Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us
OPEN ACCESS

Treatment Options for Hepatocellular Carcinoma Using Immunotherapy: Present and Future

  • Hongbin Wei1,2,
  • Chunlu Dong1,2 and
  • Xun Li1,2,3,4,5,6,* 
Journal of Clinical and Translational Hepatology   2024;12(4):389-405

doi: 10.14218/JCTH.2023.00462

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Wei H, Dong C, Li X. Treatment Options for Hepatocellular Carcinoma Using Immunotherapy: Present and Future. J Clin Transl Hepatol. 2024;12(4):389-405. doi: 10.14218/JCTH.2023.00462.

Abstract

Hepatocellular carcinoma (HCC) is a common cancer, and the body’s immune responses greatly affect its progression and the prognosis of patients. Immunological suppression and the maintenance of self-tolerance in the tumor microenvironment are essential responses, and these form part of the theoretical foundations of immunotherapy. In this review, we first discuss the tumor microenvironment of HCC, describe immunosuppression in HCC, and review the major biomarkers used to track HCC progression and response to treatment. We then examine antibody-based therapies, with a focus on immune checkpoint inhibitors (ICIs), monoclonal antibodies that target key proteins in the immune response (programmed cell death protein 1, anti-cytotoxic T-lymphocyte associated protein 4, and programmed death-ligand 1) which have transformed the treatment of HCC and other cancers. ICIs may be used alone or in conjunction with various targeted therapies for patients with advanced HCC who are receiving first-line treatments or subsequent treatments. We also discuss the use of different cellular immunotherapies, including T cell receptor (TCR) T cell therapy and chimeric antigen receptor (CAR) T cell therapy. We then review the use of HCC vaccines, adjuvant immunotherapy, and oncolytic virotherapy, and describe the goals of future research in the development of treatments for HCC.

Keywords

Hepatocellular carcinoma, Liver cancer, Immunotherapy, Vaccines

Introduction

Liver cancer is among the most common cancers worldwide, and is becoming increasingly prevalent in Western nations.1,2 Hepatocellular carcinoma (HCC) is the main type of liver cancer, and it usually occurs due to persistent liver damage from infection by the hepatitis B virus (HBV) or hepatitis C virus (HCV), alcoholism, or metabolic syndrome.3 The most common serum biomarker for HCC is α-fetoprotein (AFP). Tumor load, location, and comorbidities influence the choice of treatment, which may consist of transplantation, resection with percutaneous ablation, trans-arterial chemoembolization (TACE), and radio-embolization.4

Effective anticancer immune surveillance occurs due to the interplay between the adaptive and innate immune responses. Immune evasion occurs when there are dysfunctional interactions between the body’s defense system and a tumor. More specifically, the immune system may have weakened detection of tumor-associated antigens (TAAs) or there may be an immune-suppressive tumor microenvironment (TME).5,6 Alterations in peptide or antigen processing, post-transcriptional inactivation, epigenetics, and other changes can impair the identification of TAAs by different components of the immune system.7 Deficient immune regulation by regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), inhibitory B cells, and M2-polarized tumor-associated macrophages (TAMs) can stimulate cancer progression. Increased regulation of co-inhibitory lymphocyte signals and increased levels of tolerogenic enzymes also contribute to cancer development and progression.8 Targeting the body’s defense system by immunotherapy is therefore a potentially effective general strategy for the treatment of many malignancies.

Liver tumors use specific ligands and receptors to enable communication of tumor cells with stromal cells, and to bypass anti-tumor immune responses.9 Effector lymphocytes generate several co-inhibitory compounds at immune checkpoints, such as lymphocyte-activation gene 3 (LAG3), cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), PD-1, T cell immunoglobulin, and mucin domain containing-3 (TIM3), and this prevents over-activation of the immune response.10 Activated T cells and Tregs express high levels of CTLA-4, and this down-regulates the immune response and blocks the activation of effector T cells.11 Activated T cells, Tregs, monocytes, natural killer (NK) cells, dendritic cells (DCs), and MDSCs all express programmed cell death protein-1 (PD-1), and a variety of stromal and cancer cells express programmed cell death-ligand 1 (PD-L1). PD-1 decreases the activity of effector T cells, leading to suppression of effector functions. Many studies that used specific monoclonal antibodies (mAbs) as immune checkpoint inhibitors (ICIs) demonstrated the benefits of an efficient immune response that eliminates different types of cancerous cells. ICIs can prevent the deactivation of T cells by blocking the binding of checkpoint proteins with their ligands.12 and are the first immunotherapy drugs with demonstrated efficacy against HCC.

Tumor immunotherapy has become a promising strategy for preventing the spread, recurrence, and development of many different tumors.13 Immunotherapy agents trigger tumor-specific immune responses and prevent immunological tolerance. Cancer immunotherapy has the potential to provide systemic and long-term anti-tumor activities, making it an appealing treatment option for metachronous and multicentric HCC. The US Food and Drug Administration (FDA) has licensed eight different ICIs that target PD-1, CTLA-4, or PD-L1 for the treatment of a variety of malignancies, including HCC.14,15 Other immunotherapeutic methods (such as the administration of immune cells with chimeric antigen receptors, cancer vaccines, adoptive cell therapy, and specially formulated cytokines) are currently under development and offer fresh optimism for the treatment of patients with HCC.16,17

Immune microenvironment of HCC

Antigenicity

Antigen expression is the first event in the establishment of a T cell response to a tumor. Uncontrolled expression of cancer testis antigens and oncofetal antigens during hepatocarcinogenesis can trigger a spontaneous immune response.18 Blood and tumor samples from HCC patients exhibit tumor-specific T-cell reactions, such as the production of CD8+ T lymphocytes, and these can target AFP, melanoma-associated gene 1 (MAGE-A1), glypican 3 (GPC3), and New York esophageal squamous cell carcinoma 1 (NY-ESO1). Genetic changes during hepatocarcinogenesis may lead to amino acid modifications of proteins and the formation of cancer neoantigens.19 These amino acid modifications may improve the peptide’s ability to bind to human leukocyte antigens (HLA), attract T cells toward the novel epitope, or create novel structures with T cell receptors (TCRs), and this allows T cells to recognize the novel epitope without being blocked by immunological tolerance.20 Many neoantigens derive from mutations of tumor suppressor proteins, such as tumor protein p53 (TP53), and are present in a variety of tumors. However, most of these novel epitopes are private neoepitopes (on a single HLA) caused by somatic mutations, and are considered to be passenger mutations.21,22

Next-generation sequencing (NGS) technology has allowed researchers to identify the mutational landscapes of numerous tumors.23 The tumor mutational burden (TMB, somatic mutations per Mb) is widely employed as a proxy for neoantigens due to its correlation with the number of T cells specific for neoantigens.24 The TMB varies widely among different types of tumors, and is low in pancreatic tumors (<1 mut/Mb) but high in many other tumors (>20 mut/Mb).25 HCC often has a low-to-moderate TMB compared to other tumors, with a mean TMB of about 5 mut/Mb, corresponding to about 60 non-synonymous changes.26 Theoretically, a tumor is more likely to become antigenic in the presence of a high TMB. Neoantigens are common in HCC, but their pathological significance is uncertain.

Immunological profile

The liver performs a variety of functions, such as blood transportation via the hepatic artery and portal vein, filtration of intestinal infectious agents, and excretion of harmful substances, and is therefore exposed to numerous foreign antigens.27,28 The hepatic reticulo-endothelial system is comprised of sinusoids, liver sinusoidal endothelial cells (LSECs), and Kupffer cells, which present antigens to innate T-cells. This system induces an immunological reaction that is tolerogenic in healthy individuals.29 Liver inflammation due to infection by HBV or HCV leads to the recruitment of cytokines and other immune molecules that can promote cancer proliferation.30 Innate and adaptive immune cells in the TME and TAMs stimulate PD-1 and CTLA-4.31 CTLA-4 blocks T-cell activation by competing with CD28 in its binding to CD80/86 on antigen-presenting cells (APC); PD-1 regulates T-cell collapse and hinders T-cell stimulation.32 The different components of the TME function in several complex processes, such as decreasing the detection of TAAs, interacting with immunological checkpoints, and forming immune suppressive cells that provide a balanced and immunotolerant status.33 The immunotolerance of HCC is also associated with the production of numerous cytokines and other regulatory molecules, including the immunosuppressive transforming growth factor beta (TGF-β).34

Immune cell microenvironment

The liver also down-regulates immune system activity, and this promotes tolerance to foreign antigens that are benign, such as those in the diet.35,36 The maintenance of a tolerogenic environment in humans requires interactions of non-parenchymal liver cells, such as Kupffer cells, LSECs, and hepatic stellate cells (HSCs). Kupffer cells can function as APCs in conjunction with LSECs and HSCs.37 Kupffer cells also produce inhibitory molecules, including indoleamine 2,3-dioxygenase (IDO), prostaglandins, and interleukin 10 (IL-10),38 and increase the activation of Tregs.39 PD-L1 has high expression in LSECs, and is responsible for the TGF-β-mediated initiation of Tregs. HSCs secrete hepatocyte growth factor (HGF), and this leads to the accumulation of MDSCs and Tregs in the liver, followed by PD-L1-mediated T cell death.40–42

The TME of HCC contains a mixture of cancer cells, cancer-associated fibroblasts, and non-parenchymal hepatic cells. Activation of the TME and the presence of defective tumor-infiltrating lymphocytes (TILs) are manifestations of a muted adaptive immune response to HCC.18 Suppression of the innate immune response, expression of inhibitory receptors.43,44 MDSC-mediated immune suppression,45 and an increase in the number of defective NK cells contribute to the activation of the TME.46 TGF-β controls immune cells in the liver, and promotes a balance between immune adaptability and stimulation under normal conditions.47 However, the production of excessive TGF-β within the TME can disrupt this balance and promote cancer growth, because of its pathological effects on a range of important cell types that control innate and adaptive immunity (Fig. 1).

Schematic illustration of the various approaches targeting TGF-β pathways for Hepatocellular carcinoma (HCC).
Fig. 1  Schematic illustration of the various approaches targeting TGF-β pathways for Hepatocellular carcinoma (HCC).

The effect of TGF-β controls both immune evasion and antitumor immunity. TGF-β, transforming growth factor beta.

Mechanism of immunosuppression in HCC

Co-stimulatory molecules (CMs) must be produced on T cells and APCs for TCRs to bind the major histocompatibility (MHC) peptides on APCs. The downregulation of MHC class I on cancer cells hampers antigen conversion.48,49 Additionally, the decreased development of CMs, such as B7-1 and B7-2, in HCC can cause T cell anergy.50 In healthy humans, immune checkpoints defend against unchecked autologous immunity by blocking excessive T-cell activation. However, tumor cells can overexpress immune checkpoint compounds that bind to TCRs and prevent T cell stimulation. Thus, activation of immune checkpoint pathways in HCC compromises the effector function of cellular immune reactions.51,52

The impaired CD4+ T cells in HCC patients can also suppress the body’s defense system.53 In the absence of appropriate CMs, expansion of MHC class II leads to inactive CD4+ T cells.54 Additionally, immunosuppressive components, such as Tregs.55 MDSCs, and regulatory DCs,56 have significant immunosuppressive effects in cancer patients. An increase in the number of immunosuppressive cells (such as Tregs), may promote cancer progression and lead to a poor prognosis. For example, previous research showed that HCC patients with venous metastases experienced changes in the Th1/Th2 balance in the hepatic microenvironment.57 More specifically, abnormal immune responses in the TME are an important indicator of HCC metastasis, and these manifest as the up-regulation of Th2-like cytokines (which are immunosuppressive and anti-inflammatory) and the down-regulation of Th1-like cytokines (which are pro-inflammatory and immunogenic) in adjacent non-cancerous hepatic tissues.58,59Figure 2 summarizes the bidirectional interactions between HCC tumor cells and the immunosuppressive component of the TME.

The bidirectional interactions between Hepatocellular carcinoma (HCC) tumor cells (image drawn in the center) and the immunosuppressive component including MDSCs, M2 macrophages, ILCs, N2 cells, NK2, Th2, Treg, NKT of the tumor microenvironment.
Fig. 2  The bidirectional interactions between Hepatocellular carcinoma (HCC) tumor cells (image drawn in the center) and the immunosuppressive component including MDSCs, M2 macrophages, ILCs, N2 cells, NK2, Th2, Treg, NKT of the tumor microenvironment.

MDSCs, myeloid-derived suppressor cells; ILCs, Innate lymphoid cells; NK2, Natural killer; Th2, T helper 2; Treg, regulatory T cells; NKT, natural killer T.

Immunotherapies for HCC

The liver collects blood from the hepatic artery and portal vein, and blood from the portal vein contains nutrients and gut bacteria that are exposed to Kupffer cells (macrophages), NK cells, and innate T lymphocytes in the hepatic sinusoids.60,61 Immune adaptability in the liver, coordinated by Tregs and immunosuppressive inflammatory mediators, is essential because there is a need to prevent excessive immune responses against harmless antigens and bacteria.62 Immunotherapeutic approaches to hepatic cancer can be especially effective because the liver’s defense system promotes an immunosuppressive landscape that can encourage the development and impede the immune capture of cancerous hepatic cells.

Various factors that increase liver inflammation (e.g., toxins, non-alcoholic hepatic steatosis, and viruses such as HBV and HCV) are also risk factors for the onset and progression of HCC. HCC can occur as a consequence of cirrhosis, which is characterized by abnormal interactions among angiogenic cells, fibroblasts, and defense cells.63 HCC is mostly influenced by disruptions in the balance of immune-suppressive and immune-activating cells, and these alterations in the tissue microenvironment can affect prognosis. For example, in vitro studies showed that increased expression of Tregs was linked to more advanced stages of HCC.64 Additionally, clinical studies showed that increased expression of Tregs was associated with a worse prognosis and an increased risk of metastasis.65,66 Tregs impede the invasion of CD8+ effector T cells and decrease the activities of granzyme and perforin.64 Thus, increasing the level of checkpoint blockers (PD-1, PD-L1, Tregs, and MDSCs) decreases antiviral immune reactions.67,68 In particular, PD-L1 overexpression decreases cytokine production, increases the growth of Tregs, and decreases effector T cell cytotoxicity.69 PD-L1 expression is associated with a worse prognosis, a more advanced tumor stage, and an increased likelihood of tumor recurrence.67,70 Several in vitro studies demonstrated that PD-L1 blockade decreased viral density, halted cancer-derived immunosuppression, and prevented cancer growth.71

Biomarkers for tracking responses to immunotherapies in HCC

Many ongoing clinical trials are examining the application of immune therapy for HCC (Table 1).72–74 Some of these studies focused on PD-L1 expression as an indicator of the response to immunotherapy, and examined the effects of novel combinations of ICIs. Early studies found that PD-L1 production is associated with poor prognosis in patients with advanced HCC.75–77 In the ORIENT-32 trial, sintilimab combined with IBI305 showed significant overall survival and progression-free survival benefits compared with first-line treatment with sorafenib in patients with unresectable HBV-related hepatocellular carcinoma.72 Early studies found that the OS benefit of tislelizumab is non-inferior to that of sorafenib, with a higher objective response rate and more durable response, while the median progression-free survival of sorafenib is longer.73 There are also considerable inter-institutional variations in the techniques used to assess PD-L1 production, and this may explain the disparate results of multicenter clinical trials. Ideally, cooperating institutions should establish standardized methods for defining PD-L1 production. It is also crucial to analyze sub-groups of patients with advanced HCC who have similar tumor loads, because PD-L1 expression changes as the disease progresses. The best time for immunohistochemical analysis of HCC tissue specimens and measurement of PD-L1 production is unknown; it is also unknown whether PD-L1 should be measured in stromal tissues, tumor tissues, or both. The inconsistent immuno-histochemical methods used to measure PD-L1 expression make it more difficult to interpret results that assess responses to immunotherapy.78

Table 1

Significant immunotherapy trials are still being conducted to treat advanced hepatocellular carcinoma

Trial namePhaseSettingTargetInterventionReferences
LEAP-002Phase IIILenvatinib vs. the initial therapy for patients who have not received treatmentTKI, PD-1 inhibitorLenvatinib+pembrolizumab74
ORIENT-32Phase II/IIIComparison of sorafenib and first-line therapy for treatment-naive patientsPD-1 inhibitor, VEGFSintilimab+IBI30872
RATIONALE-301Phase IIIComparison of sorafenib and first-line therapy for treatment-naive patientsPD-1 inhibitorTislelizumab73

High intra-tumoral concentrations of CD3+ and CD8+ T cells are associated with longer recurrence-free survival, and the clinical outcomes following treatment with nivolumab (PD-1 inhibitor) and ipilimumab (CTLA-4 inhibitor) are associated with the penetration of CD8+ cells.79 However, this relationship might not apply to patients with chronic hepatitis C, because T-cell entry generally increases in the presence of chronic viral disease. Thus, more research is required to assess the beneficial effects of ICIs in patients with HCC.79 The TMB can also indicate a tumor’s immunogenic potential.80,81 The generally low TMB in HCC (average of 5 mut/Mb) may mean that this metric has limited application for HCC, although TMB is increasingly used as a biomarker in a variety of other malignancies. A thorough genomic profile analysis of 755 patients with late-stage HCC showed that the median overall TMB was only about 4 mut/Mb, and that there was no link between TMB and response to treatment, disease progression, or stable disease (SD). This underlines the limited usefulness of TMB as a biomarker for HCC.82

The organization of cancers into molecular features is contrary to individual types of indicators, because HCC tumors are variable and have unique TMEs. A study of HCC found that the most prevalent genetic “clusters” were for interferon predominance, lymphocyte deficiency, inflammation, and wound healing. An improved understanding of the role of different gene clusters in the development of particular immune escape mechanisms may help to develop more effective combination therapies.83 This may allow researchers to specify patient groups that would benefit most from specific combination therapies according to their genetic profiles.

Antibody-based therapies for HCC

Treatments with a single ICI

Effector immune cells express different immunological checkpoints, and activation of these checkpoints prevents an overactive immune response. There are numerous endogenous inhibitors of these checkpoints, including the T cell immunoreceptor with Ig and ITIM domains (TIGIT), B and T lymphocyte attenuator, LAG3, and TIM3.84,85 HCC exploits this checkpoint system to prevent anti-tumor immune responses.86 ICIs are mAbs that block these checkpoints and restore immune responses. The immune response against tumors can also be enhanced by preventing the deactivation of T cells and the reactivation of immune targets. PD-1, CTLA-4, and PD-L1 are currently the main targets of approved ICIs.87 The vast majority of immune cells (including MDSCs) mainly stimulated T-cells, Treg, DCs, NK cells, and monocytes, express PD-1, which is a member of the CD28 family. PD-1, PD-L1, and PD-L2 inhibit T cells, and this activates HCC and allows it to evade the immune system.88

The US FDA approved nivolumab (PD-1 inhibitor) in 2017 for use as a second-line therapy for patients with severe HCC after the receipt of sorafenib, a tyrosine kinase inhibitor (TKI).89 Several trials have also examined the use of other ICIs as treatments for HCC, and pembrolizumab (PD-1 inhibitor) and atezolizumab (PD-L1 inhibitor) were approved as clinical treatments for HCC in various countries. Nivolumab and pembrolizumab can provide an objective remission rate of 15 to 20%, and a complete recovery rate of 1 to 5%. More specifically, for the 48 patients in the CheckMate 040 trial, nivolumab treatment led to a median response duration of 17 months, and 80% of responders had a survival time of 2 years or more.90 The KEYNOTE-240 study, a phase III trial of 413 patients that compared pembrolizumab after sorafenib with placebo after sorafenib, found a significantly longer survival time in the pembrolizumab group. Based on overall survival (OS) and progression-free survival (PFS), pembrolizumab appears to provide long-term benefit for some patients.91 The phase III CheckMate 459 trial of 743 patients who had not used systemic medicines also evaluated nivolumab and sorafenib. In comparison with the sorafenib group, the nivolumab group had a longer median survival time.92 The extended continuation period in the CheckMate 459 trial supported the superiority of nivolumab over sorafenib in terms of long-term survival.93

Tislelizumab (PD-1 inhibitor) also provided long-term benefits and was readily accepted by patients who received prior systemic treatment for unresectable HCC. Tislelizumab and sorafenib were used as a first-line treatment in a large, randomized phase III trial of adults with unresectable HCC (NCT03412773).94 The majority of CD28 family member CTLA-4-expressing T cells and DCs that have been stimulated do so. CTLA-4 (which is related to CD28) downregulates the immune response after binding to B7.95 Ipilimumab (CTLA-4 inhibitor) was approved in 2011 and was the first ICI approved by the US FDA for the treatment of advanced skin tumors. Tremelimumab (CTLA-4 inhibitor) was approved in 2022, and is the most recently approved ICI.96

Ipilimumab (CTLA-4 inhibitor and an IgG1 mAb) and tremelimumab (CTLA-4 inhibitor and an IgG2 mAb) have distinct antibody-dependent cell-mediated cytotoxicities and complement-dependent cytotoxicities.97 Clinical trials showed that tremelimumab had potent anti-HCC effects, with a 17.6% partial response (PR) rate and a 76.4% illness control rate.98 Some research suggests that the efficacy of CTLA-4 inhibitors can be attributed to the targeted removal of Tregs from cancers.99 HCC impairs the ability of T cells to function as effector cells due to the occurrence of TIM3 on TAMs and TILs, and there is a corresponding increased expression of tumor suppressor genes by Tregs.100 The excess production of TIM3 is related to a less distinguished HCC.101 Compared to other immune system components, LAG3 is considerably more abundant on tumor-specific CD4+ and CD8+ TILs in individuals with HCC. Fibrinogen-like protein 1, which is produced by hepatocytes, is another soluble ligand for LAG3.102 There is evidence that Siglec-15 (a lectin that binds to sialic acid) prevents the lysosomal destruction of CD44, leading to increased migration of liver cancer cells.102,103 TIGIT is also affected by T-cell immunoreceptors that have immunoglobulin and ITIM domains.104 To prevent the activation of T cells, DCs produce more IL-10 and less IL-12 by the activation of the TIGIT/CD155 pathway.105 Recent clinical trials showed that individuals who received ICIs alone had inadequate responses. Hence, future trials should examine combinations of different ICIs with other treatments.

The combination of atezolizumab (PD-L1 inhibitor) with bevacizumab (anti-angiogenic antibody) significantly decreased the risk of death in patients with unresectable HCC.106 The overall response rate of 46% was achieved by combining pembrolizumab with lenvatinib (TKI), and patients with unresectable HCC had a complete response (CR) rate of 11% and a partial response (PR) rate of 35%.107 Other recent preclinical and clinical studies showed that co-administration of ICIs with radiation, radiofrequency ablation (RFA), or TACE improved therapeutic efficacy.108,109 A phase Ib/II clinical study of patients with advanced HCC is examining the effect of camrelizumab (investigational PD-1 inhibitor) with FOLFOX4 chemotherapy.110

The HIMALAYA phase III trial showed that treatment with a single PD-1 inhibitor or a single PD-L1 inhibitor led to significant antitumor activity.111 A study that administered tremelimumab (CTLA-4 inhibitor) monotherapy to patients who previously received sorafenib and experienced intolerable toxic effects or who rejected sorafenib, showed that this ICI provided demonstrable protection; however, the combination of tremelimumab with durvalumab (PD-L1 inhibitor) led to greater benefit.112Figure 3 lists the numerous ICIs that have been used to treat HCC and Table 2 summarizes the clinical studies that examined the use of ICIs for the treatment of HCC during the past three years.75,90,105,112–115

Inhibitors of immune checkpoints in hepatocellular carcinoma.
Fig. 3  Inhibitors of immune checkpoints in hepatocellular carcinoma.

CTLA-4, Cytotoxic T-lymphocyte antigen 4.

Table 2

Hepatocellular carcinoma immune therapy with immune checkpoint blockers: A reported clinical trial

TreatmentPatients, nORR%OS in moReferences
Camrelizumab21715 (0)13.8115
Pembrolizumab27818 (2)13.9105
Durvalumab10411 (0)13.6113
Tremelimumab697 (0)15.1113
Durvalumab and tremelimumab1599.5–24.0 (1–2)11.3–18.7113
Pembrolizumab and lenvatinib10036 (1)22112
Nivolumab and ipilimumab14831–32 (0–8)12.5–22.875
Atezolizumab and bevacizumab33627 (6)NE112
Nivolumab and cabozantinib3614 (3)21.575
Nivolumab, ipilimumab and cabozantinib3531 (6)NE75
Atezolizumab5917 (5)NA115
Nivolumab37115(4)16.490

Combination treatments with two ICIs

PD-1 with CTLA-4 blockers

Treatments for advanced HCC are currently focusing on the use of CTLA-4 inhibitors and PD-1 inhibitors together. For example, the CheckMate 040 study examined the effect of ipilimumab (CTLA-4 inhibitor) with nivolumab (PD-1 inhibitor) in 148 individuals who developed advanced HCC after sorafenib treatment.75,116 This trial found that the median response time was 17 months, the overall response rate was 31%, and the disease control rate was 49%. These results led the US FDA to approve the combination of ipilimumab and nivolumab for these patients. The CheckMate 9DW phase III trial (NCT04039607) is currently assessing this combination treatment as first-line therapy for advanced HCC.

ICIs with vascular endothelial growth factor inhibitors

The IMbrave150 phase III study examined the combination of a PD-1 inhibitor (atezolizumab) with a vascular endothelial growth factor (VEGF) inhibitor that inhibits angiogenesis (bevacizumab) as a unique approach for the treatment of advanced HCC.117,118 This treatment provided good protection and antitumor efficacy in individuals with untreated late-stage HCC.119 In particular, relative to sorafenib, this combination reduced the risk of death by 42%. The IMbrave150 trial led to the approval of treatment that combined atezolizumab and bevacizumab, instead of TKIs (sorafenib or lenvatinib), as a first-line therapy for unresectable HCC in the United States and Europe. The efficacy of this new treatment is most likely due to the synergistic effects of these two drugs, which inhibit PD-L1 (stimulating the immune system, chiefly T-effector cells) and also inhibit VEGF (promoting T-cell infiltration, reducing VEGF-mediated immunosuppression, and inhibiting angiogenesis).113

ICIs with tyrosine kinase inhibitors

The combination of ICIs with TKIs (rather than an anti-VEGF antibody) is an alternative approach. The multi-cohort COSMIC-021 phase Ib trial (NCT03170960) examined the effect of cabozantinib (TKI) and atezolizumab (PD-L1 inhibitor) for the treatment of HCC.120,121 This study also compared the effect of cabozantinib monotherapy to sorafenib as a secondary outcome. A planned interim analysis demonstrated no significant difference in OS. However, a phase Ib trial showed that the combination of lenvatinib (TKI) with pembrolizumab (PD-1 inhibitor) led to acceptable outcomes in 104 patients with unresectable HCC who did not receive a previous systemic therapy.107 The LEAP002 phase III trial (NCT03713593) compared this combination with lenvatinib monotherapy.122 Another study examined the effect of camrelizumab (investigational PD-1 inhibitor) with apatinib (TKI) and reported the overall response rate was 50%.123 Moreover, an ongoing phase III study (NCT03764293) is evaluating camrelizumab with apatinib vs. sorafenib as a first-line treatment for advanced HCC.

Recent research indicated that ICI combination therapies, including those with TKIs, have increased anticancer efficacy due to their immunomodulatory effects on the TME and their pro-angiogenic effects on certain pro-tumor immune cells. Before the IMbrave150 trial, there were promising response rates to TKI+ICI combinations compared to individual therapies. In particular, lenvatinib decreased the level of tumor-associated macrophages and increased the level of CD8+ T cells.124 Regorafenib (TKI), which targets VEGFR, EGFR, PDGFR, and FGFR, promoted antitumor immunity by regulating macrophages and enhancing CD8+ T cell proliferation. Preclinical and clinical studies of cabozantinib showed it had synergistic effects when combined with an ICI; the effect on tumor antigens (such as TAMs) reduced tumor vascularity, and bevacizumab significantly restored an immune-supportive TME.125 The use of PD-1 inhibitors with TKIs and TACE was also an effective treatment for patients with unresectable HCC, and the ORR surpassed that achieved by the monotherapies.126,127 Thus, combining an ICI with anti-angiogenic drugs can reverse the immunosuppressive character of the TME, but the most effective and safe TKI to be used with an ICI has not yet been established.

Cellular therapies for HCC

Therapies with non-genetically altered cells

Cytokine-induced killer cells

Cytokine-induced killer cells (CIKs) are CD3+CD56+ NK-like T-cells that develop from peripheral blood mononuclear cells or cord blood following ex vivo incubation with anti-CD3 mAb, IL-2, IFN-γ, and IL-1α.128,129 CIKs have non-MHC-restricted cytotoxic and anti-proliferation effects.130 A phase I clinical study with adjuvant CIKs found there was decreased tumor recurrence of graft-versus-host disease (GvHD) after surgery in patients with stage A/B liver cancer (staging based on the Barcelona Clinic Liver Cancer system).131,132 Three of these 13 patients had good tolerance to the CIKs, and this treatment reduced the HBV burden and slowed tumor development.133 A second phase I basket study examined 12 patients who received 3 cycles of CIKs over 3 weeks (median dosage: 28×109 cells, range: 6 to 61×109 cells) for the treatment of late-stage HCC, resistant renal cell carcinoma, or lymphoma. After an average follow-up time of 33 months, the CIKs were well tolerated, 3 patients attained CR, and 2 patients had SD.134

Another study of patients with unresectable HCC assessed the effect of CIKs paired with TACE.135 The CIK+TACE group had a PFS of 6 months in 72.2% of patients, 1 year in 40.4% of patients, and 2 years in 25.3% of patients, and these numbers were better than in the TACE-alone group (34.8%, 7.7%, and 2.6%, respectively). The median OS in the CIK+TACE group was 31 months (95% CI: 27–35), but was only 10 months (95% CI: 7–13) in the TACE-alone group. These findings indicate the potential benefits of combining an immunotherapy strategy with TACE for treating HCC.135 A subsequent clinical study of 64 patients with HCC applied TACE and RFA sequentially in patients who did (n=33) or did not (n=31) receive prior CIK treatment.129 These researchers administered 8 doses of CIKs into the hepatic artery or a peripheral vein at intervals of 4 weeks. After 1 year, 68% of the control group and 88% of the CIK group were free of recurrence.129 Another study evaluated a DC+CIK combination therapy in a basket trial, and showed that DCs enhanced the activation of CIKs.136 There is also evidence that a DC+CIK regimen can control tumor development and increase OS.136,137 An in vitro study of HCC cells (BEL27402) compared to DC+CIK alone, sorafenib alone, and CIK alone, and DC+CIK with sorafenib, showed the greatest efficacy in the DC+CIK with sorafenib group.138

Tumor infiltrating lymphocytes

TILs are also used in autologous therapies. In particular, TILs are developed ex vivo from polyclonal, tumor-targeting T-cells that are generated against a patient’s tumor. In patients with immunogenic tumors, such as metastatic melanoma, these treatments led to an overall response rate (ORR) of 49 to 72%, a CR rate of 10 to 20%, and persistent responses in 40% of all patients.139,140 Terminally differentiated TILs, with a predominance of immune-suppressive components (Tregs), can result from protracted ex vivo processing. To prevent this, tumor-reactive/neoantigen-responsive cytotoxic T-cells are now grown non-selectively in large quantities of TILs,141 and clinical investigations of these treatments are currently being conducted for patients with non-small cell lung cancer and melanoma. HCC TILs, by phenotype worn out in preclinical models, have increased production of TIM-3 and LAG-3, but reduced production of inflammatory mediators.142,143 Preclinical studies demonstrated that TIGIT and PD-1 co-blockade enhanced the proliferation of these cells and the release of cytokines.143

Therapies with genetically altered cells

The capacity of the immune system to identify TAAs is a prerequisite for effective anti-tumor responses in immunotherapeutic cancer treatments. A functional immune system has self-tolerance, and because all tumors consist of self-tissues, it is difficult to induce a strong anti-tumor response. Therapies that use chimeric antigen receptor (CAR) engineered T cells and T cell receptor (TCR) engineered T cells use synthetic receptors developed by genetic engineering to alter immune cells and increase the detection of TAAs and tumor-specific antigens (TSAs).144

Chimeric antigen receptor T cells

Immune cells can be genetically reprogrammed to recognize and attack cells that express specific TAAs using artificial cell surface receptors (CARs) that are independent of the MHC system.145,146 A typical CAR structure consists of an external, antibody-derived single-chain variable fragment (scFv) antigen-binding domain that is linked to an intracellular signaling endodomain, which has (at a minimum) CD3ζ signaling capabilities.147 First-generation CARs with isolated CD3ζ signaling were mostly replaced by CARs with CD28 or 4-1BB receptors, leading to enhanced CAR-T cell growth and increased cancer cell death in vitro and in vivo.148,149 The most significant development in hematological malignancies during the past ten years was the development of CARs that target CD19 in the subsequent phase, and have 4-1BB150 and CD28151 co-stimulatory endodomains. There is now global approval of autologous CD19-targeting CAR-T cells for relapsed and refractory B-cell lymphoma. Notably, third-generation CARs, which contain two intracellular signaling domains, such as 4-1BB and CD28, are also under development.152–155 Ongoing research is examining the effect of CAR-T cell therapies for HCC, and there are several promising CAR-T cell tumor targets, including AFP, c-MET, GPC3, Mucin 1, and NK group 2D ligand (NKG2DL).

Glypican-3 biological system and CAR-T cells

The placenta contains GPC3, a 65 kD (580 amino acid) heparin sulfate proteoglycan that is anchored to glycosylphosphatidylinositol and normally functions in morphogenesis by activating the Wnt pathway.129,156,157 Several solid tumors, including 72% of HCCs, carries a negative prediction.158,159 Crucially, GPC3 is only weakly expressed in noncancerous tissues, such as healthy tissues and cirrhotic liver tissues.160,161 About 53% of HCC patients have soluble GPC3, and this protein is therefore under investigation as a disease biomarker.159 GPC3 appears to affect the onset and progression of HCC.162,163 Studies of primary HCC cells reported that siRNA-mediated GPC3 silencing reduced proliferation, increased apoptosis, and impaired the migration of tumor cells.164

Just like central- or stem-cell memory T-cells, GPC3-CAR-T item characterization showed advancement for final differentiation, CD45RA+ re-expressing effector memory T-cells (78.2%) and effector memory T-cells (14.1%).165 Several studies showed that CD19-CAR-T activity was negatively affected by terminally differentiated T-cells, and this was likely to occur in HCC.166,167 Leukapheresis could possibly improve the efficacy of GPC3-CAR-T cell therapy.168

Even the responses to these therapies were inadequate, the toxicities were generally manageable, and this trial laid the groundwork for future GPC3-CAR-T strategies. For example, current trials are examining ‘armored’ GPC3-CAR-T models, with components designed for 41BBL and IL-15/IL-21 (NCT02932956),169 a combination of techniques using TKIs or IPIs (NCT03980288), and administration via hepatic artery perfusion (NCT03993743). It is not yet clear how soluble GPC3 can affect GPC3-CAR-T function. However, soluble GPC3 might prevent accessibility to cell-surface GPC3.170 Future studies should consider this when creating and testing the preclinical versions of the next-generation GPC3-CAR-T cell therapies.

Others

AFP can occur as a serum protein or an intracellular protein, and a modified version can occur as a cell surface protein on MHC class I molecules. AFP is a biomarker for HCC, its release into the serum stimulates the growth of tumors, and a high serum level is associated with poor prognosis.171,172 A phase I clinical trial, whose results have not yet been published, is examining the effect of intravenous and intrahepatic arterial administration of AFP-CAR-T cells in AFP+ patients with HCC (NCT03349255).

After processing and presenting short antigenic peptide fragments on HLA class I and II molecules, engineered TCRs can recognize intracellular TAAs and TSAs.173 The ability of TCR-T cells to recognize and connect intracellular antigens encoded on HLA, despite a low target density, can provide a significant benefit.174,175 Two disadvantages of this approach are that this treatment is limited to a small percentage of patients, primarily those who are HLA-A*0201 positive,174 and it is difficult to engineer high-affinity, synthetic αβ-TCR for TAA-specific targeting, while avoiding the challenges of TCR chain mispairing and low TCR production.176 Additionally, there is a chance that production in normal and cancerous tissues will overlap at low antigen densities. TCR-T cell binder development could be advanced by adjusting TCR affinity with physiochemical and in silico approaches to facilitate discrimination of cancerous and non-cancerous cells.177 There are now seven phase I/II TCR-T cell clinical studies of HCC that are accepting participants.153 Additional trials listed on clinicaltrials.gov that have ‘status unknown’ or ‘not yet recruiting’ include three studies of patients with HCC relapse following liver transplantation (NCT02686372, NCT04677088, and NCT02719782).

Clinical studies utilizing an AFP epitope (AFP158–166, [NCT03132792]), AFP is managed and displayed on HLA and reflects a useful seek for TCR-T methods.178 Studies that performed in vitro testing described three novel HLA class I epitopes (AFP542–550, AFP137–145, and AFP325–354) that were cytotoxic and induced IFN- production against in HLA-A*0201+/AFP+ tumor cells. Other research used a transgenic mouse model to investigate the immunogenicity of different AFP epitopes.179,180 Altogether, these results of these studies demonstrate the presence of four immunogenic AFP epitopes that could be used as targets of TCR-T cells.

The initial clinical findings of a study that used affinity-boosted autologous specific peptide enhanced affinity receptor (SPEAR) T-cells targeting AFP (NCT 03132792) were released in 2019.129 This study examined patients with the HLA-A*02:01 or HLA-A*02:642 haplotypes who had serum AFP levels of 400 ng/mL or more or had positive immunohistochemical staining for AFP (≥1+ in 20% or more HCC cells). Following a fludarabine/cyclophosphamide chemotherapy regimen, SPEAR T cells (102 to 103 million) were given to cohort-one (n=5) to determine the maximum tolerated dose (MTD). All five patients had SD as the optimal reaction. The MTD for cohort two (n=3) was set at 5×109 SPEAR T- cells (range: 5.0 to 5.6×109). One patient achieved PR, and the other 2 experienced progressive disease, and the treatment was well-tolerated.178

HBV is responsible for about 80% of all cases of HCC in Asia because of persistent hepatic inflammation, and the virus is incorporated into the hepatic cell genome.181,182 Because the likelihood of on-target, off-tumor toxic effects decreases and the low rate of target production lends itself to TCR-T targeting, attacking viral, non-self-antigens is an appealing approach for TCR-T cell treatments. HBV infections typically induce high-affinity TCRs, which have significant therapeutic potential because they can lyse HBV-infected cells. However, the main disadvantage of this approach is that non-malignant liver tissues are also likely to have HBV antigens, raising the possibility of life-threatening liver injury.183

Gehring et al. presented a liver transplant patient who had extra-hepatic HCC, and HBV-DNA integration resulted in surface HBV (HBsAg), but there was no detectable HBV-DNA in the blood.183,184 The HCC cells displayed HLAA0201/HBV peptide complexes, and their expression was uniform across the tumor. To produce autologous, patient-derived HBV-TCR-T cells, these researchers identified a specific TCR that targeted the HLA-A-*0201/HBs183-91 mixture and cloned this gene. Without lympho-depletion, the individual received only one dosage of 1.2×104 HBV-TCR-T cells/kg. These cells multiplied and decreased the blood levels of HbsAg, and there was no evidence of harm to healthy tissues.183 However, the recurrence rate from hepatitis B is substantial (50% within five years)13 and the blood HbsAg level is linked to recurrence. An important question raised by this instance is if HBV-TCR-T might be utilized greater frequently for HCC patients who relapse after liver transplantation.185 In particular, after liver transplantation in HbsAg+ patients, TCR-T cells could be administered as prophylactic to avert relapse.

Bispecific antibodies (BsAbs), produced using recombinant DNA technologies, can precisely and simultaneously bind two antigens or epitopes.186,187 To alter immune-suppression in the tumor environment, a BsAb can be used to target immune checkpoints and TAAs, thereby enhancing the function of immune cells. Therefore, because BsAbs have two effects, they are potentially more effective than mAbs. BsAbs typically function as a “bridge”; they can recruit and activate immune cells to target cancer cells.188 Solitomab (AMG110, MT110) is one example of a humanized EpCAM/CD3 BsAb. The in vitro binding of Gamma-Delta T cells with the bispecific T-cell engager (BITE) leads to the nearly complete lysis of HCC cells. This treatment is characterized by the attachment of the anti-epithelial cell adhesion molecule (EpCAM), single-chain variable fragment (scFv), and the anti-CD3 single-chain variable fragment (scFv) by a Gly4Ser linker.189 A distinct BsAb named GPC3/CD3 BITE was designed to attract CTL and target GPC3+ HCC cells. This BsAb used flexible linker peptides to unite two anti-GPC3 Fab fragments to an asymmetric Fab-sized binding module, leading to an IgG-shaped Tri-Fab that activated two antigens sequentially, so that it could be used for the targeted delivery of different payloads.190

HCC vaccines

Antigenic compounds can elicit tumor-specific immune responses, leading to a reduced tumor load and prevention of tumor reversion. HCC vaccines can be developed from cancer cells, DCs, peptides, and DNA, and some of these vaccines have successfully prevented tumor spread and recurrence. Peptides are widely used as cancer vaccines, and the most appropriate peptide for generating a cancer vaccine is determined by the type of tumor and the immunologic characteristics of the patient.191,192 The search for “HCC vaccines” identified six general types of research (Table 3).193

Table 3

The clinical trial of cancer vaccines targeting HCC193

TargetPhaseStartEndPeptideMethods/combinationDescriptions
DNAJB1- PRKACA1Apr., 20DNAJB1-PRKACANivolumab and IpilimumabThe trial’s main goal is to determine the vaccine’s safety and tolerability.
16 common cancer antigens1 and 2Sep., 17Dec., 1916 newly identified, excessively expressed tumor-related peptidesNovel RNAFor the treatment of (hepatocellular carcinoma), a new adjuvant called CV8102 is paired with a new cancer vaccination called IMA970A.
VEGFR1, VEGFR2120072013VEGFR1, VEGFR2This study aims to evaluate the side effects of angiogenic peptide vaccine therapy in patients with advanced hepatocellular carcinoma who are HLA-A*2402 restricted.
AFP1 and 2Jan., 01Oct., 08Four HLA-A*0201-restricted immunodominant AFP peptides [hAFP137-145 (PLFQVPEPV), hAFP158-166 (FMNKFIYEI), hAFP325-334 (GLSPNLNRFL), and hAFP542-550 (GVALQTMKQ)]Dendritic cellsPhase I/II study to examine the efficacy of vaccination treatment in the management of patients with liver cancer.
AFP1 and 2Jul., 09Jun., 024- HLA-A*0201-restricted immunodominant AFP peptides [hAFP137-145 (PLFQVPEPV), hAFP158-166 (FMNKFIYEI), hAFP325-334 (GLSPNLNRFL), and hAFP542-550 (GVALQTMKQ)]IntradermalPhase I/II study to examine the efficacy of vaccination treatment in the management of patients with liver cancer.
Ras mutation2Oct., 07May., 07Mutated Ras Peptides Specific for TumorsIL2 or GM- CSFAdults with metastatic solid tumors will be treated in a phase II trial to see whether vaccination therapy combined with interleukin-2 and/or sargramostim is beneficial.

Cellular vaccines

Autologous or allogenic HCC cells or extracts that have been physically or chemically killed or damaged so that they are not pathogenic can be used as antigens to induce tumor-specific defense reactions. A phase I trial examined 8 patients with advanced HCC that tested bi-shRNA/granulocyte-macrophage colony-stimulating factor (GM-CSF) boosted autologous tumor cells. The long-term follow-up showed that 3 patients had clear immune responses to the reinfused cancer cells, and the long-term follow-up demonstrated survival times were 319, 729, 784, 931+, and 1,043+ days after treatment. However, the effectiveness of HCC vaccines remains unknown due to their poor immunogenicity.194

Antigen peptide vaccines

Several studies used peptide-based vaccines to treat HCC, and these vaccines utilized AFP, GPC3, SSX-2, NY-ESO-1, human telomerase reverse transcriptase (hTERT), human carcinoma-associated antigen (HCA587), and melanoma antigen gene-A (MAGE-A) as TAAs.195,196 Embryonic liver cells normally produce AFP, but this protein is also overexpressed on the surface of HCC cells. However, the development of acquired immunological tolerance during development limits immune responses to this excess AFP in patients with HCC. A recent study used recombinant rat AFP to trigger cross-reactions between xenografts and endogenous molecules in mice to overcome this immunological tolerance and reported minor cellular and humoral immune responses.197 A phase II trial examined 25 patients who received a GPC3-derived peptide vaccine for HCC. The treatment consisted of 10 injections over a 1-year period following surgery. Relapse was less common in patients who received surgery and vaccination relative to those who received surgery alone (24% vs. 48% at 1 year [p=0.047] and 52.4% vs. 61.9% at 2 years [p=0.387]), demonstrating the effectiveness of this vaccine.198

Many other clinical investigations have investigated the use of HCC vaccines (Fig. 4).193 The outcomes of one trial that examined a peptide vaccine developed using GPC3 (which typically has elevated expression in HCC) were published in 2011.199 These patients had advanced HCC, were from the National Cancer Center Hospital East (Kashiwa, Japan), and were recruited into this phase I trial to assess the protection and immunogenic response elicited by the vaccine.200 This study demonstrated a relationship between the peptide-specific cytotoxic T lymphocyte level and patient privacy concerns regarding the GPC3 peptide vaccine utilization because RFA influences a specific T cell’s improvement against HCC-related antigens or GPC3. The same team conducted a single-arm Phase II trial in which some patients received adjuvant treatment with a GPC3-derived peptide vaccine.201 This GPC3 peptide vaccine induced a CTL response that efficiently destroyed cancer cells that expressed GPC3, so that GPC3-negative cells proliferated. This proof-of-concept utilizing the GPC3 peptide and additional peptides was effective, and hence opened the door for studies of other peptide and antigen treatments.

Diagram illustrating the GPC3-targeting peptide vaccination.
Fig. 4  Diagram illustrating the GPC3-targeting peptide vaccination.

The picture shows the various steps involved in creating the vaccine. GPC3, glypican 3.193

DC vaccines

DCs are the most potent APCs, and function in the absorption, digestion, and presentation of tumor antigens. These cells have significant levels of MHC and Cas ligands with multiple Src homology (SH) 3 domains (CMS), such as B7-1 and B7-2. They also induce primary T cells and release IFN-γ, a cytokine that inhibits tumor angiogenesis and creates immunological memory. Altogether, DCs therefore have many anticancer effects.202,203

During the creation of a vaccine against HCC, DCs were first stimulated by specific mediators (e.g., rhGM-CSF and rhIL-4) then developed in the existence of TNF-α, and eventually became activated by autologous tumor cells or antigens.196 DCs with gene transfections continue to express cytokines or tumor antigens that enhance their function. A recent study of mice with HCC administered nifuroxazide (which blocks signal transduction mediated by stimulation of transcription 3 [STAT3]), together with DCs that were loaded with tumor cell lysate (TCL). This combination increased the antitumor immune response, slowed tumor development, and increased the survival time.204 A phase I/IIa trial examined the effect of tumor antigen-pulsed DCs for HCC patients who received primary treatment and showed that DC immunization was an efficient adjuvant therapy.205 Another study reported the safety and tolerability of DC vaccinations in HCC patients.206

Adjuvant immunotherapies for HCC

Numerous clinical studies are examining the use of adjuvant immunotherapies for patients with intermediate-stage HCC. The preliminary results suggest that tremelimumab with TACE or RFA has the potential for use in patients with early-stage HCC.109,207 This approach was also examined in the IMbrave150 trial, a randomized, open-label, phase III study that examined HCC patients after curative resection or RFA and compared atezolizumab with bevacizumab to active monitoring.208 Additionally, the phase III EMERALD-1 and EMERALD-2 trials are comparing durvalumab with or without bevacizumab to placebo for patients with intermediate-stage HCC who received TACE or RFA.93,209

Unlike many other cancers, HCC can be treated using locoregional therapies (LRTs), including TACE or RFA. Consequently, complementary therapies administered after an LRT may improve clinical outcomes. Sorafenib was approved in 2007, and it remains the only treatment option for advanced HCC. Additional systemic medications for advanced HCC have recently been examined, including new TKIs (e.g., lenvatinib) as a front-line therapy and regorafenib or cabozantinib as a second-line therapy. In an effort to improve clinical outcomes, several researchers are now examining a combination of TKIs and LRTs. Nevertheless, sorafenib followed by resection or ablation (STORM study)210 failed to increase recurrence-free survival compared to placebo. Likewise, multiple earlier trials showed that TKIs following TACE failed to improve clinical outcomes.211 For unresectable HCC, an ICI such as nivolumab is now recommended, although recent trials demonstrated that ICI monotherapies did not significantly improve survival in HCC (in contrast to other tumors).212 As a result, other studies have investigated different strategies for overcoming the inadequate response to ICIs. As a first-line treatment, a combined regimen of atezolizumab with bevacizumab provided significantly greater clinical benefit than sorafenib.107 Additionally, another combination treatment—pembrolizumab with lenvatinib—was recently tested in clinical studies and has shown good clinical efficacy during the early stages of treatment.117 Combining LRTs with ICIs could be an important development in the treatment of HCC, and could also significantly improve the prognosis of these patients.

Oncolytic virotherapy

Therapeutic oncolytic viruses are engineered viruses or wild-type viruses that reproduce and destroy cancer tissues or other pathological tissues without adversely affecting healthy tissues.213,214 Because a tumor’s defenses against viral infection are compromised, most viruses can easily spread to cancer cells.215 Furthermore, the stimulation of immune responses against neighboring cancer cells can be facilitated by the presence of tumor antigens and viruses within cell lysates.216,217 One advantage of oncolytic virotherapy is that it may not be subject to some of the limitations of conventional cancer therapies, such as chemotherapy and radiation therapy. For example, certain cancer cells can become resistant to chemotherapy or radiotherapy, but viruses can persist over time and still infect and kill tumor cells.218 In addition to directly killing cancer cells, oncolytic viruses can also stimulate the immune system to target the cancer cells because lysed cancer cells release TSAs that can stimulate an immune response. This approach therefore enhances the body’s ability to detect and eliminate tumor cells,219 and is particularly suitable for treating HCC because the liver has a high degree of immune surveillance.60

Oncolytic viruses can target cancer cells through several different mechanisms. First, several wild-type viruses can infect tumors by different mechanisms that evolved in nature, such as Sindbis viruses, reoviruses, and varicella viruses.220 Second, genetic engineering can also be used to create oncolytic viruses by eliminating viral genes that are essential for replication in healthy cells but have no functionality in cancerous cells.221 Third, the targeted transcription of viruses in cancer cells can be achieved by inserting tumor-specific promoters, including the human telomerase reverse transcriptase promoter, upstream of essential viral genes.222 Finally, viruses can target tumor cells after alteration by TAA-specific receptors. For instance, the tumor-specific inhibition of tumor angiogenesis can be achieved using an oncolytic vaccinia virus that is engineered with anti-angiogenic genes.223

Previous research examined the effectiveness of a progressive tumor-favoring modified vaccinia virus (CVV) in an animal model of metastatic HCC. In this study, groups of rats were randomly given sorafenib, the CVV, or sorafenib plus the CVV. In comparison to the sorafenib-only group, the other two groups had smaller metastatic areas. These findings indicate the potential use of CVV as a treatment for metastatic HCC.224 JX-594 is a modified vaccinia virus that is particularly hazardous to cancer cells, but is stable and safe for people. This virus has a mutated TK gene (which regulates cancer cell-specific reproduction) and an insertion in the human GM-CSF gene (which boosts antitumor immune reactions).225 A phase II randomized open-label trial of patients with HCC examined the effectiveness and safety of oncolytic virotherapy using JX-594. The results showed that the intrahepatic reaction rate was 62%, 1 patient achieved CR, the therapy was well tolerated at high and low dosages, and the OS was greater in the high-dose group than in the low-dose group.226 Numerous other studies have investigated other oncolytic viruses for the treatment of HCC, including GLV-1h68 and G47delta.227 Important safety considerations related to this approach are the risk of viral disease and the development of insertional mutations that stimulate oncogenes or disrupt tumor suppressor genes.

Conclusions and future prospects

HCC is a complex disease that can escape immune responses by various mechanisms, suggesting great potential for treatments that use different or multiple immunotherapy approaches. The range of immunotherapy treatments for HCC has expanded significantly during the past 10 years, and ICIs are now widely used for patients with advanced-stage HCC. The development of novel medicines and combination therapies is being shaped by the greater understanding of the molecular pathways responsible for cancer initiation and termination of the body’s anti-tumor immune responses in the TME. Although many trials have demonstrated the possible efficacy of different immunotherapies for HCC, only a few have been formally licensed. The identification of more focused immunological targets (such as TAAs/TSAs and new immune checkpoints) and the use of oncolytic viruses require further research. It is also important to accelerate the enrollment of patients in these clinical studies and to consider the effectiveness and safety of novel medications. The development of more individualized treatment programs may also increase the effectiveness of immune therapies.

HCC immunotherapy has progressed greatly, and although ICIs were initially used to treat other cancers, they are now commonly used to treat HCC. Our update on the use of immunotherapy in HCC primarily describes developments in the methodologies used in clinical trials.74 It is likely that the recently developed neoadjuvant treatments for patients with resectable or non-resectable HCC will soon provide benefits to patients, in terms of decreased cancer progression and mortality. ICI-based therapies may also boost the efficiency of locoregional and radical treatments for HCC. The expansion of novel immunotherapies, such as immunostimulatory mAbs, BsAbs, tailored cytokines, antibody-drug conjugates, adoptive T cell therapies, and vaccination with neoantigens, will be important future developments. It is crucial to consider the molecular aspects of the responses to these treatments and the development of tolerance to specific drugs or mixtures, and to use relevant biomarkers to monitor patient responses to personalized immunotherapies. Clinical studies and other research should aim to incorporate the correlative findings from other investigations, and provide the results to other researchers while safeguarding the concerns and rights of patients and organizations.74

The various mechanisms that contribute to resistance to different ICI treatments may be overcome by the use of novel immunotherapies and targeted combination treatments. In addition to the results that were published at the time of the current review, the results of other ongoing clinical studies of HCC will be available soon. It is likely that patients who received more advanced ICI regimens will respond favorably to these new combination treatments. Certain combinations of VEGF inhibitors, ICIs, and TKIs are effective in patients with other types of tumors who previously received ICIs, and these approaches may be extended to HCC.114 For example, a phase Ib/II trials of lenvatinib (a TKI that inhibits angiogenesis) with pembrolizumab appears to have great potential for the treatment of patients with metastatic HCC.115

Abbreviations

AFP: 

α-fetoprotein

APC: 

antigen-presenting cell

BsAbs: 

bispecific antibodies

CAR: 

chimeric antigen receptor

CIKs: 

cytokine-induced killer cells

CMs: 

Co-stimulatory molecules

CR: 

complete response

CTLA-4: 

cytotoxic T lymphocyte-associated antigen 4

DCs: 

dendritic cells

GM-CSF: 

granulocyte-macrophage colony-stimulating factor

GPC3: 

glypican 3

HBV: 

hepatitis B virus

HCC: 

hepatocellular carcinoma

HCV: 

hepatitis C virus

HLA: 

human leukocyte antigens

HSCs: 

hepatic stellate cells

ICIs: 

immune checkpoint inhibitors

LAG3: 

lymphocyte-activation gene 3

LSECs: 

liver sinusoidal endothelial cells

MDSCs: 

myeloid-derived suppressor cells

MHC: 

major histocompatibility

NK: 

natural killer

OS: 

overall survival

PD-1: 

programmed cell death protein-1

PD-L1: 

programmed cell death-ligand 1

RFA: 

radiofrequency ablation

SPEAR: 

specific peptide enhanced affinity receptor

TAAs: 

tumor-associated antigens

TACE: 

trans-arterial chemoembolization

TAMs: 

tumor-associated macrophages

TCR: 

T cell receptor

TILs: 

tumor-infiltrating lymphocytes

TIM3: 

T cell immunoglobulin, and mucin domain containing-3

TKI: 

tyrosine kinase inhibitor

TMB: 

tumor mutational burden

TME: 

tumor microenvironment

Tregs: 

regulatory T cells

TSAs: 

tumor-specific antigens

VEGF: 

vascular endothelial growth factor

Declarations

Funding

This study was supported by the Natural Science Foundation of Gansu Province (21JR7RA374, 20JR10RA674), Gansu Provincial Major Science and Technology Project(23ZDWA003), the Provincial-level special funds for military-civilian integration development from the Department of Finance of Gansu Province, the First Hospital of Lanzhou University Intramural Fund (ldyyyn2020-20, ldyyyn2019-45), and the National Natural Science Foundation of China (82060119).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

HW and XL designed the study and HW drafted the manuscript. HW and CD reviewed and edited the manuscript. All authors revised and approved the final manuscript.

References

  1. Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol 2023;79(2):516-537 View Article PubMed/NCBI
  2. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019;144(8):1941-1953 View Article PubMed/NCBI
  3. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2016;14(2):16018 View Article PubMed/NCBI
  4. Galle PR, Forner A, Llovet JM, Mazzaferro V, Piscaglia F, Raoul J-L, et al. EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol 2018;69(1):182-236 View Article PubMed/NCBI
  5. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 2007;25:267-296 View Article PubMed/NCBI
  6. Raskov H, Orhan A, Christensen JP, Gogenur I. Cytotoxic CD8(+) T cells in cancer and cancer immunotherapy. Br J Cancer 2021;124(2):359-367 View Article PubMed/NCBI
  7. Cai L, Michelakos T, Yamada T, Fan S, Wang X, Schwab JH, et al. Defective HLA class I antigen processing machinery in cancer. Cancer Immunol Immunother 2018;67(6):999-1009 View Article PubMed/NCBI
  8. Velcheti V, Schalper K. Basic Overview of Current Immunotherapy Approaches in Cancer. Am Soc Clin Oncol Educ Book 2016;35:298-308 View Article PubMed/NCBI
  9. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013;13(4):227-242 View Article PubMed/NCBI
  10. He X, Xu C. Immune checkpoint signaling and cancer immunotherapy. Cell Res 2020;30(8):660-669 View Article PubMed/NCBI
  11. Jain N, Nguyen H, Chambers C, Kang J. Dual function of CTLA-4 in regulatory T cells and conventional T cells to prevent multiorgan autoimmunity. Proc Natl Acad Sci U S A 2010;107(4):1524-1528 View Article PubMed/NCBI
  12. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359(6382):1350-1355 View Article PubMed/NCBI
  13. Lai Q, Avolio AW, Lerut J, Singh G, Chan SC, Berloco PB, et al. Recurrence of hepatocellular cancer after liver transplantation: the role of primary resection and salvage transplantation in East and West. J Hepatol 2012;57(5):974-979 View Article PubMed/NCBI
  14. Cheng AL, Hsu C, Chan SL, Choo SP, Kudo M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol 2020;72(2):307-319 View Article PubMed/NCBI
  15. Federico P, Petrillo A, Giordano P, Bosso D, Fabbrocini A, Ottaviano M, et al. Immune Checkpoint Inhibitors in Hepatocellular Carcinoma: Current Status and Novel Perspectives. Cancers (Basel) 2020;12(10):3025-3025 View Article PubMed/NCBI
  16. Kole C, Charalampakis N, Tsakatikas S, Vailas M, Moris D, Gkotsis E, et al. Immunotherapy for Hepatocellular Carcinoma: A 2021 Update. Cancers (Basel) 2020;12(10):2859-2859 View Article PubMed/NCBI
  17. Sangro B, Sarobe P, Hervas-Stubbs S, Melero I. Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2021;18(8):525-543 View Article PubMed/NCBI
  18. Lv Y, Wang Z, Yuan K. Role of Noncoding RNAs in the Tumor Immune Microenvironment of Hepatocellular Carcinoma. J Clin Transl Hepatol 2023;11(3):682-694 View Article PubMed/NCBI
  19. Yarchoan M, Johnson Iii BA, Lutz ER, Laheru DA, Jaffee EM. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer 2017;17(4):209-222 View Article PubMed/NCBI
  20. Łuksza M, Riaz N, Makarov V, Balachandran VP, Hellmann MD, Solovyov A, et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017;551(7681):517-520 View Article PubMed/NCBI
  21. Efremova M, Finotello F, Rieder D, Trajanoski Z. Neoantigens generated by individual mutations and their role in cancer immunity and immunotherapy. Front Immunol 2017;8:1679-1679 View Article PubMed/NCBI
  22. Malekzadeh P, Yossef R, Cafri G, Paria BC, Lowery FJ, Jafferji M, et al. Antigen Experienced T Cells from Peripheral Blood Recognize p53 Neoantigens. Clin Cancer Res 2020;26(6):1267-1276 View Article PubMed/NCBI
  23. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SAJR, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature 2013;500(7463):415-421 View Article PubMed/NCBI
  24. Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol 2019;30(1):44-56 View Article PubMed/NCBI
  25. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348(6230):69-74 View Article PubMed/NCBI
  26. Fujimoto A, Furuta M, Totoki Y, Tsunoda T, Kato M, Shiraishi Y, et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat Genet 2016;48(5):500-509 View Article PubMed/NCBI
  27. Nishida N, Kudo M. Immunological Microenvironment of Hepatocellular Carcinoma and Its Clinical Implication. Oncology 2017;92(Suppl 1):40-49 View Article PubMed/NCBI
  28. Tarin D. Causes of Cancer and Mechanisms of Carcinogenesis. Berlin: Springer; 2023, 229-279 View Article PubMed/NCBI
  29. Crispe IN. Hepatic T cells and liver tolerance. Nat Rev Immunol 2003;3(1):51-62 View Article PubMed/NCBI
  30. Liu Z, Zhang Y, Shi C, Zhou X, Xu K, Jiao D, et al. A novel immune classification reveals distinct immune escape mechanism and genomic alterations: implications for immunotherapy in hepatocellular carcinoma. J Transl Med 2021;19(1):5 View Article PubMed/NCBI
  31. Fu Y, Liu S, Zeng S, Shen H. From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma. J Exp Clin Cancer Res 2019;38(1):396 View Article PubMed/NCBI
  32. Kurebayashi Y, Ojima H, Tsujikawa H, Kubota N, Maehara J, Abe Y, et al. Landscape of immune microenvironment in hepatocellular carcinoma and its additional impact on histological and molecular classification. Hepatology 2018;68(3):1025-1041 View Article PubMed/NCBI
  33. Kakumu S, Ito S, Ishikawa T, Mita Y, Tagaya T, Fukuzawa Y, et al. Decreased function of peripheral blood dendritic cells in patients with hepatocellular carcinoma with hepatitis B and C virus infection. J Gastroenterol Hepatol 2000;15(4):431-436 View Article PubMed/NCBI
  34. Chen J, Gingold JA, Su XJ. Immunomodulatory TGF-β signaling in hepatocellular carcinoma. Trends Mol Med 2019;25(11):1010-1023 View Article PubMed/NCBI
  35. Kim IS, Yang WS, Kim CH. Beneficial Effects of Soybean-Derived Bioactive Peptides. Int J Mol Sci 2021;22(16):8570-8570 View Article PubMed/NCBI
  36. Stewart KL, Lephart ED. Overview of BPH: Symptom Relief with Dietary Polyphenols, Vitamins and Phytochemicals by Nutraceutical Supplements with Implications to the Prostate Microbiome. Int J Mol Sci 2023;24(6):5486 View Article PubMed/NCBI
  37. Ebrahimkhani MR, Mohar I, Crispe IN. Cross-presentation of antigen by diverse subsets of murine liver cells. Hepatology 2011;54(4):1379-1387 View Article PubMed/NCBI
  38. Krenkel O, Tacke F. Liver macrophages in tissue homeostasis and disease. Nat Rev Immunol 2017;17(5):306-321 View Article PubMed/NCBI
  39. Ormandy LA, Hillemann T, Wedemeyer H, Manns MP, Greten TF, Korangy F. Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res 2005;65(6):2457-2464 View Article PubMed/NCBI
  40. Höchst B, Schildberg FA, Sauerborn P, Gäbel YA, Gevensleben H, Goltz D, et al. Activated human hepatic stellate cells induce myeloid derived suppressor cells from peripheral blood monocytes in a CD44-dependent fashion. J Hepatol 2013;59(3):528-535 View Article PubMed/NCBI
  41. Yu MC, Chen CH, Liang X, Wang L, Gandhi CR, Fung JJ, et al. Inhibition of T-cell responses by hepatic stellate cells via B7-H1–mediated T-cell apoptosis in mice. Hepatology 2004;40(6):1312-1321 View Article PubMed/NCBI
  42. Dunham RM, Thapa M, Velazquez VM, Elrod EJ, Denning TL, Pulendran B, et al. Hepatic stellate cells preferentially induce Foxp3+ regulatory T cells by production of retinoic acid. J Immunol 2013;190(5):2009-2016 View Article PubMed/NCBI
  43. Cariani E, Pilli M, Barili V, Porro E, Biasini E, Olivani A, et al. Natural killer cells phenotypic characterization as an outcome predictor of HCV-linked HCC after curative treatments. Oncoimmunology 2016;5(8):e1154249-e1154249 View Article PubMed/NCBI
  44. Cariani E, Pilli M, Zerbini A, Rota C, Olivani A, Zanelli P, et al. HLA and Killer Immunoglobulin-like Receptor Genes as Outcome Predictors of Hepatitis C Virus–Related Hepatocellular CarcinomaKIR and HLA Loci in HCV-Related Hepatocellular Carcinoma. Clin Cancer Res 2013;19(19):5465-5473 View Article PubMed/NCBI
  45. Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 2009;50(3):799-807 View Article PubMed/NCBI
  46. Zhang QF, Yin WW, Xia Y, Yi YY, He QF, Wang X, et al. Liver-infiltrating CD11b(-)CD27(-) NK subsets account for NK-cell dysfunction in patients with hepatocellular carcinoma and are associated with tumor progression. Cell Mol Immunol 2017;14(10):819-829 View Article PubMed/NCBI
  47. Chulpanova DS, Rizvanov AA, Solovyeva VV. The Role of Cancer Stem Cells and Their Extracellular Vesicles in the Modulation of the Antitumor Immunity. Int J Mol Sci 2022;24(1):395 View Article PubMed/NCBI
  48. Tsuchiya N, Sawada Y, Endo I, Uemura Y, Nakatsura T. Potentiality of immunotherapy against hepatocellular carcinoma. World J Gastroenterol 2015;21(36):10314-10326 View Article PubMed/NCBI
  49. van Weverwijk A, de Visser KE. Mechanisms driving the immunoregulatory function of cancer cells. Nat Rev Cancer 2023;23(4):193-215 View Article PubMed/NCBI
  50. Fujiwara K, Higashi T, Nouso K, Nakatsukasa H, Kobayashi Y, Uemura M, et al. Decreased expression of B7 costimulatory molecules and major histocompatibility complex class-I in human hepatocellular carcinoma. J Gastroenterol Hepatol 2004;19(10):1121-1127 View Article PubMed/NCBI
  51. Arihara F, Mizukoshi E, Kitahara M, Takata Y, Arai K, Yamashita T, et al. Increase in CD14+HLA-DR -/low myeloid-derived suppressor cells in hepatocellular carcinoma patients and its impact on prognosis. Cancer Immunol Immunother 2013;62(8):1421-1430 View Article PubMed/NCBI
  52. Chen Z, Shen S, Peng B, Tao J. Intratumoural GM-CSF microspheres and CTLA-4 blockade enhance the antitumour immunity induced by thermal ablation in a subcutaneous murine hepatoma model. Int J Hyperthermia 2009;25(5):374-382 View Article PubMed/NCBI
  53. Alisa A, Ives A, Pathan AA, Navarrete CV, Williams R, Bertoletti A, et al. Analysis of CD4+ T-cell responses to a novel α-fetoprotein-derived epitope in hepatocellular carcinoma patients. Clin Cancer Res 2005;11(18):6686-6694 View Article PubMed/NCBI
  54. Crispe IN, Giannandrea M, Klein I, John B, Sampson B, Wuensch S. Cellular and molecular mechanisms of liver tolerance. Immunol Rev 2006;213(1):101-118 View Article PubMed/NCBI
  55. Chen K-J, Lin S-Z, Zhou L, Xie H-Y, Zhou W-H, Taki-Eldin A, et al. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS One 2011;6(9):e24671 View Article PubMed/NCBI
  56. Han Y, Chen Z, Yang Y, Jiang Z, Gu Y, Liu Y, et al. Human CD14+ CTLA-4+ regulatory dendritic cells suppress T-cell response by cytotoxic T-lymphocyte antigen-4-dependent IL-10 and indoleamine-2, 3-dioxygenase production in hepatocellular carcinoma. Hepatology 2014;59(2):567-579 View Article PubMed/NCBI
  57. Budhu A, Forgues M, Ye QH, Jia HL, He P, Zanetti KA, et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell 2006;10(2):99-111 View Article PubMed/NCBI
  58. Buonaguro L, Petrizzo A, Tagliamonte M, Tornesello ML, Buonaguro FM. Challenges in cancer vaccine development for hepatocellular carcinoma. J Hepatol 2013;59(4):897-903 View Article PubMed/NCBI
  59. Buonaguro FM, Buonaguro L. Cancer vaccines for hepatocellular carcinoma: future directions. Immunotherapy 2016;8(4):391-393 View Article PubMed/NCBI
  60. Jenne CN, Kubes P. Immune surveillance by the liver. Nat Immunol 2013;14(10):996-1006 View Article PubMed/NCBI
  61. Xiang Z, Wu J, Li J, Zheng S, Wei X, Xu X. Gut microbiota modulation: a viable strategy to address medical needs in hepatocellular carcinoma and liver transplantation. Engineering 2023 View Article PubMed/NCBI
  62. Doherty DG. Immunity, tolerance and autoimmunity in the liver: A comprehensive review. J Autoimmun 2016;66:60-75 View Article PubMed/NCBI
  63. Neuzillet C, Rousseau B, Kocher H, Bourget P, Tournigand C. Unravelling the pharmacologic opportunities and future directions for targeted therapies in gastro-intestinal cancers Part 1: GI carcinomas. Pharmacol Ther 2017;174:145-172 View Article PubMed/NCBI
  64. Fu J, Xu D, Liu Z, Shi M, Zhao P, Fu B, et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 2007;132(7):2328-2339 View Article PubMed/NCBI
  65. Trehanpati N, Vyas AK. Immune regulation by T regulatory cells in hepatitis B virus-related inflammation and cancer. Scand J Immunol 2017;85(3):175-181 View Article PubMed/NCBI
  66. Wang BJ, Bao JJ, Wang JZ, Wang Y, Jiang M, Xing MY, et al. Immunostaining of PD-1/PD-Ls in liver tissues of patients with hepatitis and hepatocellular carcinoma. World J Gastroenterol 2011;17(28):3322-3329 View Article PubMed/NCBI
  67. Shrestha R, Prithviraj P, Anaka M, Bridle KR, Crawford DHG, Dhungel B, et al. Monitoring immune checkpoint regulators as predictive biomarkers in hepatocellular carcinoma. Front Oncol 2018;8:269-269 View Article PubMed/NCBI
  68. Zeng Z, Shi F, Zhou L, Zhang M-N, Chen Y, Chang X-J, et al. Upregulation of circulating PD-L1/PD-1 is associated with poor post-cryoablation prognosis in patients with HBV-related hepatocellular carcinoma. PloS one 2011;6(9):e23621 View Article PubMed/NCBI
  69. Langhans B, Nischalke HD, Kramer B, Dold L, Lutz P, Mohr R, et al. Role of regulatory T cells and checkpoint inhibition in hepatocellular carcinoma. Cancer Immunol Immunother 2019;68(12):2055-2066 View Article PubMed/NCBI
  70. Rotte A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J Exp Clin Cancer Res 2019;38(1):255 View Article PubMed/NCBI
  71. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006;439(7077):682-687 View Article PubMed/NCBI
  72. Ren Z, Xu J, Bai Y, Xu A, Cang S, Du C, et al. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2-3 study. Lancet Oncol 2021;22(7):977-990 View Article PubMed/NCBI
  73. Qin S, Kudo M, Meyer T, Bai Y, Guo Y, Meng Z, et al. Tislelizumab vs Sorafenib as First-Line Treatment for Unresectable Hepatocellular Carcinoma: A Phase 3 Randomized Clinical Trial. JAMA Oncol 2023;9(12):1651-1659 View Article PubMed/NCBI
  74. Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2015;12(12):681-700 View Article PubMed/NCBI
  75. Yau T, Kang YK, Kim TY, El-Khoueiry AB, Santoro A, Sangro B, et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncol 2020;6(11):e204564 View Article PubMed/NCBI
  76. Yin S, Chen Z, Chen D, Yan D. Strategies targeting PD-L1 expression and associated opportunities for cancer combination therapy. Theranostics 2023;13(5):1520-1544 View Article PubMed/NCBI
  77. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol 2018;19(7):940-952 View Article PubMed/NCBI
  78. Brunnström H, Johansson A, Westbom-Fremer S, Backman M, Djureinovic D, Patthey A, et al. PD-L1 immunohistochemistry in clinical diagnostics of lung cancer: inter-pathologist variability is higher than assay variability. Mod Pathol 2017;30(10):1411-1421 View Article PubMed/NCBI
  79. Kaseb AO, Vence L, Blando J, Yadav SS, Ikoma N, Pestana RC, et al. Immunologic Correlates of Pathologic Complete Response to Preoperative Immunotherapy in Hepatocellular CarcinomaHCC with Complete Response after Immunotherapy. Cancer Immunol Res 2019;7(9):1390-1395 View Article PubMed/NCBI
  80. Büttner R, Longshore JW, López-Ríos F, Merkelbach-Bruse S, Normanno N, Rouleau E, et al. Implementing TMB measurement in clinical practice: considerations on assay requirements. ESMO Open 2019;4(1):e000442 View Article PubMed/NCBI
  81. Xie F, Bai Y, Yang X, Long J, Mao J, Lin J, et al. Comprehensive analysis of tumour mutation burden and the immune microenvironment in hepatocellular carcinoma. Int Immunopharmacol 2020;89(Pt A):107135 View Article PubMed/NCBI
  82. Ang C, Miura JT, Gamblin TC, He R, Xiu J, Millis SZ, et al. Comprehensive multiplatform biomarker analysis of 350 hepatocellular carcinomas identifies potential novel therapeutic options. J Surg Oncol 2016;113(1):55-61 View Article PubMed/NCBI
  83. Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The Immune Landscape of Cancer. Immunity 2018;48(4):812-830.e814 View Article PubMed/NCBI
  84. Hoppenz P, Els-Heindl S, Beck-Sickinger AG. Peptide-Drug Conjugates and Their Targets in Advanced Cancer Therapies. Front Chem 2020;8:571 View Article PubMed/NCBI
  85. Pan M, Zhao H, Jin R, Leung PSC, Shuai Z. Targeting immune checkpoints in anti-neutrophil cytoplasmic antibodies associated vasculitis: the potential therapeutic targets in the future. Front Immunol 2023;14:1156212 View Article PubMed/NCBI
  86. Abreu TR, Fonseca NA, Goncalves N, Moreira JN. Current challenges and emerging opportunities of CAR-T cell therapies. J Control Release 2020;319:246-261 View Article PubMed/NCBI
  87. Li S, Yang F, Ren X. Immunotherapy for hepatocellular carcinoma. Drug Discov Ther 2015;9(5):363-71 View Article PubMed/NCBI
  88. Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol 2007;19(7):813-824 View Article PubMed/NCBI
  89. Wang J, Li J, Tang G, Tian Y, Su S, Li Y. Clinical outcomes and influencing factors of PD-1/PD-L1 in hepatocellular carcinoma. Oncol Lett 2021;21(4):279 View Article PubMed/NCBI
  90. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017;389(10088):2492-2502 View Article PubMed/NCBI
  91. Kudo M. Pembrolizumab for the Treatment of Hepatocellular Carcinoma. Liver Cancer 2019;8(3):143-154 View Article PubMed/NCBI
  92. Yau T, Park JW, Finn RS, Cheng AL, Mathurin P, Edeline J, et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol 2022;23(1):77-90 View Article PubMed/NCBI
  93. Sangro B, Chan SL, Meyer T, Reig M, El-Khoueiry A, Galle PR. Diagnosis and management of toxicities of immune checkpoint inhibitors in hepatocellular carcinoma. J Hepatol 2020;72(2):320-341 View Article PubMed/NCBI
  94. Ducreux M, Abou-Alfa G, Ren Z, Edeline J, Li Z, Assenat E, et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol 2021;23(1):77-90 View Article PubMed/NCBI
  95. Liu Y, Zheng P. Preserving the CTLA-4 Checkpoint for Safer and More Effective Cancer Immunotherapy. Trends Pharmacol Sci 2020;41(1):4-12 View Article PubMed/NCBI
  96. Graziani G, Tentori L, Navarra P. Ipilimumab: a novel immunostimulatory monoclonal antibody for the treatment of cancer. Pharmacol Res 2012;65(1):9-22 View Article PubMed/NCBI
  97. Furness AJ, Vargas FA, Peggs KS, Quezada SA. Impact of tumour microenvironment and Fc receptors on the activity of immunomodulatory antibodies. Trends Immunol 2014;35(7):290-298 View Article PubMed/NCBI
  98. Sangro B, Gomez-Martin C, de la Mata M, Inarrairaegui M, Garralda E, Barrera P, et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol 2013;59(1):81-88 View Article PubMed/NCBI
  99. Du X, Tang F, Liu M, Su J, Zhang Y, Wu W, et al. A reappraisal of CTLA-4 checkpoint blockade in cancer immunotherapy. Cell Res 2018;28(4):416-432 View Article PubMed/NCBI
  100. Gautron AS, Dominguez-Villar M, de Marcken M, Hafler DA. Enhanced suppressor function of TIM-3+ FoxP3+ regulatory T cells. European journal of immunology 2014;44(9):2703-2711 View Article PubMed/NCBI
  101. Wolf Y, Anderson AC, Kuchroo VK. TIM3 comes of age as an inhibitory receptor. Nat Rev Immunol 2020;20(3):173-185 View Article PubMed/NCBI
  102. Wang J, Sun J, Liu LN, Flies DB, Nie X, Toki M, et al. Siglec-15 as an immune suppressor and potential target for normalization cancer immunotherapy. Nat Med 2019;25(4):656-666 View Article PubMed/NCBI
  103. Liu W, Ji Z, Wu B, Huang S, Chen Q, Chen X, et al. Siglec-15 promotes the migration of liver cancer cells by repressing lysosomal degradation of CD44. FEBS Lett 2021;595(17):2290-2302 View Article PubMed/NCBI
  104. Zheng Q, Xu J, Gu X, Wu F, Deng J, Cai X, et al. Immune checkpoint targeting TIGIT in hepatocellular carcinoma. Am J Transl Res 2020;12(7):3212-3224 View Article PubMed/NCBI
  105. Yu X, Harden K, Gonzalez LC, Francesco M, Chiang E, Irving B, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol 2009;10(1):48-57 View Article PubMed/NCBI
  106. Galle PR, Finn RS, Qin S, Ikeda M, Zhu AX, Kim TY, et al. Patient-reported outcomes with atezolizumab plus bevacizumab versus sorafenib in patients with unresectable hepatocellular carcinoma (IMbrave150): an open-label, randomised, phase 3 trial. Lancet Oncol 2021;22(7):991-1001 View Article PubMed/NCBI
  107. Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, et al. KEYNOTE-240 investigators. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial. J Clin Oncol 2020;38(3):193-202 View Article PubMed/NCBI
  108. Cui J, Wang N, Zhao H, Jin H, Wang G, Niu C, et al. Combination of radiofrequency ablation and sequential cellular immunotherapy improves progression-free survival for patients with hepatocellular carcinoma. Int J Cancer 2014;134(2):342-351 View Article PubMed/NCBI
  109. Duffy AG, Ulahannan SV, Makorova-Rusher O, Rahma O, Wedemeyer H, Pratt D, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol 2017;66(3):545-551 View Article PubMed/NCBI
  110. Li H, Qin S, Liu Y, Chen Z, Ren Z, Xiong J, et al. Camrelizumab combined with FOLFOX4 regimen as first-line therapy for advanced hepatocellular carcinomas: a sub-cohort of a multicenter phase Ib/II study. Drug Des Devel Ther 2021;15:1873-1882 View Article PubMed/NCBI
  111. Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, et al. Plain language summary of the HIMALAYA study: tremelimumab and durvalumab for unresectable hepatocellular carcinoma (liver cancer). Future Oncol (London, England) 2023;19(38):2505-2516 View Article PubMed/NCBI
  112. Kelley RK, Sangro B, Harris W, Ikeda M, Okusaka T, Kang YK, et al. Safety, Efficacy, and Pharmacodynamics of Tremelimumab Plus Durvalumab for Patients With Unresectable Hepatocellular Carcinoma: Randomized Expansion of a Phase I/II Study. J Clin Oncol 2021;39(27):2991-3001 View Article PubMed/NCBI
  113. Hegde PS, Wallin JJ, Mancao C. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin Cancer Biol 2018;52(Pt 2):117-124 View Article PubMed/NCBI
  114. Motzer RJ, Robbins PB, Powles T, Albiges L, Haanen JB, Larkin J, et al. Avelumab plus axitinib versus sunitinib in advanced renal cell carcinoma: biomarker analysis of the phase 3 JAVELIN Renal 101 trial. Nature medicine 2020;26(11):1733-1741 View Article PubMed/NCBI
  115. Taylor MH, Lee CH, Makker V, Rasco D, Dutcus CE, Wu J, et al. Phase IB/II Trial of Lenvatinib Plus Pembrolizumab in Patients With Advanced Renal Cell Carcinoma, Endometrial Cancer, and Other Selected Advanced Solid Tumors. J Clin Oncol 2020;38(11):1154-1163 View Article PubMed/NCBI
  116. Sun LY, Zhang KJ, Xie YM, Liu JW, Xiao ZQ. Immunotherapies for advanced hepatocellular carcinoma. Front Pharmacol 2023;14:1138493 View Article PubMed/NCBI
  117. Finn RS, Ikeda M, Zhu AX, Sung MW, Baron AD, Kudo M, et al. Phase Ib Study of Lenvatinib Plus Pembrolizumab in Patients With Unresectable Hepatocellular Carcinoma. J Clin Oncol 2020;38(26):2960-2970 View Article PubMed/NCBI
  118. Gryziak M, Wozniak K, Kraj L, Rog L, Stec R. The immune landscape of hepatocellular carcinoma-where we are?. Oncol Lett 2022;24(5):1-13 View Article PubMed/NCBI
  119. Gao X, Zhao R, Ma H, Zuo S. Efficacy and safety of atezolizumab plus bevacizumab treatment for advanced hepatocellular carcinoma in the real world: a single-arm meta-analysis. BMC Cancer 2023;23(1):635 View Article PubMed/NCBI
  120. Foerster F, Gairing SJ, Ilyas SI, Galle PR. Emerging immunotherapy for HCC: A guide for hepatologists. Hepatology 2022;75(6):1604-1626 View Article PubMed/NCBI
  121. Kelley RK, Sangro B, Harris WP, Ikeda M, Okusaka T, Kang Y-K, et al. Efficacy, tolerability, and biologic activity of a novel regimen of tremelimumab (T) in combination with durvalumab (D) for patients (pts) with advanced hepatocellular carcinoma (aHCC). J Clin Oncol 2020;38:4508 View Article PubMed/NCBI
  122. Llovet JM, Kudo M, Cheng A-L, Finn RS, Galle PR, Kaneko S, et al. Lenvatinib (len) plus pembrolizumab (pembro) for the first-line treatment of patients (pts) with advanced hepatocellular carcinoma (HCC): Phase 3 LEAP-002 study. J Clin Oncol 2019;37:TPS4152 View Article PubMed/NCBI
  123. Xu J, Zhang Y, Jia R, Yue C, Chang L, Liu R, et al. Anti-PD-1 Antibody SHR-1210 Combined with Apatinib for Advanced Hepatocellular Carcinoma, Gastric, or Esophagogastric Junction Cancer: An Open-label, Dose Escalation and Expansion Study. Clin Cancer Res 2019;25(2):515-523 View Article PubMed/NCBI
  124. Yang J, Guo Z, Song M, Pan Q, Zhao J, Huang Y, et al. Lenvatinib improves anti-PD-1 therapeutic efficacy by promoting vascular normalization via the NRP-1-PDGFRbeta complex in hepatocellular carcinoma. Front Immunol 2023;14:1212577 View Article PubMed/NCBI
  125. Stefanini B, Ielasi L, Chen R, Abbati C, Tonnini M, Tovoli F, et al. TKIs in combination with immunotherapy for hepatocellular carcinoma. Expert review of anticancer therapy 2023;23(3):279-291 View Article PubMed/NCBI
  126. Xie D, Sun Q, Wang X, Zhou J, Fan J, Ren Z, et al. Immune checkpoint inhibitor plus tyrosine kinase inhibitor for unresectable hepatocellular carcinoma in the real world. Annals of translational medicine 2021;9(8):652 View Article PubMed/NCBI
  127. Li H, Su K, Guo L, Jiang Y, Xu K, Gu T, et al. PD-1 Inhibitors Combined with Antiangiogenic Therapy with or Without Transarterial Chemoembolization in the Treatment of Hepatocellular Carcinoma: A Propensity Matching Analysis. Journal of hepatocellular carcinoma 2023;10:1257-1266 View Article PubMed/NCBI
  128. Alvarnas JC, Linn YC, Hope EG, Negrin RS. Expansion of cytotoxic CD3+ CD56+ cells from peripheral blood progenitor cells of patients undergoing autologous hematopoietic cell transplantation. Biol Blood Marrow Transplant 2001;7(4):216-222 View Article PubMed/NCBI
  129. Roddy H, Meyer T, Roddie C. Novel Cellular Therapies for Hepatocellular Carcinoma. Cancers (Basel) 2022;14(3):504-504 View Article PubMed/NCBI
  130. Nishimura R, Baker J, Beilhack A, Zeiser R, Olson JA, Sega EI, et al. In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood 2008;112(6):2563-2574 View Article PubMed/NCBI
  131. Cao J, Kong FH, Liu X, Wang XB. Immunotherapy with dendritic cells and cytokine-induced killer cells for hepatocellular carcinoma: A meta-analysis. World J Gastroenterol 2019;25(27):3649-3663 View Article PubMed/NCBI
  132. Li YC, Zhao L, Wu JP, Qu CX, Song QK, Wang RB. Cytokine-induced killer cell infusion combined with conventional treatments produced better prognosis for hepatocellular carcinoma patients with barcelona clinic liver cancer B or earlier stage: A systematic review and meta-analysis. Cytotherapy 2016;18(12):1525-1531 View Article PubMed/NCBI
  133. Shi M, Zhang B, Tang Z-R, Lei Z-Y, Wang H-F, Feng Y-Y, et al. Autologous cytokine-induced killer cell therapy in clinical trial phase I is safe in patients with primary hepatocellular carcinoma. World J Gastroenterol 2004;10(8):1146 View Article PubMed/NCBI
  134. Olioso P, Giancola R, Di Riti M, Contento A, Accorsi P, Iacone A. Immunotherapy with cytokine induced killer cells in solid and hematopoietic tumours: a pilot clinical trial. Hematol Oncol 2009;27(3):130-139 View Article PubMed/NCBI
  135. Hao MZ, Lin HL, Chen Q, Ye YB, Chen QZ, Chen MS. Efficacy of transcatheter arterial chemoembolization combined with cytokine-induced killer cell therapy on hepatocellular carcinoma: a comparative study. Chin J Cancer 2010;29(2):172-177 View Article PubMed/NCBI
  136. Chen F, Yang M, Song Q, Wu J, Wang X, Zhou X, et al. Enhanced antitumor effects and improved immune status of dendritic cell and cytokine-induced killer cell infusion in advanced cancer patients. Mol Clin Oncol 2017;7(5):903-910 View Article PubMed/NCBI
  137. Mosińska P, Gabryelska A, Zasada M, Fichna J. Dual functional capability of dendritic cells–cytokine-induced killer cells in improving side effects of colorectal cancer therapy. Front Pharmacol 2017;8:126 View Article PubMed/NCBI
  138. Zhang D, He JT. In vitro cytotox icity effects of cocultured DC-C IK cells combined with sorafenib against hepa to cellular carcinoma. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2011;27(6):664-667 View Article PubMed/NCBI
  139. Besser MJ, Shapira-Frommer R, Itzhaki O, Treves AJ, Zippel DB, Levy D, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: Intent-to-Treat analysis and efficacy after failure to prior immunotherapiesintent-to-Treat analysis of til act and impact of ipilimumab. Clin Cancer Res 2013;19(17):4792-4800 View Article PubMed/NCBI
  140. Monberg TJ, Borch TH, Svane IM, Donia M. TIL Therapy: Facts and Hopes. Clin Cancer Res 2023;29(17):3275-3283 View Article PubMed/NCBI
  141. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351(6280):1463-1469 View Article PubMed/NCBI
  142. Gao F, Xie K, Xiang Q, Qin Y, Chen P, Wan H, et al. The density of tumor-infiltrating lymphocytes and prognosis in resectable hepatocellular carcinoma: a two-phase study. Aging (Albany NY) 2021;13(7):9665-9678 View Article PubMed/NCBI
  143. Ge Z, Zhou G, Campos Carrascosa L, Gausvik E, Boor PPC, Noordam L, et al. TIGIT and PD1 Co-blockade Restores ex vivo Functions of Human Tumor-Infiltrating CD8(+) T Cells in Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol 2021;12(2):443-464 View Article PubMed/NCBI
  144. Liu Q, Li J, Zheng H, Yang S, Hua Y, Huang N, et al. Adoptive cellular immunotherapy for solid neoplasms beyond CAR-T. Mol Cancer 2023;22(1):28 View Article PubMed/NCBI
  145. Davenport AJ, Jenkins MR, Cross RS, Yong CS, Prince HM, Ritchie DS, et al. CAR-T Cells Inflict Sequential Killing of Multiple Tumor Target Cells. Cancer Immunol Res 2015;3(5):483-494 View Article PubMed/NCBI
  146. Illán Cortadelles C. Genotype of immune response inhibitory molecules as predictors of clinical outcome after CAR T-cell therapy: a prospective-multicenter cohort study. 2023. Available from: https://dugi-doc.udg.edu/handle/10256/23049 View Article PubMed/NCBI
  147. Davenport AJ, Jenkins MR, Cross RS, Yong CS, Prince HM, Ritchie DS, et al. CAR-T Cells Inflict Sequential Killing of Multiple Tumor Target Cells. Cancer Immunol Res 2015;3(5):483-494 View Article PubMed/NCBI
  148. Brocker T, Karjalainen K. Signals through T cell receptor-zeta chain alone are insufficient to prime resting T lymphocytes. J Exp Med 1995;181(5):1653-1659 View Article PubMed/NCBI
  149. Gong MC, Latouche JB, Krause A, Heston WD, Bander NH, Sadelain M. Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen. Neoplasia 1999;1(2):123-127 View Article PubMed/NCBI
  150. Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, Geiger TL, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 2004;18(4):676-684 View Article PubMed/NCBI
  151. Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor. Nat Biotechnol 2002;20(1):70-75 View Article PubMed/NCBI
  152. Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer 2016;16(9):566-581 View Article PubMed/NCBI
  153. Ghorashian S, Pule M, Amrolia P. CD19 chimeric antigen receptor T cell therapy for haematological malignancies. British journal of haematology 2015;169(4):463-478 View Article PubMed/NCBI
  154. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371(16):1507-1517 View Article PubMed/NCBI
  155. Roddie C, Dias J, O’Reilly MA, Abbasian M, Cadinanos-Garai A, Vispute K, et al. Durable Responses and Low Toxicity After Fast Off-Rate CD19 Chimeric Antigen Receptor-T Therapy in Adults With Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia. J Clin Oncol 2021;39(30):3352-3363 View Article PubMed/NCBI
  156. Nakatsura T, Yoshitake Y, Senju S, Monji M, Komori H, Motomura Y, et al. Glypican-3, overexpressed specifically in human hepatocellular carcinoma, is a novel tumor marker. Biochem Biophys Res Commun 2003;306(1):16-25 View Article PubMed/NCBI
  157. Zhou F, Shang W, Yu X, Tian J. Glypican-3: A promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev 2018;38(2):741-767 View Article PubMed/NCBI
  158. Capurro M, Martin T, Shi W, Filmus J. Glypican-3 binds to Frizzled and plays a direct role in the stimulation of canonical Wnt signaling. J Cell Sci 2014;127(Pt 7):1565-1575 View Article PubMed/NCBI
  159. Capurro M, Wanless IR, Sherman M, Deboer G, Shi W, Miyoshi E, et al. Glypican-3: a novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology 2003;125(1):89-97 View Article PubMed/NCBI
  160. Aviel-Ronen S, Lau SK, Pintilie M, Lau D, Liu N, Tsao MS, et al. Glypican-3 is overexpressed in lung squamous cell carcinoma, but not in adenocarcinoma. Mod Pathol 2008;21(7):817-825 View Article PubMed/NCBI
  161. Stadlmann S, Gueth U, Baumhoer D, Moch H, Terracciano L, Singer G. Glypican-3 expression in primary and recurrent ovarian carcinomas. Int J Gynecol Pathol 2007;26(3):341-344 View Article PubMed/NCBI
  162. Li W, Guo L, Rathi P, Marinova E, Gao X, Wu MF, et al. Redirecting T Cells to Glypican-3 with 4-1BB Zeta Chimeric Antigen Receptors Results in Th1 Polarization and Potent Antitumor Activity. Hum Gene Ther 2017;28(5):437-448 View Article PubMed/NCBI
  163. Phung Y, Gao W, Man Y-G, Nagata S, Ho M. High-affinity monoclonal antibodies to cell surface tumor antigen glypican-3 generated through a combination of peptide immunization and flow cytometry screening. MAbs 2012;4(5):592-9 View Article PubMed/NCBI
  164. Montalbano M, Rastellini C, McGuire JT, Prajapati J, Shirafkan A, Vento R, et al. Role of Glypican-3 in the growth, migration and invasion of primary hepatocytes isolated from patients with hepatocellular carcinoma. Cell Oncol (Dordr) 2018;41(2):169-184 View Article PubMed/NCBI
  165. Shi D, Shi Y, Kaseb AO, Qi X, Zhang Y, Chi J, et al. Chimeric Antigen Receptor-Glypican-3 T-Cell Therapy for Advanced Hepatocellular Carcinoma: Results of Phase I TrialsPhase I Trials of CAR-GPC3 T Cells for Advanced HCC. Clin Cancer Res 2020;26(15):3979-3989 View Article PubMed/NCBI
  166. Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nature medicine 2018;24(5):563-571 View Article PubMed/NCBI
  167. Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016;30(2):492-500 View Article PubMed/NCBI
  168. Roddie C, O’Reilly MA, Marzolini MAV, Wood L, Pinto JDA, Abbasian M, et al. Automated manufacture of matched donor-derived allogeneic CD19 CAR T-cells for relapsed/refractory B-ALL following allogeneic stem cell transplantation: toxicity, efficacy and the important role of lymphodepletion. Blood 2019;134(9):776-781 View Article PubMed/NCBI
  169. Steffin DHM, Batra SA, Rathi P, Guo L, Li W, Courtney AN, et al. A phase I clinical trial using armored GPC3 CAR T cells for children with relapsed/refractory liver tumors. J Clin Oncol 2019;37:TPS2647 View Article PubMed/NCBI
  170. Sun L, Gao F, Gao Z, Ao L, Li N, Ma S, et al. Shed antigen-induced blocking effect on CAR-T cells targeting Glypican-3 in Hepatocellular Carcinoma. J Immunother Cancer 2021;9(4):e001875 View Article PubMed/NCBI
  171. Sideras K, Bots SJ, Biermann K, Sprengers D, Polak WG, JN IJ, et al. Tumour antigen expression in hepatocellular carcinoma in a low-endemic western area. Br J Cancer 2015;112(12):1911-1920 View Article PubMed/NCBI
  172. Zhang L, He T, Cui H, Wang Y, Huang C, Han F. Effects of AFP gene silencing on apoptosis and proliferation of a hepatocellular carcinoma cell line. Discov Med 2012;14(75):115-124 View Article PubMed/NCBI
  173. Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. Structure of the human class I histocompatibility antigen, HLA-A2. Nature 1987;329(6139):506-512 View Article PubMed/NCBI
  174. Huang J, Brameshuber M, Zeng X, Xie J, Li QJ, Chien YH, et al. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4(+) T cells. Immunity 2013;39(5):846-857 View Article PubMed/NCBI
  175. Teschner AC. Use of TCR antibody fusion proteins as bispecific agents for NK and T cell-mediated immunotherapy [Dissertation]. Heidelberg: Heidelberg University; 2023 View Article PubMed/NCBI
  176. Li D, Li X, Zhou WL, Huang Y, Liang X, Jiang L, et al. Genetically engineered T cells for cancer immunotherapy. Signal Transduct Target Ther 2019;4(1):35 View Article PubMed/NCBI
  177. Docta RY, Ferronha T, Sanderson JP, Weissensteiner T, Pope GR, Bennett AD, et al. Tuning T-Cell Receptor Affinity to Optimize Clinical Risk-Benefit When Targeting Alpha-Fetoprotein–Positive Liver Cancer. Hepatology 2019;69(5):2061-2075 View Article PubMed/NCBI
  178. Goyal L, Frigault M, Meyer T, Feun LG, Bruix J, El-Khoueiry A, et al. Initial safety of AFP SPEAR T-cells in patients with advanced hepatocellular carcinoma. Cancer Res 2019;79(13_Supplement):3183 View Article PubMed/NCBI
  179. Butterfield LH, Ribas A, Meng WS, Dissette VB, Amarnani S, Vu HT, et al. T-cell responses to HLA-A* 0201 immunodominant peptides derived from α-fetoprotein in patients with hepatocellular cancer. Clin Cancer Res 2003;9(16):5902-5908 View Article PubMed/NCBI
  180. Want MY, Bashir Z, Najar RA. T Cell Based Immunotherapy for Cancer: Approaches and Strategies. Vaccines (Basel) 2023;11(4):835-835 View Article PubMed/NCBI
  181. Costante F, Stella L, Santopaolo F, Gasbarrini A, Pompili M, Asselah T, et al. Molecular and Clinical Features of Hepatocellular Carcinoma in Patients with HBV-HDV Infection. J Hepatocell Carcinoma 2023;10:713-724 View Article PubMed/NCBI
  182. Wang SH, Yeh SH, Chen PJ. Unique Features of Hepatitis B Virus-Related Hepatocellular Carcinoma in Pathogenesis and Clinical Significance. Cancers (Basel) 2021;13(10):2454 View Article PubMed/NCBI
  183. Qasim W, Brunetto M, Gehring AJ, Xue SA, Schurich A, Khakpoor A, et al. Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. J Hepatol 2015;62(2):486-491 View Article PubMed/NCBI
  184. Gehring AJ, Xue S-A, Ho ZZ, Teoh D, Ruedl C, Chia A, et al. Engineering virus-specific T cells that target HBV infected hepatocytes and hepatocellular carcinoma cell lines. J Hepatol 2011;55(1):103-110 View Article PubMed/NCBI
  185. Faria LC, Gigou M, Roque–Afonso AM, Sebagh M, Roche B, Fallot G, et al. Hepatocellular carcinoma is associated with an increased risk of hepatitis B virus recurrence after liver transplantation. Gastroenterology 2008;134(7):1890-1899 View Article PubMed/NCBI
  186. Hosseini SS, Khalili S, Baradaran B, Bidar N, Shahbazi MA, Mosafer J, et al. Bispecific monoclonal antibodies for targeted immunotherapy of solid tumors: Recent advances and clinical trials. Int J Biol Macromol 2021;167:1030-1047 View Article PubMed/NCBI
  187. Yao Y, Hu Y, Wang F. Trispecific antibodies for cancer immunotherapy. Immunology 2023;169(4):389-399 View Article PubMed/NCBI
  188. Hoseini SS, Cheung NV. Immunotherapy of hepatocellular carcinoma using chimeric antigen receptors and bispecific antibodies. Cancer Lett 2017;399:44-52 View Article PubMed/NCBI
  189. Hoh A, Dewerth A, Vogt F, Wenz J, Baeuerle PA, Warmann SW, et al. The activity of γδ T cells against paediatric liver tumour cells and spheroids in cell culture. Liver Int 2013;33(1):127-136 View Article PubMed/NCBI
  190. Mayer K, Baumann A-L, Grote M, Seeber S, Kettenberger H, Breuer S, et al. TriFabs—trivalent IgG-shaped bispecific antibody derivatives: design, generation, characterization and application for targeted payload delivery. Int J Mol Sci 2015;16(11):27497-27507 View Article PubMed/NCBI
  191. Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 2019;4(1):7 View Article PubMed/NCBI
  192. Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023;355:760-778 View Article PubMed/NCBI
  193. Charneau J, Suzuki T, Shimomura M, Fujinami N, Nakatsura T. Peptide-Based Vaccines for Hepatocellular Carcinoma: A Review of Recent Advances. J Hepatocell Carcinoma 2021;8:1035-1054 View Article PubMed/NCBI
  194. Nakajima M, Hazama S, Tokumitsu Y, Shindo Y, Matsui H, Matsukuma S, et al. A phase I study of a novel therapeutic vaccine as perioperative treatment for patients with surgically resectable hepatocellular carcinoma: the YCP02 trial. Hepatol Res 2023;53(7):649-660 View Article PubMed/NCBI
  195. Feola S, Chiaro J, Cerullo V. Integrating immunopeptidome analysis for the design and development of cancer vaccines. Semin Immunol 2023;67:101750 View Article PubMed/NCBI
  196. Sun TY, Yan W, Yang CM, Zhang LF, Tang HL, Chen Y, et al. Clinical research on dendritic cell vaccines to prevent postoperative recurrence and metastasis of liver cancer. Genetics and molecular research. GMR 2015;14(4):16222-16232 View Article PubMed/NCBI
  197. Zhang W, Liu J, Wu Y, Xiao F, Wang Y, Wang R, et al. Immunotherapy of hepatocellular carcinoma with a vaccine based on xenogeneic homologous α fetoprotein in mice. Biochem Biophys Res Commun 2008;376(1):10-14 View Article PubMed/NCBI
  198. Sawada Y, Yoshikawa T, Ofuji K, Yoshimura M, Tsuchiya N, Takahashi M, et al. Phase II study of the GPC3-derived peptide vaccine as an adjuvant therapy for hepatocellular carcinoma patients. Oncoimmunology 2016;5(5):e1129483 View Article PubMed/NCBI
  199. Yoshikawa T, Nakatsugawa M, Suzuki S, Shirakawa H, Nobuoka D, Sakemura N, et al. HLA-A2-restricted glypican-3 peptide-specific CTL clones induced by peptide vaccine show high avidity and antigen-specific killing activity against tumor cells. Cancer Sci 2011;102(5):918-925 View Article PubMed/NCBI
  200. Sawada Y, Yoshikawa T, Nobuoka D, Shirakawa H, Kuronuma T, Motomura Y, et al. Phase I Trial of a Glypican-3–Derived Peptide Vaccine for Advanced Hepatocellular Carcinoma: Immunologic Evidence and Potential for Improving Overall SurvivalPhase I Trial of GPC3 Peptide Vaccine in HCC Patients. Clin Cancer Res 2012;18(13):3686-3696 View Article PubMed/NCBI
  201. Grisaru-Tal S, Itan M, Grass DG, Torres-Roca J, Eschrich SA, Gordon Y, et al. Primary tumors from mucosal barrier organs drive unique eosinophil infiltration patterns and clinical associations. Oncoimmunology 2021;10(1):1859732 View Article PubMed/NCBI
  202. Takakura K, Kajihara M, Ito Z, Ohkusa T, Gong J, Koido S. Dendritic-tumor fusion cells in cancer immunotherapy. Discov Med 2015;19(104):169-174 View Article PubMed/NCBI
  203. Zhou X, Li C, Chen T, Li W, Wang X, Yang Q. Targeting RNA N6-methyladenosine to synergize with immune checkpoint therapy. Mol Cancer 2023;22(1):36 View Article PubMed/NCBI
  204. Zhao T, Jia H, Cheng Q, Xiao Y, Li M, Ren W, et al. Nifuroxazide prompts antitumor immune response of TCL-loaded DC in mice with orthotopically-implanted hepatocarcinoma. Oncol Rep 2017;37(6):3405-3414 View Article PubMed/NCBI
  205. Lee JH, Lee Y, Lee M, Heo MK, Song JS, Kim KH, et al. A phase I/IIa study of adjuvant immunotherapy with tumour antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Br J Cancer 2015;113(12):1666-1676 View Article PubMed/NCBI
  206. El Ansary M, Mogawer S, Elhamid SA, Alwakil S, Aboelkasem F, Sabaawy HE, et al. Immunotherapy by autologous dendritic cell vaccine in patients with advanced HCC. J Cancer Res Clin Oncol 2013;139(1):39-48 View Article PubMed/NCBI
  207. Bosi C, Rimini M, Casadei-Gardini A, Giorgio E. Understanding the causes of recurrent HCC after liver resection and radiofrequency ablation. Expert Rev Anticancer Ther 2023;23(5):503-515 View Article PubMed/NCBI
  208. Hack SP, Spahn J, Chen M, Cheng AL, Kaseb A, Kudo M, et al. IMbrave 050: a Phase III trial of atezolizumab plus bevacizumab in high-risk hepatocellular carcinoma after curative resection or ablation. Future Oncol (London, England) 2020;16(15):975-989 View Article PubMed/NCBI
  209. Knox J, Cheng A, Cleary S, Galle P, Kokudo N, Lencioni R, et al. A phase 3 study of durvalumab with or without bevacizumab as adjuvant therapy in patients with hepatocellular carcinoma at high risk of recurrence after curative hepatic resection or ablation: EMERALD-2. Ann Oncol 2019;30:iv59-60 View Article PubMed/NCBI
  210. Bruix J, Takayama T, Mazzaferro V, Chau GY, Yang J, Kudo M, et al. Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol 2015;16(13):1344-1354 View Article PubMed/NCBI
  211. Meyer T, Fox R, Ma YT, Ross PJ, James MW, Sturgess R, et al. Sorafenib in combination with transarterial chemoembolisation in patients with unresectable hepatocellular carcinoma (TACE 2): a randomised placebo-controlled, double-blind, phase 3 trial. Lancet Gastroenterol Hepatol 2017;2(8):565-575 View Article PubMed/NCBI
  212. Pinter M, Scheiner B, Peck-Radosavljevic M. Immunotherapy for advanced hepatocellular carcinoma: a focus on special subgroups. Gut 2021;70(1):204-214 View Article PubMed/NCBI
  213. Bahreyni A, Liu H, Mohamud Y, Xue YC, Fan YM, Zhang YL, et al. A combination of genetically engineered oncolytic virus and melittin-CpG for cancer viro-chemo-immunotherapy. BMC Med 2023;21(1):193 View Article PubMed/NCBI
  214. Bourke MG, Salwa S, Harrington KJ, Kucharczyk MJ, Forde PF, de Kruijf M, et al. The emerging role of viruses in the treatment of solid tumours. Cancer Treat Rev 2011;37(8):618-632 View Article PubMed/NCBI
  215. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol 2005;5(5):375-386 View Article PubMed/NCBI
  216. Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 2015;14(9):642-662 View Article PubMed/NCBI
  217. Schirrmacher V. Oncolytic Newcastle disease virus as a prospective anti-cancer therapy. A biologic agent with potential to break therapy resistance. Expert Opin Biol Ther 2015;15(12):1757-1771 View Article PubMed/NCBI
  218. Shalhout SZ, Miller DM, Emerick KS, Kaufman HL. Therapy with oncolytic viruses: progress and challenges. Nat Rev Clin Oncol 2023;20(3):160-177 View Article PubMed/NCBI
  219. Hemminki O, Dos Santos JM, Hemminki A. Oncolytic viruses for cancer immunotherapy. J Hematol Oncol 2020;13(1):84 View Article PubMed/NCBI
  220. Stanford MM, Bell JC, Vaha-Koskela MJ. Novel oncolytic viruses: riding high on the next wave?. Cytokine Growth Factor Rev 2010;21(2-3):177-183 View Article PubMed/NCBI
  221. Chiocca EA, Rabkin SD. Oncolytic viruses and their application to cancer immunotherapy. Cancer Immunol Res 2014;2(4):295-300 View Article PubMed/NCBI
  222. Zhang W, Ge K, Zhao Q, Zhuang X, Deng Z, Liu L, et al. A novel oHSV-1 targeting telomerase reverse transcriptase-positive cancer cells via tumor-specific promoters regulating the expression of ICP4. Oncotarget 2015;6(24):20345-20355 View Article PubMed/NCBI
  223. Breitbach CJ, Arulanandam R, De Silva N, Thorne SH, Patt R, Daneshmand M, et al. Oncolytic vaccinia virus disrupts tumor-associated vasculature in humans. Cancer Res 2013;73(4):1265-1275 View Article PubMed/NCBI
  224. Yoo SY, Jeong SN, Kang DH, Heo J. Evolutionary cancer-favoring engineered vaccinia virus for metastatic hepatocellular carcinoma. Oncotarget 2017;8(42):71489-71499 View Article PubMed/NCBI
  225. Parato KA, Breitbach CJ, Le Boeuf F, Wang J, Storbeck C, Ilkow C, et al. The oncolytic poxvirus JX-594 selectively replicates in and destroys cancer cells driven by genetic pathways commonly activated in cancers. Mol Ther 2012;20(4):749-758 View Article PubMed/NCBI
  226. Breitbach CJ, Moon A, Burke J, Hwang TH, Kirn DH. A Phase 2, Open-Label, Randomized Study of Pexa-Vec (JX-594) Administered by Intratumoral Injection in Patients with Unresectable Primary Hepatocellular Carcinoma. Methods Mol Biol 2015;1317:343-357 View Article PubMed/NCBI
  227. Wang J, Xu L, Zeng W, Hu P, Zeng M, Rabkin SD, et al. Treatment of human hepatocellular carcinoma by the oncolytic herpes simplex virus G47delta. Cancer Cell Int 2014;14(1):83 View Article PubMed/NCBI