Next Article in Journal
Influence of Extracellular Vesicles on Lung Stromal Cells during Breast Cancer Metastasis
Next Article in Special Issue
Combined Administration of Escitalopram Oxalate and Nivolumab Exhibits Synergistic Growth-Inhibitory Effects on Liver Cancer Cells through Inducing Apoptosis
Previous Article in Journal
A New Role for Yeast Cells in Health and Nutrition: Antioxidant Power Assessment
Previous Article in Special Issue
The Synergistic Effect of Interventional Locoregional Treatments and Immunotherapy for the Treatment of Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Predicting Outcomes of Atezolizumab and Bevacizumab Treatment in Patients with Hepatocellular Carcinoma

1
The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
2
Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(14), 11799; https://doi.org/10.3390/ijms241411799
Submission received: 31 May 2023 / Revised: 13 July 2023 / Accepted: 20 July 2023 / Published: 22 July 2023

Abstract

:
A combination of atezolizumab with bevacizumab (AB) is the first regimen that has shown superiority compared to sorafenib and is now being used as the systemic treatment of choice for hepatocellular carcinoma (HCC) patients with Barcelona Liver Cancer Clinic stage C. However, a considerable number of patients do not achieve survival or significant responses, indicating the need to identify predictive biomarkers for initial and on-treatment decisions in HCC patients receiving AB. In this manuscript, we summarized the current data from both experimental and clinical studies. This review will be beneficial for both clinicians and researchers in clinical practice as well as those designing experimental, translational, or clinical studies.

1. Introduction

Hepatocellular carcinoma (HCC) ranks as the sixth most common cancer and the third leading cause of cancer-related deaths worldwide [1]. The prevalence of this cancer is expected to increase by 55% from 2020 to 2040 [2]. Even though surgical and locoregional treatments can be used in some cases, it is estimated that systemic therapies might be the chosen treatment for 50–60% of HCC patients [1]. Multi-targeted tyrosine kinase inhibitors (TKIs) including sorafenib, lenvatinib, regorafenib, and cabozantinib, which target various molecules, can be used as first- or later-line systemic treatments. These agents commonly target the vascular endothelial growth factor receptor (VEGFR) and also have various molecular targets depending on each drug. However, other than sorafenib and lenvatinib, no drugs have been approved for first-line systemic treatment of advanced HCC, as trials have not demonstrated a significant clinical benefit compared to sorafenib.
Recent breakthroughs have led to a new era in systemic therapies, as immune-checkpoint inhibitors (ICIs) have proven to be effective in patients with HCC. However, the use of ICIs as a monotherapy has demonstrated limited efficacy with a response rate between 15% and 20%, which benefits only a small subgroup of HCC patients in a second-line setting. There are several suggested mechanisms related to the resistance to ICI treatment in HCC, including tumor-intrinsic and extrinsic factors [3]. Thus, numerous efforts to overcome this resistance and improve the clinical outcome of HCC patients have been made, and combinations of other regimens to ICIs have also been tried.
In 2020, the results of the IMbrave150 trial, which enrolled 501 treatment-naïve patients with advanced HCC and assigned them randomly to receive either atezolizumab combined with bevacizumab (AB) or sorafenib monotherapy, were published [4]. AB is the first agent that has shown superiority compared to sorafenib as a first-line systemic treatment, and can be administered at a dose of atezolizumab 1200 mg plus bevacizumab 15 mg/kg IV every 3 weeks, with target concentrations of 6 μg/mL for atezolizumab [5] and 140 μg/mL for bevacizumab [6]. The AB group exhibited a median progression-free survival (PFS) of 6.8 months, whereas the sorafenib group had a median PFS of 4.3 months. At the 12-month follow-up, 67.2% of patients in the AB group and 54.6% in the sorafenib group survived. The objective response rate (ORR) was 33.2% in the AB group, and 13.3% in the sorafenib group, which represented an improved, but insufficient clinical benefit.
Atezolizumab is a monoclonal antibody of the IgG1 isotype that acts on PD-L1 [7], which is present in immune cells or tumor cells within the tumor, blocking its interaction with receptors on the programmed cell death protein 1 (PD-1) and B7-1 (CD80) [4]. The interactions between PD-L1 and PD-1 inhibit T cell proliferation, cytokine secretion, and cytotoxic action, leading to T cell de-activation or exhaustion [8]. Atezolizumab reactivates tumor-specific cytotoxic T cells by disrupting the interaction between PD-1 and PD-L1. Bevacizumab is a monoclonal antibody of IgG1 isotype that targets VEGF, which is a key factor in angiogenesis [7]. Angiogenesis is the process of new blood vessel formation and is regulated by a balance between pro- and anti-angiogenic factors. Representative pro-angiogenic factors encompass the VEGF family, angiopoietins, epidermal growth factors (EGFs), and fibroblast growth factors (FGFs) [9]. Inflammatory cytokines such as interleukin (IL)-6 and IL-8 also participate in angiogenesis [10]. Anti-angiogenic therapies facilitate vascular normalization, impede tumor blood supply, and induce hypoxia and nutrient deficiency, consequently resulting in tumor cell death. Furthermore, they enable a more efficient delivery of therapeutic agents and immune cells to the tumor site. However, there is no randomized trial showing the clinical benefits of bevacizumab monotherapy in HCC.
The AB combination treatment may possess a potential synergistic effect in cancer treatment, enhancing their combined therapeutic efficacies. Anti-VEGF therapies counteract VEGF-induced immunosuppression within tumors and their microenvironments, potentially boosting anti-PD-1 and anti-PD-L1 effectiveness by reversing VEGF-driven immunosuppression and enhancing T cell infiltration, thereby enhancing antitumor immune responses [11].
Although the combination regimen targets two different molecules, programmed cell death-ligand 1 (PD-L1) and vascular endothelial growth factor (VEGF), previous reviews have only focused on biomarkers for ICIs, and have not considered the point that this regimen further targets VEGF signaling. In this manuscript, we aim to reconcile the current knowledge on this topic and review both experimental/translational and clinical studies. This review will be beneficial for both clinicians and researchers in terms of making decisions and planning clinical, translational, and experimental studies. Brief introductions of the mechanisms of action and currently reported biomarkers for AB treatment in HCC patients are presented in Figure 1.

2. Clinico-Radiological Parameters

2.1. Clinical Parameters

2.1.1. Etiology

Previous studies that have identified clinical factors associated with outcomes following AB treatment are summarized in Table 1. A recent experimental study showed that pathologic CD8+PD-1+ T cells might be associated with the limited role of anti-PD-1 treatment in NASH-related HCC [12]. Another study showed that hepatitis B virus (HBV)-infected subjects have distinct upregulation of peripheral blood inflammatory cytokine profiles, compared to the other etiologies including hepatitis C virus (HCV), NASH, and alcoholic liver diseases, suggesting different peripheral, intrahepatic, and intratumoral immune environments across the etiologies of HCC [13]. The tendency of better clinical outcomes in patients with viral etiologies have been reported in association with nivolumab [14], cabozantinib plus atezolizumab [15], and tremelimumab plus durvalumab [16] regimens.
In updated efficacy and safety data from IMbrave150, AB treatment in patients with viral etiologies including HBV and HCV had superior OS and PFS compared to those treated with sorafenib, although the subgroup analysis that only included AB treatment was not presented [17]. A recent meta-analysis, which included 3 large randomized phase III trials of nivolumab, AB, and pembrolizumab, suggested that the ICI regimen might be superior to sorafenib in terms of OS in HBV- and HCV-related HCC [12]. In addition, a recent network meta-analysis showed that patients with viral etiology showed significant survival benefits with the AB regimen compared to the TKIs [18]. However, such research has provided glimpses into the role of etiology as a predictive marker because these studies only showed the benefits of the AB regimen compared to the TKIs.
A reduction in AFP levels (≥75%) at 6 weeks following the start of therapy can serve as a potential biomarker for HCC patients receiving the AB treatment to predict improved OS and PFS, particularly in those with HBV etiology, but not in the HCV and non-viral etiologies in the recent report analyzing 440 patients who were included in the Phase Ib and III trials of IMbrave150 [19]. A small-sized (n = 66) recent real-world study also showed that patients with viral etiologies have better OS and PFS than patients with non-viral etiology [20]. Another small-sized retrospective study (n = 23) also showed that patients with viral etiology have higher ORR than those with non-viral etiology receiving the AB treatment [21]. However, other real-world studies did not find differences between the two groups, therefore larger nationwide studies are needed to validate the previous data. In addition, whether there might be a difference between HBV and HCV in the clinical outcome of AB treatment and its related mechanism also needs to be clarified.

2.1.2. Tumor Burden

A larger tumor burden is associated with the more immunosuppressive tumor microenvironment (TME) contributed by regulatory T cells (Tregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), as well as immunosuppressive cytokines such as tumor growth factor-beta (TGF-β) and IL-10 [22]. In HCC patients treated with nivolumab, an intrahepatic tumor size of more than 10 cm was associated with poor OS and PFS [23]. However, there is a possibility that the VEGF inhibitor may have a complementary effect, and therefore, the impact of tumor burden on the clinical outcome of the AB treatment remains unclear. A recent retrospective report (n = 121) showed that macrovascular invasion was associated with poor OS in AB treatment [24]. In addition, the presence of extrahepatic spread was associated with poor PFS in AB treatment in a recent real-world study (n = 433) [25]. The accumulation of data from further studies will confirm this association.

2.1.3. Liver Function Parameters

Although most clinical trials, including IMbrave150, enrolled patients with good liver function of Child–Pugh A, liver function status in real-world practice might be variable and can change dynamically from the baseline status. Liver function plays a critical role in determining the prognosis and treatment options for HCC patients, particularly in patients receiving the AB treatment. A small-sized retrospective study (n = 100) showed that patients with Child–Pugh B had comparable ORR compared to those with Child–Pugh A, but had shorter OS and PFS [26]. A recent real-world study (n = 66) also showed that Child–Pugh A was a significant favorable factor for OS [20]. In addition, there was no difference between Child A and B patients in ORR, but PFS and OS were significantly better in the Child A group in a retrospective study (n = 457) [27], which indicates that liver function might be related to the prognosis rather than reflecting the therapeutic efficacy of the AB treatment.
In addition to the Child–Pugh score, albumin–bilirubin (ALBI) grade is also an important indicator of liver function, and several studies have investigated the associations between ALBI and clinical outcomes in AB treatment. Recent small-sized retrospective studies have demonstrated that ALBI grade [28,29] and Eastern Cooperative Oncology Group [28] scores before treatment were independent factors in predicting OS or PFS. Another real-world study (n = 28) also showed that a lower baseline modified ALBI (mALBI) predicts better ORR, and a Child–Pugh score of five and mALBI grades 1 and 2a were significantly associated with the continuation of treatment [30]. In addition to its baseline status, a worsening ALBI score within 3 weeks after the AB treatment was also significantly associated with OS in a retrospective analysis (n = 69) [31]. It can be combined with another biomarker. A recent multicenter retrospective study (n = 426) showed that the combination of mALBI grade and AFP (mALF score) significantly predicted OS and PFS [32].

2.2. Pre-Treatment Radiologic Examinations

2.2.1. Hepatobiliary Phase of Magnetic Resonance Imaging (MRI)

In gadoxetic acid-enhanced MRI, it has been suggested that the hepatobiliary phase could be an imaging biomarker for the identification of β-catenin mutations in HCC [33]. Recent studies suggest that HCC with CTNNB1 mutations, which induces activation of the Wnt/β-catenin pathway, is characterized by reduced intratumoral T cell infiltrations [34] and can also be related to the ICI response [35]. According to the ratio of relative enhancement and visual assessment of the hepatobiliary phase, HCCs could be classified into hypo- and hyperintensity types, as well as heterogeneous and homogeneous types. In a recent small-sized study (n = 35), the heterogeneous/hyperintensity type in the baseline MRI imaging had significantly shorter PFS compared to homogeneous/hypointensity types [36].

2.2.2. Perfusion Changes in Computed Tomography (CT), MRI, and Contrast-Enhanced Ultrasound (CEUS)

Changes in tumor perfusion have been observed in AB treatment in a preclinical HCC model [37], and this might be due to the immune cell infiltration by atezolizumab, as well as vascular normalization by bevacizumab. This finding implies that measuring a dynamic change in tumor perfusion might have a role in predicting the responses of AB treatment in HCC. In a recent retrospective pilot study (n = 19), perfusion change, which represents the decline in tumor-to-liver ratio in the arterial phase of CT or MRI at a mean of 9 weeks after AB treatment, was significantly associated with the disease control rate (DCR) [38]. Thus, early measures of perfusion change might help in predicting treatment response and long-term outcomes. In addition to the CT/MRI imaging, another retrospective study (n = 35) evaluated time-intensity curve (TIC) analysis using CEUS 3 to 7 days after the initial AB treatment [39]. As a result, cases without decreased blood flow showed significantly higher rates of progressive disease, compared to those with decreased blood flow. Decreased blood flow in the TIC analysis was also associated with longer PFS.

2.2.3. Positron Emission Tomography-Computed Tomography (PET-CT)

Imaging characteristics on 18F-fluorodeoxyglucose PET-CT (18F-FDG-PET-CT) have demonstrated a strong association with poorly differentiated HCC, and the presence of 18F-FDG-PET/CT-positive HCC is known as an unfavorable prognostic indicator for responses to anti-HCC treatments including lenvatinib or TACE [40]. A recent retrospective study (n = 20) evaluated the tumor-to-normal liver ratio (TLR) of FDG uptake before AB treatment and found that a baseline TLR ≥ 2 was associated with early progressive disease and poor PFS, but not with OS [41].

2.3. Adverse Events (AEs)

The appropriate monitoring and management of AEs are important in the continuation of chemotherapy and the outcome of patients. In fact, early bevacizumab interruption within 9 weeks after treatment was related to shorter PFS and OS and was associated with AEs, including liver injury, poor oral intake, proteinuria, and ascites [42]. It was also associated with a poor mALBI grade and, importantly, it also affected the implementation of later-line treatment. The following are current data regarding AEs and their impact on the outcomes of patients in AB treatment.

2.3.1. Immune-Related Adverse Events (irAEs)

Because it reinforces the immune system, ICI can cause irAEs that involve multiple organs such as the skin, gastrointestinal, respiratory, thyroid, and central nervous systems. However, it is unclear whether irAEs are associated with efficacy or survival, particularly in HCC patients receiving AB treatment. A recent retrospective study (n = 150) evaluated irAEs and their impact on the outcome of patients [43]. This study classified irAEs into endocrine, dermatologic, gastrointestinal, hepatic, hematological, pulmonary, musculoskeletal, cardiovascular, nervous system, and renal events. The authors found that total irAEs, not independent irAEs, are not associated with the ORR. However, grade 1/2 irAEs were significantly associated with favorable PFS and OS, compared to grade 3/4 irAEs or no irAEs in the multivariate analysis. Another retrospective study (n= 130) found that skin reactions were associated with longer OS [44]. These results suggest that irAEs might reflect the activation of immune function and the effectiveness of ICIs including atezolizumab, but the severe grade of irAEs might reduce this beneficial effect due to the discontinuation of treatment or their own severity.
Since most patients have liver cirrhosis (LC) or chronic hepatitis, hepatic irAEs require special attention in HCC patients receiving ICIs. Prior studies have indicated that liver damage is linked to unfavorable outcomes in cancer patients undergoing ICI therapy, and even HCC patients experiencing grade 1 or 2 liver injury during ICI treatments demonstrated a poor prognosis [45,46,47]. In patients receiving the AB treatment, liver injuries including AST, ALT, and bilirubin elevation were associated with shorter OS [44,48]. On the other hand, anti-VEGF treatment can also cause liver injury and reduce liver function as shown in a clinical trial of ramucirumab in HCC patients [49]. Therefore, further studies are needed to distinguish whether the liver injury is related to the irAE during AB treatment, and on-treatment strategies including dose modification or special management should be investigated.

2.3.2. Anti-VEGF-Related AEs

Hypertension related to anti-VEGF was linked to a better DCR and PFS in HCC patients undergoing AB therapy in a recent retrospective study (n = 286) [50]. Another study also showed that hypertension is associated with longer OS [44]. The underlying mechanism is uncertain; however, the direct relationship between VEGF inhibition and hypertension onset is evident, given the role of VEGF in maintaining normal endothelial cell function and vascular balance [51].
Proteinuria was also significantly associated with better OS in a recent Japanese real-world study (n = 286) [48]. Although its impact on bevacizumab efficacy is controversial across various types of cancer, proteinuria was correlated with VEGF signal inhibition [52].
Table 1. Previous studies investigating the association between clinico-radiological factors and outcome.
Table 1. Previous studies investigating the association between clinico-radiological factors and outcome.
Clinical MarkersRelated OutcomesStudy Design (n)Reference
Viral etiology (HBV + HCV)Favorable OS, PFS of AB compared to SORPhase III RCT- IMbrave150 (336)Cheng et al. [17]
AFP reduction (≥75%) after Tx in HBV subjectsFavorable OS, PFSPhase Ib and phase III IMbrave150 (440)Zhu et al. [19]
Viral etiology (HBV + HCV)Favorable ORRRetrospective (23)Takeda et al. [21]
Viral etiology (HBV + HCV)Favorable OS, PFSRetrospective (66)Himmelsbach et al. [20]
Macrovascular invasionUnfavorable OSRetrospective (121)Chon et al. [24]
Extrahepatic spreadUnfavorable PFSRetrospective (433)Fulgenzi et al. [25]
Child–Pugh BTreatment discontinuationRetrospective (28)Tanaka et al. [30]
Child–Pugh BUnfavorable OS, PFSRetrospective (100)Jost-Brinkmann et al. [26]
Child–Pugh BUnfavorable OS, PFSRetrospective (66)Himmelsbach et al. [20]
Child–Pugh BUnfavorable OS, PFSRetrospective (457)Tanaka et al. [27]
High ALBI and ECOGUnfavorable OS, PFSRetrospective (147)de Castro et al. [28]
High ALBIUnfavorable OS, PFSRetrospective (50)Sinner et al. [29]
High mALBIUnfavorable ORR, treatment discontinuationRetrospective (28)Tanaka et al. [30]
High mALF score (mALBI + AFP)Unfavorable OS, PFSRetrospective (426)Hatanaka et al. [32]
Worsening ALBI at wk3Unfavorable OSRetrospective (69)Unome et al. [31]
Heterogeneous/hyperintensity in HBP of MRIUnfavorable PFSRetrospective (35)Sasaki et al. [36]
Decline in TLR in the arterial phase of CT or MRI after TxFavorable DCRRetrospective (19)Onuoha et al. [38]
Decrease in blood flow in CEUS after TxFavorable DCR, PFSRetrospective (35)Takada et al. [39]
High tumor-to-normal ratio of FDG uptake in PET-CTUnfavorable PFS and DCRRetrospective (20)Kawamura et al. [41]
Grade 1/2 irAEsFavorable OS, PFSRetrospective (150)Fukushima et al. [43]
Skin reactionFavorable OSRetrospective (130)Shimose et al. [44]
Liver injuryUnfavorable OSRetrospective (130)Shimose et al. [44]
Liver injuryUnfavorable OSRetrospective (286)Takaki et al. [48]
HypertensionFavorable DCR, PFSRetrospective (286)Tada et al. [50]
HypertensionFavorable OSRetrospective (130)Shimose et al. [44]
ProteinuriaFavorable OSRetrospective (286)Takaki et al. [48]
HBV, hepatitis B virus; HCV, hepatitis C virus; OS, overall survival; PFS, progression free survival; AB, atezolizumab + bevacizumab; SOR, sorafenib; RCT, randomized controlled trial; AFP, alpha-fetoprotein; Tx, treatment; ORR, objective response rate; BCLC, Barcelona Clinic Liver Cancer; ALBI, albumin–bilirubin; ECOG, Eastern Cooperative Oncology Group; mALBI, modified albumin–bilirubin; HBP, hepatobiliary phase; MRI, magnetic resonance imaging; DCR, disease control rate; CT, computed tomography; TLR, tumor–liver ratio; CEUS, contrast-enhanced ultrasonography; FDG, fluorodeoxyglucose; PET, positron emission tomography; and irAEs, immune-related adverse events.

3. Blood-Based Biomarkers

3.1. Clinically Available Blood-Based Biomarkers

3.1.1. Alpha-Fetoprotein (AFP)

Previous studies that have investigated predictive factors using blood samples are summarized in Table 2. Higher levels of AFP correlate with lower survival and higher tumor recurrence rates across different stages of HCC. Despite its association with worse outcomes, AFP has not been confirmed as a predicting factor in trials for various first-line systemic treatments. However, AFP levels above 400 ng/mL have been linked to a poorer response for ramucirumab, showing for the first time that a biomarker that can be used to select a systemic treatment in HCC [49].
Many clinical studies have investigated whether AFP can be used as a biomarker for predicting clinical outcomes in HCC patients receiving the AB treatment, with a gradual accumulation of evidence. For example, a baseline AFP level of ≥100 ng/mL was associated with poor PFS in a recent real-world study (n = 286) [50]. Furthermore, more data have reported that its dynamic change might be associated with the clinical outcome of AB treatment. A reduction in AFP levels (≥75%) at 6 weeks may serve as a potential biomarker for predicting improved OS and PFS in HCC patients receiving the AB treatment, particularly in those with HBV etiology among 440 patients enrolled in the phase Ib and III trials of IMbrave150 [19]. Another prospective study (n = 284) additionally evaluated the optimal cut-off reduction in AFP level at week six [53]. As a result, both 20% and 50% showed a significant relationship with ORR and PFS. In another retrospective study (n = 58), AFP response at 6 weeks after AB treatment was associated with the ORR, OS, and PFS [54]. Early AFP reduction at 3 weeks also predicted a better radiological response and OS in the small-sized retrospective study (n = 75) [55]. This study also showed that an AFP ratio of 1.4 or higher at 3 weeks was related to PFS. These results suggest that the dynamic change of AFP has a role as an important biomarker for early determination of treatment continuation, although this needs to be further validated.

3.1.2. Protein Induced by Vitamin K Antagonist-II (PIVKA-II)

PIVKA-II is a tumor marker that is closely associated with the prognosis of HCC patients. The reduction in PIVKA-II was correlated with better ORR, OS, and PFS in HCC patients who underwent nivolumab treatment [56]. Several reports have observed an association between PIVKA-II level and prognosis in AB treatment. A recent retrospective study (n = 121) showed that a higher level of PIVKA-II at baseline (≥86 mAU/mL) was associated with poor OS and PFS [24]. A baseline PIVKA-II level of <400 mAU/mL was also associated with favorable PFS in a real-world study (n = 75) [57]. Another retrospective study (n = 69) also suggested that an early increase in PIVKA-II level was related to poor OS [31].

3.1.3. C-Reactive Protein (CRP)

High levels of CRP are associated with systemic inflammation and the progression of cancer [58]. It also has an immunosuppressive effect, which might be associated with the impaired efficacy of ICIs. A previous study showed that an elevated CRP level was a significant factor for poor PFS and OS in various types of cancers treated with ICIs [59]. Scoring systems using CRP and other parameters have been developed and validated in HCC patients receiving AB. A recent study showed that CRP < 1 mg/dL and AFP < 100 ng/mL at baseline are significantly associated with OS in HCC patients receiving PD-L1 immunotherapy [60]. These two markers were subsequently used to create the CRAFITY score, and the radiological response was significantly better in a lower CRAFITY score. Another retrospective study validated the CRAFITY score as a significant factor for OS and PFS in AB-treated HCC patients (n = 89) [61]. A Japanese retrospective study (n = 297) showed that the CRAFITY score significantly predicted OS and PFS, AEs, including liver injury, loss of appetite, proteinuria, fever, and fatigue [62].
A neo-Glasgow prognostic score (GPS) was reported in HCC patients who underwent surgery associated with postoperative complications [63]. Individuals exhibiting a serum CRP concentration exceeding 1.0 mg/dl in conjunction with an ALBI grade of either two or three can be classified as having a neo-GPS score of two. A recent retrospective study validated this scoring system with 421 patients receiving AB treatment and found that it was independently related to OS and DCR [64]. Studies using CRAFITY and neo-GPS systems suggest that CRP should also be considered in the outcome prediction of AB treatment in HCC.

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

A high NLR has been suggested to potentially serve as a marker for resistance to ICIs due to its association with intratumoral concentrations of MDSCs [65], TAMs [66], a serum cytokine profile encompassing pro-inflammatory and angiogenic cytokines [67], and the presence of tumor-infiltrating lymphocytes (TILs) [68]. Prior research has indicated that an increased NLR is linked to a worse prognosis in HCC patients undergoing nivolumab treatment [69,70]. Evidence on the role of the NLR in predicting outcomes of AB treatment in HCC patients has accumulated. An NLR > 3.21 was associated with poor ORR in a small-sized retrospective study (n = 40) [71]. Another retrospective study (n = 240) reported that an NLR ≥ 3 was not associated with the ORR, but it was associated with the cumulative discontinuation rate due to AEs, resulting in a shorter OS [72]. A German real-world study (n = 100) also showed that an NLR > 3.2 was the most significant factor predicting poor ORR and PFS [26]. Furthermore, an NLR ≥ 3 at baseline was an independent risk factor related to hyperprogressive disease (HPD) in a pilot study (n = 8) [73]. Patients with an NLR ≥ 5 had significantly poorer OS in a real-world study (n = 296) [74], and another retrospective study (n = 121) suggested that an NLR ≥ 2.5 at baseline was associated with poor OS and PFS [24]. The latter study also showed that an NLR decrease of 10% or more at the first response evaluation was an independent factor for longer OS. An NLR < 1.97 in the second course was associated with better ORR, OS, and PFS in a recent retrospective study (n = 110) [75]. An investigation to determine its optimal cut-off and mechanism should be performed in future studies. In addition to the NLR > 3, a PLR > 230 is a risk factors for poor PFS in a small-sized retrospective study (n = 48) [76]. Therefore, the NLR should be considered as a biomarker, and baseline and dynamic changes in its level should also be evaluated.

3.1.5. Prognostic Nutritional Index (PNI)

The PNI can be calculated with serum albumin and the absolute count of peripheral blood lymphocytes and has been used as a prognostic marker in HCC patients who have undergone liver transplantation [77]. However, its impact on HCC patients receiving ICIs, including an AB regimen, has been unclear. A recent study showed that a high PNI of 47 or more, with an AFP level lower than 100 ng/mL, were independent factors associated with better OS and PFS in a recent retrospective study (n = 286) [78].

3.2. Other Blood-Based Biomarkers Based on Experimental Research

3.2.1. Serum IL-6

IL-6 is a cytokine that is elevated in hepatitis, LC, and HCC patients [79], and also has a tumor-promoting or tumorigenesis effect that might be associated with the attenuation of T cell function and recruitment [80]. In a recent study, the association between serum IL-6 and clinical outcomes in AB-treated patients was investigated prospectively (n = 165) [81]. Among the various blood-derived biomarkers, a serum level of IL-6 was significantly elevated in patients who did not achieve favorable outcomes (complete response, partial response, or stable disease for at least 6 months), and it also correlated with poor OS and PFS. This study found that high IL-6 levels correlated with decreased interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) secretion from CD8+ T cells, which was validated by in vitro assays showing that the treatment of IL-6 inhibited cytokine secretion and expansion of CD8+ T cells. Furthermore, patients with elevated IL-6 levels displayed a non-T-cell-inflamed immunosuppressive TME in the transcriptome analysis. Results from another prospective study (n = 64) were also compatible, which showed that higher levels of serum IL-6 correlated with a poorer ORR, OS, and PFS [82].

3.2.2. Peripheral Blood PD-1 Expression on Granulocytes

In addition to the NLR, a prospective study further evaluated the expressions of PD-1 and PD-L1 of granulocytes among whole blood samples using flow cytometry in 34 patients with AB treatment [83]. Interestingly, PD-1 expression on granulocytes, but not PD-L1, was a significant factor, and a low baseline PD-1 percentage expressed on granulocytes was associated with better ORR and longer time to progression. Future translational studies investigating the characteristics of the peripheral blood immune cell population, as well as their dynamic changes, and their association with the clinical outcome in AB treatment, should be performed.

3.2.3. Factors Associated with Aberrant Angiogenesis

A recent retrospective study (n = 46) evaluated the serial changes of growth factors and found that patients who initially had disease control but later progressed had significantly higher levels of VEGF-D and ANG-2 [84]. These findings suggested that increased levels of VEGF-D and ANG-2 in the serum may contribute to the resistance of AB treatment.
Insulin-like growth factor-1 (IGF-1) is also associated with angiogenesis in addition to liver function [85]. A recent study divided HCC patients receiving AB treatment into baseline IGF-1 high, normal, and low groups among 371 patients enrolled in the phase III IMbrave150 trial, and found that the low IGF-1 group showed significantly better OS and PFS [86].
Growth hormone (GH) is known to promote tumor angiogenesis [87] and is also linked to tumorigenesis or tumor-promoting effects in various types of cancers, such as breast cancer and HCC. A small-sized previous study (n = 37) evaluated the prognostic role of GH in HCC patients receiving AB, the low-GH group showed significantly better OS than the high-GH group, but not PFS [88].
Table 2. Previous studies investigating the association between blood-based markers and outcome.
Table 2. Previous studies investigating the association between blood-based markers and outcome.
Blood MarkersRelated OutcomesPredictive/PrognosticStudy Design (n)Reference
AFP ≥ 100 ng/mLUnfavorable PFSPrognosticRetrospective (286)Tada et al. [50]
AFP < 100 ng/mLFavorable OS, PFSPrognosticRetrospective (485)Tada et al. [78]
AFP reduction (≥75%, 6 wks) in HBV subjectsFavorable OS, PFSPrognosticPhase Ib and phase III IMbrave150 (440)Zhu et al. [19]
AFP reduction (≥50% or 20%, 6 wks)Favorable ORR, PFSPredictive, prognosticProspective (284)Tamaki et al. [53]
AFP reduction, 6 wksFavorable ORR, OS, PFSPredictive, prognosticRetrospective (58)Kuzuya et al. [54]
Early AFP reduction, 3 wksFavorable ORR, OS, PFSPredictive, prognosticRetrospective (75)Campani et al. [55]
PIVKA-II ≥ 186 mAU/mLUnfavorable OS, PFSPrognosticRetrospective (121)Chon et al. [24]
PIVKA-II ≥ 400 mAU/mLUnfavorable PFSPrognosticRetrospective (75)Ochi et al. [57]
Early increase in PIVKA-IIUnfavorable OS, PFSPrognosticRetrospective (69)Unome et al. [31]
High CRAFITY (CRP + AFP) scoreUnfavorable OS, PFS, frequent AEsPrognosticRetrospective (297)Hatanaka et al. [62]
High CRAFITY (CRP + AFP) scoreUnfavorable OS, PFSPrognosticRetrospective (89)Teng et al. [61]
High Neo-GPS (CRP + ALBI) scoreUnfavorable OS, DCRPredictive, prognosticRetrospective (421)Tada et al. [64]
NLR > 3.21Unfavorable ORRPredictiveRetrospective (40)Eso et al. [71]
NLR ≥ 3Unfavorable OS, treatment discontinuationPrognosticRetrospective (240)Tada et al. [72]
NLR > 3.2Unfavorable ORR, PFSPredictiveRetrospective (100)Jost-Brinkmann et al. [26]
NLR ≥ 3Hyperprogressive diseasePrognosticRetrospective (8)Maesaka et al. [73]
NLR ≥ 5Unfavorable OSPrognosticRetrospective (296)Wu et al. [74]
NLR ≥ 2.5Unfavorable OS, PFSPrognosticRetrospective (121)Chon et al. [24]
NLR decrease ≥ 10% (at 1st response evaluation)Favorable OSPrognosticRetrospective (121)Chon et al. [24]
NLR ratio at second course < 1.97Favorable ORR, OS, PFSPredictive, prognosticRetrospective (110)Matoya et al. [75]
NLR > 3, PLR > 230Unfavorable PFSPrognosticRetrospective (48)Wang et al. [76]
PNI (albumin + peripheral lymphocyte counts) ≥ 47Favorable OS, PFSPrognosticRetrospective (286)Takaki et al. [78]
High serum interleukin-6Unfavorable DCR, OS, PFSPredictive, prognosticProspective (165)Yang et al. [81]
High serum interleukin-6Unfavorable ORR, OS, PFSPredictive, prognosticProspective (64)Myojin et al. [82]
Low baseline PD-1% on granulocytesFavorable ORR, TTPPredictiveProspective (34)Giovannini et al. [83]
Elevated VEGF-D and ANG-2Durable disease controlPredictiveRetrospective (46)Yang et al. [84]
Low IGF-1Favorable OS, PFSPrognosticPhase III IMbrave150 (371)Kaseb et al. [86]
Low growth hormoneFavorable OSPrognosticProspective (37)Mohamed et al. [88]
High anti-drug antibodies, 3 wksUnfavorable OS, PFSPrognosticProspective (174)Kim et al. [89]
High ctDNA Unfavorable ORR, OS, PFSPredictive, prognosticRetrospective (85)Matsumae et al. [90]
TERT mutation within ctDNA Unfavorable OSPrognosticRetrospective (85)Matsumae et al. [90]
Low CXCL9 within ctDNAUnfavorable DCRPredictiveRetrospective (29)Hosoda et al. [91]
AFP, alpha-fetoprotein; wks, weeks; PFS, progression-free survival; OS, overall survival; ORR, objective response rate; AEs, adverse events; DCR, disease control rate; HBV, hepatitis B virus; PIVKA-II, protein induced by vitamin K antagonist-II; GPS, Glasgow prognostic score; ALBI, albumin–bilirubin; NLR, neutrophil–lymphocyte ratio; PLR, platelet–lymphocyte ratio; PNI, prognostic nutritional index; PD-1, programmed cell death-1; VEGF, vascular endothelial growth factor; ANG-2, angiopoietin-2; IGF-1, insulin-like growth factor-1; ctDNA, circulating tumor DNA; TERT, telomerase reverse transcriptase; and CXCL9, chemokine ligand9.

4. Tissue-Driven Biomarkers

4.1. Biomarkers Related to Immune Cells

4.1.1. PD-L1 Expression

Previous studies that have investigated predictive factors using tissue samples are summarized in Table 3. In the tumor and adjacent liver tissues of HCC, PD-L1 can be expressed by tumor cells, Kupffer cells, hepatocytes, and sinusoidal cells [92]. Although its expression is variable across the studies and antibody clones used for immunohistochemistry (IHC), its expression within tumor tissues has been considered to be associated with macrovascular invasion, poor differentiation, high AFP levels, and poor prognosis in HCC, as reported previously [93,94]. Its impact on the efficacy of ICI treatment in HCC patients is controversial. For example, PD-L1 expression measured by IHC on tumor cells did not affect the outcome of HCC patients treated with nivolumab [95], but the baseline combined positive score (CPS) of PD-L1 expression was related to ORR in pembrolizumab-treated patients [96]. In the phase III IMbrave150 trial, PD-L1 expression (clone SP263) with a CPS ≥ 1% was associated with the benefits of the AB treatment in terms of PFS and ORR, compared to the sorafenib treatment [17]. However, another experimental study included 358 patients enrolled in the phase Ib and III IMbrave150 trial, which also used an SP263 clone for IHC, did not observe a difference in ORR [97]. Therefore, further studies are needed to validate the role of PD-L1 expression measured by IHC as a biomarker for AB treatment in HCC. Of note, the standardization for the antibody clone and staining methods, and adjustment of the cut-off value are mandatory as it is the key target molecule of AB treatment.

4.1.2. Immune Cell Infiltrations within Tumor Tissues

Although immune cells including various subsets of T cells, TAMs, and MDSCs among the TME of HCC might be closely related to the responses to ICI treatment, limited data have been reported, probably due to the difficulty in obtaining tumor tissues. In nivolumab-treated HCC patients, higher baseline CD3+ and CD8+ TILs measured by IHC were related to better OS [69]. Furthermore, higher CD3+ and CD8+ TILs 6 weeks after tremelimumab treatment for HCC were correlated to the ORR [98], which implicates the role of tumor biopsy and histologic examinations for immune cell populations before and after ICI treatment.
Using multiplex IHC staining, a previous study using samples from 358 patients enrolled in the phase Ib and phase III IMbrave150 trial showed that higher infiltrations of CD3+ T cells, CD8+ T cells, MHC-1+ tumor cells, and CD3+granzyme B+ T cells, within tumor tissues were associated with the ORR of AB treatment [97]. They also used xCell deconvolution analysis from transcriptome data and observed that a higher presence of CD8+ T cells, CD4+ T cells, Tregs, B cells, and dendritic cells (DCs) is associated with better ORR and PFS. Further examinations evaluating various immune cell populations within tumor tissues before and after AB treatment should be performed. In addition, the baseline evaluation and monitoring of the peripheral immune cell population should also be considered in future studies.

4.2. Atezolizumab-Bevacizumab Response Signature

The development of novel signatures by tumor tissues that can predict responses should also be investigated. Using bulk RNA sequencing analyses of tumor tissues, the authors built an atezolizumab–bevacizumab response signature (ABRS) that contains genes related to pre-existing immunity, including genes associated with high PD-L1 and effector T cell signatures using samples from patients enrolled in the phase Ib and phase III IMbrave150 trial. As a result, a high ABRS was significantly associated with better ORR and PFS [97].

5. Novel Biomarkers

5.1. Anti-Drug Antibodies (ADA)

Atezolizumab treatment has been reported to induce ADA [99]. Consequently, this might reduce the level of atezolizumab, but the impact on clinical efficacy has been unclear. Serum ADA can be measured by enzyme-linked immunosorbent assays. A recent prospective multicenter cohort study (n = 174) showed that a higher ADA level in AB-treated HCC patients at 3 weeks was associated with poor PFS and OS [89]. A high level of ADA against atezolizumab at 3 weeks after initial treatment was found in 17.4% of the patients, whereas 82.6% of the patients had low or negative results. The levels of ADAs were negatively correlated with the level of atezolizumab. A high-ADA group had significantly lower OS and PFS in discovery (HR = 2.84 and 3.30, respectively) and validation (HR = 2.52 and 5.81, respectively) studies. Furthermore, patients with higher ADA levels showed lower proliferation and cytokine-secreting functions of T cells, suggesting that ADA attenuated the antitumor immune response. Future studies might be needed to validate this study.

5.2. Liquid Biopsy

5.2.1. Circulating Tumor DNA (ctDNA) Levels and TERT Mutation

The number of ctDNA can be correlated with tumor stage and prognosis in HCC, and a recent study (n = 85) evaluated its association with prognosis in patients receiving AB [90]. As a result, a higher level of ctDNA was correlated with poorer ORR, PFS, and OS. When ultradeep sequencing was performed, the TERT promoter, tumor protein 53 (TP53), and catenin beta 1 (CTNNB1) were frequently mutated, but TERT ctDNA mutation was an independent factor predicting poor OS. CTNNB1 mutation was not a significant factor, as found in another study [100]. These findings imply that measuring and profiling ctDNA should be researched with a larger sample size in future studies.

5.2.2. CXCL9 within ctDNA

Litchfield and colleagues examined whole-exome and transcriptome data from over 1000 patients undergoing treatment with ICIs and found that a high expression of CXCL9 is among the most significant factors predicting a positive response to ICI therapy [101]. This might be due to the role of CXCL9 in attracting cytotoxic CD8+ T cells to the tumor site [102]. In a recent Japanese retrospective study (n = 29), authors performed a cytokine array analysis using ctDNA and found that low CXCL9 levels within the ctDNA were associated with early PD in discovery and validation cohorts receiving AB [91].

6. Potential but Unproven Markers

6.1. Sarcopenia and Obesity

Sarcopenia refers to the gradual decline in both the amount and power of skeletal muscles and is associated with the prognosis in HCC patients who underwent surgical resection [103] and systemic therapies including sorafenib or lenvatinib [104,105]. In a small-sized retrospective study, there was no association between sarcopenia and prognosis in AB-treated HCC patients [106]. The association between body mass index (BMI) and the clinical outcome of ICI treatment is controversial across the cancer types and studies, and the relationship between BMI and immunotherapy in HCC has not been researched extensively. One study has shown that a BMI of 25 or higher is linked to better OS, but not PFS, in patients treated with PD-1 antibody-based therapies [107]. A recent study investigated whether high BMI is associated with the clinical outcome in patients with the AB treatment, but there was no significance in OS, PFS, and ORR [108]. Nevertheless, more data are needed to confirm the role of sarcopenia and obesity in the prognosis of HCC patients treated with AB.

6.2. Tumor Mutational Burden (TMB)

TMB has been studied across various human cancers, and a high TMB has been considered to reflect a high load of tumor neoantigens and responses to the ICI treatment for cancers, which might be due to more recognition of tumor cells by T cells and enhanced antitumor immune responses [109]. However, its impact on HCC patients receiving ICIs has been controversial. A previous study showed that the frequency of patients with a high TMB was very low (0.8%, 6 of 755 patients with HCC), and was not related to the response rate [110]. In patients receiving camrelizumab and afatinib, TMB was associated with the ORR, although the sample size was small [111]. In a recent experimental study of AB treatment, TMB was not associated with PFS and ORR [17]. However, considering its impact on antitumor immune responses, larger and multicenter studies are needed to elucidate the role of TMB in HCC patients receiving the AB treatment. Importantly, the methods of sequencing and the cut-off value of TMBs should be standardized in future studies.

6.3. Gene Mutations

Alterations in genes related to the immune function or oncogenic pathways in tumor tissues have been studied in many types of cancers including HCC. However, there is a lack of data on HCC patients receiving AB treatment. The immune-exclusion class characterized by Wnt/CTNNB1 mutation has been known to be associated with resistance to ICI treatment in HCC [112]. In a previous study, a tumor biopsy occurred before AB treatment, and expression levels of glutamine synthetase and β-catenin, which are markers of Wnt/β-catenin signal activation, were measured, but there was no difference in PFS and ORR between activation and inactivation groups [113]. TP53 gene mutations, which are related to an immunosuppressive TME in HCC [114], have not been studied in patients receiving ICI or AB treatments for HCC.
Previous studies showed that telomerase reverse transcriptase (TERT) promoter mutations are associated with poor outcomes in various types of cancers, and are particularly related to the epithelial–mesenchymal transition [115] and PD-L1 expression [116]. This might be correlated with a higher TMB and better response to ICIs [115]. Furthermore, antigens from the TERT protein have been considered to be immunogenic and recognized by T cells, which is the main effector of ICI treatment [117]. Of note, a recent experimental study showed that a TERT promoter mutation is associated with the clinical benefit of AB treatment compared to sorafenib in terms of OS and PFS [97]. Further mechanistic and validation studies in AB-treated HCC patients should be performed.

6.4. Gut Microbiome

It has been reported that the diversity, composition, and function of the gut microbiome might be associated with immune responses. Zheng et al. investigated whether the gut microbiome has an impact on the responsiveness of anti-PD-1 treatment in patients with HCC [118]. As a result, responders had a higher richness of taxa and gene counts compared to non-responders, and diversity and stability were maintained after anti-PD-1 treatment whereas it is not maintained in non-responders. However, their impact on anti-VEGF treatment, as well as their role as a biomarker in the AB treatment of HCC patients, still needs to be clarified.

7. Discussion

The IMbrave150 trial represents a significant milestone in the systemic treatment for advanced HCC and has contributed to further investigations and approvals of ICI-based combination treatments. As confirmed in the meta-analysis of various types of cancers, including HCC, ICI treatment can increase the chance of a complete response compared to conventional treatments [119]. Moreover, the addition of locoregional treatments, including transarterial radioembolization to the ICI-regimen has also been tried to maximize the clinical benefit [120]. Nevertheless, a considerable number of patients do not respond to the treatment, about 70% [121], indicating the need to identify predictive biomarkers to guide initial and on-treatment decisions in HCC patients receiving AB.
Considering the mechanism of action for AB, it is crucial to develop biomarkers that accurately reflect the interplay between angiogenesis and immunosuppression within the TME. Of note, VEGF not only fosters angiogenesis in tumors but also establishes an immunosuppressive TME by attracting and inducing immunosuppressive cells, such as Tregs, TAMs, and MDSCs [11]. Moreover, VEGF hampers DC differentiation and maturation, as well as effector T cell proliferation, ultimately weakening T cell priming and targeting cell elimination [11]. In addition, VEGF drives the T cell exhaustion-specific program, which upregulates transcription factor TOX in the TME [122]. Thus, blocking VEGF signaling not only inhibits intra-tumoral angiogenesis and normalizes tumor vasculature but also transforms the TME from immunosuppressive to immune-active, which might enhance the efficacy of ICIs. These potential synergisms might improve AB efficacy in HCC compared to monotherapy-based ICIs such as nivolumab and pembrolizumab.
The analysis of biomarkers in HCC has been challenging in various treatment modalities, and no standard or definite predictive biomarkers have been identified for patients receiving AB treatment. The unique environment involving hepatitis and/or cirrhosis with different etiologies in HCC might contribute to this challenge. To predict treatment response more accurately, an integrative approach that combines clinical, histopathology, imaging, and circulating markers is necessary. The fact that this regimen contains not only ICI but also VEGF inhibitors, should be considered when developing biomarkers to optimize therapeutic strategies. Furthermore, baseline biomarkers can help identify patients who are likely to respond to treatment, whereas dynamic biomarkers, which are assessed during the therapy, can provide crucial information about treatment efficacy and the potential need for adjustments.
There are some limitations to the current review. Because many of the previous reports we have reviewed were small-sized, retrospective studies, there is a possibility of bias, misrepresentation, or inconsistency. Therefore, data should be interpreted cautiously, and most biomarkers need to be validated in larger, prospective future studies. In addition, systemic treatment and immunotherapy for HCC is a rapidly evolving field; reviews regarding its prognostic or predictive biomarkers can also quickly become outdated, necessitating regular updates.

8. Concluding Remarks

The increasing availability of real-world data from patients treated with AB is expected to facilitate the development of more accurate predictive biomarkers, ultimately improving the clinical outcome of first-line systemic treatment for HCC. In addition to the examination of peripheral blood, tumor tissues, and clinical information, liquid biopsy or microbiome analysis could also play a role in predicting responses to AB and should be investigated in future studies. Importantly, the development of biomarkers for the AB treatment of HCC should consider integrative and dynamic approaches. This comprehensive strategy may lead to better patient selection, more precise prediction of treatment response, and optimized therapeutic strategies, ultimately resulting in improved patient outcomes. Furthermore, validation tools other than patient cohorts, such as organoids or patient-derived xenograft models, could be used in the development of biomarkers in AB treatment and could also be applied to the agents for novel therapeutic targets of HCC.

Author Contributions

J.W.J. designed the study. J.W.H. and J.W.J. wrote the manuscript. J.W.H. generated the figures and tables. J.W.J. supervised the study. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program of the National Research Foundation of Korea (NRF) funded by the Ministry of Education (grant number 2022R1I1A1A01063636 to J.W.H.). This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT and Future Planning (NRF-2021R1I1A2056660 to J.W.J). This work was supported by the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT and Future Planning Korea Bio Grand Challenge Program (2018M3A9H3020844 to J.W.J.).

Institutional Review Board Statement

Ethical review and approval were waived, because this study was review article.

Informed Consent Statement

Informed consent was waived, because this study was review article.

Data Availability Statement

The data that support the findings of this review are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Llovet, J.M.; Castet, F.; Heikenwalder, M.; Maini, M.K.; Mazzaferro, V.; Pinato, D.J.; Pikarsky, E.; Zhu, A.X.; Finn, R.S. Immunotherapies for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 2022, 19, 151–172. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. De Lorenzo, S.; Tovoli, F.; Trevisani, F. Mechanisms of Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Patients with Hepatocellular Carcinoma. Cancers 2022, 14, 4616. [Google Scholar] [CrossRef] [PubMed]
  4. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef] [PubMed]
  5. Deng, R.; Bumbaca, D.; Pastuskovas, C.V.; Boswell, C.A.; West, D.; Cowan, K.J.; Chiu, H.; McBride, J.; Johnson, C.; Xin, Y.; et al. Preclinical pharmacokinetics, pharmacodynamics, tissue distribution, and tumor penetration of anti-PD-L1 monoclonal antibody, an immune checkpoint inhibitor. MAbs 2016, 8, 593–603. [Google Scholar] [CrossRef] [Green Version]
  6. Johnson, D.H.; Fehrenbacher, L.; Novotny, W.F.; Herbst, R.S.; Nemunaitis, J.J.; Jablons, D.M.; Langer, C.J.; DeVore, R.F., 3rd; Gaudreault, J.; Damico, L.A.; et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J. Clin. Oncol. 2004, 22, 2184–2191. [Google Scholar] [CrossRef]
  7. Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. PubChem 2023 update. Nucleic Acids Res. 2023, 51, D1373–D1380. [Google Scholar] [CrossRef]
  8. Han, J.W.; Yoon, S.K. Tissue-resident lymphocytes: Implications in immunotherapy for hepatocellular carcinoma. Int. J. Mol. Sci. 2020, 22, 232. [Google Scholar] [CrossRef]
  9. Liu, X.; Lu, Y.; Qin, S. Atezolizumab and bevacizumab for hepatocellular carcinoma: Mechanism, pharmacokinetics and future treatment strategies. Future Oncol. 2021, 17, 2243–2256. [Google Scholar] [CrossRef]
  10. Loosen, S.H.; Schulze-Hagen, M.; Leyh, C.; Benz, F.; Vucur, M.; Kuhl, C.; Trautwein, C.; Tacke, F.; Bruners, P.; Roderburg, C. IL-6 and IL-8 serum levels predict tumor response and overall survival after TACE for primary and secondary hepatic malignancies. Int. J. Mol. Sci. 2018, 19, 1766. [Google Scholar] [CrossRef] [Green Version]
  11. Lapeyre-Prost, A.; Terme, M.; Pernot, S.; Pointet, A.-L.; Voron, T.; Tartour, E.; Taieb, J. Immunomodulatory activity of VEGF in cancer. Int. Rev. Cell Mol. Biol. 2017, 330, 295–342. [Google Scholar]
  12. Pfister, D.; Nunez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 2021, 592, 450–456. [Google Scholar] [CrossRef]
  13. Beudeker, B.J.; Groothuismink, Z.M.; van der Eijk, A.A.; Debes, J.D.; Boonstra, A. Circulating cytokines reflect the etiology-specific immune environment in cirrhosis and HCC. Cancers 2022, 14, 4900. [Google Scholar] [CrossRef]
  14. Yau, T.; Park, J.-W.; Finn, R.S.; Cheng, A.-L.; Mathurin, P.; Edeline, J.; Kudo, M.; Harding, J.J.; Merle, P.; Rosmorduc, O. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022, 23, 77–90. [Google Scholar] [CrossRef]
  15. Kelley, R.K.; Rimassa, L.; Cheng, A.-L.; Kaseb, A.; Qin, S.; Zhu, A.X.; Chan, S.L.; Melkadze, T.; Sukeepaisarnjaroen, W.; Breder, V. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2022, 23, 995–1008. [Google Scholar] [CrossRef]
  16. Chan, L.; Kudo, M.; Sangro, B.; Kelley, R.; Furuse, J.; Park, J.; Sunpaweravong, P.; Fasolo, A.; Yau, T.; Kawaoka, T. 714P Impact of viral aetiology in the phase III HIMALAYA study of tremelimumab (T) plus durvalumab (D) in unresectable hepatocellular carcinoma (uHCC). Ann. Oncol. 2022, 33, S869–S870. [Google Scholar] [CrossRef]
  17. Cheng, A.-L.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Lim, H.Y.; Kudo, M.; Breder, V.; Merle, P. 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]
  18. Vogel, A.; Rimassa, L.; Sun, H.C.; Abou-Alfa, G.K.; El-Khoueiry, A.; Pinato, D.J.; Sanchez Alvarez, J.; Daigl, M.; Orfanos, P.; Leibfried, M.; et al. Comparative Efficacy of Atezolizumab plus Bevacizumab and Other Treatment Options for Patients with Unresectable Hepatocellular Carcinoma: A Network Meta-Analysis. Liver Cancer 2021, 10, 240–248. [Google Scholar] [CrossRef]
  19. Zhu, A.X.; Dayyani, F.; Yen, C.-J.; Ren, Z.; Bai, Y.; Meng, Z.; Pan, H.; Dillon, P.; Mhatre, S.K.; Gaillard, V.E. Alpha-fetoprotein as a potential surrogate biomarker for atezolizumab + bevacizumab treatment of hepatocellular carcinoma. Clin. Cancer Res. 2022, 28, 3537–3545. [Google Scholar] [CrossRef]
  20. Himmelsbach, V.; Pinter, M.; Scheiner, B.; Venerito, M.; Sinner, F.; Zimpel, C.; Marquardt, J.U.; Trojan, J.; Waidmann, O.; Finkelmeier, F. Efficacy and safety of atezolizumab and bevacizumab in the real-world treatment of advanced hepatocellular carcinoma: Experience from four tertiary centers. Cancers 2022, 14, 1722. [Google Scholar] [CrossRef]
  21. Takeda, S.; Namisaki, T.; Tsuji, Y.; Fujimoto, Y.; Murata, K.; Enomoto, M.; Fujinaga, Y.; Nishimura, N.; Kitagawa, K.; Takaya, H. Initial Experience with Atezolizumab Plus Bevacizumab for Unresectable Hepatocellular Carcinoma: A Real-world Retrospective Study. Anticancer Res. 2022, 42, 5465–5473. [Google Scholar] [CrossRef] [PubMed]
  22. Yu, J.W.; Bhattacharya, S.; Yanamandra, N.; Kilian, D.; Shi, H.; Yadavilli, S.; Katlinskaya, Y.; Kaczynski, H.; Conner, M.; Benson, W. Tumor-immune profiling of murine syngeneic tumor models as a framework to guide mechanistic studies and predict therapy response in distinct tumor microenvironments. PLoS ONE 2018, 13, e0206223. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, H.S.; Kim, C.G.; Hong, J.Y.; Kim, I.-h.; Kang, B.; Jung, S.; Kim, C.; Shin, S.J.; Choi, H.J.; Cheon, J. The presence and size of intrahepatic tumors determine the therapeutic efficacy of nivolumab in advanced hepatocellular carcinoma. Ther. Adv. Med. Oncol. 2022, 14, 17588359221113266. [Google Scholar] [CrossRef] [PubMed]
  24. Chon, Y.E.; Cheon, J.; Kim, H.; Kang, B.; Ha, Y.; Kim, D.Y.; Hwang, S.G.; Chon, H.J.; Kim, B.K. Predictive biomarkers of survival in patients with advanced hepatocellular carcinoma receiving atezolizumab plus bevacizumab treatment. Cancer Med. 2023, 12, 2731–2738. [Google Scholar] [CrossRef]
  25. Fulgenzi, C.A.M.; Cheon, J.; D’Alessio, A.; Nishida, N.; Ang, C.; Marron, T.U.; Wu, L.; Saeed, A.; Wietharn, B.; Cammarota, A. Reproducible safety and efficacy of atezolizumab plus bevacizumab for HCC in clinical practice: Results of the AB-real study. Eur. J. Cancer 2022, 175, 204–213. [Google Scholar] [CrossRef]
  26. Jost-Brinkmann, F.; Demir, M.; Wree, A.; Luedde, T.; Loosen, S.H.; Müller, T.; Tacke, F.; Roderburg, C.; Mohr, R. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma: Results from a German real-world cohort. Aliment. Pharmacol. Ther. 2023, 57, 1313–1325. [Google Scholar] [CrossRef]
  27. Tanaka, T.; Hiraoka, A.; Tada, T.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E.; Fukunishi, S. Therapeutic efficacy of atezolizumab plus bevacizumab treatment for unresectable hepatocellular carcinoma in patients with Child-Pugh class A or B liver function in real-world clinical practice. Hepatol. Res. 2022, 52, 773–783. [Google Scholar] [CrossRef]
  28. De Castro, T.; Jochheim, L.S.; Bathon, M.; Welland, S.; Scheiner, B.; Shmanko, K.; Roessler, D.; Ben Khaled, N.; Jeschke, M.; Ludwig, J.M. Atezolizumab and bevacizumab in patients with advanced hepatocellular carcinoma with impaired liver function and prior systemic therapy: A real-world experience. Ther. Adv. Med. Oncol. 2022, 14, 17588359221080298. [Google Scholar] [CrossRef]
  29. Sinner, F.; Pinter, M.; Scheiner, B.; Ettrich, T.J.; Sturm, N.; Gonzalez-Carmona, M.A.; Waidmann, O.; Finkelmeier, F.; Himmelsbach, V.; De Toni, E.N. Atezolizumab Plus Bevacizumab in Patients with Advanced and Progressing Hepatocellular Carcinoma: Retrospective Multicenter Experience. Cancers 2022, 14, 5966. [Google Scholar] [CrossRef]
  30. Tanaka, T.; Takata, K.; Yokoyama, K.; Fukuda, H.; Yamauchi, R.; Fukunaga, A.; Shakado, S.; Sakisaka, S.; Hirai, F. Pretreatment Modified Albumin–Bilirubin Grade Is an Important Predictive Factor Associated with the Therapeutic Response and the Continuation of Atezolizumab plus Bevacizumab Combination Therapy for Patients with Unresectable Hepatocellular Carcinoma. Curr. Oncol. 2022, 29, 4799–4810. [Google Scholar] [CrossRef]
  31. Unome, S.; Imai, K.; Takai, K.; Miwa, T.; Hanai, T.; Nishigaki, Y.; Hayashi, H.; Kochi, T.; Shimizu, S.; Nagano, J. Changes in ALBI Score and PIVKA-II within Three Months after Commencing Atezolizumab Plus Bevacizumab Treatment Affect Overall Survival in Patients with Unresectable Hepatocellular Carcinoma. Cancers 2022, 14, 6089. [Google Scholar] [CrossRef]
  32. Hatanaka, T.; Kakizaki, S.; Hiraoka, A.; Tada, T.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E. Development and validation of a modified albumin–bilirubin grade and α-fetoprotein score (mALF score) for hepatocellular carcinoma patients receiving atezolizumab and bevacizumab. Hepatol. Int. 2023, 17, 86–96. [Google Scholar] [CrossRef]
  33. Ueno, A.; Masugi, Y.; Yamazaki, K.; Komuta, M.; Effendi, K.; Tanami, Y.; Tsujikawa, H.; Tanimoto, A.; Okuda, S.; Itano, O. 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]
  34. Luke, J.J.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human CancersWNT/β-catenin–Associated Immune Exclusion across Cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [Google Scholar] [CrossRef] [Green Version]
  35. Harding, J.J.; Nandakumar, S.; Armenia, J.; Khalil, D.N.; Albano, M.; Ly, M.; Shia, J.; Hechtman, J.F.; Kundra, R.; El Dika, I. Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune TherapiesGenotyping of HCC in the Clinic. Clin. Cancer Res. 2019, 25, 2116–2126. [Google Scholar] [CrossRef] [Green Version]
  36. Sasaki, R.; Nagata, K.; Fukushima, M.; Haraguchi, M.; Miuma, S.; Miyaaki, H.; Soyama, A.; Hidaka, M.; Eguchi, S.; Shigeno, M. 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]
  37. Shigeta, K.; Datta, M.; Hato, T.; Kitahara, S.; Chen, I.X.; Matsui, A.; Kikuchi, H.; Mamessier, E.; Aoki, S.; Ramjiawan, R.R. Dual programmed death receptor-1 and vascular endothelial growth factor receptor-2 blockade promotes vascular normalization and enhances antitumor immune responses in hepatocellular carcinoma. Hepatology 2020, 71, 1247–1261. [Google Scholar] [CrossRef]
  38. Onuoha, E.; Smith, A.D.; Cannon, R.; Khushman, M.d.; Kim, H. Perfusion Change of Hepatocellular Carcinoma during Atezolizumab plus Bevacizumab Treatment: A Pilot Study. J. Gastrointest. Cancer, 2022; in press. [Google Scholar] [CrossRef]
  39. Takada, H.; Yamashita, K.; Osawa, L.; Komiyama, Y.; Nakakuki, N.; Muraoka, M.; Suzuki, Y.; Sato, M.; Takano, S.; Fukasawa, M. Prediction of Therapeutic Response Using Contrast-Enhanced Ultrasound in Japanese Patients Treated with Atezolizumab and Bevacizumab for Unresectable Hepatocellular Carcinoma. Oncology 2023, 101, 173–184. [Google Scholar] [CrossRef]
  40. Seo, S.; Hatano, E.; Higashi, T.; Hara, T.; Tada, M.; Tamaki, N.; Iwaisako, K.; Ikai, I.; Uemoto, S. Fluorine-18 fluorodeoxyglucose positron emission tomography predicts tumor differentiation, P-glycoprotein expression, and outcome after resection in hepatocellular carcinoma. Clin. Cancer Res. 2007, 13, 427–433. [Google Scholar] [CrossRef] [Green Version]
  41. Kawamura, Y.; Kobayashi, M.; Shindoh, J.; Matsumura, M.; Okubo, S.; Muraishi, N.; Fujiyama, S.; Hosaka, T.; Saitoh, S.; Sezaki, H. Pretreatment Positron Emission Tomography with 18F-Fluorodeoxyglucose May Be a Useful New Predictor of Early Progressive Disease following Atezolizumab plus Bevacizumab in Patients with Unresectable Hepatocellular Carcinoma. Oncology 2022, 100, 320–330. [Google Scholar] [CrossRef] [PubMed]
  42. Hatanaka, T.; Hiraoka, A.; Tada, T.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E.; Fukunishi, S. Association of early bevacizumab interruption with efficacy of atezolizumab plus bevacizumab for advanced hepatocellular carcinoma: A landmark analysis. Hepatol. Res. 2022, 52, 462–470. [Google Scholar] [CrossRef]
  43. Fukushima, T.; Morimoto, M.; Kobayashi, S.; Ueno, M.; Uojima, H.; Hidaka, H.; Kusano, C.; Chuma, M.; Numata, K.; Tsuruya, K. Association between Immune-Related Adverse Events and Survival in Patients with Hepatocellular Carcinoma Treated with Atezolizumab Plus Bevacizumab. Oncologist 2023, 28, oyad090. [Google Scholar] [CrossRef] [PubMed]
  44. Shimose, S.; Iwamoto, H.; Tanaka, M.; Niizeki, T.; Kajiwara, M.; Itano, S.; Moriyama, E.; Shirono, T.; Noda, Y.; Kamachi, N. Association between Adverse Events and Prognosis in Patients with Hepatocellular Carcinoma Treated with Atezolizumab Plus Bevacizumab: A Multicenter Retrospective Study. Cancers 2022, 14, 4284. [Google Scholar] [CrossRef] [PubMed]
  45. Yokohama, K.; Asai, A.; Matsui, M.; Okamoto, N.; Yasuoka, H.; Nishikawa, T.; Ohama, H.; Tsuchimoto, Y.; Inoue, Y.; Fukunishi, S. Liver dysfunction is associated with poor prognosis in patients after immune checkpoint inhibitor therapy. Sci. Rep. 2020, 10, 14470. [Google Scholar] [CrossRef]
  46. Yamamoto, T.; Ito, T.; Hase, T.; Ishigami, M.; Mizuno, K.; Yamamoto, K.; Imai, N.; Ishizu, Y.; Honda, T.; Shibata, H. Immune-related liver injury is a poor prognostic factor in patients with nonsmall cell lung cancer treated with immune checkpoint inhibitors. Cancer Investig. 2022, 40, 189–198. [Google Scholar] [CrossRef]
  47. Chan, S.L.; Yip, T.C.F.; Wong, V.W.S.; Tse, Y.K.; Yuen, B.W.Y.; Luk, H.W.S.; Lui, R.N.S.; Chan, H.L.Y.; Mok, T.S.K.; Wong, G.L.H. Pattern and impact of hepatic adverse events encountered during immune checkpoint inhibitors—A territory-wide cohort study. Cancer Med. 2020, 9, 7052–7061. [Google Scholar] [CrossRef]
  48. Takaki, S.; Kurosaki, M.; Mori, N.; Tsuji, K.; Ochi, H.; Marusawa, H.; Nakamura, S.; Tada, T.; Narita, R.; Uchida, Y. Effects on survival of the adverse event of atezolizumab plus bevacizumab for hepatocellular carcinoma: A multicenter study by the Japan Red Cross Liver Study Group. Investig. New Drugs 2023, 41, 340–349. [Google Scholar] [CrossRef]
  49. 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. 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]
  50. Tada, T.; Kumada, T.; Hiraoka, A.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E.; Fukunishi, S. Adverse events as potential predictive factors of therapeutic activity in patients with unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab. Cancer Med. 2022, 12, 7772–7783. [Google Scholar] [CrossRef]
  51. Syrigos, K.N.; Karapanagiotou, E.; Boura, P.; Manegold, C.; Harrington, K. Bevacizumab-induced hypertension: Pathogenesis and management. BioDrugs 2011, 25, 159–169. [Google Scholar] [CrossRef]
  52. Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef] [Green Version]
  53. Tamaki, N.; Tada, T.; Kurosaki, M.; Yasui, Y.; Ochi, H.; Mashiba, T.; Sakamoto, A.; Marusawa, H.; Narita, R.; Uchida, Y. Optimal threshold of alpha-fetoprotein response in patients with unresectable hepatocellular carcinoma treated with atezolizumab and bevacizumab. Investig. New Drugs 2022, 40, 1290–1297. [Google Scholar] [CrossRef]
  54. Kuzuya, T.; Kawabe, N.; Hashimoto, S.; Miyahara, R.; Sawaki, A.; Nakano, T.; Nakaoka, K.; Tanaka, H.; Miyachi, Y.; Mii, A. Early changes in alpha-fetoprotein are a useful predictor of efficacy of atezolizumab plus bevacizumab treatment in patients with advanced hepatocellular carcinoma. Oncology 2022, 100, 12–21. [Google Scholar] [CrossRef]
  55. Campani, C.; Bamba-Funck, J.; Campion, B.; Sidali, S.; Blaise, L.; Ganne-Carrié, N.; Demory, A.; Sutter, O.; Larrey, E.; Evain, M. Baseline ALBI score and early variation of serum AFP predicts outcomes in patients with HCC treated by atezolizumab–bevacizumab. Liver Int. 2023, 43, 708–717. [Google Scholar] [CrossRef]
  56. Sun, X.; Mei, J.; Lin, W.; Yang, Z.; Peng, W.; Chen, J.; Zhang, Y.; Xu, L.; Chen, M. Reductions in AFP and PIVKA-II can predict the efficiency of anti-PD-1 immunotherapy in HCC patients. BMC Cancer 2021, 21, 775. [Google Scholar] [CrossRef]
  57. Ochi, H.; Kurosaki, M.; Joko, K.; Mashiba, T.; Tamaki, N.; Tsuchiya, K.; Marusawa, H.; Tada, T.; Nakamura, S.; Narita, R. Usefulness of neutrophil-to-lymphocyte ratio in predicting progression and survival outcomes after atezolizumab–bevacizumab treatment for hepatocellular carcinoma. Hepatol. Res. 2023, 53, 61–71. [Google Scholar] [CrossRef]
  58. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  59. Iivanainen, S.; Ahvonen, J.; Knuuttila, A.; Tiainen, S.; Koivunen, J.P. Elevated CRP levels indicate poor progression-free and overall survival on cancer patients treated with PD-1 inhibitors. Esmo Open 2019, 4, e000531. [Google Scholar] [CrossRef] [Green Version]
  60. Scheiner, B.; Pomej, K.; Kirstein, M.M.; Hucke, F.; Finkelmeier, F.; Waidmann, O.; Himmelsbach, V.; Schulze, K.; von Felden, J.; Fründt, T.W. Prognosis of patients with hepatocellular carcinoma treated with immunotherapy–development and validation of the CRAFITY score. J. Hepatol. 2022, 76, 353–363. [Google Scholar] [CrossRef]
  61. Teng, W.; Lin, C.-C.; Su, C.-W.; Lin, P.-T.; Hsieh, Y.-C.; Chen, W.-T.; Ho, M.-M.; Wang, C.-T.; Chai, P.-M.; Hsieh, J.C.-H. Combination of CRAFITY score with Alpha-fetoprotein response predicts a favorable outcome of atezolizumab plus bevacizumab for unresectable hepatocellular carcinoma. Am. J. Cancer Res. 2022, 12, 1899. [Google Scholar] [CrossRef] [PubMed]
  62. Hatanaka, T.; Kakizaki, S.; Hiraoka, A.; Tada, T.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E. Prognostic impact of C-reactive protein and alpha-fetoprotein in immunotherapy score in hepatocellular carcinoma patients treated with atezolizumab plus bevacizumab: A multicenter retrospective study. Hepatol. Int. 2022, 16, 1150–1160. [Google Scholar] [CrossRef] [PubMed]
  63. Kaibori, M.; Hiraoka, A.; Matsui, K.; Matsushima, H.; Kosaka, H.; Yamamoto, H.; Yamaguchi, T.; Yoshida, K.; Sekimoto, M. Predicting complications following surgical resection of hepatocellular carcinoma using newly developed neo-Glasgow prognostic score with ALBI grade: Comparison of open and laparoscopic surgery cases. Cancers 2022, 14, 1402. [Google Scholar] [CrossRef] [PubMed]
  64. Tada, T.; Kumada, T.; Hiraoka, A.; Kariyama, K.; Tani, J.; Hirooka, M.; Takaguchi, K.; Atsukawa, M.; Fukunishi, S.; Itobayashi, E. New prognostic system based on inflammation and liver function predicts prognosis in patients with advanced unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab: A validation study. Cancer Med. 2022, 12, 6980–6993. [Google Scholar] [CrossRef] [PubMed]
  65. Ohki, S.; Shibata, M.; Gonda, K.; Machida, T.; Shimura, T.; Nakamura, I.; Ohtake, T.; Koyama, Y.; Suzuki, S.; Ohto, H. Circulating myeloid-derived suppressor cells are increased and correlate to immune suppression, inflammation and hypoproteinemia in patients with cancer. Oncol. Rep. 2012, 28, 453–458. [Google Scholar] [CrossRef] [Green Version]
  66. Sunakawa, Y.; Yang, D.; Cao, S.; Zhang, W.; Moran, M.; Astrow, S.H.; Hsiang, J.; Stephens, C.; Tsuji, A.; Takahashi, T. Immune-related genes to dominate neutrophil-lymphocyte ratio (NLR) associated with survival of cetuximab treatment in metastatic colorectal cancer. Clin. Color. Cancer 2018, 17, e741–e749. [Google Scholar] [CrossRef]
  67. Chen, Z.; Raghav, K.; Lieu, C.; Jiang, Z.; Eng, C.; Vauthey, J.; Chang, G.; Qiao, W.; Morris, J.; Hong, D. Cytokine profile and prognostic significance of high neutrophil-lymphocyte ratio in colorectal cancer. Br. J. Cancer 2015, 112, 1088–1097. [Google Scholar] [CrossRef]
  68. Pine, J.; Morris, E.; Hutchins, G.; West, N.; Jayne, D.; Quirke, P.; Prasad, K. Systemic neutrophil-to-lymphocyte ratio in colorectal cancer: The relationship to patient survival, tumour biology and local lymphocytic response to tumour. Br. J. Cancer 2015, 113, 204–211. [Google Scholar] [CrossRef] [Green Version]
  69. Sangro, B.; Melero, I.; Wadhawan, S.; Finn, R.S.; Abou-Alfa, G.K.; Cheng, A.-L.; Yau, T.; Furuse, J.; Park, J.-W.; Boyd, Z. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J. Hepatol. 2020, 73, 1460–1469. [Google Scholar] [CrossRef]
  70. Dharmapuri, S.; Özbek, U.; Lin, J.Y.; Sung, M.; Schwartz, M.; Branch, A.D.; Ang, C. Predictive value of neutrophil to lymphocyte ratio and platelet to lymphocyte ratio in advanced hepatocellular carcinoma patients treated with anti–PD-1 therapy. Cancer Med. 2020, 9, 4962–4970. [Google Scholar] [CrossRef]
  71. 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]
  72. Tada, T.; Kumada, T.; Hiraoka, A.; Hirooka, M.; Kariyama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; Itobayashi, E.; Fukunishi, S. 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]
  73. Maesaka, K.; Sakamori, R.; Yamada, R.; Tahata, Y.; Imai, Y.; Ohkawa, K.; Miyazaki, M.; Mita, E.; Ito, T.; Hagiwara, H. Hyperprogressive disease in patients with unresectable hepatocellular carcinoma receiving atezolizumab plus bevacizumab therapy. Hepatol. Res. 2022, 52, 298–307. [Google Scholar] [CrossRef]
  74. Wu, Y.L.; Fulgenzi, C.A.M.; D’Alessio, A.; Cheon, J.; Nishida, N.; Saeed, A.; Wietharn, B.; Cammarota, A.; Pressiani, T.; Personeni, N. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios as prognostic biomarkers in unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab. Cancers 2022, 14, 5834. [Google Scholar] [CrossRef]
  75. Matoya, S.; Suzuki, T.; Matsuura, K.; Suzuki, Y.; Okumura, F.; Nagura, Y.; Sobue, S.; Kuroyanagi, K.; Kusakabe, A.; Koguchi, H. The neutrophil-to-lymphocyte ratio at the start of the second course during atezolizumab plus bevacizumab therapy predicts therapeutic efficacy in patients with advanced hepatocellular carcinoma: A multicenter analysis. Hepatol. Res. 2023, 53, 511–521. [Google Scholar] [CrossRef]
  76. Wang, J.-H.; Chen, Y.-Y.; Kee, K.-M.; Wang, C.-C.; Tsai, M.-C.; Kuo, Y.-H.; Hung, C.-H.; Li, W.-F.; Lai, H.-L.; Chen, Y.-H. The prognostic value of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in patients with hepatocellular carcinoma receiving atezolizumab plus bevacizumab. Cancers 2022, 14, 343. [Google Scholar] [CrossRef]
  77. Pravisani, R.; Mocchegiani, F.; Isola, M.; Lorenzin, D.; Adani, G.L.; Cherchi, V.; De Martino, M.; Risaliti, A.; Lai, Q.; Vivarelli, M. Postoperative trends and prognostic values of inflammatory and nutritional biomarkers after liver transplantation for hepatocellular carcinoma. Cancers 2021, 13, 513. [Google Scholar] [CrossRef]
  78. Tada, T.; Kumada, T.; Hiraoka, A.; Kariyama, K.; Tani, J.; Hirooka, M.; Takaguchi, K.; Atsukawa, M.; Fukunishi, S.; Itobayashi, E. Nutritional status is associated with prognosis in patients with advanced unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab. Oncology 2023, 101, 270–282. [Google Scholar] [CrossRef]
  79. Shakiba, E.; Ramezani, M.; Sadeghi, M. Evaluation of serum interleukin-6 levels in hepatocellular carcinoma patients: A systematic review and meta-analysis. Clin. Exp. Hepatol. 2018, 4, 182–190. [Google Scholar] [CrossRef]
  80. Tsukamoto, H.; Fujieda, K.; Senju, S.; Ikeda, T.; Oshiumi, H.; Nishimura, Y. Immune-suppressive effects of interleukin-6 on T-cell-mediated anti-tumor immunity. Cancer Sci. 2018, 109, 523–530. [Google Scholar] [CrossRef] [Green Version]
  81. Yang, H.; Kang, B.; Ha, Y.; Lee, S.H.; Kim, I.; Kim, H.; Lee, W.S.; Kim, G.; Jung, S.; Rha, S.Y. High serum IL-6 correlates with reduced clinical benefit of atezolizumab and bevacizumab in unresectable hepatocellular carcinoma. JHEP Rep. 2023, 5, 100672. [Google Scholar] [CrossRef] [PubMed]
  82. Myojin, Y.; Kodama, T.; Sakamori, R.; Maesaka, K.; Matsumae, T.; Sawai, Y.; Imai, Y.; Ohkawa, K.; Miyazaki, M.; Tanaka, S. Interleukin-6 is a circulating prognostic biomarker for hepatocellular carcinoma patients treated with combined immunotherapy. Cancers 2022, 14, 883. [Google Scholar] [CrossRef] [PubMed]
  83. Giovannini, C.; Suzzi, F.; Tovoli, F.; Bruccoleri, M.; Marseglia, M.; Alimenti, E.; Fornari, F.; Iavarone, M.; Piscaglia, F.; Gramantieri, L. Low-Baseline PD1+ Granulocytes Predict Responses to Atezolizumab–Bevacizumab in Hepatocellular Carcinoma. Cancers 2023, 15, 1661. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, Z.; Suda, G.; Maehara, O.; Ohara, M.; Yoda, T.; Sasaki, T.; Kohya, R.; Yoshida, S.; Hosoda, S.; Tokuchi, Y. Changes in Serum Growth Factors during Resistance to Atezolizumab Plus Bevacizumab Treatment in Patients with Unresectable Hepatocellular Carcinoma. Cancers 2023, 15, 593. [Google Scholar] [CrossRef] [PubMed]
  85. Lin, S.; Zhang, Q.; Shao, X.; Zhang, T.; Xue, C.; Shi, S.; Zhao, D.; Lin, Y. IGF-1 promotes angiogenesis in endothelial cells/adipose-derived stem cells co-culture system with activation of PI 3K/Akt signal pathway. Cell Prolif. 2017, 50, e12390. [Google Scholar] [CrossRef] [Green Version]
  86. Kaseb, A.O.; Guan, Y.; Gok Yavuz, B.; Abbas, A.R.; Lu, S.; Hasanov, E.; Toh, H.C.; Verret, W.; Wang, Y. Serum IGF-1 Scores and Clinical Outcomes in the Phase III IMbrave150 Study of Atezolizumab Plus Bevacizumab versus Sorafenib in Patients with Unresectable Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2022, 9, 1065–1079. [Google Scholar] [CrossRef]
  87. Brunet-Dunand, S.E.; Vouyovitch, C.; Araneda, S.; Pandey, V.; Vidal, L.J.-P.; Print, C.; Mertani, H.C.; Lobie, P.E.; Perry, J.K. Autocrine human growth hormone promotes tumor angiogenesis in mammary carcinoma. Endocrinology 2009, 150, 1341–1352. [Google Scholar] [CrossRef] [Green Version]
  88. Mohamed, Y.I.; Duda, D.G.; Awiwi, M.O.; Lee, S.S.; Altameemi, L.; Xiao, L.; Morris, J.S.; Wolff, R.A.; Elsayes, K.M.; Hatia, R.I. Plasma growth hormone is a potential biomarker of response to atezolizumab and bevacizumab in advanced hepatocellular carcinoma patients. Oncotarget 2022, 13, 1314–1321. [Google Scholar] [CrossRef]
  89. Kim, C.; Yang, H.; Kim, I.; Kang, B.; Kim, H.; Kim, H.; Lee, W.S.; Jung, S.; Lim, H.Y.; Cheon, J. 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]
  90. Matsumae, T.; Kodama, T.; Myojin, Y.; Maesaka, K.; Sakamori, R.; Takuwa, A.; Oku, K.; Motooka, D.; Sawai, Y.; Oshita, M. Circulating cell-free DNA profiling predicts the therapeutic outcome in advanced hepatocellular carcinoma patients treated with combination immunotherapy. Cancers 2022, 14, 3367. [Google Scholar] [CrossRef]
  91. Hosoda, S.; Suda, G.; Sho, T.; Ogawa, K.; Kimura, M.; Yang, Z.; Yoshida, S.; Kubo, A.; Tokuchi, Y.; Kitagataya, T. Low baseline CXCL9 predicts early progressive disease in unresectable HCC with atezolizumab plus bevacizumab treatment. Liver Cancer 2022, 12, 156–170. [Google Scholar] [CrossRef]
  92. Yamazaki, T.; Akiba, H.; Iwai, H.; Matsuda, H.; Aoki, M.; Tanno, Y.; Shin, T.; Tsuchiya, H.; Pardoll, D.M.; Okumura, K. Expression of programmed death 1 ligands by murine T cells and APC. J. Immunol. 2002, 169, 5538–5545. [Google Scholar] [CrossRef] [Green Version]
  93. Calderaro, J.; Rousseau, B.; Amaddeo, G.; Mercey, M.; Charpy, C.; Costentin, C.; Luciani, A.; Zafrani, E.S.; Laurent, A.; Azoulay, D. Programmed death ligand 1 expression in hepatocellular carcinoma: Relationship with clinical and pathological features. Hepatology 2016, 64, 2038–2046. [Google Scholar] [CrossRef]
  94. Gao, Q.; Wang, X.-Y.; Qiu, S.-J.; Yamato, I.; Sho, M.; Nakajima, Y.; Zhou, J.; Li, B.-Z.; Shi, Y.-H.; Xiao, Y.-S. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin. Cancer Res. 2009, 15, 971–979. [Google Scholar] [CrossRef] [Green Version]
  95. Yau, T.; Kang, Y.-K.; Kim, T.-Y.; El-Khoueiry, A.B.; Santoro, A.; Sangro, B.; Melero, I.; Kudo, M.; Hou, M.-M.; Matilla, A. Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: The CheckMate 040 randomized clinical trial. JAMA Oncol. 2020, 6, e204564. [Google Scholar] [CrossRef]
  96. Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A. 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]
  97. 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. 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]
  98. Duffy, A.G.; Ulahannan, S.V.; Makorova-Rusher, O.; Rahma, O.; Wedemeyer, H.; Pratt, D.; Davis, J.L.; Hughes, M.S.; Heller, T.; ElGindi, M. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J. Hepatol. 2017, 66, 545–551. [Google Scholar] [CrossRef] [Green Version]
  99. Wu, B.; Sternheim, N.; Agarwal, P.; Suchomel, J.; Vadhavkar, S.; Bruno, R.; Ballinger, M.; Bernaards, C.A.; Chan, P.; Ruppel, J. Evaluation of atezolizumab immunogenicity: Clinical pharmacology (part 1). Clin. Transl. Sci. 2022, 15, 130–140. [Google Scholar] [CrossRef]
  100. Ogawa, K.; Kanzaki, H.; Chiba, T.; Ao, J.; Qiang, N.; Ma, Y.; Zhang, J.; Yumita, S.; Ishino, T.; Unozawa, H. Effect of Atezolizumab plus Bevacizumab in Patients with Hepatocellular Carcinoma Harboring CTNNB1 Mutation in Early Clinical Experience. J. Cancer 2022, 13, 2656. [Google Scholar] [CrossRef]
  101. Litchfield, K.; Reading, J.L.; Puttick, C.; Thakkar, K.; Abbosh, C.; Bentham, R.; Watkins, T.B.; Rosenthal, R.; Biswas, D.; Rowan, A. Meta-analysis of tumor-and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 2021, 184, 596–614.e514. [Google Scholar] [CrossRef] [PubMed]
  102. Gorbachev, A.V.; Kobayashi, H.; Kudo, D.; Tannenbaum, C.S.; Finke, J.H.; Shu, S.; Farber, J.M.; Fairchild, R.L. CXC chemokine ligand 9/monokine induced by IFN-γ production by tumor cells is critical for T cell-mediated suppression of cutaneous tumors. J. Immunol. 2007, 178, 2278–2286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Itoh, S.; Shirabe, K.; Matsumoto, Y.; Yoshiya, S.; Muto, J.; Harimoto, N.; Yamashita, Y.-i.; Ikegami, T.; Yoshizumi, T.; Nishie, A. Effect of body composition on outcomes after hepatic resection for hepatocellular carcinoma. Ann. Surg. Oncol. 2014, 21, 3063–3068. [Google Scholar] [CrossRef] [PubMed]
  104. Takada, H.; Kurosaki, M.; Nakanishi, H.; Takahashi, Y.; Itakura, J.; Tsuchiya, K.; Yasui, Y.; Tamaki, N.; Takaura, K.; Komiyama, Y. Impact of pre-sarcopenia in sorafenib treatment for advanced hepatocellular carcinoma. PLoS ONE 2018, 13, e0198812. [Google Scholar] [CrossRef] [PubMed]
  105. Uojima, H.; Chuma, M.; Tanaka, Y.; Hidaka, H.; Nakazawa, T.; Iwabuchi, S.; Kobayashi, S.; Hattori, N.; Ogushi, K.; Morimoto, M. Skeletal muscle mass influences tolerability and prognosis in hepatocellular carcinoma patients treated with lenvatinib. Liver Cancer 2020, 9, 193–206. [Google Scholar] [CrossRef]
  106. Toshida, K.; Itoh, S.; Tomiyama, T.; Morinaga, A.; Kosai, Y.; Tomino, T.; Kurihara, T.; Nagao, Y.; Morita, K.; Harada, N. Comparison of the prognostic effect of sarcopenia on atezolizumab plus bevacizumab and lenvatinib therapy in hepatocellular carcinoma patients. JGH Open 2022, 6, 477–486. [Google Scholar] [CrossRef]
  107. Akce, M.; Liu, Y.; Zakka, K.; Martini, D.J.; Draper, A.; Alese, O.B.; Shaib, W.L.; Wu, C.; Wedd, J.P.; Sellers, M.T. Impact of sarcopenia, BMI, and inflammatory biomarkers on survival in advanced hepatocellular carcinoma treated with anti-PD-1 antibody. Am. J. Clin. Oncol. 2021, 44, 74–81. [Google Scholar] [CrossRef]
  108. Vithayathil, M.; D’Alessio, A.; Fulgenzi, C.A.M.; Nishida, N.; Schönlein, M.; von Felden, J.; Schulze, K.; Wege, H.; Saeed, A.; Wietharn, B. Impact of body mass index in patients receiving atezolizumab plus bevacizumab for hepatocellular carcinoma. Hepatol. Int. 2023; in press. [Google Scholar] [CrossRef]
  109. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancerstmb predicts response to immunotherapy in diverse cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [Green Version]
  110. Liu, L.; Bai, X.; Wang, J.; Tang, X.-R.; Wu, D.-H.; Du, S.-S.; Du, X.-J.; Zhang, Y.-W.; Zhu, H.-B.; Fang, Y. Combination of TMB and CNA Stratifies Prognostic and Predictive Responses to Immunotherapy Across Metastatic CancerPrognostic and Predictive of the Combination of TMB and CNA. Clin. Cancer Res. 2019, 25, 7413–7423. [Google Scholar] [CrossRef] [Green Version]
  111. Xu, J.; Zhang, Y.; Jia, R.; Yue, C.; Chang, L.; Liu, R.; Zhang, G.; Zhao, C.; Zhang, Y.; Chen, C. Anti-PD-1 antibody SHR-1210 combined with apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: An open-label, dose escalation and expansion study. Clin. Cancer Res. 2019, 25, 515–523. [Google Scholar] [CrossRef] [Green Version]
  112. Pinyol, R.; Sia, D.; Llovet, J.M. Immune Exclusion-Wnt/CTNNB1 Class Predicts Resistance to Immunotherapies in HCCCTNNB1-Mutated HCC Tumors Are Resistant to Immunotherapy. Clin. Cancer Res. 2019, 25, 2021–2023. [Google Scholar] [CrossRef] [Green Version]
  113. 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]
  114. Guo, G.; Yu, M.; Xiao, W.; Celis, E.; Cui, Y. Local Activation of p53 in the Tumor Microenvironment Overcomes Immune Suppression and Enhances Antitumor ImmunityLocal p53 Activation in the TME Enhances Antitumor Immunity. Cancer Res. 2017, 77, 2292–2305. [Google Scholar] [CrossRef] [Green Version]
  115. Stern, J.L.; Hibshman, G.; Hu, K.; Ferrara, S.E.; Costello, J.C.; Kim, W.; Tamayo, P.; Cech, T.R.; Huang, F.W. Mesenchymal and MAPK Expression Signatures Associate with Telomerase Promoter Mutations in Multiple CancersMesenchymal/MAPK Signatures of TERT Promoter Mutants. Mol. Cancer Res. 2020, 18, 1050–1062. [Google Scholar] [CrossRef] [Green Version]
  116. Mak, M.P.; Tong, P.; Diao, L.; Cardnell, R.J.; Gibbons, D.L.; William, W.N.; Skoulidis, F.; Parra, E.R.; Rodriguez-Canales, J.; Wistuba, I.I. A Patient-Derived, Pan-Cancer EMT Signature Identifies Global Molecular Alterations and Immune Target Enrichment Following Epithelial-to-Mesenchymal TransitionPan-Cancer EMT Molecular and Immune Alterations. Clin. Cancer Res. 2016, 22, 609–620. [Google Scholar] [CrossRef] [Green Version]
  117. Zanetti, M. A second chance for telomerase reverse transcriptase in anticancer immunotherapy. Nat. Rev. Clin. Oncol. 2017, 14, 115–128. [Google Scholar] [CrossRef]
  118. Zheng, Y.; Wang, T.; Tu, X.; Huang, Y.; Zhang, H.; Tan, D.; Jiang, W.; Cai, S.; Zhao, P.; Song, R. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. Immunother. Cancer 2019, 7, 193. [Google Scholar] [CrossRef] [Green Version]
  119. Santoni, M.; Rizzo, A.; Kucharz, J.; Mollica, V.; Rosellini, M.; Marchetti, A.; Tassinari, E.; Monteiro, F.S.M.; Soares, A.; Molina-Cerrillo, J.; et al. Complete remissions following immunotherapy or immuno-oncology combinations in cancer patients: The MOUSEION-03 meta-analysis. Cancer Immunol. Immunother. 2023, 72, 1365–1379. [Google Scholar] [CrossRef]
  120. Di Federico, A.; Rizzo, A.; Carloni, R.; De Giglio, A.; Bruno, R.; Ricci, D.; Brandi, G. Atezolizumab-bevacizumab plus Y-90 TARE for the treatment of hepatocellular carcinoma: Preclinical rationale and ongoing clinical trials. Expert Opin. Investig. Drugs 2022, 31, 361–369. [Google Scholar] [CrossRef]
  121. Rizzo, A.; Cusmai, A.; Gadaleta-Caldarola, G.; Palmiotti, G. Which role for predictors of response to immune checkpoint inhibitors in hepatocellular carcinoma? Expert Rev. Gastroenterol. Hepatol. 2022, 16, 333–339. [Google Scholar] [CrossRef] [PubMed]
  122. Kim, C.G.; Jang, M.; Kim, Y.; Leem, G.; Kim, K.H.; Lee, H.; Kim, T.-S.; Choi, S.J.; Kim, H.-D.; Han, J.W. VEGF-A drives TOX-dependent T cell exhaustion in anti–PD-1–resistant microsatellite stable colorectal cancers. Sci. Immunol. 2019, 4, eaay0555. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Predicting clinical outcomes in patients with HCC receiving Atezolizumab and Bevacizumab combination treatment. Atezolizumab targets PD-L1 resulting in the inhibition of PD-1 mediated T cell exhaustion pathway and T cell restoration. Bevacizumab targets VEGF and inhibits aberrant angiogenesis of tumor, as well as improving immunosuppressive TME via affecting Tregs, MDSCs, and TAMs. Predicting clinical outcomes of AB treatment can be performed by analyzing clinical factors such as medical records, laboratory tests, imaging tests, and the presence of AEs. They are also able to be performed by blood examinations, which include tumor markers, inflammatory markers, cytokines, and angiogenic factors. Liquid biopsy is also under investigation. Tissue studies include PD-L1 staining, whole genome or RNA sequencing, and staining for immune cell infiltration. Gut microbiome, CTCs, analysis for PBMCs, and various cytokines and chemokines should also be studied.
Figure 1. Predicting clinical outcomes in patients with HCC receiving Atezolizumab and Bevacizumab combination treatment. Atezolizumab targets PD-L1 resulting in the inhibition of PD-1 mediated T cell exhaustion pathway and T cell restoration. Bevacizumab targets VEGF and inhibits aberrant angiogenesis of tumor, as well as improving immunosuppressive TME via affecting Tregs, MDSCs, and TAMs. Predicting clinical outcomes of AB treatment can be performed by analyzing clinical factors such as medical records, laboratory tests, imaging tests, and the presence of AEs. They are also able to be performed by blood examinations, which include tumor markers, inflammatory markers, cytokines, and angiogenic factors. Liquid biopsy is also under investigation. Tissue studies include PD-L1 staining, whole genome or RNA sequencing, and staining for immune cell infiltration. Gut microbiome, CTCs, analysis for PBMCs, and various cytokines and chemokines should also be studied.
Ijms 24 11799 g001
Table 3. Previous studies investigating the association between tissue-based markers and outcome.
Table 3. Previous studies investigating the association between tissue-based markers and outcome.
Tissue MarkersRelated OutcomesStudy Design (n)Reference
PD-L1 expression CPS ≥ 1% (IHC, clone SP263)Favorable ORR, PFS of AB compared to SORPhase III RCT- IMbrave150 (336)Cheng et al. [17]
Signature associated with the PD-L1 expression (RNA sequencing)Favorable ORR, PFSPhase Ib and phase III IMbrave150 (358)Zhu et al. [97]
TERT promoter mutation (whole exome sequencing)Favorable ORR, PFS of AB compared to SORPhase Ib and phase III IMbrave150 (358)Zhu et al. [97]
High atezolizumab–bevacizumab response signature (ABRS)
-PD-L1 and effector T cell signatures (RNA sequencing)
Favorable ORR, PFSPhase Ib and phase III IMbrave150 (358)Zhu et al. [97]
High infiltration of CD3/8+ T cells, Granzyme+ T cells, and MHC-I+ tumor cells (IHC)Favorable ORRPhase Ib and phase III IMbrave150 (358)Zhu et al. [97]
High expression of CD4/8+ T cells, Treg cells, B cells, and DCs (RNA sequencing)Favorable ORR, PFSPhase Ib and phase III IMbrave150 (358)Zhu et al. [97]
PD-L1, programmed cell death-ligand 1; CPS, combined positive score; IHC, immunohistochemistry; ORR, objective response rate; PFS, progression-free survival; AB, atezolizumab + bevacizumab; SOR, sorafenib; TERT, telomerase reverse transcriptase; MHC, major histocompatibility complex; Treg, regulatory T; and DC, dendritic cell.
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

Han, J.W.; Jang, J.W. Predicting Outcomes of Atezolizumab and Bevacizumab Treatment in Patients with Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 11799. https://doi.org/10.3390/ijms241411799

AMA Style

Han JW, Jang JW. Predicting Outcomes of Atezolizumab and Bevacizumab Treatment in Patients with Hepatocellular Carcinoma. International Journal of Molecular Sciences. 2023; 24(14):11799. https://doi.org/10.3390/ijms241411799

Chicago/Turabian Style

Han, Ji Won, and Jeong Won Jang. 2023. "Predicting Outcomes of Atezolizumab and Bevacizumab Treatment in Patients with Hepatocellular Carcinoma" International Journal of Molecular Sciences 24, no. 14: 11799. https://doi.org/10.3390/ijms241411799

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