Next Article in Journal
Efficacy of Thermal Ablation for Small-Size (0–3 cm) versus Intermediate-Size (3–5 cm) Colorectal Liver Metastases: Results from the Amsterdam Colorectal Liver Met Registry (AmCORE)
Next Article in Special Issue
Advancements in the Treatment Landscape of Hepatocellular Carcinoma
Previous Article in Journal
Whole Breast Irradiation in Comparison to Endocrine Therapy in Early Stage Breast Cancer—A Direct and Network Meta-Analysis of Published Randomized Trials
Previous Article in Special Issue
Bilobar Radioembolization Carries the Risk of Radioembolization-Induced Liver Disease in the Treatment of Advanced Hepatocellular Carcinoma: Safety and Efficacy Comparison to Systemic Therapy with Atezolizumab/Bevacizumab
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Predictive Biomarkers of Systemic Drug Therapy for Hepatocellular Carcinoma: Anticipated Usefulness in Clinical Practice

Department of Hepatology, Iizuka Hospital, 3-83 Yoshio-machi, Iizuka, Fukuoka 820-8505, Japan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(17), 4345; https://doi.org/10.3390/cancers15174345
Submission received: 7 July 2023 / Revised: 21 August 2023 / Accepted: 25 August 2023 / Published: 30 August 2023

Abstract

:

Simple Summary

A number of agents, including immune checkpoint inhibitors, have become available for the treatment of hepatocellular carcinoma, but the objective response rate of these drugs is currently only 30% to 40%. Therefore, the identification of new predictive biomarkers and an increased knowledge of the mechanisms of response or resistance to systemic chemotherapies are required.

Abstract

In the systemic drug treatment of hepatocellular carcinoma, only the tyrosine kinase inhibitor (TKI) sorafenib was available for a period. This was followed by the development of regorafenib as a second-line treatment after sorafenib, and then lenvatinib, a new TKI, proved non-inferiority to sorafenib and became available as a first-line treatment. Subsequently, cabozantinib, another TKI, was introduced as a second-line treatment, along with ramucirumab, the only drug proven to be predictive of therapeutic efficacy when AFP levels are >400 ng/mL. It is an anti-VEGF receptor antibody. More recently, immune checkpoint inhibitors have become the mainstay of systemic therapy and can now be used as a first-line standard treatment for HCC. However, the objective response rate for these drugs is currently only 30% to 40%, and there is a high incidence of side effects. Additionally, there are no practical biomarkers to predict their therapeutic effects. Therefore, this review provides an overview of extensive research conducted on potential HCC biomarkers from blood, tissue, or imaging information that can be used in practice to predict the therapeutic efficacy of systemic therapy before its initiation.

1. Introduction

Liver cancer is a major cause of death worldwide, and the number of people diagnosed with liver cancer is expected to increase [1]. As the principal histologic type of liver cancer, hepatocellular carcinoma (HCC) is responsible for the majority (approximately 75%) of liver cancer diagnoses and deaths [2]. Treatment options for localized HCC, such as surgical resection, ablation, liver transplantation, and transarterial chemoembolization, were established in the 20th century, but effective drug therapy for advanced HCC did not emerge until 2007 [3]. Although many clinical trials of potential drug therapies for unresectable HCC were conducted before the introduction of tyrosine kinase inhibitors (TKIs), no chemotherapeutic drugs demonstrated any significant survival benefit, as shown in the meta-analysis by Mathurin et al. [4]. Subsequently, an advanced understanding of the mechanisms of tumor cell proliferation and angiogenesis supported the development of the TKI, sorafenib [5]. In the Sorafenib HCC Assessment Randomized Protocol (SHARP) clinical trial of unresectable HCC, sorafenib showed a clear survival benefit over placebo and became the standard treatment for unresectable HCC in 2007 [6]. Subsequently, the results of the RESORSE trial, a clinical trial limited to unresectable HCC patients who tolerated sorafenib, led to the approval of regorafenib as a second-line therapy after sorafenib treatment in 2017 [7]. In 2018, lenvatinib was demonstrated to be non-inferior to sorafenib in the REFLECT trial, leading to a choice of sorafenib or lenvatinib as the first-line therapy [8]. In 2019, based on the results of the REACH-2 trial of cases with alpha-fetoprotein (AFP) levels of 400 ng/mL or more after sorafenib treatment, ramucirumab (an anti-vascular endothelial growth factor (VEGF) receptor antibody) became available as a second-line therapy [9]. Additionally, cabozantinib, a TKI, was developed as a second-line or later therapy [10].
In the following years, immune checkpoint inhibitors (ICIs) emerged. In 2017, nivolumab demonstrated promising results in the Checkmate040 phase I/II trial [11], leading to its approval by the Food and Drug Administration. Additionally, pembrolizumab was approved by the Food and Drug Administration as a first-line and second-line treatment based on the results of the phase II KEYNOTE-224 [12] and phase III KEYNOTE-240 trials [13].
In 2020, a combination therapy of atezolizumab and bevacizumab, which surpassed sorafenib in clinical trials, was introduced [14]. As a result, both sorafenib and lenvatinib were relegated to second-line or later therapies. Furthermore, a combination therapy of two ICIs, durvalumab and tremelimumab (STRIDE regimen), outperformed sorafenib in the treatment results of the HIMALAYA trial. Durvalumab monotherapy also showed non-inferiority to sorafenib [15], allowing these therapeutics to be added as new treatment options.
Through these trials, a number of agents, including ICIs, have become available for the treatment of HCC. However, the objective response rate (ORR) of these drugs is currently only 30% to 40%, with a high incidence of side effects [6,7,8,9,10,11,12,13,14,15]. Other than ramucirumab, which requires an AFP level of 400 ng/mL or more, there are no practical biomarkers to predict the therapeutic effects of these treatment methods.
Most of the systemic therapies for HCC are administered in general hospitals without research facilities. In such hospitals, imaging tests such as computed tomography and magnetic resonance imaging (MRI), and pathological diagnoses, including tumor tissue sampling and immunohistochemistry (IHC) staining, can be conducted. However, the introduction of advanced technologies, such as the analysis of tumor genomic or transcriptomic profiles, miRNA evaluation, cell cultures, and the identification of driver gene mutations, is often challenging. These limitations arise due to constraints in facilities, personnel, and financial implications.
In this review, we have provided an overview of potential biomarkers that, with further prospective validation, could be translated into clinical applications in the future. The aim of our review is to offer readers a comprehensive perspective, enabling them to assess which of these potential biomarkers might be feasibly implemented within their specific hospital environments.

2. Exploring Potential Biomarkers to Predict the Therapeutic Effects of TKIs (Table 1)

2.1. Potential Biomarkers for Sorafenib

The first TKI, sorafenib, has remained the first drug of choice for a decade, leading to numerous early studies being conducted on biomarkers to predict its therapeutic effect.
One of the objectives of the phase III SHARP trial was to investigate plasma biomarkers to predict prognoses and therapeutic effects, with 10 plasma biomarkers measured at baseline and after 12 weeks of treatment [16]. As a result, baseline angiopoietin 2 (Ang-2) and VEGF concentrations were found to be independent predictors of survival in the entire advanced HCC patient population. However, these were not unique to the sorafenib cohort and were similar to the placebo cohort [16].
In a real-world study in Japan, Miyahara et al. measured the serum levels of eight pro-angiogenic cytokines (Ang-2, follistatin (FST), granulocyte colony-stimulating factor (G-CSF), hepatocyte growth factor (HGF), leptin, platelet-derived growth factor-BB (PDGF-BB), platelet endothelial cell adhesion molecule-1 (PECAM-1), and VEGF) in 120 consecutive HCC patients treated with sorafenib. They reported that high expression of Ang-2 or three or more pro-angiogenic cytokines was associated with poor progression-free survival (PFS) and overall survival (OS) in patients treated with sorafenib [17]. In addition, the presence of macrovascular invasion (MVI) was also shown to be associated with poor OS related to clinical parameters [17].
In an analysis of pooled data from the SHARP and Asia Pacific (AP) phase III trials, a significantly greater OS benefit compared with placebo was observed in patients without extrahepatic spread (EHS; hazard ratio (HR), 0.55 vs. 0.84), with hepatitis C virus (HCV) (HR, 0.47 vs. 0.81), and a low neutrophil-to-lymphocyte ratio (NLR) (HR, 0.59 vs. 0.84) [18]. In this analysis, the NLR was divided into >3.0 and <3.0, which was the median in the sorafenib administration group. Although this was a retrospective study, it described certain factors that had significant differences compared with the placebo group. However, these findings were strictly predictions of the survival period and not of drug effectiveness.
The NLR reflects the inflammatory response to cancer, and its elevation is recognized as an indicator of poor prognosis [19,20]. Qi et al. conducted a systematic review and meta-analysis of 20,475 HCC patients from 90 articles to explore the prognostic role of NLR in HCC and reported that a lower baseline NLR was significantly associated with the survival period (HR, 1.80, 95% confidence interval (CI): 1.59–2.04, p < 0.00001) [21]. This study also conducted a subgroup meta-analysis for cases treated with sorafenib, and although it was not compared with a placebo group, a low NLR was associated with better survival in 170 cases between high and low NLR groups [21].
The GIDEON trial, a large prospective observational registration study focused on assessing the safety of sorafenib treatment in the real world, showed better OS in Child–Pugh A patients compared with Child–Pugh B patients. Moreover, a univariate Cox regression analysis of each factor of the Child–Pugh score showed that albumin and bilirubin, which form the ALBI score, strongly influenced OS [22]. The authors did not utilize anything other than descriptive statistics that considered the impact of selection bias. Additionally, this study lacked a control group and was not randomized. However, liver functional reserve can clearly affect the patient’s survival period after treatment with sorafenib for HCC. For example, several studies have shown that in advanced HCC cases related to HCV, OS is extended as the liver functional reserve improves following HCV eradication with interferon (IFN) or direct-acting antivirals [23,24].
For pathological biomarkers in tumor tissues, phase II trial results indicated that phosphorylated ERK might be a useful biomarker to predict the prognosis of patients treated with sorafenib. This protein is located downstream of Raf kinase in the MAPK cascade, which is a major target of sorafenib [25]. Although there have been studies in clinical settings that have shown favorable efficacy [26], several have also shown unfavorable efficacy [27,28]. Therefore, no consensus has been reached regarding phosphorylated ERK.
Arao et al. reported that in 13 cases with significant tumor shrinkage after sorafenib treatment, fibroblast growth factor (FGF)3/FGF4 amplification was observed in the tumor genome. Additionally, multiple lung metastases in poorly differentiated histological types were seen as clinical pathological features. Although the sample size was relatively small, FGF3/FGF4-amplified tumors were frequently observed in responders to sorafenib [29]. Although this study examined biomarkers for sorafenib treatment efficacy, no further research with additional cases to support these findings was conducted.
Tumor tissues can also be used to investigate microRNA (miRNA) expression. miRNAs are small endogenous non-coding RNAs that inhibit translation or support cleavage of mRNAs to negatively regulate gene expression. These molecules are highly stable and can be reliably detected in stored clinical samples and cell cytology specimens, making them ideal biomarker candidates [30,31,32]. Various miRNAs are also mechanistically involved in the development, proliferation, and progression of HCC and can be detected in serum and plasma samples, suggesting they might be used as diagnostic markers [33].
Gyöngyösi et al. investigated the expression levels of 14 miRNAs in 20 HCC cases where tumor tissue samples were collected by fine needle aspiration before sorafenib administration, with the data demonstrating the high expression of miR-224 was associated with increased PFS and OS rates [31].
Vaira et al. conducted a comprehensive profiling of approximately 700 miRNAs in a series of 26 HCC patients treated with sorafenib (training set) using tumor tissues collected prior to treatment, then verified the results in an independent series of 58 patients (validation set) [34]. As a result, six miRNAs were found to be significantly associated with clinical variables in the training set. Of these, only miR-425-3p was significant in the validation set, with high miR-425-3p levels being associated with a longer time to progression (TTP) and PFS [34]. However, no follow-up studies have been conducted to date. In general, miRNA-related cancer research is primarily focused on the development of therapies that target specific miRNAs or the use of these molecules as a tool for early cancer detection [35].
Other studies have constructed high-throughput assay systems in completely different ways. Qiu et al. created a Liver Cancer Model Repository (LIMORE) panel of 81 cell lines by creating 50 patient-derived liver cancer cell lines, in addition to 31 existing cell lines, to model HCC heterogeneity. The authors examined the sensitivity of these cells to a total of 90 drugs. By using this panel, which has verified gene mutations and gene expression characteristics, it is possible to identify gene–drug interactions of therapeutic methods and biomarker candidates. When predicting the effect of sorafenib treatment, Dickkopf-1 (DKK-1) was identified as a potentially useful biomarker [36]. Interestingly, DKK-1 is a secreted protein that antagonizes Wnt signal transduction, which is known to affect ICI efficacy [37]. Because DKK-1 is a serum protein, it may be relatively easy to verify its potential as a biomarker in existing cohorts with preserved serum samples.

2.2. Potential Biomarkers for Regorafenib

Although insufficient results were obtained from the biomarker studies of the phase III SHARP trial for sorafenib, a more comprehensive exploratory biomarker analysis was conducted for patients in the RESORCE trial at the DNA, RNA, and protein levels [38]. Of the 266 proteins studied in baseline plasma samples, decreases in five, Ang-1, cystatin B, the latency-associated peptide of transforming growth factor beta 1 (LAP TGF-β1), oxidized low-density lipoprotein receptor 1 (LOX-1), and C-C motif chemokine ligand 3 (MIP-1α), were found to be associated with extended TTP and OS. Moreover, nine plasma miRNAs, miR-30a, miR-122, miR-125b, miR-200a, miR-374b, miR-15b, miR-107, miR-320, and miR-645, were related to OS, although none were associated with TTP. Furthermore, there was no apparent correlation between the AFP or c-MET protein expression levels and the OS or TTP benefits of regorafenib treatment, causing them to be excluded as potential predictive biomarkers [38]. Currently, with treatments including ICIs becoming the standard of care as first-line treatments, the number of cases where regorafenib is used after sorafenib treatment is likely to be low. This could make planning prospective validation studies challenging. However, as these potential biomarkers are plasma proteins and miRNAs, it is hoped that they can be validated in existing cohorts using blood samples that have been preserved.

2.3. Signaling Pathways as Biomarkers for TKIs: Insights from Trials with mTOR and MET Inhibitors

In several cancer types, other than HCC, driver gene mutations in signaling pathways strongly promote tumor growth. This has led to established biomarker-driven treatment concepts for drug selection and predicting treatment outcomes [39,40,41]. In HCC, signaling pathways such as RAS, mammalian target of rapamycin (mTOR), MET, and FGF-19 have been considered potential therapeutic targets. Several clinical trials were conducted using inhibitors targeting these pathways, but unfortunately, many ended with disappointing results [42]. For example, mTOR signaling is activated in about half of all HCC cases and is associated with worse outcomes [43]. Despite this strong theoretical basis for using the mTOR inhibitor everolimus to treat HCC, the final results from a phase III trial did not suggest any trend of prolonged OS (everolimus vs. placebo, 7.6 vs. 7.3 months) [44].
The potential cause of failure in these clinical trials may be the inclusion of all patients with unresectable HCC. It is considered desirable to incorporate molecular selection factors into prospective research, as in clinical trials for other cancers [45]. One of the drugs for which there was hope for biomarker-driven treatment in HCC was tivantinib, an MET inhibitor. In a phase II trial against placebo as a second-line treatment, tivantinib improved survival rates in patients with high tumor MET expression levels although no significant effect was observed in all cases [46]. From these results, phase III trials (METIV-HCC, JET-HCC) were conducted comparing tivantinib and placebo only in patients with high MET expression [47,48]. However, in both trials, no statistically significant treatment effect was observed for tivantinib compared with placebo.
Cabozantinib, which targets several TKs including MET, VEGF, and AXL, was successful in a phase III trial (CELESTIAL) [10]. In this trial, baseline plasma levels of MET, AXL, VEGFR2, HGF, GAS6, VEGF-A, PlGF, IL-8, EPO, ANG2, IGF-1, VEGF-C, and c-KIT were evaluated as biomarkers; however, none of these predicted the treatment effect of cabozantinib on OS or PFS [49].
The cause of HCC is diverse, including viral infection, toxin exposure, and metabolic disorders. From the results of large-scale genomic analyses, it has become clear that gene mutations in HCC are centered on diverse non-drug targetable mutations, such as TERT, CTNNB1, and TP53 [50,51]. Furthermore, HCC is heterogeneous even within an individual patient, and sequencing analysis of a single lesion cannot fully characterize the genomic features of HCC in certain cases [52]. In clinical trials of tyrosine kinase inhibitors, the failure to successfully use the expression of specific therapeutic target molecules or the activation of signaling pathways as biomarkers suggests that hepatocellular carcinoma may have low dependency on these signaling pathways, reflecting its underlying heterogeneity and diversity [53].

2.4. Potential Biomarkers for Lenvatinib

Lenvatinib is a multi-kinase inhibitor that inhibits vascular endothelial growth factor receptors (VEGFR) 1–3, fibroblast growth factor receptors (FGFR) 1–4, platelet-derived growth factor receptor (PDGFR) α, and oncogenes RET and KIT [54]. Preclinical studies have shown that lenvatinib has strong anti-angiogenic activity, primarily through inhibition of the VEGF and FGF signaling pathways [55].
In a subgroup analysis of the REFLECT trial, patients with HBV infection or alcohol as underlying factors showed better PFS rates with lenvatinib treatment than with sorafenib [8]. However, no biomarker exploration beyond the subgroup analysis was planned in the REFLECT trial.
Tada et al. focused on the associations between outcomes in HCC patients treated with lenvatinib and the NLR. In a multivariate analysis of a cohort of 237 individuals, an NLR ≥ 4 was independently associated with OS and PFS. There was also a significant difference in the disease control rate between patients with low NLR (<4) and high NLR (≥4) (85.5% vs. 67.3%, p = 0.007). A spline curve analysis showed that an NLR of approximately 3.0 to 4.5 was an appropriate cutoff value related to OS [56]. In addition, in the retrospective RELEVANT study from 23 other facilities, data were collected for 1325 patients treated with lenvatinib. In the multivariate analysis of OS, HBsAg positivity, NLR > 3, and AST > 38 were independently associated with poor prognosis in all three groups. Furthermore, nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH)-related etiology was independently associated with a good prognosis. The multivariate analysis showed that NAFLD/NASH, Barcelona Clinic Liver Cance (BCLC) stage, NLR, and AST were independent prognostic factors for PFS in cases treated with lenvatinib [57]. However, these studies did not make comparisons with placebo or other drug treatments. Of note, a control group or placebo group is necessary to identify predictors of the therapeutic effect of a certain drug; thus, it is necessary to consider the limitations of such single-arm observational studies when evaluating predictive markers.
Qiu et al., who identified DKK-1 as a potential predictor of sorafenib effectiveness, used a panel of 81 HCC cell lines from the Liver Cancer Model Repository to show that FGFR inhibitors including lenvatinib had a favorable effect on HCC strains with the amplification of FGFR and FGF. They suggested that the amplification of FGF19 and FGFR might be biomarkers for lenvatinib effectiveness [36].
Myojin et al. developed a new HCC mouse model that reproduced the diversity of tumor driver genes by introducing a pooled cancer gene cDNA library using transposon-based intrahepatic delivery. This could be used to simultaneously evaluate the individual effects of various genetic drivers on TKI sensitivity in HCC in vivo. This model revealed that tumors expressing FGF19 were sensitive to lenvatinib in vivo. They comprehensively evaluated tumor secretory proteins to discover biomarkers for FGF19-driven HCC, identifying a correlation between FGF19 and the secretory protein ST6 β-galactoside α-2,6-sialyltransferase 1 (ST6GAL1) in HCC cells. This provided clinical evidence that ST6GAL1 may be a useful serum biomarker for the selection of HCC patients who may derive more benefit from lenvatinib than sorafenib treatment [58]. This study strongly focused on the exploration of biomarkers for the therapeutic effect of lenvatinib in cancer cells, but validation in a prospective cohort is necessary for clinical application. Because ST6GAL1 protein levels can be measured in serum samples, it would be a very useful biomarker if validated.
Lenvatinib was reported to have a higher selectivity for FGFR compared with other kinase inhibitors [54,59]. Therefore, biomarkers related to FGF-FGFR signaling may be more promising than those related to other signaling pathways that have been previously studied.
Table 1. Factors influencing patient prognosis or efficacy when treating hepatocellular carcinoma with tyrosine kinase inhibitors.
Table 1. Factors influencing patient prognosis or efficacy when treating hepatocellular carcinoma with tyrosine kinase inhibitors.
TherapeuticsStudy DesignNumber of CasesPrognostic and Predictive
Factors
OutcomeStatistical Analysis
HR (95% CI)
p-ValueAuthors
[Reference No.]
SorafenibRetrospective, single-arm120High serum Ang-2PFS ↓Univariate
1.84 (1.21–2.81)
0.004Miyahara K et al.
[17]
OS ↓Multivariate
1.83 (1.12–2.98)
0.014
High angiogenic group *
*: patients with >three serum cytokines
(Ang-2, FST, G-CSF, HGF, Leptin,
PDGF-BB, PECAM-1, or VEGF)
PFS ↓Univariate
1.98 (1.30–3.06)
0.001
OS ↓Multivariate
1.76 (1.07–2.94)
0.023
MVI (present)OS ↓Multivariate
2.27 (1.36–3.72)
0.001
SorafenibRetrospective pooled analysis of two phase 3 trials (vs. placebo)Sorafenib 448
Placebo 379
Without EHSOS ↑Multivariate
0.55 (0.42–0.72)
0.015Bruix J et al.
[18]
With HCVOS ↑Multivariate
0.47 (0.32–0.69)
0.035
Low NLROS ↑Multivariate
0.59 (0.46–0.77)
0.0497
SorafenibSubgroup meta-analyses,
single-arm
170Low NLROS ↑Univariate
1.49 (1.17–1.91)
0.001Qi X et al.
[20]
SorafenibObservational registry, single-arm3371Child–Pugh AOS ↑Kaplan–MeierN/AMarrero JA et al.
[22]
BilirubinOSUnivariate
1.71 (1.57–1.86)
N/A
AlbuminOSUnivariate
1.76 (1.63–1.89)
N/A
SorafenibRetrospective, single-arm,
HCV patients only
103HCV eradicationOS ↑Multivariate
0.46 (0.26–0.78)
0.004Kuwano A et al.
[23]
ALBI scoreOSMultivariate
2.29 (1.20–4.37)
0.012
SorafenibPopulation-based retrospective cohort,
HCV patients only, single-arm
1684DAA userOS ↑Univariate
PSM univariate
<0.0001
<0.0001
Tsai H-Y et al.
[24]
SorafenibRetrospective, single-arm55FGF3/FGF4 amplification
(Frozen tumor tissue)
CR/PR ↑Fisher’s exact0.006Arao T et al.
[29]
Multiple lung metastasesCR/PR ↑Fisher’s exact0.006
SorafenibRetrospective, single-arm20High miR-224 expression
(FFPE tumor tissue)
PFS ↑Univariate
0.28 (0.09–0.92)
0.029Gyöngyösi B et al. [31]
OS ↑Univariate
0.24 (0.07–0.79)
0.012
SorafenibRetrospective, single-armTraining 26
Validation 58
High miR-425-3p expression
(FFPE tumor tissue)
TTP ↑Multivariate
0.4 (0.1–0.7)
0.002Vaira V et al.
[34]
PFS ↑Multivariate
0.3 (0.1–0.7)
0.0012
SorafenibRetrospective validation
of the pharmacogenomics panel, single-arm
54High serum DKK-1PFS ↑Univariate0.0396Qiu Z et al.
[36]
OS ↑Univariate0.0171
RegorafenibRetrospective pooled analysis of the phase 3 trial (vs. placebo)Protein cohort
Regorafenib 332
Placebo 167
Plasma ANG-1
(1 ng/mL increase)
OS ↓Multivariate
1.12 (1.05–1.19)
0.019Teufel M et al.
[38]
TTP ↓Multivariate
1.10 (1.04–1.17)
0.017
Low plasma Cystatin-B
(2-fold increase)
OS ↓Multivariate
1.46 (1.15–1.85)
0.04
TTP ↓Multivariate
1.42 (1.14–1.77)
0.018
Low plasma LAP TGF-β1
(2-fold increase)
OS ↓Multivariate
1.36 (1.12–1.65)
0.04
TTP ↓Multivariate
1.41 (1.18–1.68)
0.004
Low plasma LOX-1
(1 ng/mL increase)
OS ↓Multivariate
1.35 (1.16–1.57)
0.009
TTP ↓Multivariate
1.78 (1.33–2.39)
0.003
Low plasma MIP-1α
(1 pg/mL increase)
OS ↓Multivariate
1.02 (1.01–1.04)
0.04
TTP ↓Multivariate
1.02 (1.00–1.03)
0.043
miRNA cohort
Regorafenib 234
Placebo 109
Decreased miR-15bOS ↑Multivariate
0.37 (0.20–0.70)
0.002
Decreased miR-107OS ↑Multivariate
0.54 (0.37–0.81)
0.003
Decreased miR-320bOS ↑Multivariate
0.57 (0.41–0.81)
0.001
Increased miR-122OS ↑Multivariate
1.35 (1.14–1.60)
0.0004
Increased miR-374bOS ↑Multivariate
1.36 (1.11–1.65)
0.002
Increased miR-200aOS ↑Multivariate
1.39 (1.15–1.68)
0.001
Increased miR-30aOS ↑Multivariate
1.47 (1.14–1.88)
0.003
Increased miR-125bOS ↑Multivariate
1.54 (1.19–1.99)
0.001
Absence miR-645 *
(* dichotomized analysis, not vs. placebo)
OS ↑Multivariate
3.16 (1.52–6.55)
0.002
LenvatinibSubgroup analysis of the open-label phase 3 trial
(vs. sorafenib)
Lenvatinib 478
(HBV 251,
Alcohol 36)
sorafenib 476
(HBV 228,
Alcohol 21)
HBVPFS ↑Univariate
0.62 (0.50–0.75)
N/AKudo M et al.
[8]
AlcoholPFS ↑Univariate
0.27 (0.11–0.66)
N/A
LenvatinibRetrospective, single-arm237NLR ≥ 4OS ↓Multivariate
1.87 (1.10–3.12)
0.021Tada T et al.
[56]
PFS ↓Multivariate
1.90 (1.27–2.84)
0.002
DCR ↓Chi-square test?0.007
AFP ≥ 400 ng/mLOS ↓Multivariate
1.97 (1.19–3.27)
0.009
mALBI grade 2b or 3OS ↓Multivariate
2.12 (1.27–3.56)
0.004
BCLC stage ≥ CPFS ↓Multivariate
1.52 (1.03–2.24)
0.036
LenvatinibRetrospective, single-arm1325HBVOS ↓Multivariate
1.56 (1.13–2.17) *
0.0071 *Casadei-Gardini A et al.
[57]
*: Data are from the model 1 of 3 multivariate analyses.
NAFLD/NASHOS ↑Multivariate
0.58 (0.33–0.98) *
0.0044 *
PFS ↑Multivariate
0.87 (0.75–0.93)
0.0090
BCLC stage COS ↓Multivariate
1.64 (1.19–2.27) *
0.0027 *
PFS ↓Multivariate
1.33 (1.14–1.55)
0.0002
NLR > 3OS ↓Multivariate
1.95 (1.46–2.60) *
<0.0001 *
PFS ↓Multivariate
1.16 (1.01–1.36)
0.0482
AST > 38OS ↓Multivariate
1.52 (1.08–2.13) *
0.0167 *
PFS ↓Multivariate
1.21 (1.01–1.45)
0.0365
LenvatinibRetrospective validation of the experimentally identified biomarker
(vs. sorafenib)
Lenvatinib 65
(ST6GAL1 high 22,
low 43)
sorafenib 31
(ST6GAL1 high 12,
low 19)
Serum ST6GAL1 highOS ↑Univariate<0.05Myojin Y et al.
[58]
HR, hazard ratio; CI, confidence interval; PFS, progression-free survival; OS, overall survival; CR, complete response; PR, partial response; TTP, time to progression; DCR, disease control rate; HBV, hepatitis B virus; HCV, hepatitis C virus; MVI, macrovascular invasion; EHS, extrahepatic spread; NLR, neutrophil to lymphocyte ratio. Note: In the “Statistical analysis” section, “univariate” typically refers to the Kaplan–Meier method and log-rank test, and the inclusion of HR indicates the use of Cox regression. Additionally, ‘multivariate’ typically refers to the utilization of the multivariate Cox regression model.

3. AFP as an Approved Predictive Biomarker for Ramucirumab Treatment

VEGF and VEGFR2 signaling pathways have a crucial role in angiogenesis and tumor growth [60]. Multi-kinase inhibitors, such as sorafenib and lenvatinib, which have been shown to be effective against HCC, target VEGFR2. Ramucirumab is a human IgG1 monoclonal antibody that inhibits the ligand activation of VEGFR2 [61]. In a phase II trial of ramucirumab as a first-line therapy for HCC, ramucirumab demonstrated an ORR and OS that surpassed the sorafenib administration group in the SHARP trial [62]. In this trial, an exploratory study of biomarkers measured circulating VEGF, soluble VEGFR1 (sVEGFR1), sVEGFR2, and several cytokines and growth factors in serum samples after ramucirumab administration. Among them, a potential correlation was suggested between reduced serum sVEGFR1 levels until day 8 post-administration and prolonged PFS and OS [61]. However, in the REACH trial, a phase III trial of ramucirumab vs. placebo as a second-line therapy after sorafenib treatment, a significant improvement in OS was not achieved in the ramucirumab group compared with the placebo group [63].
Apart from the initially explored biomarkers, a subgroup analysis of the REACH trial revealed that OS in the ramucirumab group was significantly better when limited to cases with an AFP level ≥ 400 ng/mL [63]. Therefore, the REACH-2 trial was planned, which was a phase III trial of ramucirumab vs. placebo as a second-line therapy restricted to cases with an AFP level ≥ 400 ng/mL after sorafenib treatment [9]. As expected, the REACH-2 trial results indicated that OS was significantly extended in the ramucirumab treatment group compared with the placebo group. This trial became the first successful phase III trial for advanced HCC treatment that selected target cases using a biomarker [9].
Because AFP was shown to be a predictive biomarker for the therapeutic effect of ramucirumab, an analysis of 520 HCC cases with known baseline AFP values was conducted to investigate the molecular profile differences of tumors using AFP levels. The data suggested that tumors in cases with an AFP level > 400 ng/mL showed significant activation of VEGF signaling [64].

4. Exploration of Potential Biomarkers to Predict the Therapeutic Efficacy of Single-Agent ICIs and Combined Immunotherapy (Table 2)

The pharmacotherapy of HCC has shifted from being dominated by TKIs to ICIs and combined immunotherapies. Correspondingly, research into biomarkers to predict therapeutic effectiveness has transitioned from focusing on those related to tumor growth signals to those focusing on the tumor microenvironment and tumor immune environment [65].
Studies of biomarkers to predict the therapeutic effects of ICIs for HCC began with the validation of biomarkers discovered in other cancer types, such as melanoma, non-small cell lung cancer, and colorectal cancer. However, despite a demonstrated response rate of about 20% for HCC cases to single-agent therapies such as nivolumab and pembrolizumab, these treatments did not become standard first-line or second-line therapies [10,11,12,66,67]. The established combination therapy of atezolizumab and bevacizumab, which are anti-PD-L1 and anti-VEGF-A antibodies, respectively, became the standard treatment. This resulted in the research of biomarkers to predict the therapeutic effects of ICIs to focus on this type of combined immunotherapy.

4.1. Known Candidate Predictive Markers of the Efficacy of Single-Agent ICI and Combined Immunotherapies for HCC: PD-L1 Expression, Tumor Mutation Burden (TMB), and Microsatellite Instability (MSI)

The discovery of immune checkpoint proteins, such as PD-1/PD-L1 and CTLA-4, represents a significant breakthrough in the cancer immunotherapy field [68,69]. Currently, anti-PD-1 antibodies, anti-PD-L1 antibodies, and anti-CTLA-4 antibodies are used to treat HCC, including in combination therapies. For other types of cancer, PD-L1 expression, TMB, and MSI have been reported as biomarkers to predict the therapeutic effects of these ICIs [70]. However, when considering the practicality of these biomarkers, the frequency of PD-L1 expression, TMB-High, and MSI-High becomes an issue. According to a large cohort study by Ang et al., the incidence of MSI-High cases in HCC was extremely limited, with only one case among 542 patients. Additionally, only six cases (0.8%) among 755 cases had a TMB of 20 mutations/Mb or more [71].
Zhu et al. conducted comprehensive analyses of transcriptomics, genomics, and IHC staining of patient samples collected in the phase Ib GO30140 and phase III IMbrave150 trials to explore biomarkers for atezolizumab and bevacizumab combination therapy [72]. Whole exome sequencing (WES) or FoundationOne panel profiling was performed to evaluate TMB, resulting in median TMBs of 5.6 mutations/Mb and 4.4 mutations/Mb, respectively. TMB was categorized as low, medium, or high, and its associations with response rates and survival times were verified. However, no relationship between TMB and response rate or survival benefit was observed in the GO30140 trial arm A and IMbrave 150 [72].
PD-L1 expression merits further investigation. In the phase I/II CheckMate 040 trial of nivolumab monotherapy for advanced HCC, PD-L1 expression in tumor tissues was examined in 174 out of 214 cases in the dose-expansion phase. Of these, 34 cases (20%) showed PD-L1 expression ≥ 1% in tumor cells via IHC, and these cases demonstrated an ORR of 9/34 (26%; 95% CI 13–44). However, even in 140 cases with PD-L1 < 1%, an ORR was observed in 26/140 cases (19%; 95% CI 13–26), suggesting that therapeutic responses were observed regardless of PD-L1 expression status [11].
In the phase III Checkmate459 trial of nivolumab vs. sorafenib, PD-L1 expression ≥ 1% in tumor cells was found in 71 of 366 cases (19%) in the nivolumab group and 64 of 362 cases (18%) in the sorafenib group. In patients administered nivolumab, a higher ORR was indicated if they had PD-L1 ≥ 1% vs. PD-L1 < 1% (PD-L1 ≥ 1% ORR 20/71 (28%; 18–40); PD-L1 < 1% ORR 36/295 (12%; 9–17)). However, in the sorafenib group, there was no difference in ORR between those with PD-L1 ≥ 1% or PD-L1 < 1% (PD-L1 ≥ 1% ORR 6/64 (9%; 4–19); PD-L1 < 1% ORR 20/300 (7%; 4–10)). A comparison between those with PD-L1 ≥ 1% in the nivolumab group and those with PD-L1 ≥ 1% in the sorafenib group indicated a trend that favored nivolumab with a median OS of 16.1 months (95% CI 8.4–22.3) vs. 8.6 months (95% CI 5.7–16.3), but the difference was not statistically significant (HR 0.80 (0.54–1.19)) [66].
In the phase II Keynote224 trial of pembrolizumab monotherapy, the conventional positive cell rate of PD-L1 in tumor cells (tumor proportion score (TPS)) and the combined positive score (combined positive score (CPS)) were calculated by dividing the number of PD-L1 positive cells in tumor cells and immune cells by the total number of surviving tumor cells and multiplying by 100 [73]. Of 52 cases, 22 (42%) were CPS positive and only seven cases (13%) were TPS positive. Significant differences were observed in the response rates and PFS between CPS positive and negative cases, but not between TPS positive and negative cases [73].
According to the comprehensive analysis by Zhu et al., patients with high CD274 (PD-L1 mRNA) expression had a longer PFS with the atezolizumab–bevacizumab combination therapy than those with low expression. However, IHC data for PD-L1 protein levels indicated there was only a potential correlation between PD-L1 expression and response [72].
As these results suggest, PD-L1 expression is somewhat related to the efficacy of anti-PD-1/PD-L1 immunotherapy in HCC and might be a biomarker to predict therapeutic effects. However, there are some uncertainties in its benefit because it is difficult to definitively make these predictions at the protein level, whether to use TPS or CPS scoring has not been determined, and there is an issue of heterogeneity associated with IHC staining assays [74].

4.2. NASH as a Background Liver Disease

One potential biomarker that might predict the lack of efficacy of ICI monotherapy is NASH/NAFLD as a background liver disease. Pfister et al. suggested that in a mouse model of NASH-induced HCC, CD8+/PD-1+ T cells promoted the progression of NASH. The administration of ICIs “released the brakes” on these NASH-promoting cells, resulting in a potential exacerbation of NASH and increased HCC occurrence [75]. The authors conducted a meta-analysis of the cohorts from three phase III trials where ICIs were administered, namely Checkmate 459, IMbrave 150, and KEYNOTE-240. This analysis showed that although ICI treatment significantly prolonged OS compared with the control in HBV-related and HCV-related HCC cases, the prognosis did not improve in non-viral HCC cases. Furthermore, in two separate retrospective cohorts treated with anti-PD-1 or anti-PD-L1 antibodies, HCC cases caused by NAFLD showed reduced OS compared with those with other etiologies [75]. As demonstrated in this study, a subgroup analysis of the phase III IMbrave 150 trial of atezolizumab–bevacizumab combination therapy and sorafenib showed non-viral HCC including NASH did not show superiority, with a median OS of 17.0 months in the atezolizumab–bevacizumab combination group compared with 18.1 months in the sorafenib group [76].
Moreover, in a multicenter study involving 36 facilities in four countries (Italy, Japan, South Korea, and the UK), a retrospective analysis of 759 cases of advanced non-viral HCC revealed that when lenvatinib and atezolizumab + bevacizumab treatments were compared, lenvatinib had significantly better OS and PFS rates in non-viral HCC overall. When non-viral HCC was divided into NAFLD/NASH and non-NAFLD/NASH, lenvatinib treatment was associated with a significant survival benefit compared with atezolizumab + bevacizumab in patients with NAFLD/NASH HCC [77].
However, not all evidence suggests ICIs are less active in patients with a non-viral etiology. In a recent post hoc analysis of IMbrave150, clinical data from 279 out of 336 patients were obtained and analyzed, with etiologies categorized into HBV, HCV, alcohol, and NAFLD. However, no significant differences in OS, PFS, or ORR were observed between the different etiologies [78]. Etiology seems to have a role in modulating the response to ICIs, and currently, no recommendations regarding different treatments based on the underlying liver disease have been made.

4.3. Wnt/β-Catenin Mutations as a Biomarker and MRI Findings as Imaging Biomarkers

Spranger et al. reported the presence of Wnt/β-catenin mutations in melanoma resulted in the exclusion of T cell infiltration and resistance to ICIs [79]. Using The Cancer Genome Atlas (TCGA), Luke et al. demonstrated that tumors lacking the genetic expression signature of T cell-mediated inflammation, including 31 types of solid cancers including melanoma, had activated Wnt/β-catenin signaling [80]. Furthermore, Harding et al. conducted a genomic analysis of 127 HCC tumor tissues and reported that Wnt/β-catenin mutations were present in 45% of cases. Although the presence or absence of these mutations did not affect PFS with sorafenib treatment, their presence significantly shortened PFS with ICI treatment (2.0 vs. 7.4 months, p < 0.0001) [81]. In addition, in a comprehensive study by Zhu et al., patients with a wild-type CTNNBI genotype in the IMbrave150 trial showed a greater therapeutic effect with atezolizumab + bevacizumab compared with sorafenib treatment, but no significant difference was observed between the treatments in cases with CTNNBI mutations [72].
Ueno et al. focused on the differences in HCC findings in the hepatobiliary phase of gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid (Gd-EOB-DTPA)-enhanced MRI, comprehensively examined the transporter of Gd-EOB-DTPA and analyzed the molecular regulatory mechanism. Using clinical samples, they demonstrated that high expression levels of OATP1B3 were strongly correlated with greater enhancement in the hepatobiliary phase of Gd-EOB-DTPA-enhanced MRI. Additionally, activated Wnt/β-catenin signaling was closely associated with OATP1B3 expression in HCC cell cultures [82].
Aoki et al. analyzed 18 HCC cases that had received anti-PD-1 or anti-PD-L1 monotherapy and had Gd-EOB-DTPA-enhanced MRI taken before treatment. As a result, in cases with high signal nodules in the hepatobiliary phase (n = 8), the median PFS was 2.7 months, whereas in cases with low signal nodules (n = 10), it was 5.8 months (p = 0.007). There was also a significant difference in the period until tumor enlargement, indicating that the hepatobiliary phase of Gd-EOB-DTPA-enhanced MRI is a promising imaging biomarker to predict the therapeutic effect of anti-PD-1/PD-L1 monotherapy [83].
Moreover, it should be noted that multiple studies have reported that the efficacy of lenvatinib treatment for HCC was not affected by the signal intensity of the hepatobiliary phase of EOB-MRI [84,85].
Murai et al. focused on increasingly prevalent non-viral HCC cases. They extracted genomic DNA and total RNA from tumor tissues for profiling and then compared them with pathological findings to identify sensitivity to immunotherapy. Steatotic HCC accounted for 23% of non-viral HCC cases, which showed an immune-rich, yet immune-exhausted, tumor immune microenvironment characterized by T cell exhaustion, infiltration of M2 macrophages and cancer-associated fibroblasts (CAFs), high expression of immune PD-L1, and activation of TGF-β signaling. Histological fatty deposition of resected HCC tissue and Fat Fraction Corrected for Spectral Complexity and Inhomogeneities (FFCSI) measured by MRI were strongly correlated. The retrospective review of 30 HCC patients evaluated by MRI before atezolizumab–bevacizumab combination therapy confirmed a significantly longer PFS in patients with steatotic HCC [86].
Whether MRI findings are useful as imaging biomarkers to predict the efficacy of systemic therapy for HCC remains to be demonstrated with prospective validation studies. However, as the number of cohorts of systemic therapy using ICIs, such as atezolizumab–bevacizumab combination therapy, increases in clinical practice, we expect a consensus to be formed.

4.4. Problems with Wnt/β-Catenin Mutations as a Biomarker and MRI Findings as Imaging Biomarkers

Sasaki et al. reported that HCC patients receiving combination therapy of atezolizumab and bevacizumab with a high-intensity EOB-MRI hepatobiliary phase suggesting Wnt/β-catenin signal activation, had a shorter PFS than the low-intensity HCC patients in the atezolizumab + bevacizumab group. This MRI finding was not associated with the treatment effect of lenvatinib [87]. However, in a study by Kuwano et al. that used pretreatment tumor biopsies rather than EOB-MRI, there was no significant difference in the treatment effect or PFS of those receiving atezolizumab/bevacizumab combination therapy that depended on the presence or absence of Wnt/β-catenin activation [88]. These discrepancies may be caused by biases resulting from each study being retrospective and having a small number of cases, but other research results suggest otherwise.
Previous research results indicated there might be a discrepancy between EOB-MRI hepatocellular phase uptake findings and Wnt/β-catenin mutations. The transcription factor HNF4α maintains mature hepatocyte function and was decreased in dedifferentiated HCC, leading to the decreased expression of OATP1B3 regardless of Wnt/β-catenin mutations. This resulted in a loss of gadoxetic acid uptake in the hepatocyte phase, which may cause a mismatch [89,90].
Another question is whether the presence of Wnt/β-catenin mutations in HCC always results in a suppressed immune response. Sia et al. reported that about 25% of HCC cases had a subtype of an immune class characterized by immune activation, with overexpression of adaptive immune response genes, such as CD8A, CD3E, IFNG, CXCL9, and others, termed the active immune response subtype. Additionally, immunosuppressive signals, including TGF-β, and M2 macrophages were present in the exhausted immune response subtype. The authors also stated that a better response to ICI treatment was expected for this immune class [91].
This research group further investigated the immune characteristics of HCC cases outside this immune class. They found that about 10% of HCC cases had an immune-like class characterized by high IFN signaling, cytokines, and a diverse T cell repertoire, despite the significant activation of Wnt/β-catenin signaling by CTNNB1 mutation. This led them to classify HCC into an inflamed class, which includes the immune class and the immune-like class, as well as other non-inflamed classes. They suggested an “inflamed signature” consisting of 20 genes that accurately indicated the inflamed class and confirmed a significant overexpression of this signature in a group of patients who showed a partial response (PR) with ICI treatment in an external cohort compared with a group of patients who had stable disease (SD) or progressive disease (PD) [92].
When evaluating the tumor microenvironment, immunostaining in the inflamed class showed enrichment of intratumoral CD8+ T cells (CD8 ≥ 1%, 58% vs. 30%, p = 0.08) and PD-L1 (PD-L1 ≥ 1%, 21% vs. 4%, p = 0.19) compared with the non-inflamed class. Additionally, an analysis using CIBERSORT, which estimates the presence and ratios of immune cell subsets within tissues from gene expression data, showed a significantly higher proportion of CD8+ T cells (p = 3.51 × 10−7) and M1 macrophages (p = 1.82 × 10−4). However, in the immune-like class, M2 macrophages were significantly excluded (p = 1.78 × 10−6) [92].
Furthermore, the authors created a 13-protein signature as a liquid biopsy-based biomarker to identify the inflamed class using a cohort with blood samples. They suggested that when treating HCC, whether to distinguish between the inflamed and non-inflamed classes using the 20-gene signature in tumor tissues or the liquid biopsy-based signature should be considered [92].
Interestingly, a study using a dataset of over 9000 solid cancer cases across 31 types from TCGA found that activation of the Wnt/β-catenin pathway was often associated with reduced T cell infiltration in most human cancers. A significant inverse correlation was observed between β-catenin protein levels and T cell inflammatory gene expression in 177 HCC cases [80]. According to this study, the degree of the inverse correlation between β-catenin protein levels and T cell inflammatory gene expression varied by cancer type. Furthermore, in certain types of cancer, such as colorectal and rectal cancers, examples where T cell inflammation and activation of the Wnt/β-catenin pathway coexist are common [80]. This suggests that although activation of the Wnt/β-catenin pathway often hinders T cell inflammation in human cancers, this is not always the case. However, it appears that subclasses, such as the immune-like class, have not been proposed for other types of cancers.
Several studies have suggested that the hepatocyte phase of EOB-MRI may not match Wnt/β-catenin mutations and that some HCC cases of the immune-like class do not suppress immune responses against tumors, even when they have Wnt/β-catenin mutations. Therefore, findings from the hepatobiliary phase of EOB-MRI might be useful as biomarkers to predict the therapeutic effect of ICIs or the combination therapy of atezolizumab and bevacizumab. However, these findings do not necessarily indicate the presence or absence of Wnt/β-catenin mutations. When using the presence or absence of Wnt/β-catenin mutations as biomarkers to predict the therapeutic effect of ICIs, it is necessary to consider the existence of the immune-like class.

4.5. Potential Biomarkers to Predict the Therapeutic Effect of ICI Therapy

Spahn et al. reported that baseline levels of AFP 400 μg/L at the beginning of ICI therapy (anti-PD-1 antibodies) were linked to higher rates of PR or CR as the best responses and lower rates of PD. Furthermore, AFP levels below 400 μg/L were linked to considerably prolonged PFS and OS [93]. It was also reported that early AFP decline was associated with a favorable response to ICI therapy [94]. This report suggests that early AFP level changes are important biomarkers of ICI therapy efficacy.
Scheiner et al. created a training set of 190 cases and a validation set of 102 cases from a database of HCC cases in Europe that had received PD-L1/PD-1-based immunotherapy. Seventy-five cases (40%) in the training set and 25 cases (25%) in the validation set were patients who had received atezolizumab and bevacizumab combination therapy. In the training set, the investigated baseline parameters were etiology, whether immunotherapy was primary or after other treatments, Child–Pugh class, Eastern Cooperative Oncology Group performance status, radiological criteria, including the presence of major vessel invasion and extrahepatic metastasis, and serum AFP and C-reactive protein (CRP) levels. Serum AFP < 100 vs. ≥100 ng/mL and CRP < 1 vs. ≥1 mg/dL were identified as independent prognostic factors in a multivariate analysis, and the CRAFITY score was developed using these values [95].
Patients with a score of 0 (CRAFITY-low: AFP < 100 ng/mL and CRP < 1 mg/dL) had the longest OS, followed by those with a score of 1 (CRAFITY-intermediate: either AFP ≥ 100 ng/mL or CRP ≥ 1 mg/dL), and those with a score of 2 (CRAFITY-high: both AFP ≥ 100 ng/mL and CRP ≥ 1 mg/dL). Similarly, the best treatment effect was seen in patients with a low CRAFITY score. This study also validated a cohort of 204 cases of sorafenib administration. The CRAFITY score was associated with the survival of the individuals, but not with the therapeutic effect [95]. C-statistics, a statistical indicator that was used to evaluate the performance and predictive ability of the model in the CRAFITY score, was 0.62 for the derivation and validation cohorts. Although not highly accurate, it is very simple to use in routine practice and may be useful when predicting responses to ICI.
In a multi-institutional retrospective study in Japan, the CRAFITY score of 297 patients who received atezolizumab and bevacizumab combination therapy was analyzed. The median PFS in the CRAFITY score 0, 1, and 2 groups was 11.8, 6.5, and 3.2 months, respectively (p < 0.001). The median OS in patients with CRAFITY scores of 0, 1, or 2 was not reached, 14.3 months, and 11.6 months, respectively. This study showed the CRAFITY score might be useful for predicting therapeutic outcomes [96].
The pre-treatment NLR reflects the inflammatory response to cancer and was reportedly associated with patient prognosis and response to ICI treatment in various tumors [97,98,99,100,101]. Eso et al. analyzed the course of 40 HCC patients who received atezolizumab and bevacizumab combination therapy and found that the NLR value was significantly lower in the complete response (CR), PR, and SD groups than in the PD group (2.47 vs. 4.48, p = 0.013). Using the optimal NLR cut-off value (3.21) determined by receiver operating characteristic curve analysis for predicting responses, they also found that patients with an NLR ≤ 3.21 had significantly better PFS than patients with an NLR > 3.21 [101].
A similar examination was conducted in a multi-institutional joint study in Japan, where the cumulative OS rate was significantly different between patients with low NLR (<3.0) and high NLR (≥3.0) (p = 0.001). Conversely, there was no difference in the cumulative PFS or response between patients with low and high NLR values. In Cox proportional hazard modeling analysis using inverse probability weighting, an NLR of at least 3.0 was significantly associated with OS [102].
Because the CRAFITY score and NLR are parameters that can be easily obtained from blood samples, it is necessary to rigorously validate whether they should be used as biomarkers in routine practice.
Myojin et al. measured the levels of 34 baseline plasma proteins in patients with advanced HCC who received atezolizumab + bevacizumab therapy and found that plasma IL-6 levels were a significant predictor of non-response to this therapy. They confirmed that the PFS and OS were significantly shorter in the high IL-6 group than in the low IL-6 group [103].
Matsumae et al. evaluated pretreatment cfDNA in HCC patients with atezolizumab and bevacizumab. Patients with high cfDNA levels had a significantly lower ORR than those with low cfDNA levels. The PFS and OS were also significantly shorter in patients with high cfDNA levels than in patients with low cfDNA levels. Pretreatment cfDNA may be useful for predicting the therapeutic outcome of HCC patients treated with atezolizumab and bevacizumab [104]. Further investigation is needed to determine whether cfDNA concentration and composition affect immunotherapy responses to HCC.
Kim et al. investigated whether elevated levels of anti-drug antibodies in atezolizumab and bevacizumab affected therapeutic efficacy and T cell function. Patients with high ADA levels treated with atezolizumab and bevacizumab had a lower response rate and worse PFS and OS than patients with low ADA levels. Patients with high ADA levels had lower serum atezolizumab concentrations, reduced CD8-positive T cell proliferation, and lower IFN-γ and tumor necrosis factor-α production from CD8-positive T cells than patients with low ADA levels [105]. Therefore, elevated ADA levels after atezolizumab and bevacizumab administration may be considered a poor prognostic factor.
As will be discussed below, the results of the HIMALAYA trial have made it possible to administer a combination of durvalumab and tremelimumab as a first-line treatment. However, comparisons of treatment outcomes have indicated that the first-line treatment of choice for unresectable HCC cases is currently atezolizumab and bevacizumab combination therapy [106,107].
Several studies have reported gene expression profiles as biomarkers of responses to ICI therapy. Gene expression profiling of tissues revealed that PD-1 and PD-L1 expressions, biomarkers of inflammation (CD3 and CD8), and inflammatory gene signatures (CD274, CD8A, LAG3, STAT1) tended to be associated with improved survival and responses of HCC patients treated with anti-PD1 antibody [108]. Haber et al. reported that IFN signaling and major histocompatibility complex-related genes were key molecular features of HCCs that responded to anti-PD1 [109].
As mentioned previously, Zhu et al. used transcriptome analysis to derive an atezolizumab + bevacizumab response signature (ABRS) comprised of 10 genes associated with a response to atezolizumab + bevacizumab (defined as CR or PR). High expression of the ABRS, as well as the existing immune gene CD274 (PD-L1 mRNA) or the Teff sign (CXCL9, PRFI, and GZMB), were associated with longer PFS in patients treated with atezolizumab + bevacizumab. The Treg signature (CCR8, BATF, CTSC, TNFRSF4, FOXP3, TNFRSF18, IKZF2, and IL2RA) was also related to improved PFS and OS when the Treg/Teff signature ratio was low in the IMbrave150 study, which compared atezolizumab + bevacizumab with sorafenib treatment [72].
Multiplex IHC analysis in this study showed that in GO30140 cohort A, responding patients (CR/PR) had a higher density of infiltrating CD8+ T cells, CD3+ T cells, and GZMB+/CD3+ T cells in tumor areas than non-responders (SD/PD). The density of tumor-infiltrating CD8+ T cells in IMbrave150 baseline tumor samples was analyzed and patients with a high density of tumor-infiltrating CD8+ T cells (defined by the split median) had significantly longer OS when treated with atezolizumab + bevacizumab combination therapy compared with those treated with sorafenib [73]. These studies highlight an important point: the state of T cell immunity in the tumor microenvironment before treatment ultimately influences the treatment effect of atezolizumab + bevacizumab combination therapy for HCC.
Combination therapy with the anti-PD-L1 antibody durvalumab with anti-CTLA-4 antibody tremelimumab for unresectable HCC cases has surpassed the control drug sorafenib in the HIMALAYA trial and has been approved as a first-line therapy [15]. Among the factors studied as potential biomarkers in the HIMALAYA trial, the only results currently available relate to the PD-L1 status of the tumor prior to therapy. According to the subgroup analysis, there was no difference in benefit for the combination of two ICIs compared with sorafenib, regardless of whether PD-L1 expression was positive or negative [15].
Kuwano et al. examined the relationship between the tumor infiltration of CD8+ T cells detected by IHC staining of liver tumor biopsies before treatment initiation and the therapeutic effect of drug therapy. In cases with a high level of CD8+ T cell tumor infiltration, the PFS of patients treated with atezolizumab + bevacizumab combination therapy was significantly extended, and the response rate was also significantly improved compared with cases with low levels. However, in patients receiving lenvatinib, there was no association between CD8+ T cell tumor infiltration and the response rate or PFS [110]. Although this study included a limited number of patients, it suggests that evaluating CD8+ T cell tumor infiltration alone, without any transcriptomics analysis or genomic profiling, may serve as a useful biomarker to help decide the treatment method choice and predict the therapeutic response to drug therapy in HCC.
Another interesting finding was that the combination therapy showed clear advantages over sorafenib treatment in cases of HBV-related and non-viral HCC, but not in cases of HCV-related HCC. A similar trend was observed with durvalumab monotherapy [15]. As mentioned in the sorafenib section of this review, this may be because sorafenib has greater benefits for HCV-related HCC [18]. Nevertheless, there is no clear biomarker candidate for the durvalumab and tremelimumab combination therapy. The CRAFITY score and NLR can be easily validated, but to date, no reports have confirmed their effectiveness with this treatment.
Lee et al. reported a relationship between microbiota and the efficacy of ICIs for HCC patients. Lachnospiraceae and Veillonellaceae were enriched in the feces of patients with OR. In contrast, apparent increases in Prevotellaceae and Enterobacteriaceae, but a reduced abundance of Lachnospiraceae and Veillonellaceae, were observed in patients with PD after immunotherapy [111]. Spahn et al. revealed systemic antibiotics were associated with worse outcomes in HCC patients undergoing anti-PD-1 treatment [93]. This result indicates that gut dysbiosis adversely affects HCC ICI therapy. The composition of gut microbiota might be a biomarker for the therapeutic efficacy of immunotherapy for patients with HCC and may be targeted as a treatment to enhance the efficacy of ICI therapy.
ICI therapy often induces inflammatory reactions termed irAEs, which can affect various organs, including the skin, gastrointestinal, liver, respiratory, thyroid, and pituitary glands, and occur at any time. Recently, several studies reported that irAEs were associated with the efficacy of ICI therapy in patients with HCC [112]. Fukushima et al. reported that low-grade irAEs were strongly correlated with the PFS and OS in HCC patients treated with atezolizumab and bevacizumab [113]. IrAEs, particularly low-grade irAEs, are markers that predict better ICI therapy efficiency in HCC patients.
Table 2. Factors Influencing Prognosis/Efficacy in Hepatocellular Carcinoma with Immune Checkpoint Inhibitors.
Table 2. Factors Influencing Prognosis/Efficacy in Hepatocellular Carcinoma with Immune Checkpoint Inhibitors.
Therapeutics Study DesignNumber of CasesPrognostic and Predictive FactorsOutcomeStatistical Analysis
HR (95% CI)
p-ValueAuthor
[Reference No.]
Anti-PD-(L)1-based immunotherapyMeta-analyses of
3 phase 3 trials:
Checkmate 459
(nivolumab vs.
sorafenib),
IMbrave 150
(Atezo/Beva vs.
sorafenib),
KEYNOTE-240
(Pembrolizumab vs. Placebo)
ICI 985
Nivolumab 371
Pembrolizumab 278
Atezo/Beva 336
Control 672
Sorafenib 372 + 165
Placebo 135
HBVOS ↑Univariate
0.64 (0.49–0.83)
0.0008Pfister D et. al. [75]
HCVOS ↑Univariate
0.68 (0.48–0.97)
0.04
Retrospective
(ICI single arm)
exploratory cohort 130
validation cohort 118
NAFLDOS ↓Multivariate
2.6. (1.2–5.6)
0.017
Atezo/Beva
Lenvatinib
(Sorafenib)
Retrospective Non-viral cohort
Atezo/Beva 190
Lenvatinib 569
LenvatinibOS ↑Multivariate
0.65 (0.44–0.95)
0.0268Rimini M et al.
[77]
PFS ↑Multivariate
0.67 (0.51–0.86)
0.035
NAFLD/NASH cohort
Atezo/Beva 82
Lenvatinib 254
LenvatinibOS ↑Multivariate
0.46 (0.26–0.84)
0.011
PFS ↑Multivariate
0.55 (0.38–0.82)
0.031
Anti-PD-(L)1 monotherapyRetrospective,
single arm
18Hyperintensity tumor
(RER ‡ ≥ 0.9) on EOB-MRI
PFS ↓Multivariate
7.78 (1.59–38.1)
0.011Aoki T et. al.
[83]
Atezo/BevaRetrospective
validation based on
multiomics study,
single arm
Non-viral HCC 30Steatotic HCCPFS ↑Univariate<0.05Murai H et.al.
[86]
Atezo/Beva
Lenvatinib
Retrospective,
separate single arm
(not vs. lenvatinib)
Atezo/Beva 35Heterogeneous tumor
on EOB-MRI
PFS ↓Univariate0.007Sasaki R et.al.
[87]
Hyperintensity tumor
(RER ‡ ≥ 0.9) on EOB-MRI
PFS ↓Univariate0.012
Lenvatinib 33(no significant factor) -
Anti-PD-(L)1-based immunotherapyRetrospective,
single arm
2420 gene inflamed signature
(CCL5, CD2, CD3D, CD48, CD52, CD53, CXCL9, CXCR4, FYB, GZMA, GZMB, GZMK, IGHG1, IGHG3, LAPTM5, LCP2, PTPRC, SLA, TRAC, TRBC2)
PR ↑Wilcoxon rank sum0.047Montironi C et.al.
[92]
Anti-PD-1 monotherapyRetrospective,
single arm
99AFP < 400OS ↑Univariate
2.81 (1.56–4.97)
<0.0001Spahn et. al.
[93]
PFS ↑Univariate
1.33 (0.86–2.06)
<0.05
Anti-PD-1 monotherapyRetrospective,
single arm
60AFP response
as a >20% decline
OS ↑Univariate
0.09 (0.02–0.44)
<0.001Shao et. al.
[94]
PFS ↑Univariate
0.13 (0.04–0.39)
0.003
Anti-PD-(L)1-based
immunotherapy

Sorafenib
Retrospective,
separate single arm
(not vs. sorafenib)
Anti-PD-(L)1-based immunotherapy:
training cohort 190
(anti-PD-(L)1
mono 110,
Atezo/Beva 75,
Others 5)
validation cohort 102
(anti-PD-(L)1
mono 68,
Atezo/Beva 25,
Anti-PD-(L)1 + TKI 7,
Others 2)
Child–Pugh AOS ↑Multivariate
2.3 (1.5–3.4)
<0.001Scheiner B et.al.
[95]
ECOG PS 0OS ↑Multivariate
2.1 (1.4–3.2)
<0.001
AFP < 100OS ↑Multivariate
1.7 (1.2–2.6)
0.007
CRP < 1OS ↑Multivariate
1.7 (1.2–2.6)
0.007
CRAFITY score †OSUnivariate0.001
CRAFITY low1
CRAFITY int.2.0 (1.1–3.4)
CRAFITY high3.6 (2.1–6.2)
CRAFITY score †ORRChi-square 0.001
DCRChi-square <0.001
CRAFITY score †OSUnivariate0.001
DCRChi-square 0.037
Sorafenib 204CRAFITY score †OSUnivariate<0.001
Ate/BevRetrospective,
single arm
297AFP < 100PFS ↑Multivariate<0.001Hatanaka T et.al. [96]
OS ↑Multivariate0.028
CRP < 1PFS ↑Multivariate<0.001
OS ↑Multivariate0.032
CRAFITY score †PFSUnivariate<0.001
OSUnivariate
DCRChi-square 0.029
Ate/BevRetrospective,
single arm
40NLR ≤ 3.21PFS ↑Univariate<0.0001Eso Y et.al
[101]
Ate/BevRetrospective,
single arm
249NLR > 3OS ↓Multivariate
3.37 (1.02–11.08)
0.001Tada T et.al.
[102]
Atezo/Beva

Sorafenib
Retrospective
pooled analysis of
the phase 1b GO30140 (single arm) and the phase 3 trial
IMbrave 150
(Atezo/Beva
vs. sorafenib)
GO30140 arm A
cohort
Atezo/Beva 90
(single arm)
<Transcriptome analyses> Zhu AX et. al.
[72]
ABRS a highPFS ↑Univariate
0.51 (0.3–0.87)
0.013
CD274 b highPFS ↑Univariate
0.42 (0.25–0.72)
0.0011
Teff c highPFS ↑Univariate
0.46 (0.27–0.78)
0.0035
<In situ analyses>
CD8+ T cell densityCR/PR ↑Student T 0.007
CD3+ T cell densityCR/PR ↑Student T 0.039
CD3+ GZMB +
T cell density
CR/PR ↑Student T 0.044
MHC1 + tumor cellsCR/PR ↑Student T 0.0087
IMbrave 150
(Atezo/Beva 119
sorafenib 58)
<Transcriptome analyses>
ABRS a highPFS ↑Multivariate
0.49 (0.25–0.97)
0.041
OS ↑Multivariate
0.26 (0.11–0.58)
0.0012
CD274 b highPFS ↑Multivariate
0.46 (0.25–0.86)
0.015
OS ↑Multivariate
0.3 (0.14–0.64)
0.002
Teff c highPFS ↑Multivariate
0.52 (0.28–0.99)
0.047
OS ↑Multivariate
0.24 (0.11–0.5)
0.0002
Treg d/Teff c lowPFS ↑Multivariate
0.42 (0.22–0.79)
0.007
OS ↑Multivariate
0.24 (0.11–0.54)
0.0006
GPC3 lowPFS ↑Multivariate
0.47 (0.27–0.81)
0.006
OS ↑Multivariate
0.29 (0.13–0.62)
0.002
AFP lowPFS ↑Multivariate
0.49 (0.28–0.87)
0.014
OS ↑Multivariate
0.32 (0.14–0.73)
0.007
<In situ analyses>
CD8+ T cell high dens.OS ↑Multivariate
0.29 (0.14–0.61)
0.0011
PFS ↑Multivariate
0.54 (0.29–1.00)
0.053
<Genetic profiling>
CTNNB1 WTOS ↑Multivariate
0.42 (0.19–0.91)
3 × 10−4
PFS ↑Multivariate
0.45 (0.27–0.86)
0.0086
TERT MutOS ↑Multivariate
0.38 (0.16–0.89)
7.8 × 10−5
PFS ↑Multivariate
0.61 (0.33–1.10)
0.047
Atezo/BevaRetrospective,
single arm
34High plasma IL-6PFS ↑Univariate<0.05Myojin Y et.al.
[103]
Multivariate
2.785 (1.216–6.38)
0.01
OS ↑Univariate<0.05
Atezo/Beva

Lenvatinib
Retrospective,
separate single arm
(not vs. lenvatinib)
Atezo/Beva 24High-level CD8+ TILsPFS ↑Univariate0.041Kuwano A et.al.
[110]
ORR ↑Chi-square 0.012
DCR ↑Chi-square0.031
Lenvatinib 15(No significant factor)
Anti-PD-1 monotherapyCheckMate 040 trial
(nivolumab)
37<Genetic profiling>
Inflammatory gene
signatures (CD274, CD8A, LAG3, STAT1)
OS ↑Univariate0.01Sangro et.al.
[108]
PD-L1OS ↑Univariate0.032
Anti-PD-1 monotherapyRetrospective,
single arm
99Antibiotic treatmentPD ↑Chi-square<0.05Spahn et.al.
[93]
PFS ↓Univariate
1.65 (0.9–3.0)
<0.05
Atezo/BevaRetrospective,
single arm
85Cell free DNA lowOS ↑Univariate0.018Matsumae et.al
[104]
PFS ↑Univariate0.021
Atezo/BevaRetrospective,
single arm
174Anti-drug antibodiesOS ↓Univariate
5.81 (2.7–12.5)
0.001Kim et.al.
[105]
PFS ↓Univariate
2.52 (1.27–5.01)
0.006
Atezo/BevaRetrospective,
single arm
150Grade 1/2 irAEsOS ↑Multivariate
0.09 (0.01–0.64)
0.017Fukushima ey.al.
[113]
PFS ↑Multivariate
0.34 (0.17–0.69)
0.003
HR, hazard ratio; CI, confidence interval; PFS, progression-free survival; OS, overall survival; PD-1, programmed death receptor 1; PD-L1, programmed cell death ligand 1; HBV, hepatitis B virus; HCV, hepatitis C virus; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; RER, relative enhancement ratio; EOB-MRI, gadolinium ethoxybenzyl diethylenetriaminepentaacetic acid-enhanced magnetic resonance imaging; HCC, hepatocellular carcinoma; PR, partial response; ECOG, Eastern Cooperative Oncology Group; AFP, α-fetoprotein; CRP, C-reactive protein; ORR, overall response rate; DCR, disease control rate; NLR, neutrophil to lymphocyte ratio; CR, complete response; irAE, immune-related adverse events. Note: In the “Statistical analysis” section, “univariate” typically refers to the Kaplan–Meier method and log-rank test, and the inclusion of HR indicates the use of Cox regression. Additionally, “multivariate” typically refers to the utilization of the multivariate Cox regression model. ‡: (nodule SI/parenchyma SI on hepatobiliary phase images)/(nodule SI/parenchyma SI on precontract images). SI: signal intensity. †: CRAFITY-low: AFP < 100 and CRP < 1, intermediate: AFP ≥ 100 ng/mL or CRP ≥ 1 mg/dL, high: AFP ≥ 100 ng/mL and CRP ≥ 1 mg/dL. a: ABRS, atezolizumab + bevacizumab response signature (including CXCR2P1, ICOS, TIMD4, CTLA4, PAX5, KLRC3, FCRL3, AIM2, GBP5, and CCL4). b: CD274, PD-L1 mRNA. c: Teff, T effector (including CXCL9, PRF1, and GZMB). d: Treg, T regulatory (including CCR8, BATF, CTSC, TNFRSF4, FOXP3, TNFRSF18, IKZF2, and IL2RA).

5. Conclusions and Future Directions

In this review, we discussed published research findings on biomarkers to predict the therapeutic effects of drugs available for unresectable HCC tumors. As TKIs will continue to be used as secondary therapies, the search for biomarkers must continue. Among the factors mentioned, hepatic function, underlying hepatic disease, and the NLR may serve as vague indications of utility, but they are far from being decisive in drug selection. Because performing prospective trials of existing TKIs might be difficult in the future, we think it is worthwhile to proactively investigate whether biomarker candidates including DKK-1, ST6GAL1, and regorafenib have predictive value, because they have been indicated in several studies. We encourage this investigation within existing cohorts that have retained blood samples.
Prognostic and predictive factors for ICI therapy are listed in Table 3. For the current standard treatment of atezolizumab and bevacizumab combination therapy, routine examinations such as underlying hepatic disease, CRAFITY score, and the NLR seem to provide some guidance. Furthermore, imaging diagnostics, such as the evaluation of fat deposition throughout the EOB-MRI hepatobiliary phase or FFCSI and information from MRI examinations, can be useful. At present, the most important factor is thought to be an accurate assessment of the state of T cell immunity in the tumor microenvironment prior to treatment. The literature suggests that evaluating the tumor microenvironment and immune environment is more achievable compared with evaluating the diversity and heterogeneity of tumor cells. Assessing CD8+ T cell infiltration by collecting tumor tissues can be performed relatively easily, even in general hospitals. Thus, this has a high potential to become a practical biomarker.
Currently, there are no approved biomarkers, except for AFP and ramucirumab, for the prediction of response/resistance to systemic therapy using ICIs and TKIs. In general hospitals that treat HCC patients, it is common to administer drug therapy for advanced HCC cases based on guidelines informed solely by the results of blood tests and computed tomography scans. Although this approach is not inherently flawed, based on our perspective, gathering diagnostic materials such as blood and tissue samples, coupled with performing MRI image evaluations before treatment, can offer deeper insights into the state within the tumor tissue.

Author Contributions

Writing—original draft preparation, K.M. and A.K.; writing—review and editing, K.M., A.K., Y.K. and K.T.; revised scientific literature research, K.M., M.Y., and A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We are grateful to Y. Ishibashi for assistance with manuscript preparation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rumgay, H.; Arnold, M.; Ferlay, J.; Lesi, O.; Cabasag, C.J.; Vignat, J.; Laversanne, M.; McGlynn, K.A.; Soerjomataram, I. Global burden of primary liver cancer in 2020 and predictions to 2040. J. Hepatol. 2022, 77, 1598–1606. [Google Scholar] [CrossRef] [PubMed]
  2. McGlynn, K.A.; Petrick, J.L.; El-Serag, H.B. Epidemiology of Hepatocellular Carcinoma. Hepatology 2021, 73 (Suppl. S1), 4–13. [Google Scholar] [CrossRef] [PubMed]
  3. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef]
  4. Mathurin, P.; Rixe, O.; Carbonell, N.; Bernard, B.; Cluzel, P.; Bellin, M.F.; Khayat, D.; Opolon, P.; Poynard, T. Overview of medical treatments in unresectable hepatocellular carcinoma-an impossible meta-analysis? Aliment. Pharmacol. Ther. 1998, 12, 111–126. [Google Scholar] [CrossRef] [PubMed]
  5. Wilhelm, S.M.; Carter, C.; Tang, L.; Wilkie, D.; McNabola, A.; Rong, H.; Chen, C.; Zhang, X.; Vincent, P.; McHugh, M.; et al. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the raf/mek/erk pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004, 64, 7099–7109. [Google Scholar] [CrossRef] [PubMed]
  6. Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.-F.; De Oliveira, A.C.; Santoro, A.; Raoul, J.-L.; Forner, A.; et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef] [PubMed]
  7. Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.-H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 389, 56–66. [Google Scholar] [CrossRef]
  8. Kudo, M.; Finn, R.S.; Qin, S.; Han, K.-H.; Ikeda, K.; Piscaglia, F.; Baron, A.; Park, J.-W.; Han, G.; Jassem, J.; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018, 391, 1163–1173. [Google Scholar] [CrossRef]
  9. Zhu, A.X.; Kang, Y.-K.; Yen, C.-J.; Finn, R.S.; Galle, P.R.; Llovet, J.M.; Assenat, E.; Brandi, G.; Pracht, M.; Lim, H.Y.; et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019, 20, 282–296. [Google Scholar] [CrossRef]
  10. Abou-Alfa, G.K.; Meyer, T.; Cheng, A.-L.; El-Khoueiry, A.B.; Rimassa, L.; Ryoo, B.-Y.; Cicin, I.; Merle, P.; Chen, Y.; Park, J.-W.; et al. Cabozantinib in Patients with Advanced and Progressing Hepatocellular Carcinoma. N. Engl. J. Med. 2018, 379, 54–63. [Google Scholar] [CrossRef]
  11. El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.-Y.; Choo, S.-P.; Trojan, J.; Welling, T.H.; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef] [PubMed]
  12. 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]
  13. Finn, R.S.; Ryoo, B.-Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab as Second-Line Therapy in Patients with Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef]
  14. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef] [PubMed]
  15. 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, E.N.; et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022, 1, EVI-Doa2100070. [Google Scholar] [CrossRef]
  16. Llovet, J.M.; Peña, C.E.; Lathia, C.D.; Shan, M.; Meinhardt, G.; Bruix, J. Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin. Cancer Res. 2012, 18, 2290–2300. [Google Scholar] [CrossRef]
  17. Miyahara, K.; Nouso, K.; Morimoto, Y.; Takeuchi, Y.; Hagihara, H.; Kuwaki, K.; Onishi, H.; Ikeda, F.; Miyake, Y.; Nakamura, S.; et al. Pro-angiogenic cytokines for prediction of outcomes in patients with advanced hepatocellular carcinoma. Br. J. Cancer 2013, 109, 2072–2078. [Google Scholar] [CrossRef]
  18. Bruix, J.; Cheng, A.-L.; Meinhardt, G.; Nakajima, K.; De Sanctis, Y.; Llovet, J. Prognostic factors and predictors of sorafenib benefit in patients with hepatocellular carcinoma: Analysis of two phase III studies. J. Hepatol. 2017, 67, 999–1008. [Google Scholar] [CrossRef]
  19. Guthrie, G.J.K.; Charles, K.A.; Roxburgh, C.S.D.; Horgan, P.G.; McMillan, D.C.; Clarke, S.J. The systemic inflammation-based neutrophil–lymphocyte ratio: Experience in patients with cancer. Crit. Rev. Oncol. Hematol. 2013, 88, 218–230. [Google Scholar] [CrossRef]
  20. Templeton, A.J.; Mcnamara, M.G.; Šeruga, B.; Vera-Badillo, F.E.; Aneja, P.; Ocaña, A.; Leibowitz-Amit, R.; Sonpavde, G.; Knox, J.J.; Tran, B.; et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: A systematic review and meta-analysis. J. Natl. Cancer Inst. 2014, 106, dju124. [Google Scholar] [CrossRef]
  21. Qi, X.; Li, J.; Deng, H.; Li, H.; Su, C.; Guo, X. Neutrophil-to-lymphocyte ratio for the prognostic assessment of hepatocellular carcinoma: A systematic review and meta-analysis of observational studies. Oncotarget 2016, 7, 45283–45301. [Google Scholar] [CrossRef] [PubMed]
  22. Marrero, J.A.; Kudo, M.; Venook, A.P.; Ye, S.-L.; Bronowicki, J.-P.; Chen, X.-P.; Dagher, L.; Furuse, J.; Geschwind, J.-F.H.; de Guevara, L.L.; et al. Observational registry of sorafenib use in clinical practice across Child-Pugh subgroups: The GIDEON study. J. Hepatol. 2016, 65, 1140–1147. [Google Scholar] [CrossRef]
  23. Kuwano, A.; Yada, M.; Nagasawa, S.; Tanaka, K.; Morita, Y.; Masumoto, A.; Motomura, K. Hepatitis C virus eradication ameliorates the prognosis of advanced hepatocellular carcinoma treated with sorafenib. J. Viral Hepat. 2022, 29, 543–550. [Google Scholar] [CrossRef] [PubMed]
  24. Tsai, H.-Y.; Chang, H.-P.; Chen, C.-J.; Hsu, W.-L.; Huang, L.-Y.; Lee, P.-C. Effects of direct-acting antiviral therapy for patients with advanced hepatocellular carcinoma and concomitant hepatitis C-A population-based cohort study. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 7543–7552. [Google Scholar] [CrossRef] [PubMed]
  25. Abou-Alfa, G.K.; Schwartz, L.; Ricci, S.; Amadori, D.; Santoro, A.; Figer, A.; De Greve, J.; Douillard, J.-Y.; Lathia, C.; Schwartz, B.; et al. Phase II Study of Sorafenib in patients with advanced hepatocellular carcinoma. J. Clin. Oncol. 2006, 24, 4293–4300. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, D.; Zhao, P.; Li, S.-Q.; Xiao, W.-K.; Yin, X.-Y.; Peng, B.-G.; Liang, L.-J. Prognostic impact of pERK in advanced hepatocellular carcinoma patients treated with sorafenib. Eur. J. Surg. Oncol. 2013, 39, 974–980. [Google Scholar] [CrossRef] [PubMed]
  27. Negri, F.V.; Bello, B.D.; Porta, C.; Campanini, N.; Rossi, S.; Tinelli, C.; Poggi, G.; Missale, G.; Fanello, S.; Salvagni, S.; et al. Expression of pERK and VEGFR-2 in advanced hepatocellular carcinoma and resistance to sorafenib treatment. Liver Int. 2015, 35, 2001–2008. [Google Scholar] [CrossRef] [PubMed]
  28. Personeni, N.; Rimassa, L.; Pressiani, T.; Destro, A.; Ligorio, C.; Tronconi, M.C.; Bozzarelli, S.; Carnaghi, C.; Di Tommaso, L.; Giordano, L.; et al. Molecular determinants of outcome in sorafenib-treated patients with hepatocellular carcinoma. J. Cancer Res. Clin. Oncol. 2013, 139, 1179–1187. [Google Scholar] [CrossRef]
  29. Arao, T.; Ueshima, K.; Matsumoto, K.; Nagai, T.; Kimura, H.; Hagiwara, S.; Sakurai, T.; Haji, S.; Kanazawa, A.; Hidaka, H.; et al. FGF3/FGF4 amplification and multiple lung metastases in responders to sorafenib in hepatocellular carcinoma. Hepatology 2012, 57, 1407–1415. [Google Scholar] [CrossRef]
  30. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008, 18, 997–1006. [Google Scholar] [CrossRef]
  31. Gyöngyösi, B.; Végh, E.; Járay, B.; Székely, E.; Fassan, M.; Bodoky, G.; Schaff, Z.; Kiss, A. Pretreatment MicroRNA Level and Outcome in Sorafenib-treated Hepatocellular Carcinoma. J. Histochem. Cytochem. 2014, 62, 547–555. [Google Scholar] [CrossRef] [PubMed]
  32. Giordano, S.; Columbano, A. MicroRNAs: New tools for diagnosis, prognosis, and therapy in hepatocellular carcinoma? Hepatology 2012, 57, 840–847. [Google Scholar] [CrossRef] [PubMed]
  33. Ghidini, M.; Braconi, C. Non-Coding RNAs in Primary Liver Cancer. Front. Med. 2015, 2, 36. [Google Scholar] [CrossRef] [PubMed]
  34. Vaira, V.; Roncalli, M.; Carnaghi, C.; Faversani, A.; Maggioni, M.; Augello, C.; Rimassa, L.; Pressiani, T.; Spagnuolo, G.; Di Tommaso, L.; et al. MicroRNA-425-3p predicts response to sorafenib therapy in patients with hepatocellular carcinoma. Liver Int. 2014, 35, 1077–1086. [Google Scholar] [CrossRef] [PubMed]
  35. Shi, Y.; Liu, Z.; Lin, Q.; Luo, Q.; Cen, Y.; Li, J.; Fang, X.; Gong, C. MiRNAs and Cancer: Key Link in Diagnosis and Therapy. Genes 2021, 12, 1289. [Google Scholar] [CrossRef] [PubMed]
  36. Qiu, Z.; Li, H.; Zhang, Z.; Zhu, Z.; He, S.; Wang, X.; Wang, P.; Qin, J.; Zhuang, L.; Wang, W.; et al. A Pharmacogenomic Landscape in Human Liver Cancers. Cancer Cell 2019, 36, 179–193.e11. [Google Scholar] [CrossRef] [PubMed]
  37. Semënov, M.V.; Zhang, X.; He, X. DKK1 antagonizes wnt signaling without promotion of lrp6 internalization and degradation. J. Biol. Chem. 2008, 283, 21427–21432. [Google Scholar] [CrossRef]
  38. Teufel, M.; Seidel, H.; Köchert, K.; Meinhardt, G.; Finn, R.S.; Llovet, J.M.; Bruix, J. Biomarkers Associated with Response to Regorafenib in Patients with Hepatocellular Carcinoma. Gastroenterology 2019, 156, 1731–1741. [Google Scholar] [CrossRef]
  39. Piccart-Gebhart, M.J.; Procter, M.; Leyland-Jones, B.; Goldhirsch, A.; Untch, M.; Smith, I.; Gianni, L.; Baselga, J.; Bell, R.H.; Jackisch, C.; et al. Trastuzumab after adjuvant chemotherapy in her2-positive breast cancer. N. Engl. J. Med. 2005, 353, 1659–1672. [Google Scholar] [CrossRef]
  40. Cataldo, V.D.; Gibbons, D.L.; Pérez-Soler, R.; Quintás-Cardama, A. Treatment of non-small-cell lung cancer with Erlotinib or Gefitinib. N. Engl. J. Med. 2011, 364, 947–955. [Google Scholar] [CrossRef]
  41. Solomon, B.J.; Mok, T.; Kim, D.-W.; Wu, Y.-L.; Nakagawa, K.; Mekhail, T.; Felip, E.; Cappuzzo, F.; Paolini, J.; Usari, T.; et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N. Engl. J. Med. 2014, 371, 2167–2177. [Google Scholar] [CrossRef] [PubMed]
  42. Llovet, J.M.; Hernandez-Gea, V. Hepatocellular carcinoma: Reasons for phase III failure and novel perspectives on trial design. Clin. Cancer Res. 2014, 20, 2072–2079. [Google Scholar] [CrossRef] [PubMed]
  43. Villanueva, A.; Chiang, D.Y.; Newell, P.; Peix, J.; Thung, S.; Alsinet, C.; Tovar, V.; Roayaie, S.; Minguez, B.; Sole, M.; et al. Pivotal role of mTOR signaling in hepatocellular carcinoma. Gastroenterology 2008, 135, 1972–1983. [Google Scholar] [CrossRef] [PubMed]
  44. Zhu, A.X.; Kudo, M.; Assenat, E.; Cattan, S.; Kang, Y.-K.; Lim, H.Y.; Poon, R.T.P.; Blanc, J.-F.; Vogel, A.; Chen, C.-L.; et al. Effect of Everolimus on survival in advanced hepatocellular carcinoma after failure of Sorafenib. JAMA 2014, 312, 57–67. [Google Scholar] [CrossRef]
  45. Zhang, B.; Finn, R.S. Personalized Clinical Trials in Hepatocellular Carcinoma Based on Biomarker Selection. Liver Cancer 2016, 5, 221–232. [Google Scholar] [CrossRef]
  46. Santoro, A.; Rimassa, L.; Borbath, I.; Daniele, B.; Salvagni, S.; Van Laethem, J.L.; Van Vlierberghe, H.; Trojan, J.; Kolligs, F.T.; Weiss, A.; et al. Tivantinib for second-line treatment of advanced hepatocellular carcinoma: A randomised, placebo-controlled phase 2 study. Lancet Oncol. 2013, 14, 55–63. [Google Scholar] [CrossRef]
  47. Rimassa, L.; Assenat, E.; Peck-Radosavljevic, M.; Pracht, M.; Zagonel, V.; Mathurin, P.; Caremoli, E.R.; Porta, C.; Daniele, B.; Bolondi, L.; et al. Tivantinib for second-line treatment of MET-high, advanced hepatocellular carcinoma (METIV-HCC): A final analysis of a phase 3, randomised, placebo-controlled study. Lancet Oncol. 2018, 19, 682–693. [Google Scholar] [CrossRef] [PubMed]
  48. Kudo, M.; Morimoto, M.; Moriguchi, M.; Izumi, N.; Takayama, T.; Yoshiji, H.; Hino, K.; Oikawa, T.; Chiba, T.; Motomura, K.; et al. A randomized, double-blind, placebo-controlled, phase 3 study of tivantinib in Japanese patients with MET-high hepatocellular carcinoma. Cancer Sci. 2020, 111, 3759–3769. [Google Scholar] [CrossRef]
  49. Rimassa, L.; Kelley, R.K.; Meyer, T.; Ryoo, B.-Y.; Merle, P.; Park, J.-W.; Blanc, J.-F.; Lim, H.Y.; Tran, A.; Chan, Y.-W.; et al. Outcomes Based on Plasma Biomarkers for the Phase 3 CELESTIAL Trial of Cabozantinib versus Placebo in Advanced Hepatocellular Carcinoma. Liver Cancer 2021, 11, 38–47. [Google Scholar] [CrossRef]
  50. Totoki, Y.; Tatsuno, K.; Covington, K.R.; Ueda, H.; Creighton, C.J.; Kato, M.; Tsuji, S.; Donehower, L.A.; Slagle, B.L.; Nakamura, H.; et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 2014, 46, 1267–1273. [Google Scholar] [CrossRef]
  51. Schulze, K.; Imbeaud, S.; Letouzé, E.; Alexandrov, L.B.; Calderaro, J.; Rebouissou, S.; Couchy, G.; Meiller, C.; Shinde, J.; Soysouvanh, F.; et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 2015, 47, 505–511. [Google Scholar] [CrossRef] [PubMed]
  52. Xue, R.; Li, R.; Guo, H.; Guo, L.; Su, Z.; Ni, X.; Qi, L.; Zhang, T.; Li, Q.; Zhang, Z.; et al. Variable Intra-Tumor Genomic Heterogeneity of Multiple Lesions in Patients with Hepatocellular Carcinoma. Gastroenterology 2016, 150, 998–1008. [Google Scholar] [CrossRef] [PubMed]
  53. Pinter, M.; Peck-Radosavljevic, M. Review article: Systemic treatment of hepatocellular carcinoma. Aliment. Pharmacol. Ther. 2018, 48, 598–609. [Google Scholar] [CrossRef] [PubMed]
  54. Tohyama, O.; Matsui, J.; Kodama, K.; Hata-Sugi, N.; Kimura, T.; Okamoto, K.; Minoshima, Y.; Iwata, M.; Funahashi, Y. Antitumor activity of lenvatinib (E7080): An angiogenesis inhibitor that targets multiple receptor tyrosine kinases in preclinical human thyroid cancer models. J. Thyroid Res. 2014, 2014, 638747. [Google Scholar] [CrossRef] [PubMed]
  55. Matsuki, M.; Hoshi, T.; Yamamoto, Y.; Ikemori-Kawada, M.; Minoshima, Y.; Funahashi, Y.; Matsui, J. Lenvatinib inhibits angiogenesis and tumor fibroblast growth factor signaling pathways in human hepatocellular carcinoma models. Cancer Med. 2018, 7, 2641–2653. [Google Scholar] [CrossRef] [PubMed]
  56. Tada, T.; Kumada, T.; Hiraoka, A.; Michitaka, K.; Atsukawa, M.; Hirooka, M.; Tsuji, K.; Ishikawa, T.; Takaguchi, K.; Kariyama, K.; et al. Neutrophil-to-lymphocyte ratio is associated with survival in patients with unresectable hepatocellular carcinoma treated with lenvatinib. Liver Int. 2020, 40, 968–976. [Google Scholar] [CrossRef] [PubMed]
  57. Casadei-Gardini, A.; Rimini, M.; Kudo, M.; Shimose, S.; Tada, T.; Suda, G.; Goh, M.J.; Jefremow, A.; Scartozzi, M.; Cabibbo, G.; et al. Real Life Study of Lenvatinib Therapy for Hepatocellular Carcinoma: RELEVANT Study. Liver Cancer 2022, 11, 527–539. [Google Scholar] [CrossRef] [PubMed]
  58. Myojin, Y.; Kodama, T.; Maesaka, K.; Motooka, D.; Sato, Y.; Tanaka, S.; Abe, Y.; Ohkawa, K.; Mita, E.; Hayashi, Y.; et al. ST6GAL1 Is a Novel Serum Biomarker for Lenvatinib-Susceptible FGF19-Driven Hepatocellular Carcinoma. Clin. Cancer Res. 2021, 27, 1150–1161. [Google Scholar] [CrossRef]
  59. Wilson, L.J.; Linley, A.; Hammond, D.E.; Hood, F.E.; Coulson, J.M.; MacEwan, D.J.; Ross, S.J.; Slupsky, J.R.; Smith, P.D.; Eyers, P.A.; et al. New Perspectives, Opportunities, and Challenges in Exploring the Human Protein Kinome. Cancer Res. 2018, 78, 15–29. [Google Scholar] [CrossRef]
  60. Ferrara, N.; Gerber, H.-P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef]
  61. Spratlin, J.L.; Cohen, R.B.; Eadens, M.; Gore, L.; Camidge, D.R.; Diab, S.; Leong, S.; O’Bryant, C.; Chow, L.Q.M.; Serkova, N.J.; et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J. Clin. Oncol. 2010, 28, 780–787. [Google Scholar] [CrossRef] [PubMed]
  62. Zhu, A.X.; Finn, R.S.; Mulcahy, M.; Gurtler, J.; Sun, W.; Schwartz, J.D.; Dalal, R.P.; Joshi, A.; Hozak, R.R.; Xu, Y.; et al. A Phase II and biomarker study of ramucirumab, a human monoclonal antibody targeting the VEGF receptor-2, as first-line monotherapy in patients with advanced hepatocellular cancer. Clin. Cancer Res. 2013, 19, 6614–6623. [Google Scholar] [CrossRef]
  63. Zhu, A.X.; Park, J.O.; Ryoo, B.-Y.; Yen, C.-J.; Poon, R.; Pastorelli, D.; Blanc, J.-F.; Chung, H.C.; Baron, A.D.; Pfiffer, T.E.F.; et al. Ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma following first-line therapy with sorafenib (REACH): A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2015, 16, 859–870. [Google Scholar] [CrossRef]
  64. Montal, R.; Andreu-Oller, C.; Bassaganyas, L.; Esteban-Fabró, R.; Moran, S.; Montironi, C.; Moeini, A.; Pinyol, R.; Peix, J.; Cabellos, L.; et al. Molecular portrait of high alpha-fetoprotein in hepatocellular carcinoma: Implications for biomarker-driven clinical trials. Br. J. Cancer 2019, 121, 340–343. [Google Scholar] [CrossRef] [PubMed]
  65. Donne, R.; Lujambio, A. The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma. Hepatology 2023, 77, 1773–1796. [Google Scholar] [CrossRef] [PubMed]
  66. Yau, T.; Park, J.-W.; Finn, R.S.; Cheng, A.-L.; Mathurin, P.; Edeline, J.; Kudo, M.; Harding, J.J.; Merle, P.; Rosmorduc, O.; et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022, 23, 77–90. [Google Scholar] [CrossRef] [PubMed]
  67. Qin, S.; Chen, Z.; Fang, W.; Ren, Z.; Xu, R.; Ryoo, B.-Y.; Meng, Z.; Bai, Y.; Chen, X.; Liu, X.; et al. Pembrolizumab Versus Placebo as Second-Line Therapy in Patients from Asia with Advanced Hepatocellular Carcinoma: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2023, 41, 1434–1443. [Google Scholar] [CrossRef]
  68. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 Blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef]
  69. Iwai, Y.; Ishida, M.; Tanaka, Y.; Okazaki, T.; Honjo, T.; Minato, N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297. [Google Scholar] [CrossRef]
  70. Shiravand, Y.; Khodadadi, F.; Kashani, S.M.A.; Hosseini-Fard, S.R.; Hosseini, S.; Sadeghirad, H.; Ladwa, R.; O’byrne, K.; Kulasinghe, A. Immune Checkpoint Inhibitors in Cancer Therapy. Curr. Oncol. 2022, 29, 3044–3060. [Google Scholar] [CrossRef]
  71. 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] [PubMed]
  72. 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]
  73. Kudo, M. Pembrolizumab for the Treatment of Hepatocellular Carcinoma. Liver Cancer 2019, 8, 143–154. [Google Scholar] [CrossRef] [PubMed]
  74. Pinato, D.J.; Mauri, F.A.; Spina, P.; Cain, O.; Siddique, A.; Goldin, R.; Victor, S.; Pizio, C.; Akarca, A.U.; Boldorini, R.L.; et al. Clinical implications of heterogeneity in PD-L1 immunohistochemical detection in hepatocellular carcinoma: The Blueprint-HCC study. Br. J. Cancer 2019, 120, 1033–1036. [Google Scholar] [CrossRef] [PubMed]
  75. 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]
  76. 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]
  77. Rimini, M.; Rimassa, L.; Ueshima, K.; Burgio, V.; Shigeo, S.; Tada, T.; Suda, G.; Yoo, C.; Cheon, J.; Pinato, D.; et al. Atezolizumab plus bevacizumab versus lenvatinib or sorafenib in non-viral unresectable hepatocellular carcinoma: An international propensity score matching analysis. ESMO Open 2022, 7, 100591. [Google Scholar] [CrossRef] [PubMed]
  78. Espinoza, M.; Muquith, M.; Lim, M.; Zhu, H.; Singal, A.G.; Hsiehchen, D. Disease Etiology and Outcomes After Atezolizumab Plus Bevacizumab in Hepatocellular Carcinoma: Post-Hoc Analysis of IMbrave150. Gastroenterology 2023, 165, 286–288.e4. [Google Scholar] [CrossRef]
  79. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
  80. Luke, J.J.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [Google Scholar] [CrossRef]
  81. 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]
  82. Ueno, A.; Masugi, Y.; Yamazaki, K.; Komuta, M.; Effendi, K.; Tanami, Y.; Tsujikawa, H.; Tanimoto, A.; Okuda, S.; Itano, O.; et al. OATP1B3 expression is strongly associated with Wnt/β-catenin signalling and represents the transporter of gadoxetic acid in hepatocellular carcinoma. J. Hepatol. 2014, 61, 1080–1087. [Google Scholar] [CrossRef]
  83. Aoki, T.; Nishida, N.; Ueshima, K.; Morita, M.; Chishina, H.; Takita, M.; Hagiwara, S.; Ida, H.; Minami, Y.; Yamada, A.; et al. Higher Enhancement Intrahepatic Nodules on the Hepatobiliary Phase of Gd-EOB-DTPA-Enhanced MRI as a Poor Responsive Marker of Anti-PD-1/PD-L1 Monotherapy for Unresectable Hepatocellular Carcinoma. Liver Cancer 2021, 10, 615–628. [Google Scholar] [CrossRef] [PubMed]
  84. Kubo, A.; Suda, G.; Kimura, M.; Maehara, O.; Tokuchi, Y.; Kitagataya, T.; Ohara, M.; Yamada, R.; Shigesawa, T.; Suzuki, K.; et al. Characteristics and Lenvatinib Treatment Response of Unresectable Hepatocellular Carcinoma with Iso-High Intensity in the Hepatobiliary Phase of EOB-MRI. Cancers 2021, 13, 3633. [Google Scholar] [CrossRef] [PubMed]
  85. Kuwano, A.; Tanaka, K.; Yada, M.; Nagasawa, S.; Morita, Y.; Masumoto, A.; Motomura, K. Therapeutic efficacy of lenvatinib for hepatocellular carcinoma with iso-high intensity in the hepatobiliary phase of Gd-EOB-DTPA-MRI. Mol. Clin. Oncol. 2022, 16, 53. [Google Scholar] [CrossRef]
  86. Murai, H.; Kodama, T.; Maesaka, K.; Tange, S.; Motooka, D.; Suzuki, Y.; Shigematsu, Y.; Inamura, K.; Mise, Y.; Saiura, A.; et al. Multiomics identifies the link between intratumor steatosis and the exhausted tumor immune microenvironment in hepatocellular carcinoma. Hepatology 2023, 77, 77–91. [Google Scholar] [CrossRef] [PubMed]
  87. Sasaki, R.; Nagata, K.; Fukushima, M.; Haraguchi, M.; Miuma, S.; Miyaaki, H.; Soyama, A.; Hidaka, M.; Eguchi, S.; Shigeno, M.; et al. Evaluating the Role of Hepatobiliary Phase of Gadoxetic Acid-Enhanced Magnetic Resonance Imaging in Predicting Treatment Impact of Lenvatinib and Atezolizumab plus Bevacizumab on Unresectable Hepatocellular Carcinoma. Cancers 2022, 14, 827. [Google Scholar] [CrossRef] [PubMed]
  88. Kuwano, A.; Yada, M.; Narutomi, F.; Nagasawa, S.; Tanaka, K.; Kurosaka, K.; Ohishi, Y.; Masumoto, A.; Motomura, K. Therapeutic efficacy of atezolizumab plus bevacizumab for hepatocellular carcinoma with WNT/β-catenin signal activation. Oncol. Lett. 2022, 24, 216. [Google Scholar] [CrossRef] [PubMed]
  89. Yamashita, T.; Kitao, A.; Matsui, O.; Hayashi, T.; Nio, K.; Kondo, M.; Ohno, N.; Miyati, T.; Okada, H.; Yamashita, T.; et al. Gd-EOB-DTPA-enhanced magnetic resonance imaging and alpha-fetoprotein predict prognosis of early-stage hepatocellular carcinoma. Hepatology 2014, 60, 1674–1685. [Google Scholar] [CrossRef] [PubMed]
  90. Hagiwara, S.; Nishida, N.; Kudo, M. Advances in Immunotherapy for Hepatocellular Carcinoma. Cancers 2023, 15, 2070. [Google Scholar] [CrossRef]
  91. Sia, D.; Jiao, Y.; Martinez-Quetglas, I.; Kuchuk, O.; Villacorta-Martin, C.; de Moura, M.C.; Putra, J.; Camprecios, G.; Bassaganyas, L.; Akers, N.; et al. Identification of an Immune-specific Class of Hepatocellular Carcinoma, Based on Molecular Features. Gastroenterology 2017, 153, 812–826. [Google Scholar] [CrossRef] [PubMed]
  92. Montironi, C.; Castet, F.; Haber, P.K.; Pinyol, R.; Torres-Martin, M.; Torrens, L.; Mesropian, A.; Wang, H.; Puigvehi, M.; Maeda, M.; et al. Inflamed and non-inflamed classes of HCC: A revised immunogenomic classification. Gut 2023, 72, 129–140. [Google Scholar] [CrossRef] [PubMed]
  93. Spahn, S.; Roessler, D.; Pompilia, R.; Gabernet, G.; Gladstone, B.P.; Horger, M.; Biskup, S.; Feldhahn, M.; Nahnsen, S.; Hilke, F.J.; et al. Clinical and Genetic Tumor Characteristics of Responding and Non-Responding Patients to PD-1 Inhibition in Hepatocellular Carcinoma. Cancers 2020, 12, 3830. [Google Scholar] [CrossRef] [PubMed]
  94. Shao, Y.; Liu, T.; Hsu, C.; Lu, L.; Shen, Y.; Lin, Z.; Cheng, A.; Hsu, C. Early alpha-foetoprotein response associated with treatment efficacy of immune checkpoint inhibitors for advanced hepatocellular carcinoma. Liver Int. 2019, 39, 2184–2189. [Google Scholar] [CrossRef] [PubMed]
  95. Scheiner, B.; Pomej, K.; Kirstein, M.M.; Hucke, F.; Finkelmeier, F.; Waidmann, O.; Himmelsbach, V.; Schulze, K.; von Felden, J.; Fründt, T.W.; et al. 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] [PubMed]
  96. Hatanaka, T.; Kakizaki, S.; Hiraoka, A.; Tada, T.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E.; et al. 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] [PubMed]
  97. Capone, M.; Giannarelli, D.; Mallardo, D.; Madonna, G.; Festino, L.; Grimaldi, A.M.; Vanella, V.; Simeone, E.; Paone, M.; Palmieri, G.; et al. Baseline neutrophil-to-lymphocyte ratio (NLR) and derived NLR could predict overall survival in patients with advanced melanoma treated with nivolumab. J. Immunother. Cancer 2018, 6, 74. [Google Scholar] [CrossRef]
  98. Bilen, M.A.; Dutcher, G.M.A.; Liu, Y.; Ravindranathan, D.; Kissick, H.T.; Carthon, B.C.; Kucuk, O.; Harris, W.B.; Master, V.A. Association between Pretreatment Neutrophil-to-Lymphocyte Ratio and Outcome of Patients with Metastatic Renal-Cell Carcinoma Treated with Nivolumab. Clin. Genitourin. Cancer 2018, 16, e563–e575. [Google Scholar] [CrossRef]
  99. Ogata, T.; Satake, H.; Ogata, M.; Hatachi, Y.; Inoue, K.; Hamada, M.; Yasui, H. Neutrophil-to-lymphocyte ratio as a predictive or prognostic factor for gastric cancer treated with nivolumab: A multicenter retrospective study. Oncotarget 2018, 9, 34520–34527. [Google Scholar] [CrossRef]
  100. Bagley, S.J.; Kothari, S.; Aggarwal, C.; Bauml, J.M.; Alley, E.W.; Evans, T.L.; Kosteva, J.A.; Ciunci, C.A.; Gabriel, P.E.; Thompson, J.C.; et al. Pretreatment neutrophil-to-lymphocyte ratio as a marker of outcomes in nivolumab-treated patients with advanced non-small-cell lung cancer. Lung Cancer 2017, 106, 1–7. [Google Scholar] [CrossRef]
  101. Eso, Y.; Takeda, H.; Taura, K.; Takai, A.; Takahashi, K.; Seno, H. Pretreatment Neutrophil-to-Lymphocyte Ratio as a Predictive Marker of Response to Atezolizumab Plus Bevacizumab for Hepatocellular Carcinoma. Curr. Oncol. 2021, 28, 4157–4166. [Google Scholar] [CrossRef] [PubMed]
  102. Tada, T.; Kumada, T.; Hiraoka, A.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E.; Fukunishi, S.; et al. Neutrophil–lymphocyte ratio predicts early outcomes in patients with unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab: A multicenter analysis. Eur. J. Gastroenterol. Hepatol. 2022, 34, 698–706. [Google Scholar] [CrossRef] [PubMed]
  103. 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]
  104. Matsumae, T.; Kodama, T.; Myojin, Y.; Maesaka, K.; Sakamori, R.; Takuwa, A.; Oku, K.; Motooka, D.; Sawai, Y.; Oshita, M.; et al. Circulating Cell-Free DNA Profiling Predicts the Therapeutic Outcome in Advanced Hepatocellular Carcinoma Patients Treated with Combination Immunotherapy. Cancers 2022, 11, 3367. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, C.; Yang, H.; Kim, I.; Kang, B.; Kim, H.; Kim, H.; Lee, W.S.; Jung, S.; Lim, H.Y.; Cheon, J.; et al. Association of High Levels of Antidrug Antibodies Against Atezolizumab with Clinical Outcomes and T-Cell Responses in Patients with Hepatocellular Carcinoma. JAMA Oncol. 2022, 8, 1825–1829. [Google Scholar] [CrossRef] [PubMed]
  106. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update. J. Hepatol. 2022, 76, 681–693. [Google Scholar] [CrossRef] [PubMed]
  107. Kudo, M. Durvalumab Plus Tremelimumab: A Novel Combination Immunotherapy for Unresectable Hepatocellular Carcinoma. Liver Cancer 2022, 11, 87–93. [Google Scholar] [CrossRef]
  108. 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.; et al. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J. Hepatol. 2020, 73, 1460–1469. [Google Scholar] [CrossRef]
  109. Haber, P.K.; Castet, F.; Torres-Martin, M.; Andreu-Oller, C.; Puigvehí, M.; Miho, M.; Radu, P.; Dufour, J.F.; Verslype, C.; Zimpel, C.; et al. Molecular Markers of Response to Anti-PD1 Therapy in Advanced Hepatocellular Carcinoma. Gastroenterology 2023, 164, 72–88.e18. [Google Scholar] [CrossRef]
  110. Kuwano, A.; Yada, M.; Miyazaki, Y.; Tanaka, K.; Kurosaka, K.; Ohishi, Y.; Masumoto, A.; Motomura, K. Tumor-infiltrating CD8+ T cells as a biomarker for chemotherapy efficacy in unresectable hepatocellular carcinoma. Oncol. Lett. 2023, 25, 259. [Google Scholar] [CrossRef]
  111. Lee, P.C.; Wu, C.J.; Hung, Y.W.; Lee, C.J.; Chi, C.T.; Lee, I.C.; Yu-Lun, K.; Chou, S.H.; Luo, J.C.; Hou, M.C.; et al. Gut microbiota and metabolites associate with outcomes of immune checkpoint inhibitor-treated unresectable hepatocellular carcinoma. J. Immunother. Cancer 2022, 10, e004779. [Google Scholar] [CrossRef] [PubMed]
  112. Ng, K.Y.Y.; Tan, S.H.; Tan, J.J.E.; Tay, D.S.H.; Lee, A.W.X.; Ang, A.J.S.; Wong, L.W.J.; Choo, S.P.; Tai, D.W.-M.; Lee, J.J.X. Impact of Immune-Related Adverse Events on Efficacy of Immune Checkpoint Inhibitors in Patients with Advanced Hepatocellular Carcinoma. Liver Cancer 2021, 11, 9–21. [Google Scholar] [CrossRef] [PubMed]
  113. Fukushima, T.; Morimoto, M.; Kobayashi, S.; Ueno, M.; Uojima, H.; Hidaka, H.; Kusano, C.; Chuma, M.; Numata, K.; Tsuruya, K.; et al. Association between Immune-Related Adverse Events and Survival in Patients with Hepatocellular Carcinoma Treated with Atezolizumab Plus Bevacizumab. Oncologist 2023, 28, e526–e533. [Google Scholar] [CrossRef] [PubMed]
Table 3. Potential predictive biomarkers for treating hepatocellular carcinoma with immune checkpoint inhibitors.
Table 3. Potential predictive biomarkers for treating hepatocellular carcinoma with immune checkpoint inhibitors.
Prognostic and Predictive FactorsTherapeuticsStudy DesignOutcomeStatistical Analysis
HR (95% CI)
p-ValueAuthor [Reference No.]
<Etiology>
HBVAnti-PD-(L)1-based
immunotherapy
Meta-analyses
of 3 phase 3 trials:
Checkmate 459
(nivolumab vs. sorafenib),
IMbrave 150
(Atezo/Beva vs. sorafenib),
KEYNOTE-240
(Pembrolizumab vs. placebo)
OSUnivariate
0.64 (0.49–0.83)
0.0008Pfister D et al. [75]
HCVOSUnivariate
0.68 (0.48–0.97)
0.04
NAFLDRetrospective (ICI single arm)OSMultivariate
2.6 (1.2–5.6)
0.017
<Liver function and
general condition>
Child–Pugh AAnti-PD-(L)1-based
immunotherapy
Retrospective,
separate single arm
(not vs. sorafenib)
OSMultivariate
2.3 (1.5–3.4)
<0.001Scheiner B et al. [95]
ECOG PS0OSMultivariate
2.1 (1.4–3.2)
<0.001
<Image>
hypertensive tumor
(RER ‡ ≥ 0.9) on EOB-MRI
Anti-PD-(L)1 monotherapyRetrospective, single arm PFSMultivariate
7.78 (1.59–38.1)
0.011Aoki T et al. [83]
Atezo/Beva,
lenvatinib
Retrospective, separate single arm (not vs. lenvatinib)PFSUnivariate0.012Sasaki R et al. [87]
Heterogenous tumor
on EOB-MRI
PFSUnivariate0.007
Steatotic HCCAtezo/BevaRetrospective validation
based on multiomics study,
single arm
PFSUnivariate<0.05Murai H et al. [86]
<Blood marker>
AFP(<400)Anti-PD-1
monotherapy
Retrospective, single armOSUnivariate
2.81 (1.56–4.97)
<0.0001Spahn et. al. [93]
PFSUnivariate
1.33 (0.86–2.06)
<0.05
AFP response
as a >20% decline
Anti-PD-1
monotherapy
Retrospective, single armOSUnivariate
0.09 (0.02–0.44)
<0.001
PFSUnivariate
0.13 (0.04–0.39)
0.003
AFP (<100)Anti-PD-(L)1-based
immunotherapy
Retrospective,
separate single arm
OSMultivariate
1.7 (1.2–2.6)
0.007Scheiner B et al. [95]
Atezo/BevaRetrospective, single armPFSMultivariate<0.001Hatanaka T et.al. [96]
OSMultivariate0.028
CRP (<1)Anti-PD-(L)1-based
immunotherapy
Retrospective, single armOSMultivariate
1.7 (1.2–2.6)
0.007Scheiner B et.al. [95]
Atezo/BevaRetrospective, single armPFSMultivariate<0.001Hatanaka T et.al. [96]
OSMultivariate0.032
CAFITY score †Anti-PD-(L)1-based
immunotherapy
Retrospective,
separate single arm
(not vs. sorafenib)
OSUnivariate0.001Scheiner B et.al. [95]
CAFITY low1
CAFITY int2.0 (1.1–3.4)
CAFITY high3.6 (2.1–6.2)
CAFITY score †ORRChi-square0.001
DCRChi-square<0.001
CAFITY score †Atezo/Beva Retrospective, single armPFSUnivariate<0.001Hatanaka T et.al. [96]
OSUnivariate
DCRChi-square 0.029
NLR (>3.21)Atezo/Beva Retrospective, single armPFSUnivariate<0.0001Eso Y et.al [101]
NLR (>3)Atezo/Beva Retrospective, single armOSMultivariate
3.37 (1.02–11.08)
0.001Tada T et.al. [102]
IL-6Atezo/Beva Retrospective, single armPFSUnivariate<0.05Myojin Y et.al. [103]
Multivariate
2.785 (1.216–6.38)
0.01
OSUnivariate<0.05
<Transcriptome>
ABRS aAtezo/Beva (not vs. sorafenib)Retrospective analysis of GO30140 arm A cohortPFSUnivariate
0.51 (0.3–0.87)
0.013Zhu AX et. al. [72]
Atezo/Beva vs. sorafenibRetrospective analysis of
IMbrave 150
PFSMultivariate
0.49 (0.25–0.97)
0.041
OSMultivariate
0.26 (0.11–0.58)
0.0012
CD274 bAtezo/Beva (not vs. sorafenib)Retrospective analysis of GO30140 arm A cohortPFSUnivariate
0.42 (0.25–0.72)
0.0011
Atezo/Beva vs. sorafenibRetrospective analysis of
IMbrave 150
PFSMultivariate
0.46 (0.25–0.86)
0.015
OSMultivariate
0.3 (0.14–0.64)
0.002
Teff cAtezo/Beva (not vs. sorafenib)Retrospective analysis of GO30140 arm A cohortPFSUnivariate
0.46 (0.27–0.78)
0.0035
Atezo/Beva vs. sorafenibRetrospective analysis of
IMbrave 150
PFSMultivariate
0.52 (0.28–0.99)
0.047
OSMultivariate
0.24 (0.11–0.5)
0.0002
Treg d/Teff cPFSMultivariate
0.52 (0.28–0.99)
0.047
OSMultivariate
0.24 (0.11–0.5)
0.0002
GPC3PFSMultivariate
0.47 (0.27–0.81)
0.007
OSMultivariate
0.29 (0.13–0.62)
0.002
AFPPFSMultivariate
0.49 (0.28–0.87)
0.014
OSMultivariate
0.32 (0.14–0.73)
0.007
<In situ marker>
CD8+ T cellAtezo/Beva (not vs. sorafenib)Retrospective analysis of GO30140 arm A cohortCR/PRStudent T 0.007Zhu AX et al. [72]
CD3+ T cellCR/PRStudent T 0.039
CD3 + GZMB + T cellCR/PRStudent T 0.044
MHC1 + tumor cellsCR/PRStudent T 0.0087
<Genetic marker>
CTNNB1 WTAtezo/Beva vs. sorafenibRetrospective analysis of
IMbrave 150
OSMultivariate
0.42 (0.19–0.91)
3 × 10−4Zhu AX et al. [72]
PFSmultivariate
0.45 (0.27–0.86)
0.0086
TERT MutOSmultivariate
0.38 (0.16–0.89)
7.8 × 10−5
PFSmultivariate
0.61 (0.33–1.10)
0.047
CD274, CD8A, LAG3, STAT1Anti-PD-1
monotherapy
Retrospective analysis of CheckMate 040 trial OSUnivariate0.01Sangro et al. [108]
PD-L1OSUnivariate0.032
<Other>
Cell-free DNAAnti-PD-1 monotherapyRetrospective, single armOSUnivariate0.018Matsumae et.al. [104]
PFSUnivariate0.021
HR, hazard ratio; CI, confidence interval; PFS, progression-free survival; OS, overall survival; PD-1, programmed death receptor 1; PD-L1, Programmed cell death ligand 1; HBV, hepatitis B virus; HCV, hepatitis C virus; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; RER, relative enhancement ratio; EOB-MRI, gadolinium ethoxybenzyl diethylenetriaminepentaacetic acid-enhanced magnetic resonance imaging; HCC, hepatocellular carcinoma; PR, partial response; ECOG, Eastern Cooperative Oncology Group; AFP, α-fetoprotein; CRP, C-reactive protein; ORR, overall response rate; DCR, disease control rate; NLR, neutrophil to lymphocyte ratio; CR, complete response; irAE, immune-related adverse events. Note: In the “Statistical analysis” section, “univariate” typically refers to the Kaplan–Meier method and log-rank test, and the inclusion of HR indicates the use of Cox regression. Additionally, “multivariate” typically refers to the utilization of the multivariate Cox regression model. ‡: (nodule SI/parenchyma SI on hepatobiliary phase images)/(nodule SI/parenchyma SI on precontract images). SI: signal intensity. †: CRAFITY-low: AFP < 100 and CRP < 1, intermediate: AFP ≥ 100 ng/mL or CRP ≥ 1 mg/dL, high: AFP ≥ 100 ng/mL and CRP ≥ 1 mg/dL. a: ABRS, atezolizumab + bevacizumab response signature (including CXCR2P1, ICOS, TIMD4, CTLA4, PAX5, KLRC3, FCRL3, AIM2, GBP5, and CCL4). b: CD274, PD-L1 mRNA. c: Teff, T effector (including CXCL9, PRF1, and GZMB). d: Treg, T regulatory (including CCR8, BATF, CTSC, TNFRSF4, FOXP3, TNFRSF18, IKZF2, and IL2RA).
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

Motomura, K.; Kuwano, A.; Tanaka, K.; Koga, Y.; Masumoto, A.; Yada, M. Potential Predictive Biomarkers of Systemic Drug Therapy for Hepatocellular Carcinoma: Anticipated Usefulness in Clinical Practice. Cancers 2023, 15, 4345. https://doi.org/10.3390/cancers15174345

AMA Style

Motomura K, Kuwano A, Tanaka K, Koga Y, Masumoto A, Yada M. Potential Predictive Biomarkers of Systemic Drug Therapy for Hepatocellular Carcinoma: Anticipated Usefulness in Clinical Practice. Cancers. 2023; 15(17):4345. https://doi.org/10.3390/cancers15174345

Chicago/Turabian Style

Motomura, Kenta, Akifumi Kuwano, Kosuke Tanaka, Yuta Koga, Akihide Masumoto, and Masayoshi Yada. 2023. "Potential Predictive Biomarkers of Systemic Drug Therapy for Hepatocellular Carcinoma: Anticipated Usefulness in Clinical Practice" Cancers 15, no. 17: 4345. https://doi.org/10.3390/cancers15174345

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