Next Article in Journal
Proteomic Profiling of Mouse Brain Pyruvate Kinase Binding Proteins: A Hint for Moonlighting Functions of PKM1?
Next Article in Special Issue
Synergistic Nanomedicine: Photodynamic, Photothermal and Photoimmune Therapy in Hepatocellular Carcinoma: Fulfilling the Myth of Prometheus?
Previous Article in Journal
Expression of the Populus Orthologues of AtYY1, YIN and YANG Activates the Floral Identity Genes AGAMOUS and SEPALLATA3 Accelerating Floral Transition in Arabidopsis thaliana
Previous Article in Special Issue
Navigating through the PD-1/PDL-1 Landscape: A Systematic Review and Meta-Analysis of Clinical Outcomes in Hepatocellular Carcinoma and Their Influence on Immunotherapy and Tumor Microenvironment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Predictive Biomarkers for Immune-Checkpoint Inhibitor Treatment Response in Patients with Hepatocellular Carcinoma

1
Division of Hematology and Oncology, Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
2
Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
3
Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea
4
Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
5
Division of Medical Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
6
Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
7
Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
8
Weil Cornell Medicine, Cornell University, New York, NY 14853, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(8), 7640; https://doi.org/10.3390/ijms24087640
Submission received: 22 March 2023 / Revised: 15 April 2023 / Accepted: 18 April 2023 / Published: 21 April 2023

Abstract

:
Hepatocellular carcinoma (HCC) has one of the highest mortality rates among solid cancers. Late diagnosis and a lack of efficacious treatment options contribute to the dismal prognosis of HCC. Immune checkpoint inhibitor (ICI)-based immunotherapy has presented a new milestone in the treatment of cancer. Immunotherapy has yielded remarkable treatment responses in a range of cancer types including HCC. Based on the therapeutic effect of ICI alone (programmed cell death (PD)-1/programmed death-ligand1 (PD-L)1 antibody), investigators have developed combined ICI therapies including ICI + ICI, ICI + tyrosine kinase inhibitor (TKI), and ICI + locoregional treatment or novel immunotherapy. Although these regimens have demonstrated increasing treatment efficacy with the addition of novel drugs, the development of biomarkers to predict toxicity and treatment response in patients receiving ICI is in urgent need. PD-L1 expression in tumor cells received the most attention in early studies among various predictive biomarkers. However, PD-L1 expression alone has limited utility as a predictive biomarker in HCC. Accordingly, subsequent studies have evaluated the utility of tumor mutational burden (TMB), gene signatures, and multiplex immunohistochemistry (IHC) as predictive biomarkers. In this review, we aim to discuss the current state of immunotherapy for HCC, the results of the predictive biomarker studies, and future direction.

1. Introduction

Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the third leading cause of cancer death worldwide, with 906,000 new cases and 830,000 HCC-related deaths annually [1]. Repeated necrosis and regeneration of hepatocytes caused by chronic inflammation and injury gradually progress to liver fibrosis and cirrhosis, eventually leading to HCC. Unfortunately, most patients with liver cirrhosis are asymptomatic, so HCC is often diagnosed at advanced stages. Accordingly, the mortality of HCC is high and continues to rise [2]. Patients with advanced-stage HCC may benefit from systemic therapy and, until recently, most systemic treatments for HCC comprised targeted therapy with tyrosine kinase inhibitors (TKI) such as sorafenib, lenvatinib, regorafenib, and cabozantinib [3].
The immune system can recognize foreign cells based on the proteins present on the cell surface and has the ability to eliminate different from our own body such as viruses, bacteria, and malignancies. In this process, checkpoint proteins are to limit autoimmune damage to normal tissue by preventing T-cell activation. HCC and other tumors use this mechanism to evade immune responses by expressing ligands on the tumor cell surface. In addition, tumor cells promote immune evasion by interfering with the recognition of tumor antigen presentation or generating the immunosuppressive tumor microenvironment [4]. Immune checkpoint inhibitors block the interaction between checkpoint proteins and their ligands, thereby preventing the inactivation of T cell function. Chronic viral infections such as hepatitis B and hepatitis C, which are the main causes of HCC, promote chronic inflammation of the liver, and in patients with chronic inflammatory liver disease, PD-1 overexpression in lymphocyte and PD-L1/PD-L2 overexpression in stromal cells (Kupffer cell, liver sinusoidal endothelial cells) are observed. This upregulation of checkpoint proteins suggests that ICIs may be effective in HCC. The use of immunotherapy alone or in combination with targeted agents results in improved survival with a durable response and has become the new standard of care with several successful phase 3 studies published since 2020 [5,6,7]. However, despite these inspiring results, since HCC is a heterogeneous disease with diverse immunological characteristics, immunotherapy does not guarantee a clinical benefit in all patients with HCC, and more than two-thirds of advanced cancer patients do not respond to immunotherapy [8,9]. Moreover, immunotherapy improved long-term survival in cancer treatment, but considering the trend of crossing the survival curve in randomized clinical trials for ICIs, it suggests that the early mortality rate is rather higher in the ICI treatment group compared to the control group [10]. To overcome these limitations and optimize the use of ICIs in HCC, the development of predictive biomarkers that can be used to identify individuals who are more likely to experience favorable or unfavorable effects of immunotherapy has become increasingly important [11,12]. For biomarker discovery, testing of blood or feces can be used to obtain complex data using advanced technologies such as genomics, proteomics, metabolomics, and artificial intelligence. Unfortunately, the need for the development of clinically applicable predictive biomarkers remains unmet despite efforts to identify biomarkers predicting outcomes of immunotherapy for HCC.
Herein, we aim to discuss predictive biomarkers for immunotherapy response in HCC, with a focus on clinical applications (Figure 1.)

2. Biomarkers Related to the Interaction between T Cells and Tumor Cells

2.1. DNA Damage Repair Pathway (dMMR/MSI-H)

DNA damage repair (DDR) genes play key roles in maintaining the stability of the human genome, and disruption of DNA damage repair pathways by germline or somatic mutations in DDR genes may contribute to the development of cancer [13,14]. The major DNA repair mechanisms are mismatch repair (MMR), homologous recombination (HR), polymerase proofreading (POLE/POLD1), base excision repair (BER), nucleotide excision repair, and DNA repair mediated by MGMT (O6-methylguanine–DNA methyltransferase) [15]. Disruptions to BER, HR, and MMR contribute more significantly to tumor mutational burden (TMB) or neoantigens, which have the highest levels when co-mutated. [16] In HCC, these DDR alterations showed distinct characteristics. Peng Lin et al. [17] categorized DDR alterations in HCC into two heterogeneous subtypes (the DDR-activated subtype and the DDR-suppressed subtype) and compared prognosis and clinicopathologic features between subtypes. HCC patients with the DDR-suppressed subtype tended to have longer survival. Significant activation of CD4+ T cells, central memory CD4+ T Cells, and effector memory CD4+ T cells was observed in HCC patients with the DDR-activated subtype. The authors also evaluated the relationships between DDR signatures and immunotherapy responses in an external cohort, with a high DDR subtype score observed in patients with complete response (CR) and partial response (PR) [18]. In a separate Chinese study, [19] DDR pathway/gene mutations were evaluated using next-generation sequencing (NGS) analysis in 1427 HCC patients who had undergone surgery. Among the included patients, 18.8% carried somatic mutations in DDR genes and 3.5% carried germline mutations in DDR genes. TMB was significantly higher in HCC patients who carried somatic mutations in DDR genes, while no difference in TMB was observed in HCC patients with DDR germline mutations. The results of this study indicate that somatic mutations of DDR genes contribute to high TMB, suggesting a greater response to immunotherapy in patients with DDR gene mutations. In addition, each DDR mechanism has been studied for ICI response prediction, and in particular, the MMR pathway is one of the most well-known biomarkers for cancer immunotherapy. DNA MMR maintains genomic stability by repairing mismatched bases or mispairs during DNA replication and recombination, thereby preventing genetic alteration [20]. Genetic mutations involving the DNA MMR pathway are associated with increased microsatellite instability (MSI), high numbers of somatic mutations, and a higher number of tumor-infiltrating lymphocytes. This process promotes an anti-tumor cytotoxic immune response and secretion of soluble factors that contribute to the activation of the programmed death ligand-1 (PD-L1) pathway within the tumor microenvironment [21,22]. dMMR is also associated with DNA polymerase gene epsilon/delta1 (POLE/POLD1) mutations, with increased mutation, and neoantigen load. In an analysis of 47,721 patients with solid cancers, POLE/POLD1 mutations were associated with TMB-H. dMMR, TMB-H, and high neoantigen loads are thought to be associated with immunotherapy response [23]. The dramatic response of ICI shown by patients in the dMMR/MSI-H group in other tumors led to expectations that those of dMMR/MSI-H in HCC would play a similar role, but contrary to expectation, dMMR/MSI-H in HCC is limited in its use as a predictive biomarker for immunotherapy due to their low prevalence of less of than 3% [24,25,26,27]. MSI status is considered a potential biomarker for response to immunotherapy. In 2015, the FDA granted accelerated approval for the use of pembrolizumab for unresectable solid tumors with MSI-H or dMMR based on the results of the KEYNOTE-016 study. Unfortunately, since less than 3% of patients with HCC have high MSI status, the utility of assessing MSI status in HCC is expected to be limited [25,27,28,29,30,31,32].

2.2. Tumor Antigen Presentation

2.2.1. PD-L1/PD-1 Expression

PD-1, identified by Honjo et al. [33] is expressed on the surface of T lymphocytes and is a master regulator of immune cell tolerance. The binding of PD-L1 to PD-1 inhibits T cell effector functions [34]. PD-L1 was identified by Honjo et al. as a PD-1 ligand in 2000 [35]. The following year, the expression of PD-1 ligands on tumor cell lines was demonstrated, with the blockade of PD-L1 shown to exert an anti-tumor effect [36]. PD-L1 is expressed by a range of tissues. The binding of PD-L1 expressed on tumor cells and virus-infected cells to PD-1 expressed on T cells leads to direct inhibition of T cell proliferation and effector functions such as IFN-γ production and cytotoxic activity [37].
In the liver, PD-L1 is expressed by Kupffer cells, hepatocytes, and hepatic sinusoidal cells [38]. PD-L1 expression is observed in 10–20% of HCC samples. PD-L1 overexpression in HCC has been reported to be associated with poor prognosis [39,40]. Zhou et al. [41] identified suppressive immune checkpoint molecules in patients with early HCC and highlighted the importance of PD-1 in suppressing the function of tumor-infiltrating lymphocytes.
However, the utility of PD-L1 expression as a predictive biomarker in remains controversial. The phase I/II CHECKMATE 040 study [42] investigated the role of nivolumab in HCC patients that failed to respond to sorafenib. In this study, the objective response rate (ORR) was 19% in patients with <1% PD-L1 expression in tumor biopsy samples and 26% in patients with ≥1% PD-L1 expression in tumor biopsy samples using a PD-L1 IHC 28-8 pharmDx assay; however, this difference did not reach statistical significance. In a retrospective exploratory analysis of the results of the KEYNOTE 224 trial [43] on the effect of second-line pembrolizumab in HCC, baseline PD-L1 expression on Tumor cells (TC) also did not affect response rates to pembrolizumab treatment. However, the baseline combined positive score (CPS) based on PD-L1 expression on tumor cells and immune cells (macrophages and lymphocytes) was associated with response to pembrolizumab treatment. In the IMbrave150 study [44], PD-L1 expression measured using the SP263 assay had no significant effect on immunotherapy in patients with a CPS < 1% but could predict a good effect of atezolizumab/bevacizumab treatment compared to sorafenib treatment in patients with a CPS ≥ 1% (HR, 0.52; 95% confidence interval [CI], 0.32–0.87). In the HIMALAYA study, [45] PD-L1 expression measured using the SP263 assay was not significantly correlated with the therapeutic efficacy of durvalumab monotherapy or durvalumab and tremelimumab combination therapy.
Recent prospective studies in HCC have demonstrated that the use of PD-L1 expression alone has limited accuracy in predicting the efficacy of immune checkpoint inhibitors. Moreover, the antibody assays used for immunohistochemical studies, and the cut-off values used for determining PD-L1 positivity and PD-L1 expressing cells differ between studies of PD-L1. Standardization of PD-L1 analyses is urgently required in future studies.
Circulating soluble PD-L1 levels are negatively correlated with ICI efficacy in patients with NSCLC and melanoma [46,47,48,49]. In HCC, several studies [50,51,52] have reported an association between soluble PD-L1 levels and prognosis, with high serum PD-L1 levels indicating a poor prognosis. Tissue sampling of HCC is not always feasible. Moreover, it is not easy to obtain adequate amounts of tissue for reliable genetic studies. In clinical practice, there is increasing interest in the measurements of soluble PD-L1 due to the advantages of being non-invasive and enabling repeated sampling. More clinical evidence is needed for PD-L1 using liquid biopsy in HCC.

2.2.2. TMB

TMB has been regarded as a predictive biomarker for the response to immunotherapy. A high TMB is variably defined as tumors with ≥17 or ≥20 mutations/Mb upon sequencing of at least 1.2–1.5 Mb of the tumor genome [53]. High TMB is reportedly associated with increased neoantigen load [54]. Neoantigens contribute to the immune recognition of tumor cells and the subsequent induction of anti-tumor responses, which can be boosted by the use of immune checkpoint inhibitors [55]. There is a wealth of data that demonstrated that TMB is associated with immunotherapy response, with TMB having predictive value as a biomarker in most cancer subtypes [56]. As a TMB-H is prevalent in melanoma [57] and lung cancer [58], which are closely related to exposure to mutagens such as smoking and ultraviolet light, [59] studies on the efficacy of immunotherapy were first conducted in patients with melanoma or lung cancer.
While the role of TMB as a predictive marker of immunotherapy response in patients with HCC remains to be elucidated, in a large-scale study [56] conducted on 27 tumor types, TMB was correlated with ORR to anti-PD1 therapy, with a median TMB of 4–5 mutations/Mb in the 43 patients with HCC (21 treated with nivolumab and 22 treated with durvalumab). In a separate large-scale study of 24 tumor types conducted in China, [60] the median TMB in patients with HCC was reportedly 4–5 mutations per Mb. Ang et al. reported only 0.8% (n = 6) of 755 patients with HCC had TMB-H status when a TMB-H cut-off value of 4 mutations/Mb was used. In this study, TMB-H status was not associated with response to ICI therapy, likely due to the small sample size. In a phase Ib study [61] of camrelizumab and afatinib comprising 16 HCC patients, TMB values were correlated with treatment response. An exploratory analysis of the GO30140 study demonstrated that the ORR was higher in patients with TMB-H status, while TMB was unable to predict the treatment response or progression-free survival (PFS) in patients with HCC treated with atezolizumab plus bevacizumab. In the IMbrave150 study, no correlation was observed between TMB and ORR or PFS [62].
Several factors can influence the results of TMB analysis including the sequencing panel, type of mutations, and cut-off points used to define high TMB. WGS (whole genome sequencing) is the gold standard method of TMB analysis; [63] however, WES (whole exome sequencing) predominantly using NGS panels is routinely used for TMB analysis in clinical practice due to lower cost and time requirements [64]. TMB assays are not standardized due to the use of different sequencing platforms, calculations, and cut-off values. The use of TMB as a reliable biomarker is limited by the strict technical specifications, time, and high costs. Additionally, the low proportion of patients with HCC that have TMB-H status decreases the efficiency of TMB evaluations. Moreover, not all patients with TMB-H status respond to immune checkpoint inhibitors. Mcgrail et al. demonstrated that TMB-H was not associated with CD8+ T cells or neoantigen loads in breast cancer, prostate cancer, and gliomas. In these patients, the ORR of immunotherapy was less than 20%. This result was similar in patients with TMB-L [64]. Cristescu et al. conducted a study [65] based on the KEYNOTE clinical dataset and found that the response rate to pembrolizumab was higher in patients with both TMB-H and T cell inflamed GEP-H (gene express pattern, high) statuses (37–57%) than patients with TMB-H alone (11–42%). In this study, less than 5% of patients with HCC had TMB-H and GEP-H status, which were associated with favorable prognoses. Taken together, these results indicate that TMB alone may not be an ideal biomarker for predicting treatment response to ICIs.
Finally, TMB does not appear to be associated with PD-L1/PD-1 expression. Although there are reported differences between tumor subtypes, the concordance of PD-1/PD-L1 expression and TMB-H is reportedly 32% in melanoma, 12% in non-small cell lung cancer (NSCLC), 2.4% in endometrial cancer, 2.2% in esophageal cancer, and 1.2% of colorectal cancer. For all cancer types, a low proportion of patients had both TMB-H and PD-L1 expression. [53] Given these differences in TMB status and other biomarkers between cancer subtypes, further comprehensive studies are required to determine the utility of TMB and other biomarkers in predicting responses to immunotherapy in patients with HCC.

2.2.3. Interferon-Gamma Signaling Pathway

Interferon-gamma (IFN-γ) is released by T cells activated in response to recognizing neoantigens. The binding of IFN-γ to INF-γ receptors on tumor cell membranes activates Janus Kinase 1 (JAK1) and JAK2, which are signal transducers and activators of transcription (STAT) signaling that promote the expression of IFN-related genes, including interferon regulatory factor 1 (IRF1). Expression of IRF1 induces the transcription of other genes that increase surface expression of PD-L1 and MHC molecules.
Gao et al. [66] demonstrated that loss of INF-γ signaling is associated with primary resistance to anti-cytotoxic T-lymphocyte associated protein-4 (CTLA-4) therapy, with an average of 15.33 mutations in genes related to the IFN-γ pathway in non-responders compared to an average of only one mutation in responders. Of the 12 melanoma tumors with primary resistance to ipilimumab, 75% (9/12) harbored circulating nucleic acids (CNAs) related to INF-γ pathway genes (IFNGR1, IFNGR2, IRF1, JAK2, suppressor of cytokine signaling 1 [SOCS1], and STAT4), while tumors from responders did not harbor CNAs related to INF-γ signaling (0/4, 0%). INF-γ related gene signatures have been studied as a potential biomarker for predicting response to ICIs in various tumor types including melanoma, NSCLC, and urothelial carcinoma (UCC) [67,68,69,70,71]. A study published in 2000 [72] reported that IFN-γ receptor expression in HCC was associated with escape from host immune surveillance, indicating that the IFN-γ pathway could play an important role in the immune evasion of tumors. Numata et al. confirmed that IFN-γ and interleukin 1β (IL-1β) exert synergistic effects on PD-L1 expression in HCC cells. A Chinese study identified IRF-8, one of the nine IRF members that function in regulating the IFN-γ pathway, as a potential biomarker for predicting response to anti-PD-1 treatment in HCC [73].
The JAK/STAT pathway has also been reported to be associated with resistance to ICIs. Loss of function mutations in JAK1/2 can lead to acquired and primary resistance to anti-PD-1 therapy [74,75]. The suppressor of the cytokine signaling protein (SOCS) family is a group of intracellular proteins that generally function as inhibitors of the IFN-γ signaling pathway. SOCS protein has been shown to negatively regulate tumor cell proliferation, differentiation, and immune response in HCC [76]. Identification of JAK1/2 and SOCS mutations before and during ICI therapy can help predict treatment courses and outcomes. Accordingly, gene signatures related to the IFN-γ signaling pathway may have utility as biomarkers for predicting response to immunotherapy in HCC; however, further studies are required are needed in the future.

2.2.4. Alterations in Genes Related to Immune Function

Wnt signaling is associated with immune surveillance escape, and B-catenin is known to play an important role in HCC survival by promoting EGFR signaling in the early phase of carcinogenesis [77]. Deregulation of WNT/B-catenin signaling was reportedly observed in 40–80% of patients with HCC in a preclinical study. Tumors harboring WNT/B-catenin aberrations are reportedly resistant to immunotherapy in mouse models [78,79]. James et al. identified more than 341 cancer-associated genes in 127 patients with HCC using prospective NGS. In this study, [80] Wnt/β-catenin mutations were reported to be significantly associated with resistance to immunotherapy, with a shorter median overall survival (OS, 9.1 vs. 15.2 months) and PFS (2.0 months vs. 7.4 months) compared to patients with wild-type tumors.
Dai et al. [81] posited that immune-related gene signatures have utility as predictive biomarkers of ICI efficacy in HCC and identified 11 immune-related genes through analysis of 365 HCC-samples to create the IRGPI (immune-related gene-based prognostic index). The IRGPI was shown to have utility in identifying patients with HCC who are immunogenic and more sensitive to immunotherapy. Genes related to Wnt/B-catenin signaling, a widely studied pathway in HCC, may represent good candidates for predictive biomarkers of immunotherapy.
TP53 gene aberrations, independently of CTNNB1 gene aberrations, are associated with specific IFN-γ gene signatures, higher Foxp3+ Treg infiltration, and lower CD8+ T cell infiltration in HCC [82,83,84]. Wang et al. reported that 61.8% of a Chinese HCC cohort (n = 369) had TP53 gene mutations, and TP53 mutation was associated with TMB-H and worse survival in HCC [85]. Accordingly, immune cell infiltration of tumor tissues, gene signatures, TMB-H, neoantigens, and DDR gene mutations all appear to be associated with poor ICI response. Studies on the association between TP53 gene mutations and ICI response have been conducted in several types of carcinomas [86,87,88]. KRAS is involved in the development of many human cancers. The prevalence of KRAS mutations is less than 20% in HCC tissues [89,90,91] and the contribution of KRAS mutations to the pathogenesis of HCC remains unclear. Although most previous studies of KRAS mutations have been conducted in patients with NSCLC, previous studies have reported conflicting results regarding the association between KRAS mutations and the efficacy of immunotherapy [92,93,94]. Theoretically, the high PD-L1 expression observed in cells with KRAS mutation results from activation of ERK, which mediates upregulation of PD-L1 [95]; however, the efficacy of ICIs in treating NSCLC tumors harboring EGFR mutations is low, indicating that patients with KRAS but not EGFR mutations are likely to have a good response to ICIs.
Mutations in genes that encode components of the mammalian switch/sucrose non-fermentable (mSWI/SNF) complex, which are occasionally identified in HCC, [91,96,97] appear to be associated with the efficacy of ICIs. Li et al. [98] reported that ARID1A mutation in gastrointestinal cancers is associated with TMB-H and high PD-L1 expression. The authors posited that ARID1A mutation may have utility as a biomarker for identifying patients with gastrointestinal cancer patients that are likely to respond to immunotherapy. A further study [67] based on WES data from 249 patients with solid cancers who received immunotherapy demonstrated that loss of PBRM1 is correlated with response to anti-PD 1 or anti-PD-L1 therapy. However, in a large database [99] of 1544 patients with solid cancers (848 from Memorial Sloan Kettering Cancer Center and 676 from Dana-Farber Cancer Center) treated with immunotherapy, loss of function (LOF) in genes related to mSWI/SNF (ARID1A, ARID1B, SMARCA4, SMARCB1, PBRM1, and ARID2) were not correlated with ICI response. The conflicting results regarding the associations between immunotherapy response and mutations or LOF in genes related to the SWI/SNF complex appear to require further studies.
Ross et al. [100] conducted comprehensive genomic profiling in patients with NSCLC and reported that MET and BRAF alterations were associated with an increased duration of ICI treatment regardless of TMB value. Interestingly, MET overexpression is reportedly associated with poor prognosis in HCC [101].

2.3. Biomarkers Related to the Tumor Microenvironment

Cytokines and Chemokines

Immune cells within the microenvironment can respond to signals received through their inherent receptors with their own protein-based language that will influence the cell itself and other cells throughout the organism. The language of cytokines is critical in this communication. Cytokines are small soluble factors with pleiotropic functions that are released by many cell types according to gene expression patterns. Types of human cytokines include interleukins, interferons, tumor necrosis factor (TNF), transforming growth factor-β (TGF-β), and other miscellaneous hematopoietins. [102] These cytokines have been evaluated as biomarkers of response to immunotherapy in various cancers.
The binding between PD-1 and PD-L1 leads to immune system exhaustion and increased numbers of Tregs [103]. PD-L1 expression varies depending on local concentrations of pro-inflammatory cytokines [104]. IFN-γ is one of the most actively studied inflammatory cytokines. Increased IFN-γ levels are reportedly associated with a good response to immunotherapy [105,106,107]. In addition, TNF-α, [105,108,109] IL-6, [106,109,110,111] IL-8, [105,112,113,114,115] and TGF-β [18,116,117,118] are reported to have utility as predictive biomarkers for cancer immunotherapy. TGF-β is involved in immunosuppression within the TME and tumor immune evasion [119]. The inhibitory immunological function of Tregs is a major obstacle to eliciting an effective anti-tumor response, and Treg activation is modulated by the TGF-β pathway [120]. The TGF-β signaling pathway is activated at the transcriptional level in most HCCs [121,122], with a strong association demonstrated between the TGF-β signature and the exhausted immune signature in HCC [123]. In a Phase II study [116] of pembrolizumab for HCC by Lynn et al., high baseline plasma TGF-B levels (≥200 pg/mL) were significantly correlated with poor treatment outcomes after pembrolizumab treatment. In this study, there was no association between PD-L1 expression and pembrolizumab response. IL-6 is an inflammatory cytokine that has also been studied as a biomarker in HCC. Shakiba et al. [124] demonstrated significantly higher serum IL-6 levels in patients with HCC compared to healthy controls in a meta-analysis. Real-world data have revealed that high baseline IL-6 levels are associated with poor prognosis in patients with HCC treated with atezolizumab and bevacizumab [125]. Similarly, IL-27, an anti-inflammatory cytokine, has the potential to be a predictive biomarker. Active IL-27 receptor signaling was previously shown to reduce the expression of IL-6 and other inflammatory cytokines deemed pro-tumorigenic in HCC [126] and in another study, IL-27 was shown to have a similar function as IFN-γ and tends to be inhibited by IL-6 [127]. These results suggest that IL-27 might have a role as a predictive biomarker of immunotherapy for HCC. Turan Aghayev et al. also reported that elevated IL27RA mRNA expression correlated with poor survival since the treatment in Korean, Chinese, and TCGA HCC cohorts [128].
The chemokine, subdivided into four main classes depending on the location of the first two cysteine resides in their amino acid sequence: CC, CXC, C, and CX3C, is the largest subfamily of low molecular weight chemotactic cytokine [129]. Chemokines directly and indirectly affect tumor immunity, inflammatory response, proliferation, invasion, and metastasis via modulation of various signaling pathways including the JAK/STAT, PI3K/Akt, ERK1/2 MAPK, and Wnt/β-catenin pathways among others [130,131]. Chemokines related to HCC have been actively studied, and most chemokines have been confirmed to play important roles in tumor development and survival, including neovascularization, renewal, apoptosis, invasion, and metastasis in HCC [132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153]. Lin et al. posited that CXCLs are potential therapeutic targets for regulating anti-cancer immunity in HCC and may have utility as prognostic markers of response to immunotherapy [154]. Gu et al. also suggested that CCL14 may be a potential independent prognostic biomarker for HCC [141]. Previous studies evaluating chemokines as predictive biomarkers of the response to immunotherapy have been conducted using gene signatures comprising multiple chemokines (predominantly immune-related genes such as IFN-γ) rather than specific chemokines alone. The gene signatures with utility as predictive biomarkers are described in the following section. Studies on the effect of chemokines on tumor immunity in HCC and their potential as targets for immune modulation indicate that chemokines may have utility as predictive biomarkers of response to immunotherapy in HCC.

2.4. Tumor Infiltrating Lymphocyte (TIL)

TILs are defined as all lymphocytic substances within or around tumor cells and generally refer to CD4+ and CD8+ T cells. TILs are considered to be associated with a critical role in the anti-tumor immune response [155]. A significant association between high baseline TILs density and ICIs response has already been studied in RCC, CRC, NSCLC, and breast cancer [156,157,158,159,160,161]. In HCC, increased TIL was associated with a good response to ICI treatment. In the Checkmate 040 trial, increased CD3+/CD8+ tumor-infiltrating T cells of nivolumab-treated HCC patients showed a trend toward survival prolongation, but was not significant (p = 0.08) [162]. In another study of 32 HCC patients who underwent radiofrequency ablation or chemoablation with an injection of tremelimumab, the increase in CD8+ T cells identified by a 6-week tumor biopsy was associated with improved survival time [163]. Ng et al. also reported that high intra-tumoral CD38+ cells identified by immunochemistry were associated with a good response to ICI [164].
Clinical use of TILs as a biomarker to predict immunotherapy response in HCC seems challenging in the near future. Standardization and validation of test methods, test timing, and test interpretation should be needed. Nevertheless, since the role of TIL in the adaptive immune resistance mechanism is as essential as PD-L1, so it has high potential as a predictive biomarker of immunotherapy.

3. Circulating Biomarkers

3.1. Circulating Tumor DNA and Circulating Tumor Cells

As molecular biology technology develops, interest in the use of liquid biopsies is also increasing due to a rising demand for non-invasive methods of obtaining genomic information from tumor cells. Accordingly, the evaluation of circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), also known as liquid biopsy, has been widely studied in recent years. ctDNAs are cell-free materials released by tumor cells into the bloodstream following tumor cell apoptosis or necrosis [165]. In HCC, ctDNA levels are correlated with tumor size, extrahepatic spread, and vascular invasion [166]. ctDNA levels are correlated with microvascular invasion and predict tumor recurrence of HCC. Franses et al. showed that the quality of genetic information in ctDNA is just as valuable as that in tissues. They performed ctDNA profiling using commercially available NGS assays in 136 patients with unresectable HCC from four cancer centers. In 28 patients, blood TMB (bTMB) levels were approximately three-fold higher than tissue TMB (tTMB) levels [167]. Qualitative analysis of somatic mutations in HCC-derived ctDNA has detected several oncogenes and tumor suppressor genes including RAS, TERT, TP53, PTEN, ARID2, and CTNNB1 that are consistent with the results of tissue analyses in 63% of cases [168,169]. Fu et al. [170] investigated ctDNA in preoperative blood samples from 258 HCC patients who underwent curative liver resection. The number of gene alterations detected in ctDNA was associated with early tumor relapse. In this study, patients with FAT1, or LRP1b variants but without TP53 variants had worse PFS following treatment with lenvatinib combined with ICIs. According to the NORTE STUDY group, baseline CXCL9 levels measured using a cytokine array of ctDNA were significantly lower in patients with early disease progression following treatment with atezolizumab and bevacizumab [171]. In a phase II study [172] of camrelizumab plus afatinib in HCC, ctDNA has utility in predicting pathologic response and relapse following treatment. A Japanese study investigated the potential role of cfDNA/ctDNA as biomarkers for predicting treatment response in patients with unresectable HCC who had been treated with atezolizumab and bevacizumab. High pre-treatment cfDNA levels were associated with a lower response rate and shorter PFS and OS. Further, the presence of a TERT mutation and a serum AFP levels ≥400 ng/mL were independent predictors of poor OS after treatment with atezolizumab combined with bevacizumab [173].
Circulating tumor cells are nucleated cells released into the bloodstream from tumor cells. The detection of CTCs remains challenging as CTCs are present in the blood at low concentrations and there is no standardization of testing methods [174]. Nevertheless, CTCs are considered attractive biomarkers as they have the characteristics of tumor cells. Chen et al. [175] reported that CTCs were detected in 95% of 195 patients with HCC, with a median number of 6 CTC in each 5 mL blood sample. The number of CTCs was reported to be correlated with disease stage (BCLC), metastasis, and serum AFP levels. The simple number of CTCs has been used to predict prognosis, including disease status or recurrence after surgery, in HCC [176,177,178]. Winograd et al. [179] confirmed that CTCs express immune checkpoints including PD-L1, PD-L2, and CTLA-4. PD-L1 expression in CTCs may have utility in predicting immunotherapy response in HCC [180]. However, the detection of CTCs remains challenging as CTCs are present at low concentrations in blood samples, and different methods may enrich different CTC populations, thereby affecting PD-L1 measurements. Similar to other novel biomarkers, there is an urgent need for the standardization of methods for quantifying CTCs. Prospective clinical trials of liquid biopsies for predicting the efficacy of ICIs are encouraged.

3.2. Serum Alpha-Fetoprotein (AFP) and CRP

AFP is the most widely studied and used biomarker in HCC. In vitro, AFP has been proven to have an oncogenic effect by regulating TNF cytotoxicity [181], suppressing NK cell activity, [182] and promoting tumor growth by reducing levels of FAS-associated death domain protein (FADD) [183]. Recent phase 3 studies for ICIs have reported somewhat conflicting results about the predictive role of serum AFP levels. The IMbrave150 study reported [5] in a subset of patients with serum AFP levels <400 ng/mL, immunotherapy was associated with longer survival, while and the HIMALAYA [184] and CHECKMATE 459 study [6] reported in a subset of patients with serum AFP levels ≥400 ng/mL, immunotherapy was associated with longer survival compared to control arm group. However, post-treatment AFP values appear to be consistently associated with ICI response. The measurement of serum AFP levels at six weeks after initiating treatment is a potential surrogate biomarker of prognosis in patients with HCC receiving atezolizumab and bevacizumab. Zhu et al. investigated the relationship between changes in serum AFP levels and response to treatment in patients enrolled in the GO30140 and IMbrave150 studies. Based on a ≥75% decrease in serum AFP levels at six weeks after initial treatment, the sensitivity for discriminating between responders and non-responders was 0.59 with a specificity of 0.86 in the IMbrave150 study, while serum AFP levels using the same cut-off value had a sensitivity of 0.71 and a specificity of 0.91 in the GO30140 study. Lower serum AFP levels are reportedly associated with longer OS and PFS in patients with HCC, particularly in patients with HBV-related HCC [185]. Even in real-world data from patients with HCC treated with atezolizumab and bevacizumab, early changes (three weeks after treatment) in serum AFP levels were significantly associated with an objective response to treatment. An AFP ratio (AFP levels after treatment to baseline AFP levels) of 1.4 or higher three weeks after the initiation of treatment may be an early predictor of refractoriness to atezolizumab plus bevacizumab [186].
Serum CRP levels may also predict response to PD-1 inhibition. Zhang et al. reported baseline serum CRP and AFP levels may have the potential as predictors the efficacy of PD-1 inhibitors in HCC [187]. A Japanese study also demonstrated that serum AFP and CRP levels are associated with the efficacy of immunotherapy. The CRAFITY score, composed of serum AFP, and CRP levels, reportedly has utility in predicting the treatment outcomes and side effects of immunotherapy [188]. In a previous study conducted in Europe, the CRAFITY score had utility in predicting radiologic response and survival after immunotherapy [189].

3.3. Neutrophil-to-Lymphocyte Ratio (NLR) and Platelet-to-Lymphocyte Ratio (PLR)

Circulating blood components such as platelets, granulocytes, and neutrophils are involved in tumor growth and metastasis and play a role as a pool of vascular endothelial growth factor (VEGF). In various diseases, the NLR and PLR are used as inflammatory markers and are actively studied in HCC. Elevated neutrophil and platelet count can result in elevated circulating VEGF levels and is associated with poor prognosis [190,191]. Huang et al. [192] investigated the prognostic value of blood biomarkers in 100 patients with HBV-induced HCC treated with PD-1 inhibitors. In this study, a high systemic immune inflammation index, high platelet-to-lymphocyte ratio (PLR), high neutrophil-to-lymphocyte ratio (NLR), and low lymphocyte-to-monocyte ratio were correlated with decreased OS and PFS. In a separate retrospective study [193] of 103 patients with HCC treated with nivolumab, post-treatment NLR and PLR were significantly lower in patients with PR or CR compared to patients with stable disease or PD. Post-treatment NLR and PLR were significantly associated with overall survival. NLR was also identified as a significant prognostic biomarker in three multicenter retrospective real-world studies of patients with HCC treated with atezolizumab/bevacizumab in East Asia. In a Korean study [194], the authors reported that a high baseline des-gamma-carboxy prothrombin level (≥186 mAU/mL), NLR ≥ 2.5, and a decrease in NLR ≥ 10% at first response may be useful prognostic predictors for OS and PFS. Similarly in a Japanese study, [195] high baseline NLR (>3) was significantly associated with poor survival in patients with HCC treated with atezolizumab/bevacizumab. In a Chinese study, patients with a baseline NLR ≥ 5 had significantly shorter OS and PFS compared to patients with an NLR < 5 [196].

4. Host-Related Biomarkers

4.1. Etiology

Worldwide, approximately 13% of cases of cancer are associated with infections. The four most important infectious agents associated with cancer are Helicobacter pylori, human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV), which together account for more than 90% of infection-related cancers [197]. Viral infections are estimated to contribute to the development of 15–20% of human cancers. HPV-related head and neck cancers have a good treatment response and favorable prognosis. Viral-associated cancers have distinct biological and clinical features compared to other tumor types [198]. HCC is strongly linked to viral infection, with approximately 54% of cases of HCC attributed to HBV infection (which affects 400 million individuals globally), while 31% can be attributed to HCV infection (which affects 170 million individuals globally) [199]. Liver cirrhosis, which is associated with an increased risk of HCC, occurs under the influence of inflammatory cytokines [200,201]. According to a study by Beudeker et al., patients with HBV-related liver cirrhosis and HCC had the greatest upregulation of pro-inflammatory mediators compared to patients with cirrhosis due to HCV, alcohol-related liver disease, or non-alcoholic fatty liver disease (NAFLD) [202]. Non-alcoholic steatohepatitis (NASH) and NAFLD are representative causes of non-viral HCC.
Pfister et al. confirmed the unfavorable effects of anti-PD-1 treatment on NASH in experimental mice models, providing evidence of the tissue-damaging role of CD8+PD-1+ T lymphocytes [203]. Several studies have reported inflammatory responses according to HCC etiology may represent a biomarker for predicting the response of immunotherapy; however, the results are somewhat controversial. The results of two recent meta-analyses revealed no significant difference in response to immunotherapy between patients with viral-associated HCC and non-viral HCC, with a similar response rate observed between patients with HBV and HCV infection [204,205]. Recent phase III studies [6,44,202,206,207] of ICI have demonstrated that immunotherapy tends to be more effective in cases of viral HCC. The ORR in this study of 27% with atezolizumab/bevacizumab, 12% with nivolumab, 19% with durvalumab, and 18% with durvalumab plus tremelimumab in patients with non-viral HCC compared with 32%, 19%, 14.3%, and 21.3%, respectively, for HBV-associated HCC and 30%, 17%, 22.4%, and 35.5%, respectively, for HCV-associated HCC suggest that immunotherapy has similar efficacy between non-viral HCC and viral HCC. The level of evidence for the lower efficacy of ICI treatment in non-viral HCC is very low as large-scale clinical trials have failed to produce concordant results.

4.2. Performance Status and Liver Function

As the number of elderly cancer patients increases along with the prolonged life expectancy, the number of patients with poor performance status also increases. Accordingly, interest in cancer management in these vulnerable groups has been also increasing. Performance status is one of the most reliable indicators for predicting cancer prognosis. A score greater than 2 on the ECOG performance status scale is generally accepted as a relative contraindication to systemic treatment including cytotoxic chemotherapy and treatment with immune checkpoint inhibitors. In most clinical trials, eligibility criteria require a performance status based on an ECOG score of at least 2. Even in patients with HCC, performance status plays an important role in determining treatment plans. The BCLC (Barcelona Clinic Liver Cancer) staging system, which provides a guide for first-line treatment of HCC, consists of disease extension, liver function, and performance status. According to the treatment recommendations in the BCLC staging system, BCLC-C, and BCLC-D are classified according to liver function and performance status. Systemic treatments such as atezolizumab/bevacizumab are recommended for patients with BCLC-C HCC, and the best supportive care is recommended for patients with BCLC-D HCC [208]. Scheiner et al. proposed a predictive scoring system using CRP and AFP for immunotherapy in HCC [189] and reported ECOG and Child–Pugh Class as independent prognostic factors related to OS in patients with HCC receiving immunotherapy after multivariate Cox regression analysis.
The ALBI (albumin-bilirubin) score was developed to evaluate liver function more objectively and simply by excluding subjective factors such as ascites and encephalopathy that are included in the Child–Pugh score. Accordingly, the ALBI score is also expected to predict the response to immunotherapy [209]. According to the Imbrave150 exploratory analysis [210], the response to atezolizumab/bevacizumab was better than the response to sorafenib in patients with ALBI grade 1 compared to patients with ALBI grade 2. A subgroup analysis of the HIMALAYA trial [7] demonstrated that durvalumab/tremelimumab was superior to sorafenib in patients with ALBI grades 1 and 2; however, this difference did not reach statistical significance. In real-world data [211], baseline ABLI is considered an independent predictive biomarker in patients with HCC treated with immunotherapy.

4.3. Disease Status and Tumor Burden

In clinical practice, HCC treatment is predominantly determined based on BCLC staging [208]. Systemic treatment including immunotherapy is recommended for BCLC-C. For patients with BCLC-B, an intermediate stage of disease, TACE is the preferred option and immunotherapy is considered an appropriate option for those with a larger tumor burden. Most recent phase III studies [5,7,212,213,214,215] on immunotherapy in HCC have focused on patients with BCLC-B and BCLC-C HCC. However, immunotherapy was reported to be effective in patients with BCLC-C HCC but not BCLC-B HCC compared to a control group (predominantly comprising patients treated with sorafenib), which could be due to underpowered analysis with a smaller sample size [5,7].
Total tumor burden is another factor associated with response to immunotherapy. Preclinical data demonstrate that PD-1 blockade is more effective in mice bearing smaller lung squamous cell tumors [216], and PD-L1 blockade had a greater effect in mice with ovarian tumors at an early stage [217]. This negative correlation between response to PD-L1 blockade and total tumor volume has been studied in human lung cancer and melanoma as well as in animal models [218,219,220,221,222]. Larger tumors tend to be more immunosuppressive at both the local and systemic levels compared to smaller tumors. Myeloid-derived suppressor cells, tumor-associated macrophages, and regulatory T cells (Tregs) have been shown to increasingly infiltrate the TME as tumors progress in preclinical studies [223,224]. In addition to the increase in immunosuppressive cellular components in TMEs, cytokine production is distorted to a more suppressive profile in large tumors compared to small tumors. Levels of TGF-β, which has anti-tumor effects in early-stage cancer but tumor–promoting effects in late-stage cancer, IL-10, and nitric oxide synthase 2 (NOS2) increase as tumors progress [225,226]. In a recently published Korean study reporting real-world data [227], the nivolumab response was significantly correlated with primary tumor size in 261 patients with advanced HCC. In addition, the authors reported decreasing responses to nivolumab in the order of intrahepatic tumors (ORR, 10.1%) followed by metastatic tumors in the lung (ORR, 24.2%) and LN (ORR, 37.1%). The above results are supported by a previous study demonstrating that sites of HCC metastasis have altered pathological features [228] and Tregs have distinctive functions through different mediators in other organs [229].

4.4. Gut Microbiome

The gut microbiome, which is critical for the development and regulation of innate and adaptive immunity, influences other organs including the brain, liver, and pancreas, and the development of various diseases including obesity [230], diabetes [231], and cancers [232]. In 2015, the relationship between the gut microbiome and the effect of immunotherapy was reported for the first time in preclinical mouse studies. Tumor growth, spontaneous anti-tumor immunity, and the efficacy of immune checkpoint inhibitors differ in mice depending on the composition of the gut microbiota [233,234]. Data from these preclinical studies indicate that inter-individual heterogeneity of immunotherapy efficacy may be partially caused by the gut microbiome, and studies on this effect are currently being conducted. An early mouse study [233] reported that the Bacteroides genus is associated with good anti-CTLA-4 response. However, the Bacteroides genus is reportedly associated with poor response to immunotherapy in humans [235]. Bifidobacterium longum, Dorea formicigenerans, Collinsella aerofaciens, Alistipes putredinis, and Prevotella copri are reportedly enriched in responders to anti-PD 1 treatment in melanoma [236] and non-small-cell lung cancer [237], with Ruminococcus obeum, and Roseburia intestinalis found to be more abundant in non-responders. Interestingly, the microbiome observed in responders was also associated with frequent immune-related colitis.
The use of antibiotics can lead to changes in the composition and function of the gut microbiome which may reduce microbial diversity and adversely affect the immune response. Indeed, several studies [238,239,240,241] have reported that changes in the gut microbiome following the use of antibiotics can have negative impacts on responses to immunotherapy. The importance of gut microbial diversity has also been studied in HCC. Zheng et al. [239] reported that fecal samples from patients responding to immunotherapy had higher taxa richness and more gene counts than those of non-responders. In this study, both responders, and non-responders had similar microbial composition to healthy people before treatment. However, responders still had a stable microbiome while non-responders had increased proteobacteria after treatment which became dominant over time. The results of this study indicate that gut microbial diversity and stability are associated with the response to immunotherapy.
Microbial signatures can be used to stratify patients according to the likelihood of treatment response or toxicity. Modulating the gut microbiota may represent a potential treatment strategy for cancer; however, these approaches are likely to require adaption depending on the cancer type and therapeutic drug type. Large-scale studies monitoring sequential changes in the gut microbiome following the administration of immunotherapies are required to determine the utility of microbial signatures in predicting responses to immunotherapy.

5. Conclusions

The advent of immunotherapy with immune checkpoint inhibitors has shed new light on HCC treatments. However, immunotherapy has only recently been approved as a standard frontline therapy. Accordingly, there is a lack of studies on biomarkers that are able to predict the efficacy of immunotherapy in HCC. No reliable biomarkers with utility in predicting responses to immunotherapy have been identified to date as only studies on tissue PD-L1 expression and TMB has been published in addition to the results of exploratory analyses in phase III studies (Table 1.) However, increased real-world data from patients treated with ICIs are expected which may facilitate the development of more precise and accurate predictive biomarkers that improve personalized cancer therapy. Liquid biopsy and microbiome might have a role in understanding TME and inflammation, which has a strong link with the immunotherapy response of HCC.
In addition, although not mentioned in the text there are more potential candidates as a predictive biomarker for immunotherapy in HCC. Sex and age are the most basic information that shows distinguishing features immunologically. On average, women have stronger innate and adaptive immune responses than men [244], Therefore, the benefit from immunotherapy is also expected to be small. In meta-analyses of solid cancers, survival time after immunotherapy was revealed to be longer in male patients than in female patients [245,246]. As age increases, there is a tendency to experience various side effects and more severe toxicity after immunotherapy. Indeed, in the recent phase III trials of ICI for HCC [247], an increasing population over 65 was associated with lower ORR and reduced survival. Smoking also causes chronic inflammation, which can contribute to alterations in immune response. A strong association between smoking and TMB-H has already been demonstrated in NSCLC [248,249], and Wang et al. [250] found that smoking in HBV-related HCC affects the immune response through viral activation.
In conclusion, a combinatory approach that considers the intrinsic feature of the tumor, the peritumoral microenvironment, the immune system, host factors, and their clinical and molecular analyzes is likely required for the prediction of immunotherapy response in HCC. Moreover, considering the constantly changing TME and diverse tumor biology of HCC, further research on personalized biomarkers that enable continuous monitoring in a non-invasive, and cost-effective way is needed.

Author Contributions

Draft writing and preparation, Data Gathering, manipulation, analysis, J.H.J. and J.D.Y.; Methodology, J.D.Y.; Supervision, J.H.J., S.Y.H., D.L., K.S., E.K.K., G.K.A.-A., J.D.Y.; Data collecting and curation, J.H.J., S.Y.H., D.L.; Software, J.H.J. All authors have read and agreed to the published version of the manuscript.

Funding

Dr. Yang’s research is supported by American College of Gastroenterology Junior Faculty Development Award, Department of Defense Peer Reviewed Cancer Research Program Career Development Award (CA191051) and the National Institutes of Health. (K08CA259534) Dr. Ghassan’s research is supported by Cancer Center Support Grant—P30 CA008748. Memorial Sloan Kettering Cancer Center Dr. Selwyn Vickers. R01 HL149946, R01 CA273925 grants to Ekaterina K. Koltsova.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AFPAlpha-feroprotein
ALBIAlbumin-bilirubin
BCLCBarcelona Clinic Liver Cancer
BERBase excision repair
CNACirculating nucleic acids
CPSCombined positive score
CR/PRComplete response/Partial response
CTCCirculating tumor cell
ctDNACirculating tumor DNA
CTLACytotoxic T-lymphocyte associated protein
DDRDNA damage repair
ECOGEastern Cooperative Oncology Group
FADDFAS-associated death domain
GEPGene express pattern
HBV Hepatitis B virus
HCCHepatocellular carcinoma
HCVHepatitis C virus
HPVHuman papillomavirus
HRHomologous recombination
ICIImmune checkpoint inhibitor
IFN-γInterferon-gamma
IHCImmunohistochemistry
ILInterlurkin
IRGPIImmune related gene-based prognostic index
JAKJanus kinase
MGMTO6-methylguanine–DNA methyltransferase
MMRMismatch repair
MSIMicrosatellite instability
NAFLDNon-alcoholic fatty liver disease
NASHNon-alcoholic steatohepatitis
NGSNext-generation sequencing
NLR/PLRNeutrophil to lymphocyte ratio/Platelet to lymphocyte ratio
NOS2Nitric oxide synthase 2
NSCLCNon-small cell lung cancer
ORRObjective response rate
PD-(L)1Programmed death-(ligand)1
PFSProgression-free survival
POLE/POLDPolymerase ε/Polymerase δ
SOCSSuppressor of cytokine signaling protein
STATSignal transducers and activators of transcription
TACETrans-arterial chemoembolization
TCTumor cells
TGF-βTransforming growth factor-β
TKITyrosine kinase inhibitor
TMETumor microenvironment
TMBTumor mutational burden
TNFTumor necrosis factor
TregRegulatory T cell
UCCUrothelial carcinoma
VEGFVascular endothelial growth factor
WESWhole exome sequencing
WGSWhole genome sequencing

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA A Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  3. Yang, J.D.; Heimbach, J.K. New advances in the diagnosis and management of hepatocellular carcinoma. BMJ 2020, 371, m3544. [Google Scholar] [CrossRef]
  4. Sangro, B.; Sarobe, P.; Hervás-Stubbs, S.; Melero, I. Advances in immunotherapy for hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 525–543. [Google Scholar] [CrossRef] [PubMed]
  5. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef] [PubMed]
  6. Yau, T.; Park, J.-W.; Finn, R.S.; Cheng, A.-L.; Mathurin, P.; Edeline, J.; Kudo, M.; Harding, J.J.; Merle, P.; Rosmorduc, O. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022, 23, 77–90. [Google Scholar] [CrossRef]
  7. Vogel, A.; Chan, S.; Furuse, J.; Tak, W.; Masi, G.; Varela, M.; Kim, J.; Tanasanvimon, S.; Reig, M.; Dayyani, F. O-5 Outcomes by baseline liver function in patients with unresectable hepatocellular carcinoma treated with tremelimumab and durvalumab in the phase 3 HIMALAYA study. Ann. Oncol. 2022, 33, S380–S381. [Google Scholar] [CrossRef]
  8. Ding, X.; He, M.; Chan, A.W.; Song, Q.X.; Sze, S.C.; Chen, H.; Man, M.K.; Man, K.; Chan, S.L.; Lai, P.B. Genomic and epigenomic features of primary and recurrent hepatocellular carcinomas. Gastroenterology 2020, 154, 374–389. [Google Scholar] [CrossRef]
  9. Xu, L.; He, M.; Dai, Z.; Yu, J.; Wang, J.; Li, X.; Jiang, B.; Ke, Z.; Su, T.; Peng, Z. Genomic and transcriptional heterogeneity of multifocal hepatocellular carcinoma. Ann. Oncol. 2019, 30, 990–997. [Google Scholar] [CrossRef]
  10. Viscardi, G.; Tralongo, A.C.; Massari, F.; Lambertini, M.; Mollica, V.; Rizzo, A.; Comito, F.; Di Liello, R.; Alfieri, S.; Imbimbo, M.; et al. Comparative assessment of early mortality risk upon immune checkpoint inhibitors alone or in combination with other agents across solid malignancies: A systematic review and meta-analysis. Eur. J. Cancer 2022, 177, 175–185. [Google Scholar] [CrossRef] [PubMed]
  11. FDA-NIH Biomarker Working Group. BEST (Biomarkers, Endpoints, and Other Tools) Resource. 2016. Available online: https://www.ncbi.nlm.nih.gov/books/NBK326791 (accessed on 5 March 2023).
  12. Rizzo, A.; Cusmai, A.; Gadaleta-Caldarola, G.; Palmiotti, G. Which role for predictors of response to immune checkpoint inhibitors in hepatocellular carcinoma? Expert Rev. Gastroenterol. Hepatol. 2022, 16, 333–339. [Google Scholar] [CrossRef] [PubMed]
  13. Jeggo, P.A.; Pearl, L.H.; Carr, A.M. DNA repair, genome stability and cancer: A historical perspective. Nat. Rev. Cancer 2016, 16, 35–42. [Google Scholar] [CrossRef]
  14. Helleday, T.; Petermann, E.; Lundin, C.; Hodgson, B.; Sharma, R.A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer 2008, 8, 193–204. [Google Scholar] [CrossRef]
  15. Mouw, K.W.; Goldberg, M.S.; Konstantinopoulos, P.A.; D’Andrea, A.D. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer Discov. 2017, 7, 675–693. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, Z.; Zhao, J.; Wang, G.; Zhang, F.; Zhang, Z.; Zhang, F.; Zhang, Y.; Dong, H.; Zhao, X.; Duan, J.; et al. Comutations in DNA Damage Response Pathways Serve as Potential Biomarkers for Immune Checkpoint Blockade. Cancer Res. 2018, 78, 6486–6496. [Google Scholar] [CrossRef] [PubMed]
  17. Lin, P.; Gao, R.Z.; Wen, R.; He, Y.; Yang, H. DNA Damage Repair Profiles Alteration Characterize a Hepatocellular Carcinoma Subtype With Unique Molecular and Clinicopathologic Features. Front. Immunol. 2021, 12, 715460. [Google Scholar] [CrossRef] [PubMed]
  18. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  19. Li, J.; Li, N.; Azhar, M.S.; Liu, L.; Wang, L.; Zhang, Q.; Sheng, L.; Wang, J.; Feng, S.; Qiu, Q.; et al. Analysis of mutations in DNA damage repair pathway gene in Chinese patients with hepatocellular carcinoma. Sci. Rep. 2022, 12, 12330. [Google Scholar] [CrossRef]
  20. Li, G.-M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008, 18, 85–98. [Google Scholar] [CrossRef]
  21. Lee, V.; Murphy, A.; Le, D.T.; Diaz, L.A., Jr. Mismatch Repair Deficiency and Response to Immune Checkpoint Blockade. Oncologist 2016, 21, 1200–1211. [Google Scholar] [CrossRef]
  22. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
  23. Wang, F.; Zhao, Q.; Wang, Y.N.; Jin, Y.; He, M.M.; Liu, Z.X.; Xu, R.H. Evaluation of POLE and POLD1 Mutations as Biomarkers for Immunotherapy Outcomes Across Multiple Cancer Types. JAMA Oncol. 2019, 5, 1504–1506. [Google Scholar] [CrossRef] [PubMed]
  24. Goumard, C.; Desbois-Mouthon, C.; Wendum, D.; Calmel, C.; Merabtene, F.; Scatton, O.; Praz, F. Low levels of microsatellite instability at simple repeated sequences commonly occur in human hepatocellular carcinoma. Cancer Genom. Proteom. 2017, 14, 329–339. [Google Scholar]
  25. Kawaoka, T.; Ando, Y.; Yamauchi, M.; Suehiro, Y.; Yamaoka, K.; Kosaka, Y.; Fuji, Y.; Uchikawa, S.; Morio, K.; Fujino, H. Incidence of microsatellite instability-high hepatocellular carcinoma among Japanese patients and response to pembrolizumab. Hepatol. Res. 2020, 50, 885–888. [Google Scholar] [CrossRef] [PubMed]
  26. Zhao, P.; Li, L.; Jiang, X.; Li, Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019, 12, 1–14. [Google Scholar] [CrossRef] [PubMed]
  27. Mukai, S.; Kanzaki, H.; Ogasawara, S.; Ishino, T.; Ogawa, K.; Nakagawa, M.; Fujiwara, K.; Unozawa, H.; Iwanaga, T.; Sakuma, T. Exploring microsatellite instability in patients with advanced hepatocellular carcinoma and its tumor microenvironment. JGH Open 2021, 5, 1266–1274. [Google Scholar] [CrossRef] [PubMed]
  28. Hause, R.J.; Pritchard, C.C.; Shendure, J.; Salipante, S.J. Classification and characterization of microsatellite instability across 18 cancer types. Nat. Med. 2016, 22, 1342–1350. [Google Scholar] [CrossRef] [PubMed]
  29. Cortes-Ciriano, I.; Lee, S.; Park, W.Y.; Kim, T.M.; Park, P.J. A molecular portrait of microsatellite instability across multiple cancers. Nat. Commun. 2017, 8, 15180. [Google Scholar] [CrossRef]
  30. Bonneville, R.; Krook, M.A.; Kautto, E.A.; Miya, J.; Wing, M.R.; Chen, H.Z.; Reeser, J.W.; Yu, L.; Roychowdhury, S. Landscape of Microsatellite Instability Across 39 Cancer Types. JCO Precis. Oncol. 2017, 2017, 1–15. [Google Scholar] [CrossRef]
  31. Ang, C.; Klempner, S.J.; Ali, S.M.; Madison, R.; Ross, J.S.; Severson, E.A.; Fabrizio, D.; Goodman, A.; Kurzrock, R.; Suh, J.; et al. Prevalence of established and emerging biomarkers of immune checkpoint inhibitor response in advanced hepatocellular carcinoma. Oncotarget 2019, 10, 4018–4025. [Google Scholar] [CrossRef]
  32. Eso, Y.; Shimizu, T.; Takeda, H.; Takai, A.; Marusawa, H. Microsatellite instability and immune checkpoint inhibitors: Toward precision medicine against gastrointestinal and hepatobiliary cancers. J. Gastroenterol. 2020, 55, 15–26. [Google Scholar] [CrossRef]
  33. Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef] [PubMed]
  34. Yu, M.C.; Chen, C.H.; Liang, X.; Wang, L.; Gandhi, C.R.; Fung, J.J.; Lu, L.; Qian, S. Inhibition of T-cell responses by hepatic stellate cells via B7-H1–mediated T-cell apoptosis in mice. Hepatology 2004, 40, 1312–1321. [Google Scholar] [CrossRef]
  35. Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef]
  36. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [Google Scholar] [CrossRef] [PubMed]
  37. Iwai, Y.; Hamanishi, J.; Chamoto, K.; Honjo, T. Cancer immunotherapies targeting the PD-1 signaling pathway. J. Biomed. Sci. 2017, 24, 26. [Google Scholar] [CrossRef]
  38. Yamazaki, T.; Akiba, H.; Iwai, H.; Matsuda, H.; Aoki, M.; Tanno, Y.; Shin, T.; Tsuchiya, H.; Pardoll, D.M.; Okumura, K.; et al. Expression of programmed death 1 ligands by murine T cells and APC. J. Immunol. 2002, 169, 5538–5545. [Google Scholar] [CrossRef]
  39. Gao, Q.; Wang, X.Y.; Qiu, S.J.; Yamato, I.; Sho, M.; Nakajima, Y.; Zhou, J.; Li, B.Z.; Shi, Y.H.; Xiao, Y.S.; et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin. Cancer Res. 2009, 15, 971–979. [Google Scholar] [CrossRef]
  40. Calderaro, J.; Rousseau, B.; Amaddeo, G.; Mercey, M.; Charpy, C.; Costentin, C.; Luciani, A.; Zafrani, E.S.; Laurent, A.; Azoulay, D.; et al. Programmed death ligand 1 expression in hepatocellular carcinoma: Relationship With clinical and pathological features. Hepatology 2016, 64, 2038–2046. [Google Scholar] [CrossRef]
  41. Zhou, G.; Sprengers, D.; Boor, P.P.C.; Doukas, M.; Schutz, H.; Mancham, S.; Pedroza-Gonzalez, A.; Polak, W.G.; de Jonge, J.; Gaspersz, M.; et al. Antibodies Against Immune Checkpoint Molecules Restore Functions of Tumor-Infiltrating T Cells in Hepatocellular Carcinomas. Gastroenterology 2017, 153, 1107–1119. [Google Scholar] [CrossRef] [PubMed]
  42. Yau, T.; Kang, Y.K.; Kim, T.Y.; El-Khoueiry, A.B.; Santoro, A.; Sangro, B.; Melero, I.; Kudo, M.; Hou, M.M.; Matilla, A.; et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncol. 2020, 6, e204564. [Google Scholar] [CrossRef] [PubMed]
  43. Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; 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, 940–952. [Google Scholar] [CrossRef] [PubMed]
  44. Cheng, A.L.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Lim, H.Y.; Kudo, M.; Breder, V.; Merle, P.; et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J. Hepatol. 2022, 76, 862–873. [Google Scholar] [CrossRef] [PubMed]
  45. Abou-Alfa, G.K.; Lau, G.; Kudo, M.; Chan, S.L.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.-K.; Van Dao, T.; De Toni Enrico, N.; et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022, 1, EVIDoa2100070. [Google Scholar] [CrossRef]
  46. Mazzaschi, G.; Minari, R.; Zecca, A.; Cavazzoni, A.; Ferri, V.; Mori, C.; Squadrilli, A.; Bordi, P.; Buti, S.; Bersanelli, M. 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 2020, 148, 1–11. [Google Scholar] [CrossRef]
  47. Okuma, Y.; Wakui, H.; Utsumi, H.; Sagawa, Y.; Hosomi, Y.; Kuwano, K.; Homma, S. Soluble programmed cell death ligand 1 as a novel biomarker for nivolumab therapy for non–small-cell lung cancer. Clin. Lung Cancer 2018, 19, 410–417. [Google Scholar] [CrossRef]
  48. Zhou, J.; Mahoney, K.M.; Giobbie-Hurder, A.; Zhao, F.; Lee, S.; Liao, X.; Rodig, S.; Li, J.; Wu, X.; Butterfield, L.H. Soluble PD-L1 as a Biomarker in Malignant Melanoma Treated with Checkpoint BlockadeSoluble PD-L1 in Melanoma. Cancer Immunol. Res. 2017, 5, 480–492. [Google Scholar] [CrossRef]
  49. Ugurel, S.; Schadendorf, D.; Horny, K.; Sucker, A.; Schramm, S.; Utikal, J.; Pföhler, C.; Herbst, R.; Schilling, B.; Blank, C. Elevated baseline serum PD-1 or PD-L1 predicts poor outcome of PD-1 inhibition therapy in metastatic melanoma. Ann. Oncol. 2020, 31, 144–152. [Google Scholar] [CrossRef]
  50. Chang, B.; Huang, T.; Wei, H.; Shen, L.; Zhu, D.; He, W.; Chen, Q.; Zhang, H.; Li, Y.; Huang, R. 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. 2019, 68, 353–363. [Google Scholar] [CrossRef]
  51. Finkelmeier, F.; Canli, Ö.; Tal, A.; Pleli, T.; Trojan, J.; Schmidt, M.; Kronenberger, B.; Zeuzem, S.; Piiper, A.; Greten, F.R. High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur. J. Cancer 2016, 59, 152–159. [Google Scholar] [CrossRef]
  52. Han, X.; Gu, Y.-k.; Li, S.-l.; Chen, H.; Chen, M.-s.; Cai, Q.-q.; Deng, H.-x.; Zuo, M.-x.; Huang, J.-h. 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, 303–312. [Google Scholar] [CrossRef]
  53. Luchini, C.; Bibeau, F.; Ligtenberg, M.J.L.; Singh, N.; Nottegar, A.; Bosse, T.; Miller, R.; Riaz, N.; Douillard, J.Y.; Andre, F.; et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: A systematic review-based approach. Ann. Oncol. 2019, 30, 1232–1243. [Google Scholar] [CrossRef] [PubMed]
  54. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef]
  55. Rooney, M.S.; Shukla, S.A.; Wu, C.J.; Getz, G.; Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 2015, 160, 48–61. [Google Scholar] [CrossRef] [PubMed]
  56. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
  57. Chan, T.A.; Wolchok, J.D.; Snyder, A. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2015, 373, 1984. [Google Scholar] [CrossRef] [PubMed]
  58. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef]
  59. Pfeifer, G.P. Environmental exposures and mutational patterns of cancer genomes. Genome Med. 2010, 2, 54. [Google Scholar] [CrossRef]
  60. Liu, L.; Bai, X.; Wang, J.; Tang, X.R.; Wu, D.H.; Du, S.S.; Du, X.J.; Zhang, Y.W.; Zhu, H.B.; Fang, Y.; et al. Combination of TMB and CNA Stratifies Prognostic and Predictive Responses to Immunotherapy Across Metastatic Cancer. Clin. Cancer Res. 2019, 25, 7413–7423. [Google Scholar] [CrossRef]
  61. Xu, J.; Zhang, Y.; Jia, R.; Yue, C.; Chang, L.; Liu, R.; Zhang, G.; Zhao, C.; Zhang, Y.; Chen, C.; et al. Anti-PD-1 Antibody SHR-1210 Combined with Apatinib for Advanced Hepatocellular Carcinoma, Gastric, or Esophagogastric Junction Cancer: An Open-label, Dose Escalation and Expansion Study. Clin. Cancer Res. 2019, 25, 515–523. [Google Scholar] [CrossRef]
  62. Zhu, A.X.; Abbas, A.R.; de Galarreta, M.R.; Guan, Y.; Lu, S.; Koeppen, H.; Zhang, W.; Hsu, C.H.; He, A.R.; Ryoo, B.Y.; et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat. Med. 2022, 28, 1599–1611. [Google Scholar] [CrossRef] [PubMed]
  63. Sholl, L.M.; Hirsch, F.R.; Hwang, D.; Botling, J.; Lopez-Rios, F.; Bubendorf, L.; Mino-Kenudson, M.; Roden, A.C.; Beasley, M.B.; Borczuk, A.; et al. The Promises and Challenges of Tumor Mutation Burden as an Immunotherapy Biomarker: A Perspective from the International Association for the Study of Lung Cancer Pathology Committee. J. Thorac. Oncol. 2020, 15, 1409–1424. [Google Scholar] [CrossRef] [PubMed]
  64. Stenzinger, A.; Endris, V.; Budczies, J.; Merkelbach-Bruse, S.; Kazdal, D.; Dietmaier, W.; Pfarr, N.; Siebolts, U.; Hummel, M.; Herold, S.; et al. Harmonization and Standardization of Panel-Based Tumor Mutational Burden Measurement: Real-World Results and Recommendations of the Quality in Pathology Study. J. Thorac. Oncol. 2020, 15, 1177–1189. [Google Scholar] [CrossRef] [PubMed]
  65. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018, 362, eaar3593. [Google Scholar] [CrossRef] [PubMed]
  66. Gao, J.; Shi, L.Z.; Zhao, H.; Chen, J.; Xiong, L.; He, Q.; Chen, T.; Roszik, J.; Bernatchez, C.; Woodman, S.E. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 2016, 167, 397–404. [Google Scholar] [CrossRef] [PubMed]
  67. Miao, D.; Margolis, C.A.; Vokes, N.I.; Liu, D.; Taylor-Weiner, A.; Wankowicz, S.M.; Adeegbe, D.; Keliher, D.; Schilling, B.; Tracy, A. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 2018, 50, 1271–1281. [Google Scholar] [CrossRef] [PubMed]
  68. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
  69. Sharma, P.; Retz, M.; Siefker-Radtke, A.; Baron, A.; Necchi, A.; Bedke, J.; Plimack, E.R.; Vaena, D.; Grimm, M.-O.; Bracarda, S. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017, 18, 312–322. [Google Scholar] [CrossRef]
  70. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
  71. Ribas, A.; Robert, C.; Hodi, F.S.; Wolchok, J.D.; Joshua, A.M.; Hwu, W.-J.; Weber, J.S.; Zarour, H.M.; Kefford, R.; Loboda, A. Association of response to programmed death receptor 1 (PD-1) blockade with pembrolizumab (MK-3475) with an interferon-inflammatory immune gene signature. J. Clin. Oncol. 2015, 33, 3001. [Google Scholar] [CrossRef]
  72. Nagao, M.; Nakajima, Y.; Kanehiro, H.; Hisanaga, M.; Aomatsu, Y.; Ko, S.; Tatekawa, Y.; Ikeda, N.; Kanokogi, H.; Urizono, Y. The impact of interferon gamma receptor expression on the mechanism of escape from host immune surveillance in hepatocellular carcinoma. Hepatology 2000, 32, 491–500. [Google Scholar] [CrossRef]
  73. Wu, H.; Li, Y.; Shi, G.; Du, S.; Wang, X.; Ye, W.; Zhang, Z.; Chu, Y.; Ma, S.; Wang, D. Hepatic interferon regulatory factor 8 expression suppresses hepatocellular carcinoma progression and enhances the response to anti–programmed cell death protein-1 therapy. Hepatology 2022, 76, 1602–1616. [Google Scholar] [CrossRef]
  74. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef]
  75. Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; Grasso, C.S.; Hugo, W.; Sandoval, S.; Torrejon, D.Y. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef] [PubMed]
  76. Yoshimura, A.; Naka, T.; Kubo, M. SOCS proteins, cytokine signalling and immune regulation. Nat. Rev. Immunol. 2007, 7, 454–465. [Google Scholar] [CrossRef] [PubMed]
  77. Kim, E.; Lisby, A.; Ma, C.; Lo, N.; Ehmer, U.; Hayer, K.E.; Furth, E.E.; Viatour, P. Promotion of growth factor signaling as a critical function of β-catenin during HCC progression. Nat. Commun. 2019, 10, 1909. [Google Scholar] [CrossRef]
  78. Ruiz de Galarreta, M.; Bresnahan, E.; Molina-Sánchez, P.; Lindblad, K.E.; Maier, B.; Sia, D.; Puigvehi, M.; Miguela, V.; Casanova-Acebes, M.; Dhainaut, M.; et al. β-Catenin Activation Promotes Immune Escape and Resistance to Anti-PD-1 Therapy in Hepatocellular Carcinoma. Cancer Discov. 2019, 9, 1124–1141. [Google Scholar] [CrossRef]
  79. Pinyol, R.; Sia, D.; Llovet, J.M. Immune Exclusion-Wnt/CTNNB1 Class Predicts Resistance to Immunotherapies in HCC. Clin. Cancer Res. 2019, 25, 2021–2023. [Google Scholar] [CrossRef] [PubMed]
  80. Harding, J.J.; Nandakumar, S.; Armenia, J.; Khalil, D.N.; Albano, M.; Ly, M.; Shia, J.; Hechtman, J.F.; Kundra, R.; El Dika, I.; et al. Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies. Clin. Cancer Res. 2019, 25, 2116–2126. [Google Scholar] [CrossRef] [PubMed]
  81. Dai, Y.; Qiang, W.; Lin, K.; Gui, Y.; Lan, X.; Wang, D. An immune-related gene signature for predicting survival and immunotherapy efficacy in hepatocellular carcinoma. Cancer Immunol. Immunother. 2021, 70, 967–979. [Google Scholar] [CrossRef]
  82. Hu, Z.Q.; Xin, H.Y.; Luo, C.B.; Li, J.; Zhou, Z.J.; Zou, J.X.; Zhou, S.L. Associations among the mutational landscape, immune microenvironment, and prognosis in Chinese patients with hepatocellular carcinoma. Cancer Immunol. Immunother. 2021, 70, 377–389. [Google Scholar] [CrossRef]
  83. Guo, G.; Yu, M.; Xiao, W.; Celis, E.; Cui, Y. Local Activation of p53 in the Tumor Microenvironment Overcomes Immune Suppression and Enhances Antitumor ImmunityLocal p53 Activation in the TME Enhances Antitumor Immunity. Cancer Res. 2017, 77, 2292–2305. [Google Scholar] [CrossRef] [PubMed]
  84. Blagih, J.; Buck, M.D.; Vousden, K.H. p53, cancer and the immune response. J. Cell Sci. 2020, 133, jcs237453. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, L.; Yan, K.; He, X.; Zhu, H.; Song, J.; Chen, S.; Cai, S.; Zhao, Y.; Wang, L. LRP1B or TP53 mutations are associated with higher tumor mutational burden and worse survival in hepatocellular carcinoma. J. Cancer 2021, 12, 217. [Google Scholar] [CrossRef] [PubMed]
  86. Lin, X.; Wang, L.; Xie, X.; Qin, Y.; Xie, Z.; Ouyang, M.; Zhou, C. Prognostic biomarker TP53 mutations for immune checkpoint blockade therapy and its association with tumor microenvironment of lung adenocarcinoma. Front. Mol. Biosci. 2020, 7, 602328. [Google Scholar] [CrossRef]
  87. Lyu, Q.; Lin, A.; Cao, M.; Xu, A.; Luo, P.; Zhang, J. Alterations in TP53 are a potential biomarker of bladder cancer patients who benefit from immune checkpoint inhibition. Cancer Control 2020, 27, 1073274820976665. [Google Scholar] [CrossRef] [PubMed]
  88. Shi, C.; Liu, S.; Tian, X.; Wang, X.; Gao, P. A TP53 mutation model for the prediction of prognosis and therapeutic responses in head and neck squamous cell carcinoma. BMC Cancer 2021, 21, 1035. [Google Scholar] [CrossRef] [PubMed]
  89. Turhal, N.S.; Savaş, B.; Çoşkun, Ö.; Baş, E.; Karabulut, B.; Nart, D.; Korkmaz, T.; Yavuzer, D.; Demir, G.; Doğusoy, G. Prevalence of K-Ras mutations in hepatocellular carcinoma: AA Turkish Oncology Group pilot study. Mol. Clin. Oncol. 2015, 3, 1275–1279. [Google Scholar] [CrossRef]
  90. Karnoub, A.E.; Weinberg, R.A. Ras oncogenes: Split personalities. Nat. Rev. Mol. Cell Biol. 2008, 9, 517–531. [Google Scholar] [CrossRef]
  91. Ally, A.; Balasundaram, M.; Carlsen, R.; Chuah, E.; Clarke, A.; Dhalla, N.; Holt, R.A.; Jones, S.J.; Lee, D.; Ma, Y. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 2017, 169, 1327–1341.e1323. [Google Scholar] [CrossRef]
  92. Amanam, I.; Mambetsariev, I.; Gupta, R.; Achuthan, S.; Wang, Y.; Pharaon, R.; Massarelli, E.; Koczywas, M.; Reckamp, K.; Salgia, R. Role of immunotherapy and co-mutations on KRAS-mutant non-small cell lung cancer survival. J. Thorac. Dis. 2020, 12, 5086. [Google Scholar] [CrossRef]
  93. Sciortino, C.; Viglialoro, V.; Nucci, M.; Polito, M.G.; Cortesi, E.; Gelibter, A.; Gazzaniga, P.; Nicolazzo, C.; Siringo, M.; Caponnetto, S. Response to immunotherapy in KRAS G12C mutated NSCLC: A single-centre retrospective observational study. Oncotarget 2022, 13, 686. [Google Scholar] [CrossRef] [PubMed]
  94. Dong, Z.-Y.; Zhong, W.-Z.; Zhang, X.-C.; Su, J.; Xie, Z.; Liu, S.-Y.; Tu, H.-Y.; Chen, H.-J.; Sun, Y.-L.; Zhou, Q. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin. Cancer Res. 2017, 23, 3012–3024. [Google Scholar] [CrossRef] [PubMed]
  95. Lee, M.-H.; Yanagawa, J.; Li, R.; Walser, T.C.; Krysan, K.; Wang, G.; Goldman, J.W.; Garon, E.B.G.; Zeng, G.; Sharma, S. Increased PD-L1 expression in KRAS mutated premalignant human bronchial epithelial cells is enhanced by LKB1 loss and mediated by ERK activation. J. Immunother. Cancer 2015, 3, P305. [Google Scholar] [CrossRef]
  96. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed]
  97. Endo, M.; Yasui, K.; Zen, Y.; Gen, Y.; Zen, K.; Tsuji, K.; Dohi, O.; Mitsuyoshi, H.; Tanaka, S.; Taniwaki, M.; et al. Alterations of the SWI/SNF chromatin remodelling subunit-BRG1 and BRM in hepatocellular carcinoma. Liver. Int. 2013, 33, 105–117. [Google Scholar] [CrossRef]
  98. Li, L.; Li, M.; Jiang, Z.; Wang, X. ARID1A Mutations Are Associated with Increased Immune Activity in Gastrointestinal Cancer. Cells 2019, 8, 678. [Google Scholar] [CrossRef]
  99. Abou Alaiwi, S.; Nassar, A.H.; Xie, W.; Bakouny, Z.; Berchuck, J.E.; Braun, D.A.; Baca, S.C.; Nuzzo, P.V.; Flippot, R.; Mouhieddine, T.H.; et al. Mammalian SWI/SNF Complex Genomic Alterations and Immune Checkpoint Blockade in Solid Tumors. Cancer Immunol. Res. 2020, 8, 1075–1084. [Google Scholar] [CrossRef]
  100. Ross, J.; Goldberg, M.; Albacker, L.; Gay, L.; Agarwala, V.; Elvin, J.; Vergilio, J.; Suh, J.; Ramkissoon, S.; Severson, E. Immune checkpoint inhibitor (ICPI) efficacy and resistance detected by comprehensive genomic profiling (CGP) in non-small cell lung cancer (NSCLC). Ann. Oncol. 2017, 28, v404. [Google Scholar] [CrossRef]
  101. Kim, J.H.; Kim, H.S.; Kim, B.J.; Jang, H.J.; Lee, J. Prognostic value of c-Met overexpression in hepatocellular carcinoma: A meta-analysis and review. Oncotarget 2017, 8, 90351–90357. [Google Scholar] [CrossRef]
  102. Cameron, M.J.; Kelvin, D.J. Cytokines, chemokines and their receptors. In Madame Curie Bioscience Database [Internet]; Landes Bioscience: Austin, TX, USA, 2013. [Google Scholar]
  103. Spranger, S.; Spaapen, R.M.; Zha, Y.; Williams, J.; Meng, Y.; Ha, T.T.; Gajewski, T.F. Up-regulation of PD-L1, IDO, and Tregs in the melanoma tumor microenvironment is driven by CD8+ T cells. Sci. Transl. Med. 2013, 5, ra116–ra200. [Google Scholar] [CrossRef]
  104. Kinter, A.L.; Godbout, E.J.; McNally, J.P.; Sereti, I.; Roby, G.A.; O’Shea, M.A.; Fauci, A.S. The common γ-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J. Immunol. 2008, 181, 6738–6746. [Google Scholar] [CrossRef] [PubMed]
  105. Boutsikou, E.; Domvri, K.; Hardavella, G.; Tsiouda, D.; Zarogoulidis, K.; Kontakiotis, T. Tumour necrosis factor, interferon-gamma and interleukins as predictive markers of antiprogrammed cell-death protein-1 treatment in advanced non-small cell lung cancer: A pragmatic approach in clinical practice. Ther. Adv. Med. Oncol. 2018, 10, 1758835918768238. [Google Scholar] [CrossRef] [PubMed]
  106. Yamazaki, N.; Kiyohara, Y.; Uhara, H.; Iizuka, H.; Uehara, J.; Otsuka, F.; Fujisawa, Y.; Takenouchi, T.; Isei, T.; Iwatsuki, K. Cytokine biomarkers to predict antitumor responses to nivolumab suggested in a phase 2 study for advanced melanoma. Cancer Sci. 2017, 108, 1022–1031. [Google Scholar] [CrossRef] [PubMed]
  107. McNamara, M.J.; Hilgart-Martiszus, I.; Barragan Echenique, D.M.; Linch, S.N.; Kasiewicz, M.J.; Redmond, W.L. Interferon-γ Production by Peripheral Lymphocytes Predicts Survival of Tumor-Bearing Mice Receiving Dual PD-1/CTLA-4 BlockadePredictive Biomarkers Following PD-1/CTLA-4 Blockade. Cancer Immunol. Res. 2016, 4, 650–657. [Google Scholar] [CrossRef]
  108. Tanaka, R.; Okiyama, N.; Okune, M.; Ishitsuka, Y.; Watanabe, R.; Furuta, J.; Ohtsuka, M.; Otsuka, A.; Maruyama, H.; Fujisawa, Y. Serum level of interleukin-6 is increased in nivolumab-associated psoriasiform dermatitis and tumor necrosis factor-α is a biomarker of nivolumab recativity. J. Dermatol. Sci. 2017, 86, 71–73. [Google Scholar] [CrossRef]
  109. Ozawa, Y.; Amano, Y.; Kanata, K.; Hasegwa, H.; Matsui, T.; Kakutani, T.; Koyauchi, T.; Tanahashi, M.; Niwa, H.; Yokomura, K. Impact of early inflammatory cytokine elevation after commencement of PD-1 inhibitors to predict efficacy in patients with non-small cell lung cancer. Med. Oncol. 2019, 36, 33. [Google Scholar] [CrossRef]
  110. Laino, A.S.; Woods, D.; Vassallo, M.; Qian, X.; Tang, H.; Wind-Rotolo, M.; Weber, J. Serum interleukin-6 and C-reactive protein are associated with survival in melanoma patients receiving immune checkpoint inhibition. J. Immunother. Cancer 2020, 8, e000842. [Google Scholar] [CrossRef]
  111. Tsukamoto, H.; Fujieda, K.; Miyashita, A.; Fukushima, S.; Ikeda, T.; Kubo, Y.; Senju, S.; Ihn, H.; Nishimura, Y.; Oshiumi, H. Combined blockade of IL6 and PD-1/PD-L1 signaling abrogates mutual regulation of their immunosuppressive effects in the tumor microenvironment. Cancer Res. 2018, 78, 5011–5022. [Google Scholar] [CrossRef]
  112. Agulló-Ortuño, M.T.; Gómez-Martín, Ó.; Ponce, S.; Iglesias, L.; Ojeda, L.; Ferrer, I.; García-Ruiz, I.; Paz-Ares, L.; Pardo-Marqués, V. Blood predictive biomarkers for patients with non–small-cell lung cancer associated with clinical response to Nivolumab. Clin. Lung Cancer 2020, 21, 75–85. [Google Scholar] [CrossRef]
  113. Sanmamed, M.; Perez-Gracia, J.; Schalper, K.; Fusco, J.; Gonzalez, A.; Rodriguez-Ruiz, M.; Oñate, C.; Perez, G.; Alfaro, C.; Martín-Algarra, S. Changes in serum interleukin-8 (IL-8) levels reflect and predict response to anti-PD-1 treatment in melanoma and non-small-cell lung cancer patients. Ann. Oncol. 2017, 28, 1988–1995. [Google Scholar] [CrossRef]
  114. Yuen, K.C.; Liu, L.-F.; Gupta, V.; Madireddi, S.; Keerthivasan, S.; Li, C.; Rishipathak, D.; Williams, P.; Kadel III, E.E.; Koeppen, H. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat. Med. 2020, 26, 693–698. [Google Scholar] [CrossRef] [PubMed]
  115. Schalper, K.A.; Carleton, M.; Zhou, M.; Chen, T.; Feng, Y.; Huang, S.-P.; Walsh, A.M.; Baxi, V.; Pandya, D.; Baradet, T. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 2020, 26, 688–692. [Google Scholar] [CrossRef]
  116. Feun, L.G.; Li, Y.Y.; Wu, C.; Wangpaichitr, M.; Jones, P.D.; Richman, S.P.; Madrazo, B.; Kwon, D.; Garcia-Buitrago, M.; Martin, P. Phase 2 study of pembrolizumab and circulating biomarkers to predict anticancer response in advanced, unresectable hepatocellular carcinoma. Cancer 2019, 125, 3603–3614. [Google Scholar] [CrossRef]
  117. Nonomura, Y.; Otsuka, A.; Nakashima, C.; Seidel, J.A.; Kitoh, A.; Dainichi, T.; Nakajima, S.; Sawada, Y.; Matsushita, S.; Aoki, M. Peripheral blood Th9 cells are a possible pharmacodynamic biomarker of nivolumab treatment efficacy in metastatic melanoma patients. Oncoimmunology 2016, 5, e1248327. [Google Scholar] [CrossRef]
  118. Tarhini, A.A.; Zahoor, H.; Lin, Y.; Malhotra, U.; Sander, C.; Butterfield, L.H.; Kirkwood, J.M. 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. [Google Scholar] [CrossRef]
  119. Batlle, E.; Massagué, J. Transforming growth factor-β signaling in immunity and cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef] [PubMed]
  120. Ringelhan, M.; Pfister, D.; O’Connor, T.; Pikarsky, E.; Heikenwalder, M. The immunology of hepatocellular carcinoma. Nat. Immunol. 2018, 19, 222–232. [Google Scholar] [CrossRef] [PubMed]
  121. Chen, J.; Zaidi, S.; Rao, S.; Chen, J.-S.; Phan, L.; Farci, P.; Su, X.; Shetty, K.; White, J.; Zamboni, F. Analysis of genomes and transcriptomes of hepatocellular carcinomas identifies mutations and gene expression changes in the transforming growth factor-β pathway. Gastroenterology 2018, 154, 195–210. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, J.; Gingold, J.A.; Su, X. Immunomodulatory TGF-β signaling in hepatocellular carcinoma. Trends Mol. Med. 2019, 25, 1010–1023. [Google Scholar] [CrossRef]
  123. Sia, D.; Jiao, Y.; Martinez-Quetglas, I.; Kuchuk, O.; Villacorta-Martin, C.; de Moura, M.C.; Putra, J.; Camprecios, G.; Bassaganyas, L.; Akers, N. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology 2017, 153, 812–826. [Google Scholar] [CrossRef] [PubMed]
  124. Shakiba, E.; Ramezani, M.; Sadeghi, M. Evaluation of serum interleukin-6 levels in hepatocellular carcinoma patients: A systematic review and meta-analysis. Clin. Exp. Hepatol. 2018, 4, 182–190. [Google Scholar] [CrossRef]
  125. Myojin, Y.; Kodama, T.; Sakamori, R.; Maesaka, K.; Matsumae, T.; Sawai, Y.; Imai, Y.; Ohkawa, K.; Miyazaki, M.; Tanaka, S.; et al. Interleukin-6 Is a Circulating Prognostic Biomarker for Hepatocellular Carcinoma Patients Treated with Combined Immunotherapy. Cancers 2022, 14, 883. [Google Scholar] [CrossRef]
  126. Koltsova, E.K.; Kim, G.; Lloyd, K.M.; Saris, C.J.; von Vietinghoff, S.; Kronenberg, M.; Ley, K. Interleukin-27 receptor limits atherosclerosis in Ldlr-/- mice. Circ. Res. 2012, 111, 1274–1285. [Google Scholar] [CrossRef]
  127. Rolvering, C.; Zimmer, A.D.; Kozar, I.; Hermanns, H.M.; Letellier, E.; Vallar, L.; Nazarov, P.V.; Nicot, N.; Ginolhac, A.; Haan, S. Crosstalk between different family members: IL27 recapitulates IFNγ responses in HCC cells, but is inhibited by IL6-type cytokines. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2017, 1864, 516–526. [Google Scholar] [CrossRef]
  128. Aghayev, T.; Mazitova, A.M.; Fang, J.R.; Peshkova, I.O.; Rausch, M.; Hung, M.; White, K.F.; Masia, R.; Titerina, E.K.; Fatkhullina, A.R.; et al. IL27 Signaling Serves as an Immunologic Checkpoint for Innate Cytotoxic Cells to Promote Hepatocellular Carcinoma. Cancer Discov. 2022, 12, 1960–1983. [Google Scholar] [CrossRef]
  129. Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef] [PubMed]
  130. Balkwill, F. Cancer and the chemokine network. Nat. Rev. Cancer 2004, 4, 540–550. [Google Scholar] [CrossRef]
  131. Zou, W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat. Rev. Cancer 2005, 5, 263–274. [Google Scholar] [CrossRef] [PubMed]
  132. Sun, F.; Wang, J.; Sun, Q.; Li, F.; Gao, H.; Xu, L.; Zhang, J.; Sun, X.; Tian, Y.; Zhao, Q. Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2019, 38, 449. [Google Scholar] [CrossRef] [PubMed]
  133. Shi, J.Y.; Yang, L.X.; Wang, Z.C.; Wang, L.Y.; Zhou, J.; Wang, X.Y.; Shi, G.M.; Ding, Z.B.; Ke, A.W.; Dai, Z. CC chemokine receptor-like 1 functions as a tumour suppressor by impairing CCR7-related chemotaxis in hepatocellular carcinoma. J. Pathol. 2015, 235, 546–558. [Google Scholar] [CrossRef] [PubMed]
  134. Sun, C.; Hu, A.; Wang, S.; Tian, B.; Jiang, L.; Liang, Y.; Wang, H.; Dong, J. ADAM17-regulated CX3CL1 expression produced by bone marrow endothelial cells promotes spinal metastasis from hepatocellular carcinoma. Int. J. Oncol. 2020, 57, 249–263. [Google Scholar] [CrossRef]
  135. Chen, E.-B.; Zhou, Z.-J.; Xiao, K.; Zhu, G.-Q.; Yang, Y.; Wang, B.; Zhou, S.-L.; Chen, Q.; Yin, D.; Wang, Z. The miR-561-5p/CX3CL1 signaling axis regulates pulmonary metastasis in hepatocellular carcinoma involving CX3CR1+ natural killer cells infiltration. Theranostics 2019, 9, 4779. [Google Scholar] [CrossRef]
  136. Yanru, W.; Zhenyu, B.; Zhengchuan, N.; Qi, Q.; Chunmin, L.; Weiqiang, Y. Transcriptomic analyses of chemokines reveal that down-regulation of XCR1 is associated with advanced hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2018, 496, 1314–1321. [Google Scholar] [CrossRef]
  137. Wu, Q.; Chen, J.-x.; Chen, Y.; Cai, L.-l.; Wang, X.-z.; Guo, W.-h.; Zheng, J.-f. The chemokine receptor CCR10 promotes inflammation-driven hepatocarcinogenesis via PI3K/Akt pathway activation. Cell Death Dis. 2018, 9, 232. [Google Scholar] [CrossRef]
  138. Du, D.; Liu, Y.; Qian, H.; Zhang, B.; Tang, X.; Zhang, T.; Liu, W. The effects of the CCR6/CCL20 biological axis on the invasion and metastasis of hepatocellular carcinoma. Int. J. Mol. Sci. 2014, 15, 6441–6452. [Google Scholar] [CrossRef]
  139. Tan, H.; Wang, S.; Zhao, L. A tumour-promoting role of Th9 cells in hepatocellular carcinoma through CCL20 and STAT3 pathways. Clin. Exp. Pharmacol. Physiol. 2017, 44, 213–221. [Google Scholar] [CrossRef]
  140. Singh, S.K.; Mishra, M.K.; Rivers, B.M.; Gordetsky, J.B.; Bae, S.; Singh, R. Biological and clinical significance of the CCR5/CCL5 axis in hepatocellular carcinoma. Cancers 2020, 12, 883. [Google Scholar] [CrossRef] [PubMed]
  141. Gu, Y.; Li, X.; Bi, Y.; Zheng, Y.; Wang, J.; Li, X.; Huang, Z.; Chen, L.; Huang, Y.; Huang, Y. CCL14 is a prognostic biomarker and correlates with immune infiltrates in hepatocellular carcinoma. Aging 2020, 12, 784. [Google Scholar] [CrossRef]
  142. Zhu, M.; Xu, W.; Wei, C.; Huang, J.; Xu, J.; Zhang, Y.; Zhao, Y.; Chen, J.; Dong, S.; Liu, B. CCL14 serves as a novel prognostic factor and tumor suppressor of HCC by modulating cell cycle and promoting apoptosis. Cell Death Dis. 2019, 10, 796. [Google Scholar] [CrossRef] [PubMed]
  143. Zhou, S.-L.; Zhou, Z.-J.; Hu, Z.-Q.; Li, X.; Huang, X.-W.; Wang, Z.; Fan, J.; Dai, Z.; Zhou, J. CXCR2/CXCL5 axis contributes to epithelial–mesenchymal transition of HCC cells through activating PI3K/Akt/GSK-3β/Snail signaling. Cancer Lett. 2015, 358, 124–135. [Google Scholar] [CrossRef] [PubMed]
  144. Li, X.-P.; Yang, X.-Y.; Biskup, E.; Zhou, J.; Li, H.-L.; Wu, Y.-F.; Chen, M.-L.; Xu, F. Co-expression of CXCL8 and HIF-1α is associated with metastasis and poor prognosis in hepatocellular carcinoma. Oncotarget 2015, 6, 22880. [Google Scholar] [CrossRef] [PubMed]
  145. Li, B.; Su, H.; Cao, J.; Zhang, L. CXCL13 rather than IL-31 is a potential indicator in patients with hepatocellular carcinoma. Cytokine 2017, 89, 91–97. [Google Scholar] [CrossRef] [PubMed]
  146. Ding, J.; Xu, K.; Zhang, J.; Lin, B.; Wang, Y.; Yin, S.; Xie, H.; Zhou, L.; Zheng, S. Overexpression of CXCL2 inhibits cell proliferation and promotes apoptosis in hepatocellular carcinoma. BMB Rep. 2018, 51, 630. [Google Scholar] [CrossRef] [PubMed]
  147. Croker, A.K.; Allan, A.L. Cancer stem cells: Implications for the progression and treatment of metastatic disease. J. Cell. Mol. Med. 2008, 12, 374–390. [Google Scholar] [CrossRef]
  148. Chen, Y.; Teng, F.; Wang, G.; Nie, Z. Overexpression of CXCR7 induces angiogenic capacity of human hepatocellular carcinoma cells via the AKT signaling pathway. Oncol. Rep. 2016, 36, 2275–2281. [Google Scholar] [CrossRef]
  149. Lin, L.; Han, M.; Wang, F.; Xu, L.; Yu, H.; Yang, P. CXCR7 stimulates MAPK signaling to regulate hepatocellular carcinoma progression. Cell Death Dis. 2014, 5, e1488. [Google Scholar] [CrossRef]
  150. Gao, Q.; Zhao, Y.-J.; Wang, X.-Y.; Qiu, S.-J.; Shi, Y.-H.; Sun, J.; Yi, Y.; Shi, J.-Y.; Shi, G.-M.; Ding, Z.-B. CXCR6 Upregulation Contributes to a Proinflammatory Tumor Microenvironment That Drives Metastasis and Poor Patient Outcomes in Hepatocellular CarcinomaCXCR6 Promotes HCC Metastasis. Cancer Res. 2012, 72, 3546–3556. [Google Scholar] [CrossRef]
  151. Li, L.; Xu, L.; Yan, J.; Zhen, Z.-J.; Ji, Y.; Liu, C.-Q.; Lau, W.Y.; Zheng, L.; Xu, J. CXCR2–CXCL1 axis is correlated with neutrophil infiltration and predicts a poor prognosis in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2015, 34, 129. [Google Scholar] [CrossRef] [PubMed]
  152. Lu, Y.; Li, S.; Ma, L.; Li, Y.; Zhang, X.; Peng, Q.; Mo, C.; Huang, L.; Qin, X.; Liu, Y. Type conversion of secretomes in a 3D TAM2 and HCC cell co-culture system and functional importance of CXCL2 in HCC. Sci. Rep. 2016, 6, 24558. [Google Scholar] [CrossRef]
  153. Li, C.; Kang, D.; Sun, X.; Liu, Y.; Wang, J.; Gao, P. The effect of CXC motif chemokine 13 on hepatocellular carcinoma associates with Wnt signaling. BioMed Res. Int. 2015, 2015, 345413. [Google Scholar]
  154. Lin, T.; Zhang, E.; Mai, P.-P.; Zhang, Y.-Z.; Chen, X.; Peng, L.-S. CXCL2/10/12/14 are prognostic biomarkers and correlated with immune infiltration in hepatocellular carcinoma. Biosci. Rep. 2021, 41, bsr20204312. [Google Scholar] [CrossRef]
  155. Brummel, K.; Eerkens, A.L.; de Bruyn, M.; Nijman, H.W. Tumour-infiltrating lymphocytes: From prognosis to treatment selection. Br. J. Cancer 2023, 128, 451–458. [Google Scholar] [CrossRef] [PubMed]
  156. Ficial, M.; Jegede, O.A.; Sant’Angelo, M.; Hou, Y.; Flaifel, A.; Pignon, J.-C.; Braun, D.A.; Wind-Rotolo, M.; Sticco-Ivins, M.A.; Catalano, P.J. Expression of T-Cell Exhaustion Molecules and Human Endogenous Retroviruses as Predictive Biomarkers for Response to Nivolumab in Metastatic Clear Cell Renal Cell CarcinomaPredictive Biomarkers for Nivolumab in Metastatic ccRCC. Clin. Cancer Res. 2021, 27, 1371–1380. [Google Scholar] [CrossRef] [PubMed]
  157. Gataa, I.; Mezquita, L.; Rossoni, C.; Auclin, E.; Kossai, M.; Aboubakar, F.; Le Moulec, S.; Massé, J.; Masson, M.; Radosevic-Robin, N. Tumour-infiltrating lymphocyte density is associated with favourable outcome in patients with advanced non–small cell lung cancer treated with immunotherapy. Eur. J. Cancer 2021, 145, 221–229. [Google Scholar] [CrossRef] [PubMed]
  158. Loi, S.; Michiels, S.; Adams, S.; Loibl, S.; Budczies, J.; Denkert, C.; Salgado, R. The journey of tumor-infiltrating lymphocytes as a biomarker in breast cancer: Clinical utility in an era of checkpoint inhibition. Ann. Oncol. 2021, 32, 1236–1244. [Google Scholar] [CrossRef] [PubMed]
  159. Gide, T.N.; Silva, I.P.; Quek, C.; Ahmed, T.; Menzies, A.M.; Carlino, M.S.; Saw, R.P.; Thompson, J.F.; Batten, M.; Long, G.V. Close proximity of immune and tumor cells underlies response to anti-PD-1 based therapies in metastatic melanoma patients. Oncoimmunology 2020, 9, 1659093. [Google Scholar] [CrossRef]
  160. Wong, P.F.; Wei, W.; Smithy, J.W.; Acs, B.; Toki, M.I.; Blenman, K.R.; Zelterman, D.; Kluger, H.M.; Rimm, D.L. Multiplex Quantitative Analysis of Tumor-Infiltrating Lymphocytes and Immunotherapy Outcome in Metastatic MelanomaTIL Profiles and Melanoma Anti–PD-1 Response. Clin. Cancer Res. 2019, 25, 2442–2449. [Google Scholar] [CrossRef]
  161. Loupakis, F.; Depetris, I.; Biason, P.; Intini, R.; Prete, A.A.; Leone, F.; Lombardi, P.; Filippi, R.; Spallanzani, A.; Cascinu, S. Prediction of benefit from checkpoint inhibitors in mismatch repair deficient metastatic colorectal cancer: Role of tumor infiltrating lymphocytes. Oncologist 2020, 25, 481–487. [Google Scholar] [CrossRef]
  162. Sangro, B.; Melero, I.; Wadhawan, S.; Finn, R.S.; Abou-Alfa, G.K.; Cheng, A.-L.; Yau, T.; Furuse, J.; Park, J.-W.; Boyd, Z. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J. Hepatol. 2020, 73, 1460–1469. [Google Scholar] [CrossRef]
  163. Duffy, A.G.; Ulahannan, S.V.; Makorova-Rusher, O.; Rahma, O.; Wedemeyer, H.; Pratt, D.; Davis, J.L.; Hughes, M.S.; Heller, T.; ElGindi, M. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J. Hepatol. 2017, 66, 545–551. [Google Scholar] [CrossRef]
  164. Ng, H.H.M.; Lee, R.Y.; Goh, S.; Tay, I.S.Y.; Lim, X.; Lee, B.; Chew, V.; Li, H.; Tan, B.; Lim, S. Immunohistochemical scoring of CD38 in the tumor microenvironment predicts responsiveness to anti-PD-1/PD-L1 immunotherapy in hepatocellular carcinoma. J. Immunother. Cancer 2020. [Google Scholar] [CrossRef]
  165. Maheswaran, S.; Haber, D.A. Circulating tumor cells: A window into cancer biology and metastasis. Curr. Opin. Genet. Dev. 2010, 20, 96–99. [Google Scholar] [CrossRef]
  166. Wang, J.; Huang, A.; Wang, Y.-P.; Yin, Y.; Fu, P.-Y.; Zhang, X.; Zhou, J. Circulating tumor DNA correlates with microvascular invasion and predicts tumor recurrence of hepatocellular carcinoma. Ann. Transl. Med. 2020, 8, 2. [Google Scholar] [CrossRef] [PubMed]
  167. Franses, J.W.; Lim, M.; Burgoyne, A.M.; Mody, K.; Lennerz, J.; Chang, J.; Imperial, R.; Dybel, S.N.; Dinh, T.M.; Masannat, J. Profile and Predictors of Blood Tumor Mutational Burden in Advanced Hepatocellular Carcinoma. Oncologist 2022, 27, e908–e911. [Google Scholar] [CrossRef] [PubMed]
  168. Wu, X.; Li, J.; Gassa, A.; Buchner, D.; Alakus, H.; Dong, Q.; Ren, N.; Liu, M.; Odenthal, M.; Stippel, D. Circulating tumor DNA as an emerging liquid biopsy biomarker for early diagnosis and therapeutic monitoring in hepatocellular carcinoma. Int. J. Biol. Sci. 2020, 16, 1551. [Google Scholar] [CrossRef] [PubMed]
  169. Von Felden, J.; Craig, A.J.; Garcia-Lezana, T.; Labgaa, I.; Haber, P.K.; D’Avola, D.; Asgharpour, A.; Dieterich, D.; Bonaccorso, A.; Torres-Martin, M. Mutations in circulating tumor DNA predict primary resistance to systemic therapies in advanced hepatocellular carcinoma. Oncogene 2021, 40, 140–151. [Google Scholar] [CrossRef] [PubMed]
  170. Fu, Y.; Yang, Z.; Hu, Z.; Yang, Z.; Pan, Y.; Chen, J.; Wang, J.; Hu, D.; Zhou, Z.; Xu, L. Preoperative serum ctDNA predicts early hepatocellular carcinoma recurrence and response to systemic therapies. Hepatol. Int. 2022, 16, 868–878. [Google Scholar] [CrossRef]
  171. Hosoda, S.; Suda, G.; Sho, T.; Ogawa, K.; Kimura, M.; Yang, Z.; Yoshida, S.; Kubo, A.; Tokuchi, Y.; Kitagataya, T. Low baseline CXCL9 predicts early progressive disease in unresectable HCC with atezolizumab plus bevacizumab treatment. Liver Cancer 2022, 1–15. [Google Scholar] [CrossRef]
  172. Xia, Y.; Tang, W.; Qian, X.; Li, X.; Cheng, F.; Wang, K.; Zhang, F.; Zhang, C.; Li, D.; Song, J. Efficacy and safety of camrelizumab plus apatinib during the perioperative period in resectable hepatocellular carcinoma: A single-arm, open label, phase II clinical trial. J. Immunother. Cancer 2022, 10, e004656. [Google Scholar] [CrossRef]
  173. Matsumae, T.; Kodama, T.; Myojin, Y.; Maesaka, K.; Sakamori, R.; Takuwa, A.; Oku, K.; Motooka, D.; Sawai, Y.; Oshita, M. Circulating cell-free DNA profiling predicts the therapeutic outcome in advanced hepatocellular carcinoma patients treated with combination immunotherapy. Cancers 2022, 14, 3367. [Google Scholar] [CrossRef] [PubMed]
  174. Ahn, J.C.; Teng, P.C.; Chen, P.J.; Posadas, E.; Tseng, H.R.; Lu, S.C.; Yang, J.D. Detection of circulating tumor cells and their implications as a biomarker for diagnosis, prognostication, and therapeutic monitoring in hepatocellular carcinoma. Hepatology 2021, 73, 422–436. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, J.; Cao, S.-W.; Cai, Z.; Zheng, L.; Wang, Q. Epithelial-mesenchymal transition phenotypes of circulating tumor cells correlate with the clinical stages and cancer metastasis in hepatocellular carcinoma patients. Cancer Biomark. 2017, 20, 487–498. [Google Scholar] [CrossRef] [PubMed]
  176. Fan, S.T.; Yang, Z.F.; Ho, D.W.; Ng, M.N.; Yu, W.C.; Wong, J. Prediction of posthepatectomy recurrence of hepatocellular carcinoma by circulating cancer stem cells: A prospective study. Ann. Surg. 2011, 254, 569–576. [Google Scholar] [CrossRef] [PubMed]
  177. Choi, G.H.; Kim, G.I.; Yoo, J.E.; Na, D.C.; Han, D.H.; Roh, Y.H.; Park, Y.N.; Choi, J.S. Increased expression of circulating cancer stem cell markers during the perioperative period predicts early recurrence after curative resection of hepatocellular carcinoma. Ann. Surg. Oncol. 2015, 22, 1444–1452. [Google Scholar] [CrossRef]
  178. Zhou, Y.; Wang, B.; Wu, J.; Zhang, C.; Zhou, Y.; Yang, X.; Zhou, J.; Guo, W.; Fan, J. Association of preoperative EpCAM Circulating Tumor Cells and peripheral Treg cell levels with early recurrence of hepatocellular carcinoma following radical hepatic resection. BMC Cancer 2016, 16, 506. [Google Scholar] [CrossRef]
  179. Winograd, P.; Hou, S.; Court, C.M.; Lee, Y.T.; Chen, P.J.; Zhu, Y.; Sadeghi, S.; Finn, R.S.; Teng, P.C.; Wang, J.J. Hepatocellular Carcinoma–Circulating Tumor Cells Expressing PD-L1 Are Prognostic and Potentially Associated With Response to Checkpoint Inhibitors. Hepatol. Commun. 2020, 4, 1527–1540. [Google Scholar] [CrossRef] [PubMed]
  180. Winograd, P.; Hou, S.; Sadeghi, S.; Finn, R.; DiPardo, B.; Li, Q.; Kaldas, F.; Busuttil, R.; Tomlinson, J.; Tseng, H. Evaluation of hepatocellular carcinoma circulating tumor cells expressing programmed death-ligand 1. HPB 2018, 20, S2–S3. [Google Scholar] [CrossRef]
  181. Semenkova, L.N.; Dudich, E.I.; Dudich, I.V.; Shingarova, L.N.; Korobko, V.G. Alpha-fetoprotein as a TNF resistance factor for the human hepatocarcinoma cell line HepG2. Tumor Biol. 1997, 18, 30–40. [Google Scholar] [CrossRef]
  182. Yamamoto, M.; Tatsumi, T.; Miyagi, T.; Tsunematsu, H.; Aketa, H.; Hosui, A.; Kanto, T.; Hiramatsu, N.; Hayashi, N.; Takehara, T. α-Fetoprotein impairs activation of natural killer cells by inhibiting the function of dendritic cells. Clin. Exp. Immunol. 2011, 165, 211–219. [Google Scholar] [CrossRef]
  183. Chen, T.; Dai, X.; Dai, J.; Ding, C.; Zhang, Z.; Lin, Z.; Hu, J.; Lu, M.; Wang, Z.; Qi, Y. AFP promotes HCC progression by suppressing the HuR-mediated Fas/FADD apoptotic pathway. Cell Death Dis. 2020, 11, 822. [Google Scholar] [CrossRef]
  184. Abou-Alfa, G.K.; Chan, S.L.; Kudo, M.; Lau, G.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.-K.; Dao, T.V.; De Toni, E.N. Phase 3 randomized, open-label, multicenter study of tremelimumab (T) and durvalumab (D) as first-line therapy in patients (pts) with unresectable hepatocellular carcinoma (uHCC): HIMALAYA. J. Clin. Oncol. 2022, 40, 379. [Google Scholar] [CrossRef]
  185. Zhu, A.X.; Dayyani, F.; Yen, C.-J.; Ren, Z.; Bai, Y.; Meng, Z.; Pan, H.; Dillon, P.; Mhatre, S.K.; Gaillard, V.E. Alpha-fetoprotein as a potential surrogate biomarker for atezolizumab+ bevacizumab treatment of hepatocellular carcinoma. Clin. Cancer Res. 2022, 28, OF1–OF9. [Google Scholar] [CrossRef]
  186. Kuzuya, T.; Kawabe, N.; Hashimoto, S.; Miyahara, R.; Sawaki, A.; Nakano, T.; Nakaoka, K.; Tanaka, H.; Miyachi, Y.; Mii, A. Early changes in alpha-fetoprotein are a useful predictor of efficacy of atezolizumab plus bevacizumab treatment in patients with advanced hepatocellular carcinoma. Oncology 2022, 100, 12–21. [Google Scholar] [CrossRef]
  187. Zhang, Y.; Lu, L.; He, Z.; Xu, Z.; Xiang, Z.; Nie, R.-C.; Lin, W.; Chen, W.; Zhou, J.; Yin, Y. C-reactive protein levels predict responses to PD-1 inhibitors in hepatocellular carcinoma patients. Front. Immunol. 2022, 13, 808101. [Google Scholar] [CrossRef]
  188. Hatanaka, T.; Kakizaki, S.; Hiraoka, A.; Tada, T.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E. Prognostic impact of C-reactive protein and alpha-fetoprotein in immunotherapy score in hepatocellular carcinoma patients treated with atezolizumab plus bevacizumab: A multicenter retrospective study. Hepatol. Int. 2022, 16, 1150–1160. [Google Scholar] [CrossRef]
  189. Scheiner, B.; Pomej, K.; Kirstein, M.M.; Hucke, F.; Finkelmeier, F.; Waidmann, O.; Himmelsbach, V.; Schulze, K.; von Felden, J.; Fründt, T.W. Prognosis of patients with hepatocellular carcinoma treated with immunotherapy–development and validation of the CRAFITY score. J. Hepatol. 2022, 76, 353–363. [Google Scholar] [CrossRef]
  190. Halazun, K.J.; Hardy, M.A.; Rana, A.A.; Woodland IV, D.C.; Luyten, E.J.; Mahadev, S.; Witkowski, P.; Siegel, A.B.; Brown, R.S., Jr.; Emond, J.C. Negative impact of neutrophil-lymphocyte ratio on outcome after liver transplantation for hepatocellular carcinoma. Ann. Surg. 2009, 250, 141–151. [Google Scholar] [CrossRef] [PubMed]
  191. Motomura, T.; Shirabe, K.; Mano, Y.; Muto, J.; Toshima, T.; Umemoto, Y.; Fukuhara, T.; Uchiyama, H.; Ikegami, T.; Yoshizumi, T. Neutrophil–lymphocyte ratio reflects hepatocellular carcinoma recurrence after liver transplantation via inflammatory microenvironment. J. Hepatol. 2013, 58, 58–64. [Google Scholar] [CrossRef] [PubMed]
  192. Huang, R.; Zheng, Y.; Zou, W.; Liu, C.; Liu, J.; Yue, J. Blood biomarkers predict survival outcomes in patients with hepatitis B virus-induced hepatocellular carcinoma treated with PD-1 inhibitors. J. Immunol. Res. 2022, 2022, 3781109. [Google Scholar] [CrossRef] [PubMed]
  193. Dharmapuri, S.; Özbek, U.; Lin, J.Y.; Sung, M.; Schwartz, M.; Branch, A.D.; Ang, C. 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, 4962–4970. [Google Scholar] [CrossRef] [PubMed]
  194. Chon, Y.E.; Cheon, J.; Kim, H.; Kang, B.; Ha, Y.; Kim, D.Y.; Hwang, S.G.; Chon, H.J.; Kim, B.K. Predictive biomarkers of survival in patients with advanced hepatocellular carcinoma receiving atezolizumab plus bevacizumab treatment. Cancer Med. 2022, 12, 2731–2738. [Google Scholar] [CrossRef] [PubMed]
  195. Ochi, H.; Kurosaki, M.; Joko, K.; Mashiba, T.; Tamaki, N.; Tsuchiya, K.; Marusawa, H.; Tada, T.; Nakamura, S.; Narita, R. Usefulness of neutrophil-to-lymphocyte ratio in predicting progression and survival outcomes after atezolizumab–bevacizumab treatment for hepatocellular carcinoma. Hepatol. Res. 2023, 53, 61–71. [Google Scholar] [CrossRef] [PubMed]
  196. Wu, Y.L.; Fulgenzi, C.A.M.; D’Alessio, A.; Cheon, J.; Nishida, N.; Saeed, A.; Wietharn, B.; Cammarota, A.; Pressiani, T.; Personeni, N. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios as prognostic biomarkers in unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab. Cancers 2022, 14, 5834. [Google Scholar] [CrossRef]
  197. De Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef]
  198. McLaughlin-Drubin, M.E.; Munger, K. Viruses associated with human cancer. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2008, 1782, 127–150. [Google Scholar] [CrossRef]
  199. Galle, P.R.; Forner, A.; Llovet, J.M.; Mazzaferro, V.; Piscaglia, F.; Raoul, J.-L.; Schirmacher, P.; Vilgrain, V. EASL clinical practice guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef]
  200. Wilson, C.; Jurk, D.; Fullard, N.; Banks, P.; Page, A.; Luli, S.; Elsharkawy, A.; Gieling, R.; Chakraborty, J.B.; Fox, C. NFκB1 is a suppressor of neutrophil-driven hepatocellular carcinoma. Nat. Commun. 2015, 6, 6818. [Google Scholar] [CrossRef]
  201. Liu, L.Z.; Zhang, Z.; Zheng, B.H.; Shi, Y.; Duan, M.; Ma, L.J.; Wang, Z.C.; Dong, L.Q.; Dong, P.P.; Shi, J.Y. CCL 15 Recruits Suppressive Monocytes to Facilitate Immune Escape and Disease Progression in Hepatocellular Carcinoma. Hepatology 2019, 69, 143–159. [Google Scholar] [CrossRef]
  202. Beudeker, B.J.; Groothuismink, Z.M.; van der Eijk, A.A.; Debes, J.D.; Boonstra, A. Circulating cytokines reflect the etiology-specific immune environment in cirrhosis and HCC. Cancers 2022, 14, 4900. [Google Scholar] [CrossRef]
  203. Pfister, D.; Núñez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A.; et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 2021, 592, 450–456. [Google Scholar] [CrossRef] [PubMed]
  204. Ho, W.J.; Danilova, L.; Lim, S.J.; Verma, R.; Xavier, S.; Leatherman, J.M.; Sztein, M.B.; Fertig, E.J.; Wang, H.; Jaffee, E. Viral status, immune microenvironment and immunological response to checkpoint inhibitors in hepatocellular carcinoma. J. Immunother. Cancer 2020, 8, e000394. [Google Scholar] [CrossRef] [PubMed]
  205. Ding, Z.; Dong, Z.; Chen, Z.; Hong, J.; Yan, L.; Li, H.; Yao, S.; Yan, Y.; Yang, Y.; Yang, C. Viral status and efficacy of immunotherapy in hepatocellular carcinoma: A systematic review with meta-analysis. Front. Immunol. 2021, 12, 733530. [Google Scholar] [CrossRef] [PubMed]
  206. Kelley, R.K.; Rimassa, L.; Cheng, A.-L.; Kaseb, A.; Qin, S.; Zhu, A.X.; Chan, S.L.; Melkadze, T.; Sukeepaisarnjaroen, W.; Breder, V. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2022, 23, 995–1008. [Google Scholar] [CrossRef]
  207. Chan, L.S.; Kudo, M.; Sangro, B.; Kelley, R.K.; Furuse, J.; Park, J.W.; Sunpaweravong, P.; Fasolo, A.; Yau, T.; Kawaoka, T.; et al. 714P Impact of viral aetiology in the phase III HIMALAYA study of tremelimumab (T) plus durvalumab (D) in unresectable hepatocellular carcinoma (uHCC). Ann. Oncol. 2022, 33, S869–S870. [Google Scholar] [CrossRef]
  208. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update. J. Hepatol. 2022, 76, 681–693. [Google Scholar] [CrossRef] [PubMed]
  209. Johnson, P.J.; Berhane, S.; Kagebayashi, C.; Satomura, S.; Teng, M.; Reeves, H.L.; O’Beirne, J.; Fox, R.; Skowronska, A.; Palmer, D. Assessment of liver function in patients with hepatocellular carcinoma: A new evidence-based approach—The ALBI grade. J. Clin. Oncol. 2015, 33, 550. [Google Scholar] [CrossRef]
  210. Roth, G.; Villeret, F.; Decaens, T.; Merle, P.; Nahon, P. Immunotherapy in hepatocellular carcinoma: How does underlying liver disease influence therapeutic strategy and outcomes? Liver Int. 2022, 43, 546–557. [Google Scholar] [CrossRef]
  211. Pinato, D.J.; Kaneko, T.; Saeed, A.; Pressiani, T.; Kaseb, A.; Wang, Y.; Szafron, D.; Jun, T.; Dharmapuri, S.; Naqash, A.R. Immunotherapy in hepatocellular cancer patients with mild to severe liver dysfunction: Adjunctive role of the ALBI grade. Cancers 2020, 12, 1862. [Google Scholar] [CrossRef] [PubMed]
  212. Kelley, R.; Yau, T.; Cheng, A.-L.; Kaseb, A.; Qin, S.; Zhu, A.; Chan, S.; Sukeepaisarnjaroen, W.; Breder, V.; Verset, G. VP10-2021: Cabozantinib (C) plus atezolizumab (A) versus sorafenib (S) as first-line systemic treatment for advanced hepatocellular carcinoma (aHCC): Results from the randomized phase III COSMIC-312 trial. Ann. Oncol. 2022, 33, 114–116. [Google Scholar] [CrossRef]
  213. Llovet, J.M.; Kudo, M.; Cheng, A.-L.; Finn, R.S.; Galle, P.R.; Kaneko, S.; Meyer, T.; Qin, S.; Dutcus, C.E.; Chen, E. Lenvatinib (len) plus pembrolizumab (pembro) for the first-line treatment of patients (pts) with advanced hepatocellular carcinoma (HCC): Phase 3 LEAP-002 study. J. Clin. Oncol. 2019, 37, TPS4152. [Google Scholar] [CrossRef]
  214. Qin, S.; Finn, R.S.; Kudo, M.; Meyer, T.; Vogel, A.; Ducreux, M.; Macarulla, T.M.; Tomasello, G.; Boisserie, F.; Hou, J. RATIONALE 301 study: Tislelizumab versus sorafenib as first-line treatment for unresectable hepatocellular carcinoma. Future Oncol. 2019, 15, 1811–1822. [Google Scholar] [CrossRef]
  215. Ren, Z.; Xu, J.; Bai, Y.; Xu, A.; Cang, S.; Du, C.; Li, Q.; Lu, Y.; Chen, Y.; Guo, Y. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): A randomised, open-label, phase 2–3 study. Lancet Oncol. 2021, 22, 977–990. [Google Scholar] [CrossRef] [PubMed]
  216. Guisier, F.; Cousse, S.; Jeanvoine, M.; Thiberville, L.; Salaun, M. A rationale for surgical debulking to improve anti-PD1 therapy outcome in non small cell lung cancer. Sci. Rep. 2019, 9, 16902. [Google Scholar] [CrossRef] [PubMed]
  217. Duraiswamy, J.; Freeman, G.J.; Coukos, G. Therapeutic PD-1 Pathway Blockade Augments with Other Modalities of Immunotherapy T-Cell Function to Prevent Immune Decline in Ovarian CancerPD-1 Blockade Restores Immune Function in Ovarian Cancer. Cancer Res. 2013, 73, 6900–6912. [Google Scholar] [CrossRef]
  218. Joseph, R.W.; Elassaiss-Schaap, J.; Kefford, R.; Hwu, W.-J.; Wolchok, J.D.; Joshua, A.M.; Ribas, A.; Hodi, F.S.; Hamid, O.; Robert, C. Baseline Tumor Size Is an Independent Prognostic Factor for Overall Survival in Patients with Melanoma Treated with PembrolizumabImpact of Baseline Tumor Size on Outcomes in Melanoma. Clin. Cancer Res. 2018, 24, 4960–4967. [Google Scholar] [CrossRef]
  219. Lee, J.H.; Lyle, M.; Menzies, A.M.; Chan, M.M.; Lo, S.; Clements, A.; Carlino, M.S.; Kefford, R.F.; Long, G.V. Metastasis-specific patterns of response and progression with anti-PD-1 treatment in metastatic melanoma. Pigment. Cell Melanoma Res. 2018, 31, 404–410. [Google Scholar] [CrossRef] [PubMed]
  220. Chardin, D.; Paquet, M.; Schiappa, R.; Darcourt, J.; Bailleux, C.; Poudenx, M.; Sciazza, A.; Ilie, M.; Benzaquen, J.; Martin, N. Baseline metabolic tumor volume as a strong predictive and prognostic biomarker in patients with non-small cell lung cancer treated with PD1 inhibitors: A prospective study. J. Immunother. Cancer 2020, 8, e000645. [Google Scholar] [CrossRef]
  221. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
  222. Nishino, M.; Giobbie-Hurder, A.; Ramaiya, N.H.; Hodi, F.S. Response assessment in metastatic melanoma treated with ipilimumab and bevacizumab: CT tumor size and density as markers for response and outcome. J. Immunother. Cancer 2014, 2, 40. [Google Scholar] [CrossRef]
  223. Yu, J.W.; Bhattacharya, S.; Yanamandra, N.; Kilian, D.; Shi, H.; Yadavilli, S.; Katlinskaya, Y.; Kaczynski, H.; Conner, M.; Benson, W. Tumor-immune profiling of murine syngeneic tumor models as a framework to guide mechanistic studies and predict therapy response in distinct tumor microenvironments. PLoS ONE 2018, 13, e0206223. [Google Scholar] [CrossRef] [PubMed]
  224. Clark, C.E.; Hingorani, S.R.; Mick, R.; Combs, C.; Tuveson, D.A.; Vonderheide, R.H. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 2007, 67, 9518–9527. [Google Scholar] [CrossRef] [PubMed]
  225. Colak, S.; Ten Dijke, P. Targeting TGF-β signaling in cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef] [PubMed]
  226. Hsieh, C.-L.; Chen, D.-S.; Hwang, L.-H. Tumor-induced immunosuppression: A barrier to immunotherapy of large tumors by cytokine-secreting tumor vaccine. Hum. Gene Ther. 2000, 11, 681–692. [Google Scholar] [CrossRef]
  227. Kim, H.S.; Kim, C.G.; Hong, J.Y.; Kim, I.-h.; Kang, B.; Jung, S.; Kim, C.; Shin, S.J.; Choi, H.J.; Cheon, J. The presence and size of intrahepatic tumors determine the therapeutic efficacy of nivolumab in advanced hepatocellular carcinoma. Ther. Adv. Med. Oncol. 2022, 14, 17588359221113266. [Google Scholar] [CrossRef]
  228. Woo, H.Y.; Rhee, H.; Yoo, J.E.; Kim, S.H.; Choi, G.H.; Kim, D.Y.; Woo, H.G.; Lee, H.S.; Park, Y.N. Lung and lymph node metastases from hepatocellular carcinoma: Comparison of pathological aspects. Liver Int. 2022, 42, 199–209. [Google Scholar] [CrossRef]
  229. Huppert, L.A.; Green, M.D.; Kim, L.; Chow, C.; Leyfman, Y.; Daud, A.I.; Lee, J.C. Tissue-specific Tregs in cancer metastasis: Opportunities for precision immunotherapy. Cell. Mol. Immunol. 2022, 19, 33–45. [Google Scholar] [CrossRef]
  230. Virtue, A.T.; McCright, S.J.; Wright, J.M.; Jimenez, M.T.; Mowel, W.K.; Kotzin, J.J.; Joannas, L.; Basavappa, M.G.; Spencer, S.P.; Clark, M.L.; et al. The gut microbiota regulates white adipose tissue inflammation and obesity via a family of microRNAs. Sci. Transl. Med. 2019, 11, aav1892. [Google Scholar] [CrossRef]
  231. Livanos, A.E.; Greiner, T.U.; Vangay, P.; Pathmasiri, W.; Stewart, D.; McRitchie, S.; Li, H.; Chung, J.; Sohn, J.; Kim, S.; et al. Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat. Microbiol. 2016, 1, 16140. [Google Scholar] [CrossRef]
  232. Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86. [Google Scholar] [CrossRef]
  233. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  234. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed]
  235. Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef] [PubMed]
  236. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [PubMed]
  237. Jin, Y.; Dong, H.; Xia, L.; Yang, Y.; Zhu, Y.; Shen, Y.; Zheng, H.; Yao, C.; Wang, Y.; Lu, S. The Diversity of Gut Microbiome is Associated With Favorable Responses to Anti–Programmed Death 1 Immunotherapy in Chinese Patients With NSCLC. J. Thorac. Oncol. 2019, 14, 1378–1389. [Google Scholar] [CrossRef]
  238. Tinsley, N.; Zhou, C.; Tan, G.; Rack, S.; Lorigan, P.; Blackhall, F.; Krebs, M.; Carter, L.; Thistlethwaite, F.; Graham, D.; et al. Cumulative Antibiotic Use Significantly Decreases Efficacy of Checkpoint Inhibitors in Patients with Advanced Cancer. Oncologist 2019, 25, 55–63. [Google Scholar] [CrossRef] [PubMed]
  239. Zheng, Y.; Wang, T.; Tu, X.; Huang, Y.; Zhang, H.; Tan, D.; Jiang, W.; Cai, S.; Zhao, P.; Song, R.; et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. ImmunoTherapy Cancer 2019, 7, 193. [Google Scholar] [CrossRef]
  240. Pinato, D.J.; Howlett, S.; Ottaviani, D.; Urus, H.; Patel, A.; Mineo, T.; Brock, C.; Power, D.; Hatcher, O.; Falconer, A.; et al. Association of Prior Antibiotic Treatment With Survival and Response to Immune Checkpoint Inhibitor Therapy in Patients With Cancer. JAMA Oncol. 2019, 5, 1774–1778. [Google Scholar] [CrossRef]
  241. Derosa, L.; Hellmann, M.D.; Spaziano, M.; Halpenny, D.; Fidelle, M.; Rizvi, H.; Long, N.; Plodkowski, A.J.; Arbour, K.C.; Chaft, J.E.; et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann. Oncol. 2018, 29, 1437–1444. [Google Scholar] [CrossRef]
  242. Sangro, B.; Park, J.; Finn, R.; Cheng, A.; Mathurin, P.; Edeline, J.; Kudo, M.; Han, K.; Harding, J.; Merle, P.; et al. LBA-3 CheckMate 459: Long-term (minimum follow-up 33.6 months) survival outcomes with nivolumab versus sorafenib as first-line treatment in patients with advanced hepatocellular carcinoma. Ann. Oncol. Off. J. Eur. Soc. Med Oncol. 2020, 31, S241–S242. [Google Scholar] [CrossRef]
  243. Chan, S.; Kudo, M.; Sangro, B.; Kelley, R.; Furuse, J.; Park, J.-W.; Sunpaweravong, P.; Fasolo, A.; Yau, T.; Kawaoka, T.; et al. 67O Outcomes in the Asian subgroup of the phase III HIMALAYA study of tremelimumab (T) plus durvalumab (D) in unresectable hepatocellular carcinoma (uHCC). Ann. Oncol. 2022, 33, S1456. [Google Scholar] [CrossRef]
  244. Vom Steeg, L.G.; Klein, S.L. SeXX matters in infectious disease pathogenesis. PLoS Pathog. 2016, 12, e1005374. [Google Scholar] [CrossRef] [PubMed]
  245. Conforti, F.; Pala, L.; Bagnardi, V.; De Pas, T.; Martinetti, M.; Viale, G.; Gelber, R.D.; Goldhirsch, A. Cancer immunotherapy efficacy and patients’ sex: A systematic review and meta-analysis. Lancet Oncol. 2018, 19, 737–746. [Google Scholar] [CrossRef] [PubMed]
  246. Wu, Y.; Ju, Q.; Jia, K.; Yu, J.; Shi, H.; Wu, H.; Jiang, M. Correlation between sex and efficacy of immune checkpoint inhibitors (PD-1 and CTLA-4 inhibitors). Int. J. Cancer 2018, 143, 45–51. [Google Scholar] [CrossRef] [PubMed]
  247. Lyu, N.; Yi, J.-Z.; Zhao, M. Immunotherapy in older patients with hepatocellular carcinoma. Eur. J. Cancer 2022, 162, 76–98. [Google Scholar] [CrossRef] [PubMed]
  248. Yang, H.; Ma, W.; Sun, B.; Fan, L.; Xu, K.; Hall, S.R.; Al-Hurani, M.F.; Schmid, R.A.; Peng, R.-W.; Hida, T. Smoking signature is superior to programmed death-ligand 1 expression in predicting pathological response to neoadjuvant immunotherapy in lung cancer patients. Transl. Lung Cancer Res. 2021, 10, 3807. [Google Scholar] [CrossRef]
  249. Wang, X.; Ricciuti, B.; Nguyen, T.; Li, X.; Rabin, M.S.; Awad, M.M.; Lin, X.; Johnson, B.E.; Christiani, D.C. Association between smoking history and tumor mutation burden in advanced non–small cell lung cancer. Cancer Res. 2021, 81, 2566–2573. [Google Scholar] [CrossRef]
  250. Wang, Y.H.; Chuang, Y.H.; Wu, C.F.; Jan, M.C.; Wu, W.J.; Lin, C.L.; Liu, C.J.; Yang, Y.C.; Chen, P.J.; Lin, S.M. Smoking and hepatitis B virus–related hepatocellular carcinoma risk: The mediating roles of viral load and alanine aminotransferase. Hepatology 2019, 69, 1412–1425. [Google Scholar] [CrossRef]
Figure 1. Schematic figure representing predictive biomarkers of immunotherapy in HCC.
Figure 1. Schematic figure representing predictive biomarkers of immunotherapy in HCC.
Ijms 24 07640 g001
Table 1. HR of subgroups related to predict OS in recent phase 3 studies for immunotherapy in HCC (Immunotherapy versus sorafenib).
Table 1. HR of subgroups related to predict OS in recent phase 3 studies for immunotherapy in HCC (Immunotherapy versus sorafenib).
IMBRAVE150 [44]CHECKMATE 459 [6,242]HIMALAYA [45,207,243]
(STRIDE/Durvalumab)
Age ≥ 650.69 (0.49–0.98)0.88 (0.68–1.12)0.73 (0.58–0.93)/0.83 (0.66–1.06)
Male0.64 (0.49–0.83) 0.73 (0.61–0.88)/0.86 (0.72–1.03)
Asian0.62 (0.42–0.93)0.74 (0.56–0.98)0.68 (0.52–0.89)/0.83 (0.64–1.06)
ECOG 10.56 (0.39–0.80) 0.74 (0.57–0.95)/0.85 (0.66–1.10)
BCLC C0.63 (0.48–0.82)0.78 (0.65–0.95)0.76 (0.63–0.91)/0.86 (0.72–1.03)
AFP
 < 400 ng/mL0.58 (0.42–0.81)0.98 (0.78–1.24)0.82 (0.63–1.05)/0.78 (0.61–1.01)
 ≥ 400 ng/mL0.77 (0.53–1.12)0.67 (0.51–0.88)0.64 (0.45–0.91)/0.73 (0.53–1.03)
MVI: No0.66 (0.47–0.92) 0.77 (0.63–0.93)/0.87 (0.72–1.05)
EH spread: Yes0.60 (0.44–0.81) 0.67 (0.53–0.84)/0.79 (0.64–0.98)
MVI and/orEH spread: YES0.64 (0.49–0.85)0.74 (0.61–0.90)0.73 (0.59–0.89)/0.82 (0.67–1.00)
HBV0.58 (0.40–0.83)0.79 (0.59–1.07)0.64 (0.48–0.86)/0.78 (0.58–1.04)
HCV0.43 (0.25–0.73)0.72 (0.51–1.02)1.06 (0.76–1.49)/1.05 (0.75–1.48)
PD-L1 expression
 TC or IC ≥ 1% (SP263)0.52 (0.32–0.87)
 TC ≥ 1% (28-8, Dako) 0.80 (0.54–1.19)
 TC ≥ 1% (SP263) 0.85 (0.65–1.11)/0.87 (0.66–1.13)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ji, J.H.; Ha, S.Y.; Lee, D.; Sankar, K.; Koltsova, E.K.; Abou-Alfa, G.K.; Yang, J.D. Predictive Biomarkers for Immune-Checkpoint Inhibitor Treatment Response in Patients with Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 7640. https://doi.org/10.3390/ijms24087640

AMA Style

Ji JH, Ha SY, Lee D, Sankar K, Koltsova EK, Abou-Alfa GK, Yang JD. Predictive Biomarkers for Immune-Checkpoint Inhibitor Treatment Response in Patients with Hepatocellular Carcinoma. International Journal of Molecular Sciences. 2023; 24(8):7640. https://doi.org/10.3390/ijms24087640

Chicago/Turabian Style

Ji, Jun Ho, Sang Yun Ha, Danbi Lee, Kamya Sankar, Ekaterina K. Koltsova, Ghassan K. Abou-Alfa, and Ju Dong Yang. 2023. "Predictive Biomarkers for Immune-Checkpoint Inhibitor Treatment Response in Patients with Hepatocellular Carcinoma" International Journal of Molecular Sciences 24, no. 8: 7640. https://doi.org/10.3390/ijms24087640

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

Article Metrics

Back to TopTop