Back to Journals » Journal of Hepatocellular Carcinoma » Volume 10

Mechanisms of Resistance to Immunotherapy in Hepatocellular Carcinoma

Authors Manfredi GF, Celsa C, John C, Jones C, Acuti N, Scheiner B, Fulgenzi CAM, Korolewicz J, Pinter M, Gennari A, Mauri FA, Pirisi M , Minisini R, Vincenzi F, Burlone M, Rigamonti C, Donadon M, Cabibbo G , D'Alessio A , Pinato DJ 

Received 6 June 2023

Accepted for publication 24 October 2023

Published 3 November 2023 Volume 2023:10 Pages 1955—1971

DOI https://doi.org/10.2147/JHC.S291553

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Ahmed Kaseb



Giulia Francesca Manfredi,1,2 Ciro Celsa,1,3,4 Chloe John,1 Charlotte Jones,1 Nicole Acuti,1 Bernhard Scheiner,5 Claudia Angela Maria Fulgenzi,1,6 James Korolewicz,1 Matthias Pinter,5 Alessandra Gennari,7 Francesco A Mauri,1 Mario Pirisi,2,8 Rosalba Minisini,2 Federica Vincenzi,2 Michela Burlone,8 Cristina Rigamonti,2,8 Matteo Donadon,9,10 Giuseppe Cabibbo,3 Antonio D’Alessio,1,7 David James Pinato1,7

1Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London, UK; 2Department of Translational Medicine, Università Del Piemonte Orientale, Novara, Italy; 3Section of Gastroenterology & Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, Palermo, Italy; 4Department of Surgical, Oncological and Oral Sciences (Di.chir.on.s.), University of Palermo, Palermo, Italy; 5Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; 6Department of Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy; 7Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; 8Division of Internal Medicine, AOU Maggiore della Carità, Novara, Italy; 9Department of Health Science, Università Del Piemonte Orientale, Novara, Italy; 10Department of Surgery, University Maggiore Hospital della Carità, Novara, Italy

Correspondence: David James Pinato, Clinical Senior Lecturer and Consultant in Medical Oncology, Imperial College London Hammersmith Campus, Du Cane Road, London, W12 0HS, UK, Tel +44 020 83833720, Email [email protected]

Abstract: Systemic treatment for advanced hepatocellular carcinoma (HCC) has been revolutionized over the last few years following the approval of immune checkpoint inhibitors (ICI). Despite the promising survival extension seen with ICI combination regimens, responses are not universally seen and the optimal partner for programmed cell death 1 pathway inhibitors remains to be identified. Even fewer encouraging results have been demonstrated with ICI used for monotherapy. Several mechanisms of resistance have been described so far, involving characteristics of cancer cells (intrinsic mechanisms) and of the surrounding tumor microenvironment (extrinsic mechanisms). Factors related to therapy may also contribute to the development of resistance. Increasing research efforts are being dedicated to the discovery of novel approaches and targets to overcome resistance, some of which may be introduced into clinic in the future. Herein we describe a selection of resistance mechanisms that have been involved in impairing response to ICI and propose potential therapeutic approaches to overcome resistance.

Keywords: immune checkpoint inhibitors, liver cancer, recurrence, tumor microenvironment

Introduction

Hepatocellular carcinoma (HCC) represents the most common primary liver cancer and a globally burdensome oncological diagnosis, ranking as the sixth most frequently diagnosed cancer and the third most common cause of cancer death worldwide.1 Unlike other malignancies, both incidence and mortality from HCC are expected to rise by more than 55% in the next few decades.2 HCC usually develops in the context of chronic liver disease, with Hepatitis B virus (HBV) and Hepatitis C virus (HCV) infection and alcohol abuse representing the most prevalent etiologies worldwide. However, the proportion of HCC cases arising in the setting of underlying metabolic dysfunction-associated fatty liver disease (MAFLD) is rapidly increasing, and it is expected that MALFD will become the leading risk factor for HCC in the years to come.3 Improved management of underlying chronic liver disease is likely to reduce the risk of cirrhosis decompensation and the ensuing mortality, leading to an increase in the number of patients with prolonged survival from cirrhosis but still at risk for developing HCC. On the other hand, the increasing prevalence of MAFLD, associated with the lack of effective surveillance strategies in patients with non-cirrhotic MAFLD and with the intrinsic difficulty of ultrasound surveillance in cirrhotic patients, is likely related to the persistently high number of cases diagnosed at advanced stages.4 For these patients, systemic therapy still remains the standard of care.

The advent of novel, more effective systemic therapies has markedly revolutionized the prognostic outlook of patients with advanced HCC. Monotherapies with the anti-programmed cell death protein-1 (PD-1) targeting agents nivolumab and pembrolizumab received accelerated approval by the Food and Drug Administration (FDA), based on unexpectedly high radiological response rates observed in uncontrolled early-phase clinical trials.5,6 However, PD-1 monotherapy failed in showing a significant overall survival (OS) benefit in Phase III RCTs and only pembrolizumab was granted full FDA approval.7,8 Primary disease progression on nivolumab monotherapy was reported in 37% of patients,7 suggesting that resistance to this type of therapeutic approach is common. Follow-up studies of tislelizumab and durvalumab, two monoclonal antibodies directed against PD-1 and programmed cell death ligand-1 (PD-L1), respectively, demonstrated the non-inferiority of PD-1/PD-L1 blockade against sorafenib, with meta analyses showing improved therapeutic index over tyrosine kinase inhibitors (TKIs).9

Conversely, it is now known that ICI-based combination regimens provide a significant improvement in OS compared to sorafenib in phase III RCTs, shifting the expected survival of patients with advanced disease from approximately 6 months to up to 20 months.10,11 The combinations of atezolizumab (anti-PD-L1) plus bevacizumab (anti-vascular endothelial growth factor [VEGF]), and durvalumab (anti-PD-L1) plus another ICI (tremelimumab, an anti-Cytotoxic T-Lymphocyte Antigen 4 [CTLA-4]) have obtained full FDA approval for first-line treatment, whereas the combination of nivolumab plus anti-CTLA-4 ipilimumab has been granted accelerated approval for patients who have been previously treated with sorafenib.12 The rationale behind combining ICI with anti-VEGF is that VEGF inhibition may regulate and suppress elements of the tumor immune microenvironment (TiME), including regulatory T cells (T-regs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAM). On the other hand, the combined blockade of CTLA-4 and PD-1 in ICI doublets has a distinct and non-redundant immune-biologic role within the cancer immunity cycle. CTLA-4 inhibition drives immune-suppression in tumor-antigen presenting cells and T-regs, while PD-1/PD-L1 blockade predominantly downregulates the effectiveness of the cytotoxic CD8+ T cell response.13 Unfortunately, combinations of ICI plus TKI, such as atezolizumab plus cabozantinib or pembrolizumab plus lenvatinib, did not statistically improve OS compared to TKI monotherapy in phase III RCTs.14,15 Overall, compared to ICI monotherapies, ICI-based doublets are associated with improved outcomes, and this is probably related to the added benefit of each component. However, the proportion of patients obtaining long-term survival benefit still remains limited and does not exceed 20–30%. This has led to the development of triplet regimens that have been investigated in phase I/II trials (such as nivolumab plus ipilimumab and cabozantinib) or are currently under investigation (such as atezolizumab plus bevacizumab and ipilimumab).16,17

HCC is characterized by high clinical and biological heterogeneity, translating into an extremely variable prognosis.18 The diagnosis of HCC can be performed by using validated imaging criteria, without the need for histological confirmation in patients with cirrhosis.19 This has inevitably led to a relatively limited understanding of the molecular mechanisms associated with prognosis and treatment response compared to other solid tumors. Nevertheless, it is well known that HCC is characterized by a high transcriptional and genetic heterogeneity.20 Research on molecular signatures, which could potentially predict outcomes, led to the identification of two major subclasses of HCC: one characterized by intrinsic cell proliferation, and the other defined as the non-proliferative class. The former is denoted by poor tumor cell differentiation and associated with chromosomal instability, as well as p53 tumor suppressor gene (TP53) mutations. Potentially actionable driver linked to this class is transforming growth factor beta (TGF-beta) signaling. The non-proliferative class is associated with more differentiated HCC, presence of mutations in the Wnt/beta-catenin pathway and with a better prognosis.21 Utilization of high throughput sequencing technologies led to a further deepening in the understanding of the functional characteristics recapitulated within the HCC TiME. First, the presence of a percentage of HCC with expression of markers of inflammatory response, named as immune class, was identified.22 A subsequent study improved the classification.23 Three phenotypic classes were defined: an immune class, characterized by high immune cell infiltration and PD-1/PD-L1 pathway activation, an immune intermediate class, and an immune-excluded class, which conversely is associated with low immune cell infiltration, low expression of PD-1 and PD-L1 and high frequency of chromosomal aberrations.23 A fourth subclass, defined as immune-desert pattern, has also been described; it is distinguished by an impoverishment of the immune component leading to immune ignorance and a lack of priming T cell.24

While useful in understanding the pathogenesis of HCC, implementation of these classifications in clinical practice remains limited, and prospective studies proving their predictive performance are lacking. In addition to molecular characteristics, the heterogeneity of HCC etiology has become the subject of debate with regard to its differential ability to lead to effective immune-mediated tumor rejection following immunotherapy. A meta-analysis of three RCTs administering anti-PD-1 monotherapy or in combination with anti-VEGF to patients with advanced HCC (CheckMate-459,7 IMbrave150,10 and KEYNOTE-2408) showed that patients with HBV- or HCV-related HCC benefitted from ICI, whereas patients with HCC of a non-viral etiology did not.25 Non-viral etiology was associated with shorter survival in patients with HCC undergoing anti-PD-1 therapy, even after correction for potential confounding factors relevant for prognosis, such as severity of liver damage, macrovascular tumor invasion, extrahepatic metastases, performance status, and alpha-fetoprotein.25 MAFLD-HCC appears to be characterized by a particular TiME composition, including aberrant T cell activation, leading to tissue damage and impaired immune surveillance.25

Based on data from the IMbrave150 study, corroborated by post-registration evidence, between 19% and 22% of patients treated with atezolizumab plus bevacizumab experienced early disease progression following treatment.26,27

In this continuously evolving setting, the identification of mechanisms of resistance to ICI-based treatments is crucial to allow the identification of subgroups of patients that could experience the greatest benefit from different therapeutic regimens and, ultimately, to improve patient outcomes. In this review, we discuss the mechanisms of resistance to immunotherapies in HCC and review the main intrinsic and extrinsic mechanisms that might be exploited for therapy.

Defining Therapeutic Resistance

Resistance to immunotherapy can be occurring in patients who are primary non-responders, or it can be acquired if it occurs after a period of documented response to therapy.28 The mechanisms proposed to explain the primary resistance to immunotherapy concern the reduced immunogenicity of the tumor, which can manifest itself as low expression of neoantigens, altered presentation of antigens and expression of immune co-inhibitory signals.29 HCC is generally considered a malignancy with low tumor mutational burden (TMB).30 As a consequence, the small proportion of somatic mutations leads to a lower expression of putative neoantigens. Antigen presentation in HCC is also negatively influenced by the expression of several co-inhibitory receptors that further suppress the function of T cell immunity responsible for tumor control.31

Our current understanding of acquired resistance is linked to the clonal evolution of the tumor towards a low immunogenicity phenotype after the initiation of immunotherapy.28 As with primary resistance, impaired functioning of the antigen presentation system plays a highly relevant role.32,33 Little evidence is applicable to HCC to date, where acquired resistance represents a field of active research.34

Another important conceptual characterization of resistance relies on its distinction between intrinsic, ie, linked to mechanisms that cancer cells implement to avoid immune rejection, and extrinsic, therefore external to tumor cells and linked to the characteristics of the TiME.28 In the clinic, it is accepted that these mechanisms often overlap in contributing to treatment failure. Figure 1 shows selected mechanisms of intrinsic and extrinsic resistance.

Figure 1 Selected mechanisms of resistance: extrinsic and intrinsic mechanisms are shown in panel A and B, and panel C respectively. Panel A represents the processes involved in T cell exhaustion, which are mediated by lymphocyte activation gene-3 and fibrinogen-like protein 1 (FGL-1) interaction, T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain expression, and the development of inflammation led by Kupffer cells. Panel B shows the role of activated hepatic stellate cells and fibroblasts in immunosuppression. Panel C depicts intrinsic mechanisms of resistance (from left to right): the Wnt/beta-catenin pathway, interferon γ signaling with Janus kinase / signal transducer and activator of transcription pathway activation, and FGL-1 over-expression with beta-2-microglobulin dysfunction and p53 tumor suppressor gene-mediated immunosuppression. Created with BioRender.com.

Abbreviations: LAG-3, lymphocyte activation gene-3; FGL-1, fibrinogen-like protein 1; TIGIT, T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain; TREM-1, triggering receptor expressed on myeloid cell-1; HSCs, hepatic stellate cells; TGF-β, transforming growth factor beta; EMT, epithelial-to-mesenchymal transition; β-Catenin, beta catenin; INF-γ, interferon γ; INFGR, interferon γ receptor; JAK, Janus kinase; STAT, signal transducer and activator of transcription; PD-L1, programmed cell death ligand-1; β2-microglobulin, beta-2-microglobulin; TP53, p53 tumor suppressor gene.

Tumor-Intrinsic Mechanisms of Resistance

Among the many intrinsic mechanisms exploited by cancer cells to develop resistance to ICIs, oncogenic alterations have a primary role. Dysregulation of the Wnt/beta-catenin and Janus kinase (JAK) 1/2 pathways are among the key mechanisms studied to date, alongside the dysregulation of intracellular signaling in tumor cells and alteration of immune recognition processes.

The Wnt/Beta-Catenin Pathway

Dysregulated Wnt/beta-catenin signaling is one of the major oncogenic alterations in HCC.35,36 Detailed genomic analysis has elicited that gain-of-function mutations in the CTNNB1 gene, which encodes beta-catenin, and loss-of-function mutations in AXIN1 occur in around 15–35% of HCC patients.34,36 As a result of these genetic changes, Wnt/beta-catenin signaling has been linked to progression, stemness, metastasis, and resistance in HCC patients.34,36,37 Studies have revealed that the pathway is characteristic of tumors that are enriched in cancer stem cells.38,39 Fan et al provided preclinical evidence on the effect of protein tyrosine kinase 2 hypomethylation enhancing HCC stemness via Wnt activation, ultimately causing resistance and increasing the risk of recurrence (p<0.01).38 Aberrant Wnt/beta-catenin activation has been associated with resistance to sorafenib and regorafenib.36,40 Harding et al demonstrated lower disease control rates (8.3% vs 40.2%) and progression-free (1.9 vs 5.3 months) and overall survival (2.0 vs 7.4 months) rates in HCC patients associated with over-activation of the Wnt/beta-catenin pathways.40 These poorer prognostic results were significantly associated with increased resistance rates (p<0.01).40 Similar findings have been corroborated by de Galarreta et al, who described that in a genetically engineered mouse model of HCC, the induced expression of exogenous MYC; Trp53-/- antigens causes immune evasion via upregulation of beta-catenin pathways, driving resistance to nivolumab and pembrolizumab.38 Incorporating next generation sequencing to identify patients with overactivated Wnt/beta-catenin pathways has the potential to identify HCC patients who would benefit from genome-targeted therapies. However, further studies into biomarker use are required before this can be integrated into standard clinical practice.

P53 Tumor Suppressor Gene

TP53 is frequently defective in human cancers, including HCC, and its mutations can be caused by both viruses and chemicals.41 TP53 contributes to immune surveillance by recruiting CD8+ and T-helper cells, and its perturbations could promote an immunosuppressive environment.42,43 A recent study that analyzed 240 samples of HCC showed an increased rate of TP53 mutations in a group of tumors defined as “intermediate class”, characterized by a decreased immune infiltration and an immunosuppressive microenvironment. In this setting, TP53 demonstrates a critical role for immune evasion in HCC.23

Interferon-γ Signaling Pathway

Interferon γ (INF-γ) is a cytokine secreted by T cells after the recognition of tumor neoantigens that act as a key regulator of immune response against tumor cells. The binding of INF-γ to its receptor on cancer cells induces the activation of the JAK-signal transducer and activator of transcription (STAT) signaling pathway, thus resulting in the subsequent transcriptional activation of PD-L1. This leads to an increased expression of PD-L1 on the tumor cells’ surface and subsequently, to the negative regulation of antitumor immunity.44 However, tumor intrinsic alterations of the INF-γ signaling pathway reduce the expression of PD-L1, thereby promoting resistance to ICI.45

Accordingly, mutations in genes involved in the INF-γ pathway, including loss of signaling-related genes (JAK 1/2) and amplification of suppressor genes, have been identified in patients that do not respond to anti-CTLA-4 and anti-PD-1 therapies, thereby reinforcing the suggested role of INF-γ signaling disruption in ICI resistance.46,47 Moreover, studies conducted on HCC tissues identified the presence of methylations in the suppressors of cytokine signaling genes SOCS-1 and SOCS-3, which are negative regulators of the JAK2-STAT signaling pathway. This epigenetic modification silences SOCS-3, thus leading to the constitutive activation of STAT3 in hepatoma cells. STAT3 is a transcription factor involved in the regulation of many cancer mechanisms, including resistance to apoptosis and treatment.48,49

Tumor Mutational Burden

Somatic mutations occurring within neoplastic tissue may be relevant to cancer progression beyond their functional role in the protein they encode. Changes in the protein conformation induced by somatic mutations can elicit immune responses, through the unmasking of neoantigens. The positive correlation between somatic mutations and neoantigens entails that the higher the TMB of a tumor is, the greater its capacity would be to trigger a more efficient and diverse T cell response.50 Several studies have attempted to investigate TMB as a biomarker for ICI response.51–53 Although TMB alone is not sufficient to predict outcome, higher TMB scores have been associated with higher ICI efficacy in melanoma and non-small-cell lung cancer (NSCLC).51,52,54 TMB in HCC is relatively low compared to other solid tumors.52 Etiologic factors specific for HCC, such as virally induced chronic liver disease, appear to be related to a higher TMB, leading to a better response to ICI.55 In contrast, tumors arising in the context of MAFLD show a trend toward lower TMB.56 While retrospective evidence shows that higher TMB levels positively correlate with an improved 1-year OS compared to a lower TMB, the provision of immunotherapy is not routinely informed by TMB quantification in the clinic. Lack of a universally validated cutoff to define TMB-high, as well as difficulties in harmonizing sequencing platforms and depth are among the hurdles identified in the clinical applicability of this putative biomarker in HCC.57

Dysfunction of the Antigen Presentation Machinery

The cytotoxic capacity of the host immune system against tumor cells is mediated by the effective presentation of cancer antigens and neo-antigens to CD8+ T cells. The latter is facilitated by major histocompatibility complex (MHC) proteins, which interact with T cell receptors (TCRs) on T lymphocytes and elicit cytotoxic activity.58 Downregulation of MHC proteins is related to immune escape of the tumor and could lead to ICI resistance.59 Mutations in beta-2-microglobulin, a gene associated with MHC stability on the cell surface, have been linked to ICI resistance in solid neoplasms.60 Likewise, the suppression of MHC class I proteins caused by methylation of its transactivator caspase recruitment domain 5 has been related to poor antigen presentation and reduced survival.61

Fibrinogen-Like Protein 1

Fibrinogen-like protein 1 (FGL-1) is a hepatocyte protein62 involved in liver regeneration and cell proliferation.63 It is not only present in the liver, but it also exists in plasma, where it, however, has not been implicated in coagulation.62 FGL-1 has been demonstrated as an important ligand for lymphocyte activation gene-3 (LAG-3 or CD223), a transmembrane protein that, like PD-1, represents one of several immune checkpoints considered gatekeepers of immune responses.64 The interaction between the LAG-3 extracellular domain and the FGL-1 domain is independent from the antigen presenting machinery; their binding is less effective in resting T cells that express minimal LAG-3 levels.65,66 Impediment of the FGL1-LAG-3 interaction in mouse models potentiates anti-tumor immunity.65 Using this interaction as a therapy target could allow to avoid the resistance mechanisms caused by the alteration of antigen presentation mechanisms, considering that the latter would not be directly involved. Moreover, FGL-1 expression within the tumor appeared to be correlated with the progression and prognosis of HCC, suggesting that FGL-1 could be used not only as a molecular target but also as a potential prognostic biomarker.67 More research is needed in order to understand if LAG-3 through FGL-1 blockade could have a role as a new therapeutic strategy to overcome resistance to immunotherapy. In addition to its therapeutic role, it will be important to analyze the prognostic potential of FGL as a prognostic marker. In fact, FGL-1 expression in circulating tumor cells has been related to a poor prognosis in patients with HCC who underwent resection.68

Therapy-Associated Mechanisms of Resistance

Antidrug Antibodies

The widespread use of immunomodulatory agents has drawn attention to their potential immunogenicity, as well as their related effects on the safety and efficacy of treatment.69 While the majority of monoclonal antibodies is able to induce antidrug antibody (ADA) formation, following interaction with the host immunity,70 the risk of ADA development is intrinsically drug-specific, with diverse ranges of ADAs being reported across studies. Treatment with B-cell depleting agents appears associated with a lower risk of immunogenicity, compared to agents that enhance T cells functions.69 Depending on the protein composition of the monoclonal antibody, a “vaccine-like” reaction may develop with first the generation of low-affinity IgM antibodies, followed by the appearance of high-affinity IgG antibodies; in the presence of low immunogenic monoclonal antibodies, the ADAs will mostly only be responsible of a minimal clinical effect.71 Each patient may have non-neutralizing or neutralizing ADAs,72 capable of reducing the efficacy of monoclonal antibodies by altering their bioavailability and/or accelerating their clearance from circulation.73 For most agents, the titers and percentages of ADA-positive patients are reported, but the clinical consequences of ADAs are not investigated.70 A recent observational study examining 132 patients with advanced HCC treated with atezolizumab plus bevacizumab, showed that the emergence of high titers of ADA (>1000 ng/mL) as early as 3 weeks post commencement of systemic therapy, was associated with shorter OS, even after adjusting for various confounding factors. Patients with higher ADA levels exhibited reduced systemic exposure to atezolizumab, as well as impaired proliferation and activation of peripheral CD8+ T cells.74 Treatment-related anti-tremelimumab and anti-durvalumab antibodies have also been reported in the HIMALYA trial. Different antibody percentages have been identified, depending on whether the treatment involves the combination of the two drugs or the single agent: anti-durvalumab antibodies were detected in 3.1%, 2.5%, and 4.6% of patients receiving a single dose of 300 mg tremelimumab plus 1500 mg of durvalumab every 4 weeks (STRIDE regimen), durvalumab as single agent, and 75 mg of tremelimumab every 4 weeks for four doses plus 1500 mg of durvalumab every 4 weeks (T751D), respectively; neutralizing antibodies were detected in 1.7%, 0.7%, and 0% of patients, respectively. Anti-tremelimumab antibodies were detected in 11.0% patients receiving the STRIDE regimen and 21.6% receiving T751D at any visit; neutralizing antibodies were detected in 4.4% and 15.7% patients, respectively.11 Relationship with clinical outcomes was not investigated. Considering the effectiveness of immunotherapy and its first-line indication in advanced HCC, further studies are needed to investigate prevalence and clinical significance of antidrug antibodies and their consequences on therapy efficacy. A call for the universal evaluation of ADAs across clinical trials as a mechanism of resistance is needed to ensure a better and more complete understanding of their significance in the delivery of immunotherapy.

Tumor Extrinsic Mechanisms: The Immune Contexture of the Tumor Microenvironment

In HCC, the TiME is known to play a critical role in HCC development and progression. It consists of a dynamic network of growth factors and inflammatory cytokines, extracellular matrix proteins, endothelial cells, lymphocytes, TAMs, hepatic stellate cells (HSCs), and stromal cells, including cancer-associated fibroblasts (CAFs).75,76 The TiME in HCC exhibits a high functional heterogeneity, where certain immune infiltrating cells can either promote a favorable antitumor immune response or limit effective immune surveillance.77 With often functionally opposing roles, infiltrating cells composing the TiME constitute a neoplastic niche, where the tumor can proliferate rapidly, escape from the host’s defense systems that are directed against transformed cells, and facilitate the invasive behavior of cancer cells.76 This bidirectional effect of the TiME in influencing the biological characteristics of the tumor has been termed immunoediting. It consists in a dynamic process, where the immune system not only protects against cancer cell growth but is also able to promote tumor progression, either by establishing a facilitating TiME or by selecting tumor cells that are more fit to survive.78 Immunoediting is traditionally divided into three phases: elimination, where transformed cells are destroyed by a competent immune system; equilibrium, where cells that managed to survive the immune attack start the editing process; and escape, the last phase, where the tumor grows progressively, becomes clinically apparent and establishes an immunosuppressive TiME.78 This process is not only involved in tumor development, but it can also occur in a patient undergoing immunotherapy.79 Understanding the interactions among the immune response, oncogenic signaling, and the TiME is now more than ever critical to improve the efficacy of immunotherapies, which are highly reliant on T cell immune reconstitution.22,80

A more in-depth view of the TiME organization could prove useful. A recent study by Sheng et al81 focused on the spatial characteristics of the TiME in HCC. Using the imaging mass cytometry, regional function units have been identified, each one of them with a distinct distribution of stromal and immune cells. The presence of specific regions enriched with cancer cells has been associated with worse patient OS and PFS. At the same time, a poor prognosis was related to the enrichment of Kupffer cells, which negatively affected the activation of T cells.81 Indeed, technologies able to return data on spatial organization of the TiME could prove able to discover novel biomarkers and therapeutic targets.82 Having more information on the constitution of the TME, it would be possible to act on its remodeling, which could be one of the important immunotherapeutic approaches to improve HCC treatment.83

Lymphocyte Activation Gene-3

LAG-3 is a type I transmembrane protein with four extracellular Ig-like domains that is selectively transcribed into activated NK and T lymphocytes.84 It has structural similarities with another MHC class II ligand (MHC-II), called CD4, with which it shares approximately 20% amino acid sequence identity.85 While the interaction between MHC-II and CD4 supports T helper cells activation, the overexpression of LAG-3 downregulates antigen-dependent T helper (CD4+) responses in vitro.84 LAG-3 is not instrumental in the induction phase of the immune response due to its engagement only after lymphocyte activation.86 Its expression can be induced by infective87–90 and tumor-related antigens, and its overexpression leads to the generation of exhausted T cells, which ultimately results in immune escape.91–96 Exhausted CD4+ and CD8+ tumor-infiltrating T cells are defective in cytokine production.97 LAG-3 blockade has been demonstrated to reinvigorate exhausted T cells and strengthen anti-infection immunity, although to a lesser extent, compared with that caused by PD-1 blockade.98 Preclinical studies have suggested that the co-engagement of LAG-3 and PD-1 further suppresses interleukin-2 (IL-2) production, which is directly involved in T lymphocyte differentiation, arguing that LAG-3 and PD-1 act synergistically to restrain the antigenic stimulation of CD4+ T cells.99–101 Based on these assumptions, several clinical trials involving patients with different solid neoplasms, evaluating both the safety of LAG-3 inhibitors and the expression of LAG-3 as an outcome marker, have been concluded or are currently recruiting (clinicaltrials.gov).

T Cell Immunoglobulin and Immunoreceptor Tyrosine-Based Inhibitory Motif Domain (TIGIT)

TIGIT is another immune checkpoint molecule first described in 2009, able to exert its immunosuppressive effects by binding to the poliovirus receptor (PVR or CD155), which is highly expressed in various neoplasms, and to PVR-like protein.102 TIGIT is also able to modulate cytokine production in dendritic cells.102 TIGIT is expressed on NK, NKT, CD8+, Treg, and memory CD4+T cells; in normal conditions, it protects normal cells from NK-mediated cytotoxicity and counter inhibits the NK-mediated killing of tumor cells.103 It has been hypothesized that the presence or the overexpression of TIGIT poses a metabolic barrier to T cell function104 and in preclinical studies, TIGIT blockade has been shown to enhance T cells responses.105,106 Overexpression of CD155 in HCC may lead to a defective immune response by upregulating TIGIT.107 Given these premises, it is understandable why TIGIT can be used as a therapeutic target together with other immune checkpoints, particularly in combination with PD-1 blockade: the simultaneous blockade of TIGIT and PD-1 enhances CD8+ T cell function in HCC.108,109 Both preclinical and clinical trials are actively investigating anti-TIGIT antibodies.104

CD8+ T Cell Infiltration and Functional Phenotype

CD8+ cytotoxic T cells normally play a crucial role in anti-tumor immune response. Continuous exposure to antigens, such as during chronic infections and cancer, leads to failure of efficient CD8+ T cell development and, ultimately, their exhaustion.107,110 Exhausted CD8+ T cells have both a limited effector function, caused by the loss of IL-2, tumor necrosis factor-α and interferon-γ production, as well as a high co-expression of inhibitory receptors, such as PD-1, CTLA-4, TIGIT, and LAG-3, together with extensive transcriptional changes, compared to effector or memory CD8+ T cells.111 As a result, exhausted CD8+ T cells fail to control tumor growth, but implementing effective methods to reinvigorate them can significantly impact cancer progression,112 such as with ICI. HCC more often presents with an exhausted phenotype of CD8+ T cells in the TiME, compared to effector CD8+ T cells. In particular, an increase in the number of exhausted cells has been observed in advanced stages, compared to early stages.113 Moreover, the presence of exhausted CD8+ T cells expressing high levels of PD-1 has been described as increased in parallel with tumor stages, suggesting that the severity of CD8+ T cell exhaustion is related to HCC progression.114 In accordance with what has already been said, HCC patients with high proportions of T cells expressing PD-1 showed significantly dismal postoperative survival after hepatectomy and a high risk of recurrence.114 In addition to the degree of PD-1 expression, the ratio between effector T cells and exhausted CD8+ T cells proved to be associated with disease control in anti-PD-1-treated patients.115 Recruiting more active T cell populations could allow for a greater response to ICI.

The Triggering Receptor Expressed on Myeloid Cell-1

Triggering receptors expressed on myeloid cells (TREMs) are a family of cell-surface receptors expressed broadly on myeloid cells, capable of stimulating neutrophil- and monocyte-mediated inflammatory responses.116,117 TREM-1 was first described as up-regulated in infections117–119 and inflammatory conditions,120–122 but also in autoimmune diseases.123 It was later described that cancer cells in NSCLC can directly up-regulate TREM-1 expression in the patients’ macrophages, which is associated with cancer recurrence and poor survival.124 In liver fibrosis, TREM-1 expression appeared robustly increased in Kupffer cells and monocytes/macrophages, suggesting a role in the inflammatory liver response and in fibrotic liver disease.125 The inhibition of the TREM-1 signaling pathway in a preclinical model proved to regulate the inflammatory response and attenuate the development of inflammation-associated HCC.126 Furthermore, in HCC, TREM-1 expression on HSCs was related to the aggressive tumor behaviour.127 Knowing the role of TREM-1 in activating inflammation and promoting carcinogenesis, this receptor could be studied not only as a future target for immunotherapy but also as a potential biomarker.128

Transforming Growth Factor Beta

Transforming growth factor beta (TGF-beta), mainly secreted by HSCs, plays an important role in liver carcinogenesis exerting proinflammatory/fibrotic, tumor suppressive and/or pro-metastatic effects.129 In the early stages of tumor development, high levels of TGF-beta have been associated with better prognosis, whereas increased invasiveness and dedifferentiation have been reported in advanced tumors. These effects suggest that in early stages TGF-beta exerts its tumor suppressor function, but following disease progression it contributes to the development of a malignant phenotype.129 Studies have reported that TGF-beta may induce a decrease of E-cadherin, characteristic of epithelial cells, and an up-regulation of mesenchymal genes, thus leading to an epithelial-to-mesenchymal transition.130

Moreover, it is known that TiME-derived TGF-beta is involved in the suppression of antitumor T cell response. In fact, an increase in TGF-beta signaling through SMAD3 transcriptional regulation may cause the upregulation of PD-1 expression, thus inducing T cell exhaustion and conferring resistance to PD-1 blockade therapies.131

Activated Hepatic Stellate Cells

HSCs are mesenchymal cells located in the space of Disse between hepatocytes and sinusoidal endothelial cells.132 They represent the principal source of extracellular matrix formation132 and their presence is vital to hepatocellular function, as well as the liver’s response to injury.133 The exposure to any noxious stimulus, such as viral infection or alcohol, causes HSC activation and increased deposition of the extracellular matrix, ultimately leading to fibrinogenesis.134 Scar tissue formation is aimed at protecting the liver from further damage; however, sustained activation of HSCs leads to chronic fibrosis and cirrhosis.135,136 HSCs are an important source of growth factors in the liver, but they are also sensitive to these factors.136 In HCC, HSCs can promote chemoresistance through the secretion of hepatocyte growth factor, which provokes upregulation of the cancer stem cell-like characteristics of epithelial tumor cells and epithelial-to-mesenchymal transition.137,138 The activation of HSCs also contributes to immunosuppression: they are able to convert mature peripheral blood monocytes into myeloid-derived suppressor cells,139,140 they can induce T cell apoptosis through PD-L1 expression,141 as well as Treg cell accumulation inside the liver.142 Knowing their role in the regulation of immune response, targeting these cells may improve immunotherapy efficacy.

Cancer-Associated Fibroblasts

Cancer-associated fibroblasts (CAFs) represent a constitutive group of stromal cells of the TiME, capable of promoting hepatocarcinogenesis.143,144 While their origin is not yet fully understood, and a unique marker for their identification is missing,144 it appears clear that these cells can produce several factors, such as growth factors, chemokines, and angiogenic factors that support tumor-cell proliferation and invasion.145–148 Additionally, CAFs appear to sustain a crosstalk with the TiME, recruiting dendritic cells and monocytes responsible for immune response suppression,149–152 and they also have the ability to increase PD-1 expression.153 Indeed, CAFs infiltration in the TiME is associated with a response to immunotherapy.154 Moreover, their interaction with macrophages promotes the construction of a tumor immune barrier, which is associated with the efficacy of ICI.155 Their role in immunosuppression justifies the identification of these cells as a therapeutic target. Both preclinical and clinical studies of CAF-targeted therapies are under investigation,156,157 but more experience in HCC is needed.

Influencing the TiME: The Gut Microbiome

The gut microbiome is a complex entity that comprises different microorganisms capable of performing immunomodulatory activities.158 The bacteria and their metabolites are constantly interacting with immune cells at the gut mucosa to coordinate immune responses in the absence of inflammation.159 This so-called “homeostatic immunity” is controlled by several factors, including antibody responses to commensal bacteria, innate-like T cell responses, as well as the microbiota and its metabolites. Given the role of the gut microbiome in the immune system, it is unsurprising that it has been shown to modulate T cell infiltration of solid tumors and efficacy of ICIs.160 Species, such as Bifidobacterium161 and A. Muciniphila160,162 in particular, have been the focus of extensive research. Antibiotics are well documented as having a sustained impact on gut microbiota composition,163,164 with proton-pump inhibitors165,166 and corticosteroids167 also likely to modulate the gut microbiome. There have been a number of association studies investigating the impact of these medications given prior to or concomitantly with ICIs. Concomitant use of these three drug types has been associated with poor patient outcomes in retrospective analyses,168,169 with some evidence that they have a cumulative effect on predicting OS and PFS in patients with advanced cancer receiving ICI therapy.170 It has also been shown that use of antibiotics during the 30 days prior to the start of ICI therapy significantly reduces the likelihood of treatment response and OS (independent of confounding factors).171 In HCC, use of antibiotics within 30 days before or after systemic therapy initiation was associated with a worse OS and PFS, not only in ICI recipients but also in patients treated with anti-VEGF, TKI, and placebo. These results highlight the complexity of HCC composition, its interaction with underlying hepatopathy, and the pleiotropic effects of molecular therapies for HCC.172 It is essential to establish whether this is a causative relationship mediated by changes in the gut microbiome,173 as the use of concomitant or prior medications provide a potential modifiable factor in response to ICIs.

Conclusions

ICIs have revolutionized the treatment of advanced hepatocellular carcinoma. Nevertheless, resistance to immunotherapy remains a major obstacle in the treatment of HCC. Both intrinsic and extrinsic mechanisms contribute to the development of resistance. Among the intrinsic mechanisms, TMB, which refers to the total number of somatic mutations in the tumor, plays a major role in the regulation of the immune response. Notably, a high TMB, which results in a higher presence of neoantigens, has been linked to a greater response to treatment, even if its role has not been prospectively evaluated in ICI.174 Tumor intrinsic mutations can occur in genes involved in the regulation of immune response. The overactivation of beta-catenin is related to lower CD8+ T cell infiltration and PD-L1 expression, thus resulting in resistance to ICIs.175 Moreover, inactivating mutations of TP53, an oncosuppressor gene, have been linked to tumor progression and resistance to ICIs.23,43 It has also been found that dysregulation of genes involved in the INF-γ signaling pathway, as well as defects in antigen presentation mediated by the MHC machinery, may contribute to ICI resistance.45,59 Cancer cells overexpress FGL-1, which interacts with LAG-3 on T cells, leading to T cell exhaustion and subsequently, repression of the antitumor response.65

T cell exhaustion belongs to the tumor extrinsic mechanisms of resistance, for which TiME is responsible. The suppression of T cell function may be exerted by the overactivation of TIGIT, which is caused by the overexpression of its ligand, CD155, thus creating an immunosuppressive microenvironment and inducing tumor escape.104 Additionally, studies show that increased expression of TREM-1 elicits T cell dysfunction through the modulation of PD-L1, which impairment fails to improve the related immunosuppression.176 Additionally, HSCs141,142 and CAFs152 can also contribute to the development of an immunosuppressive framework. The gut microbiome’s influence on the TiME has been widely acknowledged.160

To expand the spectrum of cancer patients that could benefit from immunotherapy, more targets and innovative approaches need to be considered in future clinical trials.66 A number of strategies have been proposed to overcome resistance to immunotherapy. In addition to using triplets ICI-regimens, also other mechanisms are being explored: among them, reinvigorating exhausted T cell response seems to be one of the most promising, with pre-clinical and clinical studies currently investigating the blockade of other immunosuppressive promoters, such as LAG-3 and TIGIT (clinicaltrials.gov). Moreover, the modification of the immunosuppressive drift of the TiME could improve ICI-efficacy. Many mechanisms are thought to be responsible for the creation of an immunosuppressed niche for the tumor, but our comprehension of these complex interactions remains incomplete.

Another proposed method to overcome resistance to immunotherapy are vaccines. Tumor vaccines are intended to facilitate tumor-specific immune responses by targeting tumor antigens.177 At the present time, the technologies investigated for vaccines in HCC so far include dendritic cell vaccines and peptide vaccines, such as AFP and glypican-3 (GPC3).178 All these strategies have been demonstrated to be safe and although monotherapy with vaccines showed limited clinical efficacy, this could be attributed to the diverse features of HCC tumors and vaccine design, or even a combination of both.179 The selection of the right target for vaccines is crucial. The risk of toxicity could be magnified by the expression of the vaccine target in non-malignant tissues, such as in the case of tumor-associated antigens, which are not specifically present only in cancer cells.180 Furthermore, the development of tolerance mechanisms is considered a possible issue.180 A new technology for vaccines could represent a future therapeutic strategy: neoantigens are new protein sequences specific for each patient, resulting from mutations appearing in tumor cells.181 Neoantigens are highly tumor-specific, as they are not present in normal cells, and they possess great immunogenic capacity, but they require great MHC affinity to be processed by APC and to then be recognized by T cells.179 The combination of neoantigen-based vaccines with ICIs has been under investigation in several solid tumors, like NSCLC, bladder cancer, and melanoma.182,183 The encouraging results from previous studies paved the way for the application in HCC: currently clinical trials involving neoantigen-based vaccines in combination with anti-PD-1 (NCT04251117, NCT05269381, and NCT04912765) or anti-PD-1 plus anti-CTLA-4 immunotherapy (NCT04248569) in HCC are recruiting, both in Phase 1 and Phase 2.

Additionally, chimeric antigen receptor (CAR) T cell therapy targeting HCC antigens may also be a promising strategy to overcome ICI resistance. CARs are engineered, non-MHC-restricted receptors that redirect the specificity, function, and metabolism of T cells, ultimately leading to an improved T cell function.184,185 They possess an extracellular region of a receptor that is able to recognize a specific antigen and, consequently, lead to cell death.186 CAR-T cell therapy has already shown promising results in some oncohematological settings,187 while its role in solid tumors still requires further research. Among the major obstacles that remain to be overcome concern the tumor mass infiltration and the presence of a particular immunosuppressive TiME.188 The ability of CAR-T to reach cancer cells in solid tumors is invalidated by physical barriers belonging both to the tumor itself, for instance, the stromal tumor structure, and to the CAR-T mobility.189 Furthermore, the lack of a single target tumor reduces the effectiveness of this treatment and increases the risk of toxicity on non-malignant cells.188,189 In addition, even after the identification of an appropriate single target, the tumor could promote an antigen escape, downregulating the target antigen expression.189 Combining multiple antigens is an investigated strategy.190 For HCC, the tumor-specific GPC3 antigen has been identified as a target for CAR-T cell therapy.191–193 Several clinical trials for advanced HCC patients are investigating GPC3 CAR-T cells (clinicaltrails.gov). The combination of CAR-T cells and ICIs could represent a future therapeutic scheme capable of, on the one hand, stimulating the immune system, and, on the other hand, effectively hitting the chosen tumor targets. However, the problem of identifying a tumor marker of HCC that allows a more targeted therapy remains open, as well as the identification of a marker that allows to predict the response to immunotherapy. Various biomarkers have already been proposed, but none of these have been validated in HCC yet, and further research is needed to determine their clinical utility.

Much remains to be understood about the mechanisms of resistance to immunotherapy; further translational studies are required to identify the patients who will respond most to this type of therapy and to avoid unnecessary side effects in patients who will not respond to immunotherapy. More research is needed to develop effective strategies to overcome immunotherapy resistance.

Acknowledgments

AD is supported by the National Institute for Health Research (NIHR) Imperial BRC, by grant funding from the European Association for the Study of the Liver (2021 Andrew Burroughs Fellowship) and from Cancer Research UK (RCCPDB- Nov21/100008). DJP is supported by grant funding from the Wellcome Trust Strategic Fund (PS3416) and acknowledges grant support from the Cancer Treatment and Research Trust (CTRT) and infrastructural support by the Cancer Research UK Imperial Centre and the NIHR Imperial Biomedical Research Centre.

Disclosure

CC received speaker fees from Eisai, MSD, and Ipsen. BS received travel support from AbbVie, AstraZeneca, Ipsen, and Gilead. JK received travel grant funds from Boehringer Ingelheim. MP received speaker honoraria from Bayer, BMS, Eisai, Lilly, MSD, and Roche; he is a consultant for AstraZeneca, Bayer, BMS, Eisai, Ipsen, Lilly, MSD, and Roche; he received travel support from Bayer, BMS, and Roche. GC has served as a consultant or on advisory boards for Bayer, Eisai, Ipsen, MSD, AstraZeneca, and Roche. AD received educational support for congress attendance and consultancy fees from Roche. DJP received lecture fees from ViiV Healthcare and Bayer Healthcare; travel expenses from BMS and Bayer Healthcare; consulting fees for Mina Therapeutics, EISAI, Roche, and Astra Zeneca; and received research funding (to institution) from MSD and BMS. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

References

1. Liver-fact-sheet. International agency for research on cancer. GLOBOCA, IARC; 2020.

2. Rumgay H, Arnold M, Ferlay J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77(6):1598–1606. doi:10.1016/j.jhep.2022.08.021

3. Vitale A, Svegliati-Baroni G, Ortolani A, et al. Epidemiological trends and trajectories of MAFLD-associated hepatocellular carcinoma 2002–2033: the ITA.LI.CA database. Gut. 2023;72(1):141–152. doi:10.1136/gutjnl-2021-324915

4. Tan DJH, Ng CH, Lin SY, et al. Clinical characteristics, surveillance, treatment allocation, and outcomes of non-alcoholic fatty liver disease-related hepatocellular carcinoma: a systematic review and meta-analysis. Lancet Oncol. 2022;23(4):521–530. doi:10.1016/S1470-2045(22)00078-X

5. El-Khoueiry AB, Sangro B, Yau T, 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. doi:10.1016/S0140-6736(17)31046-2

6. Zhu AX, Finn RS, Edeline J, 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. doi:10.1016/S1470-2045(18)30351-6

7. Yau T, Park JW, Finn RS, 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. doi:10.1016/S1470-2045(21)00604-5

8. Finn RS, Ryoo BY, Merle P, et al. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial. J Clin Oncol. 2019;38:193–202. doi:10.1200/JCO.19.01307

9. Fulgenzi CA, Scheiner B, Korolewicz J, et al. Efficacy and safety of frontline systemic therapy for advanced HCC: a network meta-analysis of landmark phase III trials. JHEP Reports. 2023;5:100702. doi:10.1016/j.jhepr.2023.100702

10. Finn RS, Qin S, Ikeda M, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. New England J Med. 2020;382(20):1894–1905. doi:10.1056/NEJMoa1915745

11. Abou-Alfa GK, Lau G, Kudo M, et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evidence. 2022;1(8). doi:10.1056/EVIDoa2100070

12. Yau T, Kang YK, Kim TY, 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 Oncology. 2020;6(11):e204564. doi:10.1001/jamaoncol.2020.4564

13. Pinato DJ, Guerra N, Fessas P, et al. Immune-based therapies for hepatocellular carcinoma. Oncogene. 2020;39(18):3620–3637. doi:10.1038/s41388-020-1249-9

14. Kelley RK, Rimassa L, Cheng AL, et al. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2022;23(8):995–1008. doi:10.1016/S1470-2045(22)00326-6

15. Finn RS, Kudo M, Merle P, et al. LBA34 - Primary results from the phase III LEAP-002 study: lenvatinib+pembrolizumab versus lenvatinib as first-line (1L) therapy for advanced hepatocellular carcinoma (aHCC). Anna Oncol. 2022;2022:S808–S869.

16. Yau T, Zagonel V, Santoro A, et al. Nivolumab plus cabozantinib with or without ipilimumab for advanced hepatocellular carcinoma: results from cohort 6 of the checkMate 040 trial. J Clin Oncol. 2022;41:1747–1757. doi:10.1200/JCO.22.00972

17. Merle P, Blanc JF, Edeline J, et al. Ipilimumab with atezolizumab-bevacizumab in patients with advanced hepatocellular carcinoma: the PRODIGE 81-FFCD 2101-TRIPLET-HCC trial. Digest Liver Dis. 2023;55(4):464–470. doi:10.1016/j.dld.2023.01.161

18. Cabibbo G, Enea M, Attanasio M, Bruix J, Craxí A, Camma C. A meta-analysis of survival rates of untreated patients in randomized clinical trials of hepatocellular carcinoma. Hepatology. 2010;51(4):1274–1283. doi:10.1002/hep.23485

19. European Association for the Study of the Liver. EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236. doi:10.1016/j.jhep.2018.03.019

20. Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 2018;15(10):599–616. doi:10.1038/s41571-018-0073-4

21. Hoshida Y, Nijman SMB, Kobayashi M, et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res. 2009;69(18):7385–7392. doi:10.1158/0008-5472.CAN-09-1089

22. Sia D, Jiao Y, Martinez-Quetglas I, et al. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology. 2017;153(3):812–826. doi:10.1053/j.gastro.2017.06.007

23. Montironi C, Castet F, Haber PK, et al. Inflamed and non-inflamed classes of HCC: a revised immunogenomic classification. Gut. 2022;72:129–140. doi:10.1136/gutjnl-2021-325918

24. Gao X, Huang H, Wang Y, et al. Tumor immune microenvironment characterization in hepatocellular carcinoma identifies four prognostic and immunotherapeutically relevant subclasses. Front Oncol. 2021;10:610513. doi:10.3389/fonc.2020.610513

25. Pfister D, Núñez NG, Pinyol R, et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature. 2021;592(7854):450–456. doi:10.1038/s41586-021-03362-0

26. Fulgenzi CAM, Cheon J, D’Alessio A, et al. Reproducible safety and efficacy of atezolizumab plus bevacizumab for HCC in clinical practice: results of the AB-real study. Eur J Cancer. 2022;175:204–213. doi:10.1016/j.ejca.2022.08.024

27. Cheng AL, Qin S, Ikeda M, et al. Updated efficacy and safety data from IMbrave150: atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76(4):862–873. doi:10.1016/j.jhep.2021.11.030

28. Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of resistance to immune checkpoint blockade: why does checkpoint inhibitor immunotherapy not work for all patients? Am Soc Clin Oncol Educ Book. 2019;39:147–164. doi:10.1200/EDBK_240837

29. Xie Q, Zhang P, Wang Y, Mei W, Zeng C. Overcoming resistance to immune checkpoint inhibitors in hepatocellular carcinoma: challenges and opportunities. Front Oncol. 2022;2022:12.

30. Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1). doi:10.1038/s41572-020-00240-3

31. Sachdeva M, Chawla YK, Arora SK. Immunology of hepatocellular carcinoma. World J Hepatol. 2015;7(17):2080–2090. doi:10.4254/wjh.v7.i17.2080

32. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. New England J Med. 2016;375(9):819–829. doi:10.1056/NEJMoa1604958

33. Gettinger S, Choi J, Hastings K, et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017;7(12):1420–1435. doi:10.1158/2159-8290.CD-17-0593

34. De A, Coste LA, Romagnolo B, et al. Somatic mutations of the-catenin gene are frequent in mouse and human hepatocellular carcinomas. Proceed Natl Acad Sci. 1998;95:1.

35. Xu C, Xu Z, Zhang Y, Evert M, Calvisi DF, Chen X. β-Catenin signaling in hepatocellular carcinoma. J Clin Invest. 2022;132(4). doi:10.1172/JCI154515

36. Khalaf AM, Fuentes D, Morshid AI, et al. Role of Wnt/β-catenin signaling in hepatocellular carcinoma, pathogenesis, and clinical significance. J Hepatocell Carcinoma. 2018;5:61–73. doi:10.2147/JHC.S156701

37. Chen J, Jin R, Zhao J, et al. Potential molecular, cellular and microenvironmental mechanism of sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2015;367(1):1–11. doi:10.1016/j.canlet.2015.06.019

38. Fan Z, Duan J, Wang L, et al. PTK2 promotes cancer stem cell traits in hepatocellular carcinoma by activating Wnt/β-catenin signaling. Cancer Lett. 2019;450:132–143. doi:10.1016/j.canlet.2019.02.040

39. de Galarreta MR, Bresnahan E, Molina-Sánchez P, et al. β-catenin activation promotes immune escape and resistance to anti–PD-1 therapy in hepatocellular carcinoma. Cancer Discov. 2019;9(8):1124–1141.

40. Harding JJ, Nandakumar S, Armenia J, et al. Prospective genotyping of hepatocellular carcinoma: clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin Cancer Res. 2019;25(7):2116–2126. doi:10.1158/1078-0432.CCR-18-2293

41. Hussain SP, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26(15):2166–2176.

42. Blagih J, Zani F, Chakravarty P, et al. Cancer-Specific Loss of p53 Leads to a Modulation of Myeloid and T Cell Responses. Cell Rep. 2020;30(2):481–496.e6. doi:10.1016/j.celrep.2019.12.028

43. Blagih J, Buck MD, Vousden KH. p53, cancer and the immune response. J Cell Sci. 2020;133:5.

44. Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol. 2020;20(1):25–39. doi:10.1038/s41577-019-0218-4

45. Vu SH, Vetrivel P, Kim J, Lee M-S. Cancer Resistance to Immunotherapy: molecular Mechanisms and Tackling Strategies. Int J Mol Sci. 2022;23(18):10906. doi:10.3390/ijms231810906

46. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–249. doi:10.1146/annurev-pathol-042020-042741

47. Gao J, Shi LZ, Zhao H, et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell. 2016;167(2):397–404.e9. doi:10.1016/j.cell.2016.08.069

48. Johnson DE, O’Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 2018;15(4):234–248. doi:10.1038/nrclinonc.2018.8

49. Fabregat I. Dysregulation of apoptosis in hepatocellular carcinoma cells. World J Gastroenterol. 2009;15(5):513. doi:10.3748/wjg.15.513

50. Jardim DL, Goodman A, de Melo Gagliato D, Kurzrock R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell. 2021;39(2):154–173. doi:10.1016/j.ccell.2020.10.001

51. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. New England J Med. 2014;371(23):2189–2199. doi:10.1056/NEJMoa1406498

52. Goodman AM, Kato S, Bazhenova L, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16(11):2598–2608. doi:10.1158/1535-7163.MCT-17-0386

53. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1). doi:10.1186/s13073-017-0424-2

54. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421. doi:10.1038/nature12477

55. Gabbia D, De Martin S. Tumor mutational burden for predicting prognosis and therapy outcome of hepatocellular carcinoma. Int J Mol Sci. 2023;24(4):3441. doi:10.3390/ijms24043441

56. Wong M, Kim JT, Cox B, et al. Evaluation of tumor mutational burden in small early hepatocellular carcinoma and progressed hepatocellular carcinoma. Hepat Oncol. 2021;8(4). doi:10.2217/hep-2020-0034

57. Zheng J, Shao M, Yang W, Ren J, Chen X, Yang H. Benefits of combination therapy with immune checkpoint inhibitors and predictive role of tumour mutation burden in hepatocellular carcinoma: a systematic review and meta-analysis. Int Immunopharmacol. 2022;2022:112.

58. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10. doi:10.1016/j.immuni.2013.07.012

59. Leone P, Shin EC, Perosa F, Vacca A, Dammacco F, Racanelli V. MHC class I antigen processing and presenting machinery: organization, function, and defects in tumor cells. J Natl Cancer Inst. 2013;105(16):1172–1187.

60. Wang C, Wang Z, Yao T, Zhou J, Wang Z. The immune-related role of beta-2-microglobulin in melanoma. Front Oncol. 2022;2022:12.

61. Yoshihama S, Roszik J, Downs I, et al. NLRC5/MHC class I transactivator is a target for immune evasion in cancer. Proc Natl Acad Sci U S A. 2016;113(21):5999–6004. doi:10.1073/pnas.1602069113

62. Yamamoto T, Gotoh M, Sasaki H, Terada M, Kitajima M, Hirohashi S. Molecular cloning and initial characterization of a novel fibrinogen-related gene, HFREP-1. Biochem Biophys Res Commun. 1993;193(2):681–687. doi:10.1006/bbrc.1993.1678

63. Hara H, Uchida S, Yoshimura H, et al. Isolation and characterization of a novel liver-specific gene, hepassocin, upregulated during liver regeneration. Biochim Biophys Acta. 2000;1492(1):31–44. doi:10.1016/S0167-4781(00)00056-7

64. He X, Xu C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 2020;30(8):660–669. doi:10.1038/s41422-020-0343-4

65. Wang J, Sanmamed MF, Datar I, et al. Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell. 2019;176(1–2):334–347.e12. doi:10.1016/j.cell.2018.11.010

66. Dutta S, Ganguly A, Chatterjee K, Spada S, Mukherjee S. Targets of immune escape mechanisms in cancer: basis for development and evolution of cancer immune checkpoint inhibitors. Biology. 2023;12(2). doi:10.3390/biology12020218

67. Hua N, Chen A, Yang C, et al. The correlation of fibrinogen-like protein-1 expression with the progression and prognosis of hepatocellular carcinoma. Mol Biol Rep. 2022;49(8):7911–7919. doi:10.1007/s11033-022-07624-6

68. Yan Q, Lin HM, Zhu K, et al. Immune checkpoint FGL1 expression of circulating tumor cells is associated with poor survival in curatively resected hepatocellular carcinoma. Front Oncol. 2022;2022:12.

69. Davda J, Declerck P, Hu-Lieskovan S, et al. Immunogenicity of immunomodulatory, antibody-based, oncology therapeutics. J Immunother Cancer. 2019;7(1). doi:10.1186/s40425-019-0586-0

70. van Brummelen EMJ, Ros W, Wolbink G, Beijnen JH, Schellens JHM. Antidrug antibody formation in oncology: clinical relevance and challenges. Oncologist. 2016;21(10):1260–1268. doi:10.1634/theoncologist.2016-0061

71. Doevendans E, Schellekens H. Immunogenicity of innovative and biosimilar monoclonal antibodies. Antibodies. 2019;8(1):21. doi:10.3390/antib8010021

72. Enrico D, Paci A, Chaput N, Karamouza E, Besse B. Antidrug antibodies against immune checkpoint blockers: impairment of drug efficacy or indication of immune activation? Clin Cancer Res. 2020;26(4):787–792. doi:10.1158/1078-0432.CCR-19-2337

73. Vaisman-Mentesh A, Gutierrez-Gonzalez M, DeKosky BJ, Wine Y, Zhu L. The molecular mechanisms that underlie the immune biology of anti-drug antibody formation following treatment with monoclonal antibodies. Front Immunol. 2020;11:11. doi:10.3389/fimmu.2020.00011

74. Kim C, Yang H, Kim I, et al. Association of high levels of antidrug antibodies against atezolizumab with clinical outcomes and T-cell responses in patients with hepatocellular carcinoma. JAMA Oncol. 2022;8(12):1825–1829. doi:10.1001/jamaoncol.2022.4733

75. Yang JD, Nakamura I, Roberts LR. The tumor microenvironment in hepatocellular carcinoma: current status and therapeutic targets. Semin Cancer Biol. 2011;21(1):35–43. doi:10.1016/j.semcancer.2010.10.007

76. Birgani MT, Carloni V. Tumor microenvironment, a paradigm in hepatocellular carcinoma progression and therapy. Int J Mol Sci. 2017;18:2.

77. Llovet JM, Castet F, Heikenwalder M, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 2022;19(3):151–172. doi:10.1038/s41571-021-00573-2

78. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases-elimination, equilibrium and escape. Curr Opin Immunol. 2014;27(1):16–25. doi:10.1016/j.coi.2014.01.004

79. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331(6024):1565–1570. doi:10.1126/science.1203486

80. Sanceau J, Gougelet A. Epigenetic mechanisms of liver tumor resistance to immunotherapy. World J Hepatol. 2021;13(9):979–1002. doi:10.4254/wjh.v13.i9.979

81. Sheng J, Zhang J, Wang L, et al. Topological analysis of hepatocellular carcinoma tumour microenvironment based on imaging mass cytometry reveals cellular neighbourhood regulated reversely by macrophages with different ontogeny. Gut. 2022;71(6):1176–1191. doi:10.1136/gutjnl-2021-324339

82. Lee RY, Ng CW, Rajapakse MP, Ang N, Yeong JPS, Lau MC. The promise and challenge of spatial omics in dissecting tumour microenvironment and the role of AI. Front Oncol. 2023;13:1172314. doi:10.3389/fonc.2023.1172314

83. Feng H, Zhuo Y, Zhang X, et al. Tumor microenvironment in hepatocellular carcinoma: key players for immunotherapy. J Hepatocell Carcinoma. 2022;9:1109–1125. doi:10.2147/JHC.S381764

84. Workman CJ, Vignali DAA. The CD4-related molecule, LAG-3 (CD223), regulates the expansion of activated T cells. Eur J Immunol. 2003;33(4):970–979. doi:10.1002/eji.200323382

85. Triebel F, Jitsukawa S, Baixeras E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171:1393–1405. doi:10.1084/jem.171.5.1393

86. Huard B, Mastrangeli R, Prigent P, et al. Characterization of the major histocompatibility complex class II binding site on LAG-3 protein. Proceed Natl Acad Sci. 1997;94:5744–5749. doi:10.1073/pnas.94.11.5744

87. Blackburn SD, Shin H, Haining WN, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37. doi:10.1038/ni.1679

88. Tian X, Zhang A, Qiu C, et al. The upregulation of LAG-3 on T cells defines a subpopulation with functional exhaustion and correlates with disease progression in HIV-infected subjects. J Immunol. 2015;194(8):3873–3882. doi:10.4049/jimmunol.1402176

89. Phillips BL, Mehra S, Ahsan MH, Selman M, Khader SA, Kaushal D. LAG3 expression in active mycobacterium tuberculosis infections. Am J Pathol. 2015;185(3):820–833. doi:10.1016/j.ajpath.2014.11.003

90. Butler NS, Moebius J, Pewe LL, et al. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat Immunol. 2012;13(2):188–195. doi:10.1038/ni.2180

91. Demeure CE, Wolfers J, Martin-Garcia N, Gaulard P, Triebel F. T Lymphocytes infiltrating various tumour types express the MHC class II ligand lymphocyte activation gene-3 (LAG-3): role of LAG-3/MHC class II interactions in cell-cell contacts. Eur J Cancer. 2001;37(13):1709–1718. doi:10.1016/S0959-8049(01)00184-8

92. Gandhi MK, Lambley E, Duraiswamy J, et al. Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients. Blood. 2006;108(7):2280–2289. doi:10.1182/blood-2006-04-015164

93. Li FJ, Zhang Y, Jin GX, Yao L, Wu DQ. Expression of LAG-3 is coincident with the impaired effector function of HBV-specific CD8+ T cell in HCC patients. Immunol Lett. 2013;150(1–2):116–122. doi:10.1016/j.imlet.2012.12.004

94. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A. 2010;107(17):7875–7880. doi:10.1073/pnas.1003345107

95. Thommen DS, Schreiner J, Müller P, et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol Res. 2015;3(12):1344–1354. doi:10.1158/2326-6066.CIR-15-0097

96. Yang ZZ, Kim HJ, Villasboas JC, et al. Expression of LAG-3 defines exhaustion of intratumoral PD-1+ T cells and correlates with poor outcome in follicular lymphoma. Oncotarget. 2017;8(37):61425–61439.

97. Annunziato F, Manetti R, Tomasévic I, et al. Expression and release of LAG-3-encoded protein by human CD4+ T cells are associated with IFN-gamma production. FASEB J. 1996;10(7):769–776. doi:10.1096/fasebj.10.7.8635694

98. Maruhashi T, Sugiura D, Okazaki IM, Okazaki T. LAG-3: from molecular functions to clinical applications. J Immunother Cancer. 2020;8(2):e001014. doi:10.1136/jitc-2020-001014

99. Okazaki T, Okazaki IM, Wang J, et al. PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J Exp Med. 2011;208(2):395–407. doi:10.1084/jem.20100466

100. Andrews LP, Marciscano AE, Drake CG, Vignali DAA. LAG3 (CD223) as a cancer immunotherapy target. Immunol Rev. 2017;276(1):80–96. doi:10.1111/imr.12519

101. Huo JL, Wang YT, Fu WJ, Lu N, Liu ZS. The promising immune checkpoint LAG-3 in cancer immunotherapy: from basic research to clinical application. Front Immunol. 2022;2022:13.

102. Yu X, Harden K, Gonzalez LC, 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. doi:10.1038/ni.1674

103. Stanietsky N, Simic H, Arapovic J, et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A. 2009;106(42):17858–17863. doi:10.1073/pnas.0903474106

104. Jantz-Naeem N, Böttcher-Loschinski R, Borucki K, et al. TIGIT signaling and its influence on T cell metabolism and immune cell function in the tumor microenvironment. Front Oncol. 2023;2023:13.

105. Joller N, Hafler JP, Brynedal B, et al. Cutting edge: TIGIT Has T cell-intrinsic inhibitory functions. J Immunol. 2011;186(3):1338–1342. doi:10.4049/jimmunol.1003081

106. Levin SD, Taft DW, Brandt CS, et al. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. Eur J Immunol. 2011;41(4):902–915. doi:10.1002/eji.201041136

107. Jin H, Qin S, He J, et al. New insights into checkpoint inhibitor immunotherapy and its combined therapies in hepatocellular carcinoma: from mechanisms to clinical trials. Int J Biol Sci. 2022;18(7):2775–2794. doi:10.7150/ijbs.70691

108. Ge Z, Zhou G, Campos Carrascosa L, et al. TIGIT and PD1 co-blockade restores ex vivo functions of human tumor-infiltrating CD8+ T cells in hepatocellular carcinoma. CMGH. 2021;12(2):443–464. doi:10.1016/j.jcmgh.2021.03.003

109. Zheng Q, Xu J, Gu X, et al. Immune checkpoint targeting TIGIT in hepatocellular carcinoma. Am J Transl Res. 2020;12(7):3212.

110. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486–499. doi:10.1038/nri3862

111. Khan O, Giles JR, McDonald S, et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature. 2019;571(7764):211–218. doi:10.1038/s41586-019-1325-x

112. Jiang W, He Y, He W, et al. Exhausted CD8+T cells in the tumor immune microenvironment: new pathways to therapy. Front Immunol. 2021;2021:11.

113. Zheng C, Zheng L, Yoo JK, et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell. 2017;169(7):1342–1356.e16. doi:10.1016/j.cell.2017.05.035

114. Ma J, Zheng B, Goswami S, et al. PD1Hi CD8+ T cells correlate with exhausted signature and poor clinical outcome in hepatocellular carcinoma. J Immunother Cancer. 2019;7(1). doi:10.1186/s40425-019-0814-7

115. Barsch M, Salié H, Schlaak AE, et al. T-cell exhaustion and residency dynamics inform clinical outcomes in hepatocellular carcinoma. J Hepatol. 2022;77(2):397–409. doi:10.1016/j.jhep.2022.02.032

116. Colonna M. The biology of TREM receptors. Nat Rev Immunol. 2023;23:580–594. doi:10.1038/s41577-023-00837-1

117. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol. 2000;164(10):4991–4995. doi:10.4049/jimmunol.164.10.4991

118. Ouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature. 2001;410(6832):1103–1107. doi:10.1038/35074114

119. Gibot S, Kolopp-Sarda MN, Béné MC, et al. Plasma level of a triggering receptor expressed on myeloid cells-1: its diagnostic accuracy in patients with suspected sepsis. Ann Intern Med. 2004;141(1):9–15. doi:10.7326/0003-4819-141-1-200407060-00009

120. Wang DY, Qin RY, Liu ZR, Gupta K, Chang Q. Expression of TREM-1 mRNA in acute pancreatitis. World J Gastroenterol. 2345;10(18):86–96.

121. Koussoulas V, Vassiliou S, Demonakou M, et al. Soluble triggering receptor expressed on myeloid cells (sTREM-1): a new mediator involved in the pathogenesis of peptic ulcer disease. Eur J Gastroenterol Hepatol. 2006;18(4):375–379. doi:10.1097/00042737-200604000-00010

122. Zysset D, Weber B, Rihs S, et al. TREM-1 links dyslipidemia to inflammation and lipid deposition in atherosclerosis. Nat Commun. 2016;7. doi:10.1038/ncomms13151

123. Choi ST, Kang EJ, Ha YJ, Song JS. Levels of plasma-soluble triggering receptor expressed on myeloid cells-1 (sTREM-1) are correlated with disease activity in rheumatoid arthritis. J Rheumatol. 2012;39(5):933–938. doi:10.3899/jrheum.111218

124. Ho CC, Liao WY, Wang CY, et al. TREM-1 expression in tumor-associated macrophages and clinical outcome in lung cancer. Am J Respir Crit Care Med. 2008;177(7):763–770. doi:10.1164/rccm.200704-641OC

125. Nguyen-Lefebvre AT, Ajith A, Portik-Dobos V, et al. The innate immune receptor TREM-1 promotes liver injury and fibrosis. J Clin Invest. 2018;128(11):4870–4883. doi:10.1172/JCI98156

126. Wu J, Li J, Salcedo R, Mivechi NF, Trinchieri G, Horuzsko A. The proinflammatory myeloid cell receptor TREM-1 controls Kupffer cell activation and development of hepatocellular carcinoma. Cancer Res. 2012;72(16):3977–3986. doi:10.1158/0008-5472.CAN-12-0938

127. Liao R, Sun TW, Yi Y, et al. Expression of TREM-1 in hepatic stellate cells and prognostic value in hepatitis B-related hepatocellular carcinoma. Cancer Sci. 2012;103(6):984–992. doi:10.1111/j.1349-7006.2012.02273.x

128. Muller M, Haghnejad V, Lopez A, et al. Triggering receptors expressed on myeloid cells 1: our new partner in human oncology? Front Oncol. 2022;2022:12.

129. Chen J, Gingold JA, Su X. Immunomodulatory TGF-β Signaling in Hepatocellular Carcinoma. Trends Mol Med. 2019;25(11):1010–1023. doi:10.1016/j.molmed.2019.06.007

130. Soukupova J, Malfettone A, Bertran E, et al. Epithelial–mesenchymal transition (Emt) induced by tgf-β in hepatocellular carcinoma cells reprograms lipid metabolism. Int J Mol Sci. 2021;22(11):5543. doi:10.3390/ijms22115543

131. Park BV, Freeman ZT, Ghasemzadeh A, et al. TGFβ1-mediated SMAD3 enhances PD-1 expression on antigen-specific T cells in cancer. Cancer Discov. 2016;6(12):1366–1381. doi:10.1158/2159-8290.CD-15-1347

132. Yin C, Evason KJ, Asahina K, Stainier DYR. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 2013;123(5):1902–1910. doi:10.1172/JCI66369

133. Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev. 2008;88(1):125–172. doi:10.1152/physrev.00013.2007

134. Li D, Friedman SL. Liver fibrogenesis and the role of hepatic stellate cells: new insights and prospects for therapy. J Gastroenterol Hepatol. 1999;14(7):618–633. doi:10.1046/j.1440-1746.1999.01928.x

135. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209–218. doi:10.1172/JCI24282

136. Friedman SL. Mechanisms of Hepatic Fibrogenesis. Gastroenterology. 2008;134(6):1655–1669. doi:10.1053/j.gastro.2008.03.003

137. Yu G, Jing Y, Kou X, et al. Hepatic stellate cells secreted hepatocyte growth factor contributes to the chemoresistance of hepatocellular carcinoma. PLoS One. 2013;8:9.

138. Quiroz Reyes AG, Lozano Sepulveda SA, Martinez-Acuña N, et al. Cancer stem cell and hepatic stellate cells in hepatocellular carcinoma. Technol Cancer Res Treat. 2023;2023:22.

139. Hsieh CC, Hung CH, Chiang M, Tsai YC, He JT. Hepatic stellate cells enhance liver cancer progression by inducing myeloid-derived suppressor cells through interleukin-6 signaling. Int J Mol Sci. 2019;20(20):5079. doi:10.3390/ijms20205079

140. Höchst B, Schildberg FA, Sauerborn P, 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. doi:10.1016/j.jhep.2013.04.033

141. Yu MC, Chen CH, Liang X, 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. doi:10.1002/hep.20488

142. Dunham RM, Thapa M, Velazquez VM, et al. Hepatic stellate cells preferentially induce Foxp3+ regulatory T cells by production of retinoic acid. J Immunol. 2013;190(5):2009–2016. doi:10.4049/jimmunol.1201937

143. Xing F, Saidou J, Watabe K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front Biosci. 2010;15(1):166–179. doi:10.2741/3613

144. Baglieri J, Brenner DA, Kisseleva T. The role of fibrosis and liver-associated fibroblasts in the pathogenesis of hepatocellular carcinoma. Int J Mol Sci. 2019;20(7):1723. doi:10.3390/ijms20071723

145. Gascard P, Tlsty TD. Carcinoma-associated fibroblasts: orchestrating the composition of malignancy. Genes Dev. 2016;30(9):1002–1019. doi:10.1101/gad.279737.116

146. Mao X, Xu J, Wang W, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021;20(1). doi:10.1186/s12943-021-01428-1

147. Liu G, Sun J, Yang ZF, et al. Cancer-associated fibroblast-derived CXCL11 modulates hepatocellular carcinoma cell migration and tumor metastasis through the circUBAP2/miR-4756/IFIT1/3 axis. Cell Death Dis. 2021;12:3.

148. Xu H, Zhao J, Li J, et al. Cancer associated fibroblast–derived CCL5 promotes hepatocellular carcinoma metastasis through activating HIF1α/ZEB1 axis. Cell Death Dis. 2022;13(5). doi:10.1038/s41419-022-04935-1

149. Cheng JT, Deng YN, Yi HM, et al. Hepatic carcinoma-associated fibroblasts induce ido-producing regulatory dendritic cells through il-6-mediated stat3 activation. Oncogenesis. 2016;5(2):e198–e198. doi:10.1038/oncsis.2016.7

150. Deng Y, Cheng J, Fu B, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–1101. doi:10.1038/onc.2016.273

151. Harper J, Sainson RCA. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin Cancer Biol. 2014;25:69–77. doi:10.1016/j.semcancer.2013.12.005

152. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol. 2018;9(MAR). doi:10.3389/fimmu.2018.00414

153. Lakins MA, Ghorani E, Munir H, Martins CP, Shields JD. Cancer-associated fibroblasts induce antigen-specific deletion of CD8 + T Cells to protect tumour cells. Nat Commun. 2018;9(1). doi:10.1038/s41467-018-03347-0

154. Bagaev A, Kotlov N, Nomie K, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021;39(6):845–865.e7. doi:10.1016/j.ccell.2021.04.014

155. Liu Y, Xun Z, Ma K, et al. Identification of a tumour immune barrier in the HCC microenvironment that determines the efficacy of immunotherapy. J Hepatol. 2023;78(4):770–82.

156. Zhang C, Fei Y, Wang H, et al. CAFs orchestrates tumor immune microenvironment—A new target in cancer therapy? Front Pharmacol. 2023;2023:14.

157. Akkız H. Emerging role of cancer-associated fibroblasts in progression and treatment of hepatocellular carcinoma. Int J Mol Sci. 2023;24(4):3941. doi:10.3390/ijms24043941

158. Geva-Zatorsky N, Sefik E, Kua L, et al. Mining the human gut microbiota for immunomodulatory organisms. Cell. 2017;168(5):928–943.e11. doi:10.1016/j.cell.2017.01.022

159. Kayama H, Okumura R, Takeda K. Interaction between the microbiota, epithelia, and immune cells in the intestine. Takeda ARjats Cls. 2020;2020:5.

160. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi:10.1126/science.aan3706

161. Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi:10.1126/science.aad1329

162. Ismail AS, Behrendt CL, Hooper LV. Reciprocal interactions between commensal bacteria and γδ Intraepithelial lymphocytes during mucosal injury. J Immunol. 2009;182(5):3047–3054. doi:10.4049/jimmunol.0802705

163. Jakobsson HE, Jernberg C, Andersson AF, Sjölund-Karlsson M, Jansson JK, Engstrand L. Short-term antibiotic treatment has differing long- term impacts on the human throat and gut microbiome. PLoS One. 2010;5(3):e9836. doi:10.1371/journal.pone.0009836

164. Scott NA, Andrusaite A, Andersen P, et al. Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci Transl Med. 2018;10(464). doi:10.1126/scitranslmed.aao4755

165. Jackson MA, Goodrich JK, Maxan ME, et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut. 2016;65(5):749–756. doi:10.1136/gutjnl-2015-310861

166. Freedberg DE, Lebwohl B, Abrams JA. The impact of proton pump inhibitors on the human gastrointestinal microbiome. Clin Lab Med. 2014;34(4):771–785. doi:10.1016/j.cll.2014.08.008

167. Huang EY, Inoue T, Leone VA, et al. Using corticosteroids to reshape the gut microbiome: implications for inflammatory bowel diseases. Inflamm Bowel Dis. 2015;21(5):963–972. doi:10.1097/MIB.0000000000000332

168. Cortellini A, Facchinetti F, Derosa L, et al. Antibiotic exposure and immune checkpoint inhibitors in patients with NSCLC: the backbone matters. J Thoracic Oncol. 2022;17(6):739–741. doi:10.1016/j.jtho.2022.03.016

169. Cortellini A, Tucci M, Adamo V, et al. Integrated analysis of concomitant medications and oncological outcomes from PD-1/PD-L1 checkpoint inhibitors in clinical practice. J Immunother Cancer. 2020;8(2):e001361. doi:10.1136/jitc-2020-001361

170. Cortellini A, Di Maio M, Nigro O, et al. Differential influence of antibiotic therapy and other medications on oncological outcomes of patients with non-small cell lung cancer treated with first-line pembrolizumab versus cytotoxic chemotherapy. J Immunother Cancer. 2021;9(4):e002421. doi:10.1136/jitc-2021-002421

171. Pinato DJ, Howlett S, Ottaviani D, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 2019;5(12):1774–1778. doi:10.1001/jamaoncol.2019.2785

172. Pinato DJ, Li X, Mishra-Kalyani P, et al. Association between antibiotics and adverse oncological outcomes in patients receiving targeted or immune-based therapy for hepatocellular carcinoma. JHEP Reports. 2023;5(6):100747. doi:10.1016/j.jhepr.2023.100747

173. Naqash AR, Kihn-Alarcón AJ, Stavraka C, et al. The role of gut microbiome in modulating response to immune checkpoint inhibitor therapy in cancer. Ann Transl Med. 2021;9(12):1034–1034. doi:10.21037/atm-20-6427

174. Rizzo A, Brandi G. Biochemical predictors of response to immune checkpoint inhibitors in unresectable hepatocellular carcinoma. Cancer Treat Res Commun. 2021;2021:27.

175. Morita M, Nishida N, Aoki T, et al. Role of β-catenin activation in the tumor immune microenvironment and immunotherapy of hepatocellular carcinoma. Cancers. 2023;15(8):2311. doi:10.3390/cancers15082311

176. Wu Q, Zhou W, Yin S, et al. Blocking triggering receptor expressed on myeloid cells-1-positive tumor-associated macrophages induced by hypoxia reverses immunosuppression and anti-programmed cell death ligand 1 resistance in liver cancer. Hepatology. 2019;70(1):198–214. doi:10.1002/hep.30593

177. Han CL, Yan YC, Yan LJ, et al. Efficacy and security of tumor vaccines for hepatocellular carcinoma: a systemic review and meta-analysis of the last 2 decades. J Cancer Res Clin Oncol. 2022;149:1425–1441. doi:10.1007/s00432-022-04008-y

178. Yang YQ, Wen ZY, Liu XY, et al. Current status and prospect of treatments for recurrent hepatocellular carcinoma. World J Hepatol. 2023;15(2):129–150. doi:10.4254/wjh.v15.i2.129

179. Repáraz D, Aparicio B, Llopiz D, et al. Therapeutic vaccines against hepatocellular carcinoma in the immune checkpoint inhibitor era: time for neoantigens? Int J Mol Sci. 2022;23(4):2022. doi:10.3390/ijms23042022

180. Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immune-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15(1):28. doi:10.1186/s13045-022-01247-x

181. Buonaguro L. New vaccination strategies in liver cancer. Cytokine Growth Factor Rev. 2017;36:125–129. doi:10.1016/j.cytogfr.2017.06.010

182. Ott PA, Hu-Lieskovan S, Chmielowski B, et al. A phase ib trial of personalized neoantigen therapy plus Anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. Cell. 2020;183(2):347–362.e24. doi:10.1016/j.cell.2020.08.053

183. Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547(7662):222–226. doi:10.1038/nature23003

184. Sadelain M, Brentjens R, Rivière I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol. 2009;21(2):215–223. doi:10.1016/j.coi.2009.02.009

185. June CH, Sadelain M. Chimeric antigen receptor therapy. New England J Med. 2018;379(1):64–73. doi:10.1056/NEJMra1706169

186. Neeson P, Shin A, Tainton KM, et al. Ex vivo culture of chimeric antigen receptor T cells generates functional CD8 T cells with effector and central memory-like phenotype. Gene Ther. 2010;17(9):1105–1116. doi:10.1038/gt.2010.59

187. Sheikh S, Migliorini D, Lang N. CAR T-based therapies in lymphoma: a review of current practice and perspectives. Biomedicines. 2022;10(8):1960. doi:10.3390/biomedicines10081960

188. Martinez M, Moon EK. CAR T cells for solid tumors: new strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front Immunol. 2019;10(FEB). doi:10.3389/fimmu.2019.00128

189. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69. doi:10.1038/s41408-021-00459-7

190. Hegde M, Mukherjee M, Grada Z, et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest. 2016;126(8):3036–3052. doi:10.1172/JCI83416

191. Haruyama Y, Kataoka H. Glypican-3 is a prognostic factor and an immunotherapeutic target in hepatocellular carcinoma. World J Gastroenterol. 2016;22(1):275–283. doi:10.3748/wjg.v22.i1.275

192. Yamauchi N, Watanabe A, Hishinuma M, et al. The glypican 3 oncofetal protein is a promising diagnostic marker for hepatocellular carcinoma. Mod Pathol. 2005;18(12):1591–1598. doi:10.1038/modpathol.3800436

193. Gao H, Li K, Tu H, et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. 2014;20(24):6418–6428. doi:10.1158/1078-0432.CCR-14-1170

Creative Commons License © 2023 The Author(s). This work is published by Dove Medical Press Limited, and licensed under a Creative Commons Attribution License. The full terms of the License are available at http://creativecommons.org/licenses/by/4.0/. The license permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.