) (2018) 391(10127):1301–14. doi: 10.1016/s0140-6736(18)30010-2

CrossRef Full Text | Google Scholar

7. Bouattour M, Mehta N, He AR, Cohen EI, Nault JC. Systemic Treatment for Advanced Hepatocellular Carcinoma. Liver Cancer (2019) 8(5):341–58. doi: 10.1159/000496439

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in Advanced Hepatocellular Carcinoma. New Engl J Med (2008) 359(4):378–90. doi: 10.1056/NEJMoa0708857

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and Safety of Sorafenib in Patients in the Asia-Pacific Region With Advanced Hepatocellular Carcinoma: A Phase III Randomised, Double-Blind, Placebo-Controlled Trial. Lancet Oncol (2009) 10(1):25–34. doi: 10.1016/s1470-2045(08)70285-7

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib Versus Sorafenib in First-Line Treatment of Patients With Unresectable Hepatocellular Carcinoma: A Randomised Phase 3 Non-Inferiority Trial. Lancet (London England) (2018) 391(10126):1163–73. doi: 10.1016/s0140-6736(18)30207-1

CrossRef Full Text | Google Scholar

11. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for Patients With Hepatocellular Carcinoma Who Progressed on Sorafenib Treatment (RESORCE): A Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet (London England) (2017) 389(10064):56–66. doi: 10.1016/s0140-6736(16)32453-9

CrossRef Full Text | Google Scholar

12. Nishida N, Kitano M, Sakurai T, Kudo M. Molecular Mechanism and Prediction of Sorafenib Chemoresistance in Human Hepatocellular Carcinoma. Digest Dis (Basel Switzerland) (2015) 33(6):771–9. doi: 10.1159/000439102

CrossRef Full Text | Google Scholar

13. Chen DS, Mellman I. Oncology Meets Immunology: The Cancer-Immunity Cycle. Immunity (2013) 39(1):1–10. doi: 10.1016/j.immuni.2013.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

14. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, 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 (London England) (2017) 389(10088):2492–502. doi: 10.1016/s0140-6736(17)31046-2

CrossRef Full Text | Google Scholar

15. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, et al. Pembrolizumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib (KEYNOTE-224): A Non-Randomised, Open-Label Phase 2 Trial. Lancet Oncol (2018) 19(7):940–52. doi: 10.1016/s1470-2045(18)30351-6

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, et al. Pembrolizumab As Second-Line Therapy in Patients With Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J Clin Oncol: Off J Am Soc Clin Oncol (2020) 38(3):193–202. doi: 10.1200/jco.19.01307

CrossRef Full Text | Google Scholar

17. Yau T, Park JW, Finn RS, Cheng AL, Mathurin P, Edeline J, et al. LBA38_PR - CheckMate 459: A Randomized, Multi-Center Phase III Study of Nivolumab (NIVO) vs Sorafenib (SOR) as First-Line (1L) Treatment in Patients (Pts) With Advanced Hepatocellular Carcinoma (aHCC). Ann Oncol (2019) 30:v874–v5. doi: 10.1093/annonc/mdz394.029

CrossRef Full Text | Google Scholar

18. Yau T, Hsu C, Kim TY, Choo SP, Kang YK, Hou MM, et al. Nivolumab in Advanced Hepatocellular Carcinoma: Sorafenib-Experienced Asian Cohort Analysis. J Hepatol (2019) 71(3):543–52. doi: 10.1016/j.jhep.2019.05.014

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Wainberg ZA, Segal NH, Jaeger D, Lee K-H, Marshall J, Antonia SJ, et al. Safety and Clinical Activity of Durvalumab Monotherapy in Patients With Hepatocellular Carcinoma (HCC). J Clin Oncol (2017) 35(15_suppl):4071–. doi: 10.1200/JCO.2017.35.15_suppl.4071

CrossRef Full Text | Google Scholar

20. Sangro B, Gomez-Martin C, de la Mata M, Iñarrairaegui M, Garralda E, Barrera P, et al. A Clinical Trial of CTLA-4 Blockade With Tremelimumab in Patients With Hepatocellular Carcinoma and Chronic Hepatitis C. J Hepatol (2013) 59(1):81–8. doi: 10.1016/j.jhep.2013.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Feun LG, Li YY, Wu C, Wangpaichitr M, Jones PD, Richman SP, et al. Phase 2 Study of Pembrolizumab and Circulating Biomarkers to Predict Anticancer Response in Advanced, Unresectable Hepatocellular Carcinoma. Cancer (2019) 125(20):3603–14. doi: 10.1002/cncr.32339

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab Plus Bevacizumab in Unresectable Hepatocellular Carcinoma. New Engl J Med (2020) 382(20):1894–905. doi: 10.1056/NEJMoa1915745

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Kelley RK. Atezolizumab Plus Bevacizumab - A Landmark in Liver Cancer. New Engl J Med (2020) 382(20):1953–5. doi: 10.1056/NEJMe2004851

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Rizzo A, Ricci AD, Brandi G. Immune-Based Combinations for Advanced Hepatocellular Carcinoma: Shaping the Direction of First-Line Therapy. Future Oncol (London England) (2021) 17(7):755–7. doi: 10.2217/fon-2020-0986

CrossRef Full Text | Google Scholar

25. Rizzo A, Ricci AD, Brandi G. Atezolizumab in Advanced Hepatocellular Carcinoma: Good Things Come to Those Who Wait. Immunotherapy (2021) 13(8):637–44. doi: 10.2217/imt-2021-0026

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Pinato DJ, Mauri FA, Spina P, Cain O, Siddique A, Goldin R, et al. Clinical Implications of Heterogeneity in PD-L1 Immunohistochemical Detection in Hepatocellular Carcinoma: The Blueprint-HCC Study. Br J Cancer (2019) 120(11):1033–6. doi: 10.1038/s41416-019-0466-x

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Gao Q, Wang XY, Qiu SJ, Yamato I, Sho M, Nakajima Y, et al. Overexpression of PD-L1 Significantly Associates With Tumor Aggressiveness and Postoperative Recurrence in Human Hepatocellular Carcinoma. Clin Cancer Research: an Off J Am Assoc Cancer Res (2009) 15(3):971–9. doi: 10.1158/1078-0432.Ccr-08-1608

CrossRef Full Text | Google Scholar

28. Calderaro J, Rousseau B, Amaddeo G, Mercey M, Charpy C, Costentin C, et al. Programmed Death Ligand 1 Expression in Hepatocellular Carcinoma: Relationship With Clinical and Pathological Features. Hepatol (Baltimore Md) (2016) 64(6):2038–46. doi: 10.1002/hep.28710

CrossRef Full Text | Google Scholar

29. Sangro B, Melero I, Wadhawan S, Finn RS, Abou-Alfa GK, Cheng AL, et al. Association of Inflammatory Biomarkers With Clinical Outcomes in Nivolumab-Treated Patients With Advanced Hepatocellular Carcinoma. J Hepatol (2020) 73(6):1460–9. doi: 10.1016/j.jhep.2020.07.026

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Hansen AR, Siu LL. PD-L1 Testing in Cancer: Challenges in Companion Diagnostic Development. JAMA Oncol (2016) 2(1):15–6. doi: 10.1001/jamaoncol.2015.4685

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-Driven Biomarkers to Guide Immune Checkpoint Blockade in Cancer Therapy. Nat Rev Cancer (2016) 16(5):275–87. doi: 10.1038/nrc.2016.36

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Vilain RE, Menzies AM, Wilmott JS, Kakavand H, Madore J, Guminski A, et al. Dynamic Changes in PD-L1 Expression and Immune Infiltrates Early During Treatment Predict Response to PD-1 Blockade in Melanoma. Clin Cancer Research: an Off J Am Assoc Cancer Res (2017) 23(17):5024–33. doi: 10.1158/1078-0432.Ccr-16-0698

CrossRef Full Text | Google Scholar

33. Abiko K, Matsumura N, Hamanishi J, Horikawa N, Murakami R, Yamaguchi K, et al. IFN-γ From Lymphocytes Induces PD-L1 Expression and Promotes Progression of Ovarian Cancer. Br J Cancer (2015) 112(9):1501–9. doi: 10.1038/bjc.2015.101

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Chan LC, Li CW, Xia W, Hsu JM, Lee HH, Cha JH, et al. IL-6/JAK1 Pathway Drives PD-L1 Y112 Phosphorylation to Promote Cancer Immune Evasion. J Clin Invest (2019) 129(8):3324–38. doi: 10.1172/jci126022

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Lee HH, Wang YN, Xia W, Chen CH, Rau KM, Ye L, et al. Removal of N-Linked Glycosylation Enhances PD-L1 Detection and Predicts Anti-PD-1/PD-L1 Therapeutic Efficacy. Cancer Cell (2019) 36(2):168–78.e4. doi: 10.1016/j.ccell.2019.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Schumacher TN, Schreiber RD. Neoantigens in Cancer Immunotherapy. Sci (New York NY) (2015) 348(6230):69–74. doi: 10.1126/science.aaa4971

CrossRef Full Text | Google Scholar

37. Rayner E, van Gool IC, Palles C, Kearsey SE, Bosse T, Tomlinson I, et al. A Panoply of Errors: Polymerase Proofreading Domain Mutations in Cancer. Nat Rev Cancer (2016) 16(2):71–81. doi: 10.1038/nrc.2015.12

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Gong J, Wang C, Lee PP, Chu P, Fakih M. Response to PD-1 Blockade in Microsatellite Stable Metastatic Colorectal Cancer Harboring a POLE Mutation. J Natl Compr Cancer Netw: JNCCN (2017) 15(2):142–7. doi: 10.6004/jnccn.2017.0016

CrossRef Full Text | Google Scholar

39. Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. New Engl J Med (2017) 377(25):2500–1. doi: 10.1056/NEJMc1713444

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor Mutational Load Predicts Survival After Immunotherapy Across Multiple Cancer Types. Nat Genet (2019) 51(2):202–6. doi: 10.1038/s41588-018-0312-8

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Shrestha R, Prithviraj P, Anaka M, Bridle KR, Crawford DHG, Dhungel B, et al. Monitoring Immune Checkpoint Regulators as Predictive Biomarkers in Hepatocellular Carcinoma. Front Oncol (2018) 8:269. doi: 10.3389/fonc.2018.00269

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Xie F, Bai Y, Yang X, Long J, Mao J, Lin J, et al. Comprehensive Analysis of Tumour Mutation Burden and the Immune Microenvironment in Hepatocellular Carcinoma. Int Immunopharmacol (2020) 89(Pt A):107135. doi: 10.1016/j.intimp.2020.107135

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ang C, Klempner SJ, Ali SM, Madison R, Ross JS, Severson EA, et al. Prevalence of Established and Emerging Biomarkers of Immune Checkpoint Inhibitor Response in Advanced Hepatocellular Carcinoma. Oncotarget (2019) 10(40):4018–25. doi: 10.18632/oncotarget.26998

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Merino DM, McShane LM, Fabrizio D, Funari V, Chen SJ, White JR, et al. Establishing Guidelines to Harmonize Tumor Mutational Burden (TMB): In Silico Assessment of Variation in TMB Quantification Across Diagnostic Platforms: Phase I of the Friends of Cancer Research TMB Harmonization Project. J Immunother Cancer (2020) 8(1):e000147. doi: 10.1136/jitc-2019-000147

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Wong CN, Fessas P, Dominy K, Mauri FA, Kaneko T, Parcq PD, et al. Qualification of Tumour Mutational Burden by Targeted Next-Generation Sequencing as a Biomarker in Hepatocellular Carcinoma. Liver International: Off J Int Assoc Study Liver (2021) 41(1):192–203. doi: 10.1111/liv.14706

CrossRef Full Text | Google Scholar

46. Jiricny J. The Multifaceted Mismatch-Repair System. Nat Rev Mol Cell Biol (2006) 7(5):335–46. doi: 10.1038/nrm1907

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Baretti M, Le DT. DNA Mismatch Repair in Cancer. Pharmacol Ther (2018) 189:45–62. doi: 10.1016/j.pharmthera.2018.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Chang L, Chang M, Chang HM, Chang F. Microsatellite Instability: A Predictive Biomarker for Cancer Immunotherapy. Appl Immunohistochem Mol Morphology: AIMM (2018) 26(2):e15–21. doi: 10.1097/pai.0000000000000575

CrossRef Full Text | Google Scholar

49. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch Repair Deficiency Predicts Response of Solid Tumors to PD-1 Blockade. Sci (New York NY) (2017) 357(6349):409–13. doi: 10.1126/science.aan6733

CrossRef Full Text | Google Scholar

50. Yamamoto H, Itoh F, Fukushima H, Kaneto H, Sasaki S, Ohmura T, et al. Infrequent Widespread Microsatellite Instability in Hepatocellular Carcinomas. Int J Oncol (2000) 16(3):543–7. doi: 10.3892/ijo.16.3.543

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Goumard C, Desbois-Mouthon C, Wendum D, Calmel C, Merabtene F, Scatton O, et al. Low Levels of Microsatellite Instability at Simple Repeated Sequences Commonly Occur in Human Hepatocellular Carcinoma. Cancer Genomics Proteomics (2017) 14(5):329–39. doi: 10.21873/cgp.20043

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying Cancers Based on T-Cell Infiltration and PD-L1. Cancer Res (2015) 75(11):2139–45. doi: 10.1158/0008-5472.Can-15-0255

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S, et al. A Transcriptionally and Functionally Distinct PD-1(+) CD8(+) T Cell Pool With Predictive Potential in Non-Small-Cell Lung Cancer Treated With PD-1 Blockade. Nat Med (2018) 24(7):994–1004. doi: 10.1038/s41591-018-0057-z

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight Into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. Cancer Discov (2016) 6(8):827–37. doi: 10.1158/2159-8290.Cd-15-1545

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Duffy AG, Ulahannan SV, Makorova-Rusher O, Rahma O, Wedemeyer H, Pratt D, et al. Tremelimumab in Combination With Ablation in Patients With Advanced Hepatocellular Carcinoma. J Hepatol (2017) 66(3):545–51. doi: 10.1016/j.jhep.2016.10.029

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Kaseb AO, Vence L, Blando J, Yadav SS, Ikoma N, Pestana RC, et al. Immunologic Correlates of Pathologic Complete Response to Preoperative Immunotherapy in Hepatocellular Carcinoma. Cancer Immunol Res (2019) 7(9):1390–5. doi: 10.1158/2326-6066.Cir-18-0605

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Ng HHM, Lee RY, Goh S, Tay ISY, Lim X, Lee B, et al. Immunohistochemical Scoring of CD38 in the Tumor Microenvironment Predicts Responsiveness to Anti-PD-1/PD-L1 Immunotherapy in Hepatocellular Carcinoma. J Immunother Cancer (2020) 8(2):e000987. doi: 10.1136/jitc-2020-000987

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Huang M, He M, Guo Y, Li H, Shen S, Xie Y, et al. The Influence of Immune Heterogeneity on the Effectiveness of Immune Checkpoint Inhibitors in Multifocal Hepatocellular Carcinomas. Clin Cancer Research: an Off J Am Assoc Cancer Res (2020) 26(18):4947–57. doi: 10.1158/1078-0432.Ccr-19-3840

CrossRef Full Text | Google Scholar

59. Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, et al. Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing. Cell (2017) 169(7):1342–56.e16. doi: 10.1016/j.cell.2017.05.035

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Berger MF, Mardis ER. The Emerging Clinical Relevance of Genomics in Cancer Medicine. Nat Rev Clin Oncol (2018) 15(6):353–65. doi: 10.1038/s41571-018-0002-6

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Pinyol R, Sia D, Llovet JM. Immune Exclusion-Wnt/CTNNB1 Class Predicts Resistance to Immunotherapies in HCC. Clin Cancer Research: an Off J Am Assoc Cancer Res (2019) 25(7):2021–3. doi: 10.1158/1078-0432.Ccr-18-3778

CrossRef Full Text | Google Scholar

62. Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, et al. Identification of an Immune-Specific Class of Hepatocellular Carcinoma, Based on Molecular Features. Gastroenterology (2017) 153(3):812–26. doi: 10.1053/j.gastro.2017.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Harding JJ, Nandakumar S, Armenia J, Khalil DN, Albano M, Ly M, et al. Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies. Clin Cancer Research: an Off J Am Assoc Cancer Res (2019) 25(7):2116–26. doi: 10.1158/1078-0432.Ccr-18-2293

CrossRef Full Text | Google Scholar

64. Ruiz de Galarreta M, Bresnahan E, Molina-Sánchez P, Lindblad KE, Maier B, Sia D, et al. β-Catenin Activation Promotes Immune Escape and Resistance to Anti-PD-1 Therapy in Hepatocellular Carcinoma. Cancer Discov (2019) 9(8):1124–41. doi: 10.1158/2159-8290.Cd-19-0074

PubMed Abstract | CrossRef Full Text | Google Scholar

65. von Felden J, Craig AJ, Garcia-Lezana T, Labgaa I, Haber PK, D’Avola D, et al. Mutations in Circulating Tumor DNA Predict Primary Resistance to Systemic Therapies in Advanced Hepatocellular Carcinoma. Oncogene (2021) 40(1):140–51. doi: 10.1038/s41388-020-01519-1

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Long J, Wang A, Bai Y, Lin J, Yang X, Wang D, et al. Development and Validation of a TP53-Associated Immune Prognostic Model for Hepatocellular Carcinoma. EBioMedicine (2019) 42:363–74. doi: 10.1016/j.ebiom.2019.03.022

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Hu ZQ, Xin HY, Luo CB, Li J, Zhou ZJ, Zou JX, et al. Associations Among the Mutational Landscape, Immune Microenvironment, and Prognosis in Chinese Patients With Hepatocellular Carcinoma. Cancer Immunol Immunother: CII (2021) 70(2):377–89. doi: 10.1007/s00262-020-02685-7

CrossRef Full Text | Google Scholar

68. Cai Y, Tian Y, Wang J, Wei W, Tang Q, Lu L, et al. Identification of Driver Genes Regulating the T-Cell-Infiltrating Levels in Hepatocellular Carcinoma. Front Genet (2020) 11:560546. doi: 10.3389/fgene.2020.560546

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Bassaganyas L, Pinyol R, Esteban-Fabró R, Torrens L, Torrecilla S, Willoughby CE, et al. Copy-Number Alteration Burden Differentially Impacts Immune Profiles and Molecular Features of Hepatocellular Carcinoma. Clin Cancer Research: An Off J Am Assoc Cancer Res (2020) 26(23):6350–61. doi: 10.1158/1078-0432.Ccr-20-1497

CrossRef Full Text | Google Scholar

70. Yang H, Sun L, Guan A, Yin H, Liu M, Mao X, et al. Unique TP53 Neoantigen and the Immune Microenvironment in Long-Term Survivors of Hepatocellular Carcinoma. Cancer Immunol Immunother: CII (2021) 70(3):667–77. doi: 10.1007/s00262-020-02711-8

CrossRef Full Text | Google Scholar

71. Peng L, Chen Z, Chen Y, Wang X, Tang N. MIR155HG is a Prognostic Biomarker and Associated With Immune Infiltration and Immune Checkpoint Molecules Expression in Multiple Cancers. Cancer Med (2019) 8(17):7161–73. doi: 10.1002/cam4.2583

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Zhang Y, Zhang L, Xu Y, Wu X, Zhou Y, Mo J. Immune-Related Long Noncoding RNA Signature for Predicting Survival and Immune Checkpoint Blockade in Hepatocellular Carcinoma. J Cell Physiol (2020) 235(12):9304–16. doi: 10.1002/jcp.29730

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Yuan M, Wang Y, Sun Q, Liu S, Xian S, Dai F, et al. Identification of a Nine Immune-Related lncRNA Signature as a Novel Diagnostic Biomarker for Hepatocellular Carcinoma. BioMed Res Int (2021) 2021:9798231. doi: 10.1155/2021/9798231

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Xu Q, Wang Y, Huang W. Identification of Immune-Related lncRNA Signature for Predicting Immune Checkpoint Blockade and Prognosis in Hepatocellular Carcinoma. Int Immunopharmacol (2021) 92:107333. doi: 10.1016/j.intimp.2020.107333

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Xiao S, Huang S, Yang J. Overexpression of GIHCG is Associated With a Poor Prognosis and Immune Infiltration in Hepatocellular Carcinoma. OncoTargets Ther (2020) 13:11607–19. doi: 10.2147/ott.S271966

CrossRef Full Text | Google Scholar

76. Ye Y, Guo J, Xiao P, Ning J, Zhang R, Liu P, et al. Macrophages-Induced Long Noncoding RNA H19 Up-Regulation Triggers and Activates the miR-193b/MAPK1 Axis and Promotes Cell Aggressiveness in Hepatocellular Carcinoma. Cancer Lett (2020) 469:310–22. doi: 10.1016/j.canlet.2019.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Huang XY, Zhang PF, Wei CY, Peng R, Lu JC, Gao C, et al. Circular RNA circMET Drives Immunosuppression and Anti-PD1 Therapy Resistance in Hepatocellular Carcinoma via the miR-30-5p/Snail/DPP4 Axis. Mol Cancer (2020) 19(1):92. doi: 10.1186/s12943-020-01213-6

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Li Y, He X, Zhang X, Xu Y, Wu Y, Xu X. Immune-Related microRNA Signature for Predicting Prognosis and the Immune Microenvironment in Hepatocellular Carcinoma. Life Sci (2021) 265:118799. doi: 10.1016/j.lfs.2020.118799

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Mo Z, Cao Z, Luo S, Chen Y, Zhang S. Novel Molecular Subtypes Associated With 5mc Methylation and Their Role in Hepatocellular Carcinoma Immunotherapy. Front Mol Biosci (2020) 7:562441. doi: 10.3389/fmolb.2020.562441

PubMed Abstract | CrossRef Full Text | Google Scholar

80. De Carvalho DD, Sharma S, You JS, Su SF, Taberlay PC, Kelly TK, et al. DNA Methylation Screening Identifies Driver Epigenetic Events of Cancer Cell Survival. Cancer Cell (2012) 21(5):655–67. doi: 10.1016/j.ccr.2012.03.045

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Yoshimi A, Lin KT, Wiseman DH, Rahman MA, Pastore A, Wang B, et al. Coordinated Alterations in RNA Splicing and Epigenetic Regulation Drive Leukaemogenesis. Nature (2019) 574(7777):273–7. doi: 10.1038/s41586-019-1618-0

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Orouji E, Federico A, Larribère L, Novak D, Lipka DB, Assenov Y, et al. Histone Methyltransferase SETDB1 Contributes to Melanoma Tumorigenesis and Serves as a New Potential Therapeutic Target. Int J Cancer (2019) 145(12):3462–77. doi: 10.1002/ijc.32432

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Giacopelli B, Zhao Q, Ruppert AS, Agyeman A, Weigel C, Wu YZ, et al. Developmental Subtypes Assessed by DNA methylation-iPLEX Forecast the Natural History of Chronic Lymphocytic Leukemia. Blood (2019) 134(8):688–98. doi: 10.1182/blood.2019000490

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Brennan K, Shin JH, Tay JK, Prunello M, Gentles AJ, Sunwoo JB, et al. NSD1 Inactivation Defines an Immune Cold, DNA Hypomethylated Subtype in Squamous Cell Carcinoma. Sci Rep (2017) 7(1):17064. doi: 10.1038/s41598-017-17298-x

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Lingohr P, Dohmen J, Semaan A, Branchi V, Dietrich J, Bootz F, et al. Clinicopathological, Immune and Molecular Correlates of PD-L2 Methylation in Gastric Adenocarcinomas. Epigenomics (2019) 11(6):639–53. doi: 10.2217/epi-2018-0149

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Liu L, Toung JM, Jassowicz AF, Vijayaraghavan R, Kang H, Zhang R, et al. Targeted Methylation Sequencing of Plasma Cell-Free DNA for Cancer Detection and Classification. Ann Oncol: Off J Eur Soc Med Oncol (2018) 29(6):1445–53. doi: 10.1093/annonc/mdy119

CrossRef Full Text | Google Scholar

87. Fröhlich A, Loick S, Bawden EG, Fietz S, Dietrich J, Diekmann E, et al. Comprehensive Analysis of Tumor Necrosis Factor Receptor TNFRSF9 (4-1BB) DNA Methylation With Regard to Molecular and Clinicopathological Features, Immune Infiltrates, and Response Prediction to Immunotherapy in Melanoma. EBioMedicine (2020) 52:102647. doi: 10.1016/j.ebiom.2020.102647

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Duruisseaux M, Martínez-Cardús A, Calleja-Cervantes ME, Moran S, Castro de Moura M, Davalos V, et al. Epigenetic Prediction of Response to Anti-PD-1 Treatment in Non-Small-Cell Lung Cancer: A Multicentre, Retrospective Analysis. Lancet Respir Med (2018) 6(10):771–81. doi: 10.1016/s2213-2600(18)30284-4

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Zhang Y, Petropoulos S, Liu J, Cheishvili D, Zhou R, Dymov S, et al. The Signature of Liver Cancer in Immune Cells DNA Methylation. Clin Epigenet (2018) 10:8. doi: 10.1186/s13148-017-0436-1

CrossRef Full Text | Google Scholar

90. Wang Y, Yang Y, Gao H, Ouyang T, Zhang L, Hu J, et al. Comprehensive Analysis of CDCAs Methylation and Immune Infiltrates in Hepatocellular Carcinoma. Front Oncol (2020) 10:566183. doi: 10.3389/fonc.2020.566183

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Llopiz D, Ruiz M, Villanueva L, Iglesias T, Silva L, Egea J, et al. Enhanced Anti-Tumor Efficacy of Checkpoint Inhibitors in Combination With the Histone Deacetylase Inhibitor Belinostat in a Murine Hepatocellular Carcinoma Model. Cancer Immunol Immunother: CII (2019) 68(3):379–93. doi: 10.1007/s00262-018-2283-0

CrossRef Full Text | Google Scholar

92. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-γ-Related mRNA Profile Predicts Clinical Response to PD-1 Blockade. J Clin Invest (2017) 127(8):2930–40. doi: 10.1172/jci91190

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Gu X, Guan J, Xu J, Zheng Q, Chen C, Yang Q, et al. Model Based on Five Tumour Immune Microenvironment-Related Genes for Predicting Hepatocellular Carcinoma Immunotherapy Outcomes. J Trans Med (2021) 19(1):26. doi: 10.1186/s12967-020-02691-4

CrossRef Full Text | Google Scholar

94. Peng Y, Liu C, Li M, Li W, Zhang M, Jiang X, et al. Identification of a Prognostic and Therapeutic Immune Signature Associated With Hepatocellular Carcinoma. Cancer Cell Int (2021) 21(1):98. doi: 10.1186/s12935-021-01792-4

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Xu Y, Wang Z, Li F. Survival Prediction and Response to Immune Checkpoint Inhibitors: A Prognostic Immune Signature for Hepatocellular Carcinoma. Trans Oncol (2021) 14(1):100957. doi: 10.1016/j.tranon.2020.100957

CrossRef Full Text | Google Scholar

96. Batlle E, Massagué J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity (2019) 50(4):924–40. doi: 10.1016/j.immuni.2019.03.024

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Ringelhan M, Pfister D, O’Connor T, Pikarsky E, Heikenwalder M. The Immunology of Hepatocellular Carcinoma. Nat Immunol (2018) 19(3):222–32. doi: 10.1038/s41590-018-0044-z

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Park BV, Freeman ZT, Ghasemzadeh A, Chattergoon MA, Rutebemberwa A, Steigner J, et al. TGFβ1-Mediated SMAD3 Enhances PD-1 Expression on Antigen-Specific T Cells in Cancer. Cancer Discov (2016) 6(12):1366–81. doi: 10.1158/2159-8290.Cd-15-1347

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Chen J, Zaidi S, Rao S, Chen JS, Phan L, Farci P, et al. Analysis of Genomes and Transcriptomes of Hepatocellular Carcinomas Identifies Mutations and Gene Expression Changes in the Transforming Growth Factor-β Pathway. Gastroenterology (2018) 154(1):195–210. doi: 10.1053/j.gastro.2017.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Chen J, Gingold JA, Su X. Immunomodulatory TGF-β Signaling in Hepatocellular Carcinoma. Trends Mol Med (2019) 25(11):1010–23. doi: 10.1016/j.molmed.2019.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ Attenuates Tumour Response to PD-L1 Blockade by Contributing to Exclusion of T Cells. Nature (2018) 554(7693):544–8. doi: 10.1038/nature25501

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Kelley RK, Gane E, Assenat E, Siebler J, Galle PR, Merle P, et al. A Phase 2 Study of Galunisertib (TGF-β1 Receptor Type I Inhibitor) and Sorafenib in Patients With Advanced Hepatocellular Carcinoma. Clin Trans Gastroenterol (2019) 10(7):e00056. doi: 10.14309/ctg.0000000000000056

CrossRef Full Text | Google Scholar

103. Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, et al. Exosomal PD-L1 Contributes to Immunosuppression and is Associated With Anti-PD-1 Response. Nature (2018) 560(7718):382–6. doi: 10.1038/s41586-018-0392-8

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-Tumor Immunity and Memory. Cell (2019) 177(2):414–27.e13. doi: 10.1016/j.cell.2019.02.016

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Mazzaschi G, Minari R, Zecca A, Cavazzoni A, Ferri V, Mori C, et al. Soluble PD-L1 and Circulating CD8+PD-1+ and NK Cells Enclose a Prognostic and Predictive Immune Effector Score in Immunotherapy Treated NSCLC Patients. Lung Cancer (Amsterdam Netherlands) (2020) 148:1–11. doi: 10.1016/j.lungcan.2020.07.028

CrossRef Full Text | Google Scholar

106. Chang B, Huang T, Wei H, Shen L, Zhu D, He W, et al. The Correlation and Prognostic Value of Serum Levels of Soluble Programmed Death Protein 1 (sPD-1) and Soluble Programmed Death-Ligand 1 (sPD-L1) in Patients With Hepatocellular Carcinoma. Cancer Immunol Immunother: CII (2019) 68(3):353–63. doi: 10.1007/s00262-018-2271-4

CrossRef Full Text | Google Scholar

107. Finkelmeier F, Canli Ö, Tal A, Pleli T, Trojan J, Schmidt M, et al. High Levels of the Soluble Programmed Death-Ligand (sPD-L1) Identify Hepatocellular Carcinoma Patients With a Poor Prognosis. Eur J Cancer (Oxford England: 1990) (2016) 59:152–9. doi: 10.1016/j.ejca.2016.03.002

CrossRef Full Text | Google Scholar

108. Han X, Gu YK, Li SL, Chen H, Chen MS, Cai QQ, et al. Pre-Treatment Serum Levels of Soluble Programmed Cell Death-Ligand 1 Predict Prognosis in Patients With Hepatitis B-Related Hepatocellular Carcinoma. J Cancer Res Clin Oncol (2019) 145(2):303–12. doi: 10.1007/s00432-018-2758-6

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, et al. T-Cell Invigoration to Tumour Burden Ratio Associated With Anti-PD-1 Response. Nature (2017) 545(7652):60–5. doi: 10.1038/nature22079

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Agdashian D, ElGindi M, Xie C, Sandhu M, Pratt D, Kleiner DE, et al. The Effect of Anti-CTLA4 Treatment on Peripheral and Intra-Tumoral T Cells in Patients With Hepatocellular Carcinoma. Cancer Immunol Immunother: CII (2019) 68(4):599–608. doi: 10.1007/s00262-019-02299-8

CrossRef Full Text | Google Scholar

111. Hung YP, Shao YY, Lee JM, Hsu C, Hsu CH, Yang MH, et al. Potential of Circulating Immune Cells as Biomarkers of Nivolumab Treatment Efficacy for Advanced Hepatocellular Carcinoma. J Chin Med Association: JCMA (2021) 84(2):144–50. doi: 10.1097/jcma.0000000000000477

CrossRef Full Text | Google Scholar

112. Dharmapuri S, Özbek U, Lin JY, Sung M, Schwartz M, Branch AD, et al. Predictive Value of Neutrophil to Lymphocyte Ratio and Platelet to Lymphocyte Ratio in Advanced Hepatocellular Carcinoma Patients Treated With Anti-PD-1 Therapy. Cancer Med (2020) 9(14):4962–70. doi: 10.1002/cam4.3135

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Lipson EJ, Velculescu VE, Pritchard TS, Sausen M, Pardoll DM, Topalian SL, et al. Circulating Tumor DNA Analysis as a Real-Time Method for Monitoring Tumor Burden in Melanoma Patients Undergoing Treatment With Immune Checkpoint Blockade. J Immunother Cancer (2014) 2(1):42. doi: 10.1186/s40425-014-0042-0

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Guibert N, Mazieres J, Delaunay M, Casanova A, Farella M, Keller L, et al. Monitoring of KRAS-Mutated ctDNA to Discriminate Pseudo-Progression From True Progression During Anti-PD-1 Treatment of Lung Adenocarcinoma. Oncotarget (2017) 8(23):38056–60. doi: 10.18632/oncotarget.16935

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Lee JH, Long GV, Menzies AM, Lo S, Guminski A, Whitbourne K, et al. Association Between Circulating Tumor DNA and Pseudoprogression in Patients With Metastatic Melanoma Treated With Anti-Programmed Cell Death 1 Antibodies. JAMA Oncol (2018) 4(5):717–21. doi: 10.1001/jamaoncol.2017.5332

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Gandara DR, Paul SM, Kowanetz M, Schleifman E, Zou W, Li Y, et al. Blood-Based Tumor Mutational Burden as a Predictor of Clinical Benefit in Non-Small-Cell Lung Cancer Patients Treated With Atezolizumab. Nat Med (2018) 24(9):1441–8. doi: 10.1038/s41591-018-0134-3

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Wang Z, Duan J, Cai S, Han M, Dong H, Zhao J, et al. Assessment of Blood Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Patients With Non-Small Cell Lung Cancer With Use of a Next-Generation Sequencing Cancer Gene Panel. JAMA Oncol (2019) 5(5):696–702. doi: 10.1001/jamaoncol.2018.7098

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Winograd P, Hou S, Court CM, Lee YT, Chen PJ, Zhu Y, et al. Hepatocellular Carcinoma-Circulating Tumor Cells Expressing PD-L1 Are Prognostic and Potentially Associated With Response to Checkpoint Inhibitors. Hepatol Commun (2020) 4(10):1527–40. doi: 10.1002/hep4.1577

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Shao YY, Liu TH, Hsu C, Lu LC, Shen YC, Lin ZZ, et al. Early Alpha-Foetoprotein Response Associated With Treatment Efficacy of Immune Checkpoint Inhibitors for Advanced Hepatocellular Carcinoma. Liver International: Off J Int Assoc Study Liver (2019) 39(11):2184–9. doi: 10.1111/liv.14210

CrossRef Full Text | Google Scholar

120. Lee PC, Chao Y, Chen MH, Lan KH, Lee CJ, Lee IC, et al. Predictors of Response and Survival in Immune Checkpoint Inhibitor-Treated Unresectable Hepatocellular Carcinoma. Cancers (2020) 12(1):182. doi: 10.3390/cancers12010182

CrossRef Full Text | Google Scholar

121. Spahn S, Roessler D, Pompilia R, Gabernet G, Gladstone BP, Horger M, et al. Clinical and Genetic Tumor Characteristics of Responding and Non-Responding Patients to PD-1 Inhibition in Hepatocellular Carcinoma. Cancers (2020) 12(12):3830. doi: 10.3390/cancers12123830

CrossRef Full Text | Google Scholar

122. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The Commensal Microbiome is Associated With Anti-PD-1 Efficacy in Metastatic Melanoma Patients. Sci (New York NY) (2018) 359(6371):104–8. doi: 10.1126/science.aao3290

CrossRef Full Text | Google Scholar

123. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut Microbiome Influences Efficacy of PD-1-Based Immunotherapy Against Epithelial Tumors. Sci (New York NY) (2018) 359(6371):91–7. doi: 10.1126/science.aan3706

CrossRef Full Text | Google Scholar

124. Pasolli E, Truong DT, Malik F, Waldron L, Segata N. Machine Learning Meta-Analysis of Large Metagenomic Datasets: Tools and Biological Insights. PloS Comput Biol (2016) 12(7):e1004977. doi: 10.1371/journal.pcbi.1004977

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Yu LX, Schwabe RF. The Gut Microbiome and Liver Cancer: Mechanisms and Clinical Translation. Nat Rev Gastroenterol Hepatol (2017) 14(9):527–39. doi: 10.1038/nrgastro.2017.72

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell (2012) 21(4):504–16. doi: 10.1016/j.ccr.2012.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Loo TM, Kamachi F, Watanabe Y, Yoshimoto S, Kanda H, Arai Y, et al. Gut Microbiota Promotes Obesity-Associated Liver Cancer Through PGE(2)-Mediated Suppression of Antitumor Immunity. Cancer Discov (2017) 7(5):522–38. doi: 10.1158/2159-8290.Cd-16-0932

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, et al. Gut Microbiome-Mediated Bile Acid Metabolism Regulates Liver Cancer via NKT Cells. Sci (New York NY) (2018) 360(6391):eaan5931. doi: 10.1126/science.aan5931

CrossRef Full Text | Google Scholar

129. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites Produced by Commensal Bacteria Promote Peripheral Regulatory T-Cell Generation. Nature (2013) 504(7480):451–5. doi: 10.1038/nature12726

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, et al. The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide. Sci (New York NY) (2013) 342(6161):971–6. doi: 10.1126/science.1240537

CrossRef Full Text | Google Scholar

131. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium Promotes Antitumor Immunity and Facilitates Anti-PD-L1 Efficacy. Sci (New York NY) (2015) 350(6264):1084–9. doi: 10.1126/science.aac4255

CrossRef Full Text | Google Scholar

132. Zheng Y, Wang T, Tu X, Huang Y, Zhang H, Tan D, et al. Gut Microbiome Affects the Response to Anti-PD-1 Immunotherapy in Patients With Hepatocellular Carcinoma. J Immunother Cancer (2019) 7(1):193. doi: 10.1186/s40425-019-0650-9

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Rosshart SP, Vassallo BG, Angeletti D, Hutchinson DS, Morgan AP, Takeda K, et al. Wild Mouse Gut Microbiota Promotes Host Fitness and Improves Disease Resistance. Cell (2017) 171(5):1015–28.e13. doi: 10.1016/j.cell.2017.09.016

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell (2018) 33(4):570–80. doi: 10.1016/j.ccell.2018.03.015

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 Blockade Induces Responses by Inhibiting Adaptive Immune Resistance. Nature (2014) 515(7528):568–71. doi: 10.1038/nature13954

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Inoue H, Park JH, Kiyotani K, Zewde M, Miyashita A, Jinnin M, et al. Intratumoral Expression Levels of PD-L1, GZMA, and HLA-A Along With Oligoclonal T Cell Expansion Associate With Response to Nivolumab in Metastatic Melanoma. Oncoimmunology (2016) 5(9):e1204507. doi: 10.1080/2162402x.2016.1204507

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Postow MA, Manuel M, Wong P, Yuan J, Dong Z, Liu C, et al. Peripheral T Cell Receptor Diversity is Associated With Clinical Outcomes Following Ipilimumab Treatment in Metastatic Melanoma. J Immunother Cancer (2015) 3:23. doi: 10.1186/s40425-015-0070-4

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Kobayashi E, Mizukoshi E, Kishi H, Ozawa T, Hamana H, Nagai T, et al. A New Cloning and Expression System Yields and Validates TCRs From Blood Lymphocytes of Patients With Cancer Within 10 Days. Nat Med (2013) 19(11):1542–6. doi: 10.1038/nm.3358

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Jokinen E, Huuhtanen J, Mustjoki S, Heinonen M, Lähdesmäki H. Predicting Recognition Between T Cell Receptors and Epitopes With TCRGP. PloS Comput Biol (2021) 17(3):e1008814. doi: 10.1371/journal.pcbi.1008814

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Lin KR, Deng FW, Jin YB, Chen XP, Pan YM, Cui JH, et al. T Cell Receptor Repertoire Profiling Predicts the Prognosis of HBV-Associated Hepatocellular Carcinoma. Cancer Med (2018) 7(8):3755–62. doi: 10.1002/cam4.1610

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Han Y, Liu X, Wang Y, Wu X, Guan Y, Li H, et al. Identification of Characteristic TRB V Usage in HBV-Associated HCC by Using Differential Expression Profiling Analysis. Oncoimmunology (2015) 4(8):e1021537. doi: 10.1080/2162402x.2015.1021537

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Pfister D, Núñez NG, Pinyol R, Govaere O, Pinter M, Szydlowska M, et al. NASH Limits Anti-Tumour Surveillance in Immunotherapy-Treated HCC. Nature (2021) 592(7854):450–6. doi: 10.1038/s41586-021-03362-0

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Terry S, Savagner P, Ortiz-Cuaran S, Mahjoubi L, Saintigny P, Thiery JP, et al. New Insights Into the Role of EMT in Tumor Immune Escape. Mol Oncol (2017) 11(7):824–46. doi: 10.1002/1878-0261.12093

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Liu F, Qin L, Liao Z, Song J, Yuan C, Liu Y, et al. Microenvironment Characterization and Multi-Omics Signatures Related to Prognosis and Immunotherapy Response of Hepatocellular Carcinoma. Exp Hematol Oncol (2020) 9:10. doi: 10.1186/s40164-020-00165-3

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Ding H, Hu H, Tian F, Liang H. A Dual Immune Signature of CD8+ T Cells and MMP9 Improves the Survival of Patients With Hepatocellular Carcinoma. Bioscience Rep (2021) 41(3):BSR20204219. doi: 10.1042/bsr20204219

CrossRef Full Text | Google Scholar

146. Oh S, Park Y, Lee HJ, Lee J, Lee SH, Baek YS, et al. A Disintegrin and Metalloproteinase 9 (ADAM9) in Advanced Hepatocellular Carcinoma and Their Role as a Biomarker During Hepatocellular Carcinoma Immunotherapy. Cancers (2020) 12(3):745. doi: 10.3390/cancers12030745

CrossRef Full Text | Google Scholar

147. Qayyum A, Hwang KP, Stafford J, Verma A, Maru DM, Sandesh S, et al. Immunotherapy Response Evaluation With Magnetic Resonance Elastography (MRE) in Advanced HCC. J Immunother Cancer (2019) 7(1):329. doi: 10.1186/s40425-019-0766-y

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Nakamura Y. Biomarkers for Immune Checkpoint Inhibitor-Mediated Tumor Response and Adverse Events. Front Med (2019) 6:119. doi: 10.3389/fmed.2019.00119

CrossRef Full Text | Google Scholar

149. Valpione S, Pasquali S, Campana LG, Piccin L, Mocellin S, Pigozzo J, et al. Sex and Interleukin-6 are Prognostic Factors for Autoimmune Toxicity Following Treatment With Anti-CTLA4 Blockade. J Trans Med (2018) 16(1):94. doi: 10.1186/s12967-018-1467-x

CrossRef Full Text | Google Scholar

150. Tarhini AA, Zahoor H, Lin Y, Malhotra U, Sander C, Butterfield LH, et al. Baseline Circulating IL-17 Predicts Toxicity While TGF-β1 and IL-10 are Prognostic of Relapse in Ipilimumab Neoadjuvant Therapy of Melanoma. J Immunother Cancer (2015) 3:39. doi: 10.1186/s40425-015-0081-1

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Diehl A, Yarchoan M, Hopkins A, Jaffee E, Grossman SA. Relationships Between Lymphocyte Counts and Treatment-Related Toxicities and Clinical Responses in Patients With Solid Tumors Treated With PD-1 Checkpoint Inhibitors. Oncotarget (2017) 8(69):114268–80. doi: 10.18632/oncotarget.23217

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Kimbara S, Fujiwara Y, Iwama S, Ohashi K, Kuchiba A, Arima H, et al. Association of Antithyroglobulin Antibodies With the Development of Thyroid Dysfunction Induced by Nivolumab. Cancer Sci (2018) 109(11):3583–90. doi: 10.1111/cas.13800

PubMed Abstract | CrossRef Full Text | Google Scholar

153. Stamatouli AM, Quandt Z, Perdigoto AL, Clark PL, Kluger H, Weiss SA, et al. Collateral Damage: Insulin-Dependent Diabetes Induced With Checkpoint Inhibitors. Diabetes (2018) 67(8):1471–80. doi: 10.2337/dbi18-0002

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline Gut Microbiota Predicts Clinical Response and Colitis in Metastatic Melanoma Patients Treated With Ipilimumab. Ann Oncol: Off J Eur Soc Med Oncol (2017) 28(6):1368–79. doi: 10.1093/annonc/mdx108

CrossRef Full Text | Google Scholar

155. Rogado J, Sánchez-Torres JM, Romero-Laorden N, Ballesteros AI, Pacheco-Barcia V, Ramos-Leví A, et al. Immune-Related Adverse Events Predict the Therapeutic Efficacy of Anti-PD-1 Antibodies in Cancer Patients. Eur J Cancer (Oxford England: 1990) (2019) 109:21–7. doi: 10.1016/j.ejca.2018.10.014

CrossRef Full Text | Google Scholar

156. Freeman-Keller M, Kim Y, Cronin H, Richards A, Gibney G, Weber JS. Nivolumab in Resected and Unresectable Metastatic Melanoma: Characteristics of Immune-Related Adverse Events and Association With Outcomes. Clin Cancer Research: an Off J Am Assoc Cancer Res (2016) 22(4):886–94. doi: 10.1158/1078-0432.Ccr-15-1136

CrossRef Full Text | Google Scholar

157. Teraoka S, Fujimoto D, Morimoto T, Kawachi H, Ito M, Sato Y, et al. Early Immune-Related Adverse Events and Association With Outcome in Advanced Non-Small Cell Lung Cancer Patients Treated With Nivolumab: A Prospective Cohort Study. J Thoracic Oncol: Off Publ Int Assoc Study Lung Cancer (2017) 12(12):1798–805. doi: 10.1016/j.jtho.2017.08.022

CrossRef Full Text | Google Scholar

Keywords: hepatocellular carcinoma, biomarkers, immune checkpoint inhibitors, immune-related adverse events, immunotherapy

Citation: He Y, Lu M, Che J, Chu Q, Zhang P and Chen Y (2021) Biomarkers and Future Perspectives for Hepatocellular Carcinoma Immunotherapy. Front. Oncol. 11:716844. doi: 10.3389/fonc.2021.716844

Received: 29 May 2021; Accepted: 18 August 2021;
Published: 06 September 2021.

Edited by:

Heng-Hong Li, Georgetown University, United States

Reviewed by:

Alessandro Rizzo, Sant’Orsola-Malpighi Polyclinic, Italy
Fengbiao Mao, University of Michigan, United States

Copyright © 2021 He, Lu, Che, Chu, Zhang and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Peng Zhang, pengzhang@tjh.tjmu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.