Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01.12.2019 | Review

From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma

verfasst von: Yaojie Fu, Shanshan Liu, Shan Zeng, Hong Shen

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2019

Abstract

Hepatocellular carcinoma (HCC) ranks the most common primary liver malignancy and the third leading cause of tumor-related mortality worldwide. Unfortunately, despite advances in HCC treatment, less than 40% of HCC patients are eligible for potentially curative therapies. Recently, cancer immunotherapy has emerged as one of the most promising approaches for cancer treatment. It has been proven therapeutically effective in many types of solid tumors, such as non-small cell lung cancer and melanoma. As an inflammation-associated tumor, it’s well-evidenced that the immunosuppressive microenvironment of HCC can promote immune tolerance and evasion by various mechanisms. Triggering more vigorous HCC-specific immune response represents a novel strategy for its management. Pre-clinical and clinical investigations have revealed that various immunotherapies might extend current options for needed HCC treatment. In this review, we provide the recent progress on HCC immunology from both basic and clinical perspectives, and discuss potential advances and challenges of immunotherapy in HCC.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACT
Adoptive cell transfer
AFP
A-fetoprotein
APCs
Antigen presenting cells
CAF
Cancer associated fibroblast
CAR-T
Chimeric antigen receptor-modified T cells
CCL2
Chemokine (C-C motif) ligand 2
CIK
Cytokine-induced killer
CSCs
Cancer stem cells
CTAs
Cancer-testis antigens
CTLA-4
Cytotoxic T lymphocyte protein 4
CTLs
Cytotoxic T lymphocytes
CXCL17
Chemokine (C-X-C motif) ligand 17
CXCR4
Chemokine (C-X-C motif) receptor 4
DCs
Dendritic cells
DEXs
Dendritic cell-derived exosomes
GPC-3
Glypican-3
HCC
Hepatocellular carcinoma
HGF
Hepatocyte growth factor
HSCs
Hepatic stellate cells
hTERT
human telomerase-reverse transcriptase
ICB
Immune-checkpoint blockade
IDO
Indoleamine-2,3-dioxygenase
IFN-γ
Interferon-γ
IL-
Interleukin-
KC
Kupffer cell
LAG-3
Lymphocyte activation gene 3
MAGE-1
Melanoma-associated antigen 1
MCP-1
Monocyte chemotactic protein 1
MDSCs
Myeloid- derived suppressor cells
MTD
Maximum-tolerated dose
NK cell
Natural killer cell
OS
Overall survival
OVs
Oncolytic viruses
P
Primary endpoint
PD-1
Programmed cell death protein-1
PDGF
Platelet-derived growth factor
PD-L1
Programmed cell death protein ligand-1
PR
Partial response
RECIST
Response evaluation criteria in solid tumors
RR
Response rate
S
Secondary endpoint
SD
Stable disease
SDF-1α
Stromal cell derived factor 1α
TAA
Tumor-associated antigens
TAMs
Tumor-associated macrophages
TANs
Tumor associated neutrophils
TEXs
Tumor cell–derived exosomes
TGF-β
Transforming growth factor-β
Tim-3
Mucin domain-containing molecule-3
TME
Tumor microenvironment
Tregs
Regulatory T cells
TTP
Time to progression
TTSP
Time to symptomatic progression
VEGF
Vascular endothelial growth factor

Background

Hepatocellular carcinoma (HCC) represents the most common type of primary liver cancer, with a global incidence of 500,000 new cases per year [1]. HCC is closely associated with chronic liver inflammation and some well-known risk factors, including chronic HBV and HCV infections, alcohol consumption, diabetes mellitus and several metabolic diseases [2]. The current therapeutic options available for HCC, such as transarterial chemoembolization (TACE), radiofrequency ablation, surgical resection and transplantation, are only curative for some patients in early stages. Other more effective approaches emerged in the past few years, such as tyrosine kinase inhibitors (TKIs) targeting angiogenesis (e.g. Sorafenib, lenvatinib, regorafenib) [3], clinically tested selective Cyclin dependent kinase 5 and 4/6 (Cdk5, Cdk4/6) inhibitors (Dinaciclib & Palbociclib) [46], and highly selective fibroblast growth factor receptor 4 (FGFR4) inhibitor H3B-6527 [7, 8], which pre-clinically and clinically show encouraging efficacy and have been rigorously pursued for advanced HCC.
The liver is a ‘tolerogenic’ organ that can arouse its immune responses to prevent undesirable pathogen attack and tumor initiation. However, as a typical inflammation-linked tumorigenesis, immune evasion is one of the features occurring during the initiation and evolution of HCC [9]. A number of immune suppressor mechanisms, including intratumoral accumulation of immunosuppressive cell populations, defective antigen presentation and activation of multiple inhibitory receptor-ligand pathways, favor tolerance over immunity, and promote progression of HCC [10, 11]. The magnitude of immune suppression in the tumor microenvironment (TME) is closely correlated with poor prognosis in HCC patients. Hence, for better arousing anti-tumor immunity, more details about suppressed immune landscape of HCC urgently needs to be elucidated.

The intricate immune network in TME of HCC

The HCC tumor microenvironment (TME) is a dynamic system, which comprises cancer cells, the intricate cytokine environment, extracellular matrix, immune cell subsets and other components [12]. It’s well established that the immune landscape of HCC has a strong suppressor feature. In this complex network, the pro-tumorigenic immune response, mediated by diverse immunosuppressive cell subsets, secretions and signaling, plays a pivotal role in driving immune evasion [13] (Fig. 1.). Moreover, ‘fatigue’ of anti-tumor immunity also contributes to tumor tolerance and progression. Here, we discuss new advances in the immunosuppressive picture of HCC.

Representative immunosuppressive components in TME of HCC

Myeloid-derived suppressor cells (MDSCs)

MDSCs is a heterogeneous population of immature myeloid cells (IMCs), which are expanded in pathological conditions and up-regulate expression of immune suppressive factors, such as arginase and inducible nitric oxide synthase (iNOS or NOS2) [14]. Various tumor originated cytokines, such as G-CSF, GM-CSF, VEGF, MCP-1 and IL-1β, have been demonstrated to induce MDSCs infiltration [15]. Cell cycle related kinase (CCRK) represents a novel signaling target for cancer immunotherapy [16]. Emerging evidence also indicates the hepatoma-intrinsic CCRK upregulates interlukin-6 (IL-6) production through EZH2/NF-κB signaling, which consequently induce MDSCs accumulation in TME [17]. Hepatic carcinoma related tumor-associated fibroblasts (TAFs), a stromal part in HCC, can induce peripheral blood monocyte migration and differentiation into CD14 + HLA-DR −/low MDSCs by SDF-1α. TAFs mediate the generation of MDSCs via IL-6/STAT3 signaling [18]. In addition, local hypoxia has been identified as a key regulator that can promote MDSCs accumulation through the Chemokine C-C motif Ligand 26 (CCL26)/CX3CR1 pathway [19]. Hypoxia-inducible factor 1α (HIF-1α) mediated ENTPD2 over-expression in HCC cells, has been proven to increase the extracellular level of 5′-AMP, which subsequently recruit MDSCs into TME [20].
As a powerful inhibitory immune modulator, infiltrated MDSCs in HCC damage effector T cells, expand immune checkpoink signaling, decrease NK cell cytotoxicity and cytokine production by diverse mechanisms [21, 22]. The MDSCs in fibrotic HCC tissue are notably correlated with reduced tumor infiltrating lymphocytes (TILs) and elevated tumorigenicity, aggressive phenotype, moreover, whose activation and infiltration contribute greatly to worse survival rate both in mouse model and HCC patients [23]. CD14+ HLA-DR −/low MDSCs can blunt HCC immunity through inducing activation of CD4+ CD25+ Foxp3+ regulatory T cells, inhibiting proliferation and IFN-γ secretion of CD3/CD28-stimulated autologous peripheral blood mononuclear cells (PBMCs) [24]. T cell function is impaired due to competition for energy resources (e.g. arginine and cysteine) with MDSCs [25], as well as involvement of several inhibitory receptor-ligand pathways in MDSCs-mediated immune evasion. Tumor-derived TGF-β triggers recruitment of MDSCs in a CXCL1/2/5- and CXCR2-dependent manner. The infiltrated MDSCs selectively suppress IFN-γ production deriving from NKT cells [26]. MDSCs also can express galectin-9 that binds to TIM-3 on T cells, inducing T-cell apoptosis [27]. In addition, it’s suggested that MDSCs in advanced HCC patients may interact with Kuppfer cells to induce PD-L1 expression, and mediate inhibited cytotoxicity and cytokine release of NK cells through the NKp30 receptor [28]. Taken together, MDSCs exert versatile immunosuppressive effects in HCC. Combined treatment with anti-PD-1/PD-L1 and concomitant targeting MDSCs (such as CCRK inhibition or p38 MAPK inhibitor) may synergistically enhance efficacy to eradicate HCC [17, 23]. In addition, recent evidence suggests radiation and IL-12 combination therapy (RT/IL-12) may elevate anti-tumor immunity in HCC by reducing MDSCs accumulation and the production of reactive oxygen species (ROS) [29]. Hence, MDSCs may serve as a potential target for resetting immunotorelant state in HCC tumors.

Tumor-associated macrophages (TAMs)

Macrophages arise from bone marrow-derived circulating monocytes, and then reside in normal tissues. The two polarizing phenotypes M1 and M2, are highly plastic in response to complex stimuli. Substantial clinical data and experimental research confirmed that alternatively activated status macrophages, the M2 phenotype, stimulate tumor initiation, progression and malignant metastasis by various mechanisms [30, 31]. In HCC, some specific populations of the immunosuppressive tumor-associated macrophages (TAMs) have emerged as a research hotspot recently. The well-identified HCC-derived cytokines, such as IL-4, IL-13, CSF-1, CCL2, CXCL12, connective tissue growth factor (CTGF) [3234], induce TAMs differentiation from CCR2+ inflammatory monocytes, alternatively activated macrophages (AAMs) infiltration, then reduce innate or adaptive immunity [34, 35]. Osteopontin (OPN) expressed by HCC cells not only has a positive association with PD-L1 expression in HCC, moreover, it facilitates alternative activation and chemotactic migration of TAMs via CSF1-CSF1R pathway in TAMs [36]. HCC cells originated HIF-1α has been recently suggested to enhance IL-1β release by TAMs via TLR4/TRIF/NF-κB signaling pathway, which fosters EMT and immune evasion of HCC [37]. Crosstalk between MDSCs and TAMs results in decreased production of IL-6, IL-12, down-expression of MHCII, and elevated production of IL-10, a strong inhibitory mediator that impairs downstream CD8+ T cell and NK cell cytotoxicity [38]. TAMs-derived IL-10 also increases intratumoral Foxp3+ Tregs frequency, which then suppresses CD4+CD25 T cells activation [38, 39]. TAMs in the peritumoral stroma of HCC have been shown to secrete multiple key proinflammatory cytokines (e.g. IL-1β, IL-6, IL-23, and TNF-α) and contribute to the expansion of interleukin-17-producing CD4+ T helper 17 cells (Th17), which suppress anti-tumor immunity by over-expressing several activation markers, such as PD-1, CTLA-4, and GITR [40]. In addition, TAMs are highly involved in other immune inhibitory regulations [41, 42]. TGF-β in the TME of HCC promotes the Tim-3 expression on TAMs, subsequently enabling the activated TAMs to facilitate tumor growth and immune tolerance via NF-κB signaling and downstream IL-6 production [43]. TAMs derived IL-6/STAT3 signaling also has been validated to sustain HCC carcinogenesis by promoting its carcinoma stem cells (CSCs)-liked characteristics [44].
Notably, recent evidence indicates that PD-1 TAMs can capture anti-PD-1 monoclonal antibodies (aPD-1 mAbs) through Fcγ receptors (FcγRs) expressed on the surface binding to drug’s Fc domain glycan [45]. This novel investigation indicates that blockade of FcγRs before aPD-1 mAbs administration may substantially improve checkpoint blockade therapy.

Tumor associated neutrophils (TANs)

Heterogeneity of TANs is a fundamental property that allows TANs to perform corresponding functions for adaptations to changing microenvironment. Similar to macrophages, neutrophils differently affect tumor biological behaviors depending on their polarization, either anti-tumoral (N1) and pro-tumoral (N2) phenotypes [46]. In some solid tumor models, such as lung cancer, metastatic renal cell cancer (mRCC) and melanoma, it was previously reported that TANs infiltration or neutrophil-lymphocyte ratio (NLR) closely correlate with tumor progression, which can serve as a significant predictor for monitoring patients with advanced tumor receiving anti-PD-1/PD-L1 immunotherapy [47, 48]. TANs activation is modulated by cytokines, such as Type I interferons (IFNs) and TGF-β [49]. TANs mainly suppress anti-tumor immunity via interacting with CD8+ T cells, inducing CD8+ T cells apoptosis through nitric oxide (NO) production mediated by tumor necrosis factor-α (TNF-α) [50].
The facilitator role of TANs in pathological progression of HCC has become a topic of growing interest in recent years. Clinically, TANs play a key role in driving progression and poor prognosis of HCC, and NLR is an independent predictor of survival after hepatectomy in patients with HCC [51, 52]. The newest discovery shows that loss of hypoxia associated factor, HAF (encoded by SART1) results in inappropriate HIF-1 activation, and overproduction of downstream HIF-1 dependent chemokine, RANTES. HIF-1/RANTES upregulation accumulates TANs infiltration, which is associated with non-alcoholic steatohepatitis (NASH) driven HCC initiation and progression [53]. Moreover, recent studies suggested that TANs mediate the intratumoral infiltration of TAMs and regulatory T cells by overproducing some chemokines, such as CCL2 and CCL17, which then contributes to HCC progression, metastasis and resistance to sorafenib treatment [54]. A newly identified positive feedback loop implies that TANs induce HCC stem cell like characteristics via upregulating expression of miR-301b-3p in cancer cells, and maintain hyper-action in NF-kB signaling, lead to higher secretion level of C-X-C motif chemokine5 (CXCL5) and in turn recruit more TANs infiltration [55]. In general, TANs are strongly connected with immunosuppression in HCC, but direct interactions between TANs and other components in HCC tissue and the exact underlying mechanisms behind this regulation in HCC are not yet clear.

Tumor-infiltrating lymphocytes (TILs)

A high density of tumor-infiltrating lymphocytes (TILs) was once thought to be the host’s immune reaction against cancer. Some early clinical data suggested postoperative HCC patients with high level lymphocytes infiltration, especially T cells, had reduced recurrence and better survival [56]. However, accumulating evidence suggests that the overall degree of TILs in HCC is not capable of mounting effective anti-tumor immunity to control tumor progression [57]. Intrahepatic immune response involves diverse lymphocyte populations, which contribute differently to HCC immune surveillance. The intratumoral balance of regulatory and cytotoxic T cells plays a key role in evaluating the immune state and progression of HCC [57, 58].

Regulatory T cell (Treg)

Regulatory T cells (Tregs) can be derived from peripheral blood T lymphocytes, resident T cells and other cellular sources. Its recruitment has been found to be induced by the CCR6 (CC chemokine receptor type 6)–CCL20 (CC motif chemokine 20) axis. Tregs activation is induced by T cell receptor (TCR) engagement concurrent with IL-10 and TGF-β signaling [59]. Apart from activation via pro-inflammatory signals, recent investigations elucidate that long noncoding RNAs (LncRNAs) may play pivotal roles in driving Tregs differentiation and implications during HCC progression [60]. Overexpressed Lnc-epidermal growth factor receptor (Lnc-EGFR) in Tregs binds to EGFR and prevents its ubiquitination by c-CBL, augmenting activation of its downstream AP-1/NF-AT1 axis in Tregs thus to promote immunosuppression in HCC [60]. Moreover, Amphiregulin (AREG), a multifunctional player, may enhance Tregs suppressive function via the EGFR motivation as well [61].
The frequencies of Tregs are associated with HCC invasiveness and have a crucial role in hampering the development of effective anti-tumor responses in HCC [57, 62]. Recent evidence indicates that CD4+CD25+ Tregs in HCC patients can trigger a compromised immune response through various mechanisms [63]. A typical subset, CD4+ CD25+ Foxp3+ Tregs, may impair CD8+ T cells killing capacity via inhibiting the release and production of granzyme A, B (GrA, B), and perforin [64], concurrently, they also selectively suppress certain molecules (such as TNF-α, IFN-γ) involved in CD8+ T cell activation [64, 65]. Additionally, high expression of IL-35 in HCC tissue has been implicated positively to correlate with another newly identified subtype, CD39+ Foxp3+ Tregs infiltration [66], which serves as a better independent predictive indicator for recurrence in HCC patients after curative resection.

CD8+ cytotoxic T lymphocytes (CTLs)

The presence of CD8+ Cytotoxic T lymphocytes (CTLs) in HCC tissue is associated with improved survival. However, the efficacy of CTLs-mediated anti-tumor immune response is functionally limited through diverse mechanisms. Physical conditions (e.g. overload of lactic acid, low pH, hypoxia) [67], severe “metabolic competition” with tumor cells, a lack of CD4+ T cells help (moreover, interact with Tregs and other suppressor cells) [64, 68], and high expression of a large amount of immunoregulatory molecules in T cells or HCC cells (e.g. IL-10, Fas/FasL, CXCL17, VEGF, indoleamine-2,3-dioxygenase and so on) [67, 6971], may be responsible for restricted tumor-associated antigens (TAAs)-specific CD8+ T cell responses and poor IFN-γ production of CTLs [72, 73]. Apart from the classic immunosuppressive cells in TME, other components critically manipulate the functions of CTLs as well. Liver fibrosis, a prominent characteristic of HCC, impairs platelet-derived CD44 recognition by CD8+ T cells, reducing effector CD8+ T cells infiltration, and adhering to liver sinusoids to perform immunosurveillance [74]. Expression of Fas/FasL in CD8+ T cells positively correlates with HCC anti-tumor immunity [69]. Recent evidence indicates that tumor-derived vascular endothelial growth factor A (VEGF-A) and prostaglandin E2 (PGE2) cooperatively induce FasL expression in endothelial cells, which leads to excessive turnover of CD8 + T cells and reduce anti-tumor immune responses [71]. CD14+ dendritic cells (CD14+ DCs), a newly discovered immune regulator of HCC, has been suggested to suppress CTLs via IL-10 and indoleamine-2,3-dioxygenase (IDO) production, and the two cytokines play central roles in various physiological and pathological immune responses and inflammatory processes [75].
Notably, immune checkpoint signaling, which involves enhancement of numerous inhibitory co-stimulatory molecules (e.g. PD-1, LAG-3, CTLA-4, Tim-3, 2B4), has been demonstrated to dramatically induce CTLs exhaustion [58, 76, 77]. More details will be discussed in the section “immune checkpoint pathways and related therapeutics”.

Innate immune players and stromal components

Natural killer (NK) cells

Natural killer (NK) cells constitute a large proportion of the innate immune system in the liver. As the first line of host defense against viral infections (e.g. HBV, HCV) and carcinogenesis, NK cells play a key role in maintaining the balance between immune defense and tolerance. Increasing evidence suggests that hypoxic stress in HCC tissue, switch of activating/inhibitory NK receptors (NKRs) and influences by immune regulatory components in TME, largely contribute to NK cells dysfunction, which significantly correlates with fatigue anti-tumor immunity and poor prognosis [78, 79].
α-Fetoprotein (AFP) overexpressed by HCC cells was demonstrated to indirectly impair interlukine-12 (IL-12) production from dendritic cells (DCs), which results in attenuated cytotoxic effector molecules release, decreased expression of natural killer group 2, member D (NKG2D), an activating receptor on NK cells, and subsequently inhibiting activation and ability of NK cells [80, 81]. A recent study also indicates AFP may exert dual effects on NK cells functions in a direct manner. Short-term exposure to AFP induces IL-2 hyperresponsive phenotype NK cells, accompanied with elevated secretion of IL-1β, IL-6 and TNF-α [82]. These pro-inflammatory cytokines were associated with a low recurrence rate and a prolonged overall survival (OS) of HBV-related HCC patients [83]. In contrast, extended effect of AFP negatively affects long-term NK cell viability [82].
Other modulators in TME of HCC also exert multiple effects on NK activities (Fig. 2.). As mentioned above, MDSCs and TAMs infiltration inhibit autologous NK cell cytotoxicity and cytokine production, and the suppression is mainly dependent on NKp30 on NK cells [28]. Tregs compete with NK cells for IL-2 availability and impair NK responses via cytokines release, such as IL-8, TGF-ß1 and IL-10, which then down-regulates expression of NKR ligands on hepatic stellate cells (HSCs) and inhibits their recognition by NKG2D [84]. Hepatocellular carcinoma-associated fibroblasts (CAFs or TAFs), has been shown to induce MDSCs generation through the IL-6/STAT3 axis and stromal cell-derived factor (SDF)-1α secretion [18]. In addition to its direct influence on immunosuppressive TME, CAFs-derived IDO and PGE2 attenuate NK cells-mediated TNF-α and IFN-γ production, which may be associated with persistent fibrosis in HCC and tumor cell immune evasion [85, 86].

Kupffer cells (KCs)

KCs have previously been demonstrated to constitute an important part in maintaining liver immune homeostasis. Some studies reported that IL-10-mediated suppression of KC-derived inflammatory TNF-α and NO production contribute to attenuation of hepatitis [87]. Although KCs were once regarded as a powerful line of defense against tumors in the liver, recently, KCs have commonly been explored as pro-carcinogenic stakeholders in the context of HCC, more underlying mechanisms about their immune regulator roles, and KCs-related innate or adaptive immune response have been gradually uncovered. Current investigations indicate that altered functions of KCs are mainly influenced by pro-inflammatory signals and other suppressive cells (e.g. MDSCs) [88]. Triggering receptors expressed on myeloid cells-1 (TREM-1) expressed by KCs, is a crucial factor in HCC initiation. New studies suggest that the potential ligand for TREM-1, high mobility group Box 1 (HMGB1) released by necrotic hepatocytes, is likely involved in activating KCs pro-inflammatory signaling and promoting HCC progression [89]. Autophagy-defective KCs, a novel non-parenchymal liver cellular degradation deficiency, has been shown to promote liver fibrosis, inflammation and hepatocarcinogenesis during the pre-neoplastic stage via enhancing the mitochondrial ROS/NF-κB/IL-1 pathway [90]. In addition, KCs-derived galectin-9, the natural ligand for the T cell immunoglobulin domain and mucin domain protein 3 (Tim-3), leads to expansion of CD4+ CD25+ FoxP3+ Tregs, contraction of CD4+ effector T cells, and apoptosis of CTLs in HCC [91]. Galectin-9 not only mediates T-cell senescence in HBV-associated HCC, significantly contributes to the inflammatory reactions and HCC immune escape [92], but notably also represents a potential biomarker of liver fibrosis and may emerge as a novel immunotherapeutic target for treating HCC and liver viral infections [92, 93].

Dendritic cells (DCs) and DC-based vaccines

Similar to NK cells, as another major player of innate immunity, DCs serve as professional antigen-presenting cells that are able to prime T-cells against tumor associated antigens (TAAs) involved in HCC progression. Recently, DCs have been an area of high interest as novel vaccines based on DCs have been developed and widely used in treating solid tumors including prostate cancer, melanoma, renal cancer and HCC [94]. DCs engineered with tumor-associated antigens (TAAs), which have been clarified by numerous in vitro and in vivo studies, are regarded as promising vaccines in HCC immunotherapy. In addition, autologous DCs pulsed ex vivo with the lysate of autologous tumor cells, HepG2 cells and telomerase peptides, have been evaluated in human clinical trials.
Recently, the Dendritic cell (DC)-derived exosomes (DEXs) and tumor cell–derived exosomes (TEXs), which elicit tumor regression in autochthonous HCC mouse models, form a new class of cell-free vaccines and extend options for HCC immunotherapeutic vaccines [95, 96] (Table 1.).
Table 1
Biological effects of DCs-based vaccines in HCC: representative in vitro and in vivo investigations
Agents
Descriptions
Trial category
Biological effects or clinical results
References
AFP and interleukin 18 engineered DCs (AFP/IL-18-DCs)
DCs co-transduced with the AFP gene and IL-18
In vitro studies
• Significantly increase the production of IFN-γ
• Promote CD4+ T cells proliferation; elevate CTLs activity against AFP-expressing HCC cells
[97]
DCs pulsed with NY-ESO-1
DCs pulsed with the recombinant NY-ESO-1 protein
In vitro studies
• Be more effective in stimulating T cell proliferation compared with immature DCs
[98]
IL-12 engineered DCs (IL-12-DCs)
Endogenous IL-12-expression by adenoviral gene transfer effectively enhances immunostimulation of DC
Translational trials with murine models
• Induce a sufficient Th1 TME allowing the recruitment of Teff to enhance anti-tumor immunity
• Improve dendritic cells (DCs)-based immunotherapy of HCC
[99]
CD40 Ligand-Expressing DCs
Transduction of TAA-pulsed DCs with CD40L-encoding adenovirus (Ad-CD40L)
Translational trials with mice models
• Promote DC immunostimulation with up-regulation of CD80/CD86 and IL-12 expression
• Increase tumor infiltration with CD4+, CD8+ T cells and NK cells
• Elevate IFN-γ release and CTLs cytotoxicity
[100]
TEXs pulsed DCs
Tumor cell derived exosomes (TEXs)-pulsed DCs
In vitro and in vivo orthotopic HCC mice models
• Increase numbers of T lymphocytes infiltration, elevate IFN-γ production; decrease IL-10, TGF-β in tumor sites
• Elicit a stronger immune response than cell lysates in vitro and in vivo
[95]
A new form vaccine: DCs-DEXs
Exosomes derived from AFP- expressing DCs
Translational investigation in mouse models
• A cell-free vaccine option for HCC immunotherapy
• Decrease Tregs infiltration, IL-10, TGF-β in tumor sites
• Reshape the TME in HCC
[96]
TAAs pulsed DCs vaccine
α-fetoprotein, glypican-3 and MAGE-1 recombinant fusion proteins pulsed DCs
A prospective phase I/II clinical study in 5 HCC patients
• Result: safe and well-tolerated
• Over 95% of DCs demonstrated highly expressed MHC class I (HLA-ABC), MHC class II (HLA-DR), and costimulatory molecules (CD86, CD80, and CD40)
• Induce Th1 immune responses with highly produced IL-12, IFN-γ
• Trigger stronger CTLs responses
[101]
TAAs pulsed DCs vaccine
α-fetoprotein, glypican-3 and MAGE-1 recombinant fusion proteins pulsed DCs
A prospective phase I/II clinical study in 12 HCC patients
• Result: safe and well-tolerated
• 1-, 2-, and 5-year cumulative RFS rates were improved
[102]
DCs pulsed with tumor cell lysate
Mature autologous DCs pulsed exvivo with HepG2 lysate
A phase II clinical trial with 35 patients with advanced HCC
• Result: safe and well-tolerated
• MS: 168 days; 6-month survival rate: 33%; 1-year survival rate 11%
• Induce stronger T cell responses and IFN-γ release
[103]
DCs pulsed with tumor cell lysate
Mature autologous DCs pulsed ex vivo with HepG2 lysate
A clinical trial with 2 groups:
Group1: 15 advanced HCC patients received DCs vaccination
Group2: control group
• Result: safe and well-tolerated
• CD8+ T cells and serum IFN-γ were elevated after DCs injection
• Partial radiological response: 13.3%; stable course: 60%; and 26.7% showed progressive disease and died at 4 months post-injection
[104]
DCs pulsed with AFP
AFP peptides pulsed onto autologous DCs
A phase I/II clinical trial in which HLA-A*0201 patients with AFP-positive HCC, 10 patients received DCs vaccination
• 6 of 10 subjects increased IFN-γ producing AFP-specific T cell responses
[105]
Notes: TAA tumor-associated antigens, MAGE-1 melanoma-associated antigen 1, GPC-3 glypican-3, IL-12 interleukin-12, AFP a-fetoprotein, TEXs tumor cell–derived exosomes, TGF-β transforming growth factor-β, TME tumor microenvironment, IFN-γ interferon-γ, DEXs dendritic cell-derived exosomes, CTLs cytotoxic T lymphocytes, Tregs regulatory T cells

Representative immune inhibitory factors and modulators

The abundance of pro-inflammatory chemokines, cytokines and immunosuppressive molecules, which orchestrates a strongly immunosuppressive tumor milieu, play critical roles in reshaping TME, mediating intercellular crosstalk, and exerting immune evasion-promoting effects of HCC. Some of their specific functions have been mentioned while discussing immune cells of HCC, here, we summarize the representative players that current studies mainly highlight (Table 2.).
Table 2
Representative molecules and signaling pathways mediated pro−/anti-tumor immunity of HCC
Cytokines/signaling molecules
Category
Description
References
IL-1β
Pro-inflammatory cytokine
• A favorable factor for prolonged OS of HBV-related HCC patients
• TAMs-secreted IL-1β in HCC contributes to HIF-1α stability, IL-1β/HIF-1α induce EMT and metastasis of HCC
[18] [37]
IL-12
Pro-inflammatory cytokine (anti-tumor immunity modulator)
• Promote cytotoxicity and IFN-γ production
• Mediate CD4+ T helper cells transformation to Th1 phenotype, enhance cell based immunity
• Up-regulate NKG2D related NKs anti-tumor immunity
[8183]
IL-8
Pro-inflammatory cytokine
• Trigger potent pro-inflammatory signals in HCC; promote HCC immune evasion and metastasis
• Enhance HCC-related fibrosis and Tregs enrichment in tumor tissue
[33, 84, 106]
IL-10
Inhibitory cytokine that involves in both innate and adaptive immunity in HCC
• Tolerogenic DCs/ FcγRIIlow/−B cells derived IL-10 induces hepatic tolerance by promoting T cell hypo-responsiveness
• Suppress CD4+ T cells activity via CTLA-4-dependent manner
• IL-10 production is associated with Foxp3+ Tregs accumulation in HCC
• Accelerate HCC progression by mediating polarization of alternatively activated M2 macrophages
[38, 39, 75] [107, 108]
IL-6/STAT3
Pro-inflammatory/carcinogenesis signaling
• Mediate MDSCs activation then result in immunosuppression
• Up-regulate IL-10, IDO expression; down-regulate IFN-γ; induce T cells dysfunction and apoptosis
[18, 109]
PD-1/PD-L1
Immune checkpoint molecules
• Impairing anti-tumor immunity and promotes CD8+ T cells exhaustion and apoptosis
• PD-1 over-expressed myeloid cells, such as DCs, suppress T cell responses in HCC
[110, 111]
LAG3
Immune checkpoint molecule
• Up-regulated on TAA-specific T cells
• Significantly impairs CD4+ and CD8+ TILs functions in HCC
[112]
CTLA-4
Immune checkpoint molecule
• Mediates immunosuppression by inducing Tregs activity and IDO and IL-10 productions in DCs
• Suppresses the proliferation of T cells
[73, 112]
Tim3/Galectin-9 pathway
Immune checkpoint signaling
• Negatively regulates Th1-mediated immune responses
• Mediates CTLs dysfunction and immunosuppressive responses in HBV-associated HCC
• Fosters HCC development by enhancing TGF-β-mediated alternative activation of macrophages
[27, 43, 76, 113]
VEGF, PDGF, HGF
Major growth factors in TME of HCC
• Enhance interactions between TAFs/HSCs and HCC cells
• Mediates recruitment of immune inhibitory cells
• Mediates other pro-inflammatory signals in TME (e.g. IL-6/STAT3 axis)
• Promotes angiogenesis and immune evasion
[18, 75]
IDO
Immunosuppressive modulator
• High level IDO expression is associated with poor prognosis and high recurrence rate in HCC patients; a potential target for HCC immunotherapy
• Enhance regulation of immune responses, such as T-cell proliferation impairment, promotion of Tregs expansion
• IDO derived from HSCs and CAFs impair cytotoxicity and cytokine production of NK cells
• CD14+CTLA-4+ regulatory DCs derived IDO suppress CTLs response; cause NKs dysfunction in HCC anti-tumor immunity
[75] [83] [85, 109] [114]
SDF-1α/CXCR4
A multiple signaling that mediates HCC immune evasion, progression and metastasis
• Enhance interactions between TAFs/HSCs and HCC cells
• Facilitate MDSCs recruitment and generation, then results in immune evasion
• Contribute to HCC fibrosis and hypoxia
• Synergize with other stroma-derived cytokines (such as HGF, VEGF, TGF-β and so on), promoting HCC growth, angiogenesis, metastasis
[18, 115]
[116]
CXCL17
119-amino acid chemokine
• An independent factor that correlates with HCC regulatory immune cells infiltration
• Predict poor prognosis of HCC
[70]
CCL2(also named MCP-1)
Multifunctional factor
• Multiple cellular resources, including HSCs, hepatocytes, macrophages and so on
• CCL2/CCR2 promotes regulatory cytokines release, M2-macrophages accumulation and polarization
• Suppress cytotoxic CD8+ T lymphocytes anti-tumor responses
• Facilitate TANs infiltration in HCC
[54, 117]
[118]
Hypoxia (HIF-1α)
Versatile modulator of TME and tumor immunotolerant state
• Promote recruitment of Treg, MDSCs.
• regulate release of multiple chemokines and inflammatory factors; Activate transcription of C-C motif ligand 26, 28 (CCL26, CCL28) and interleukines (ILs).
• contribute to immune tolerance and angiogenesis.
[19, 119, 120]
CXCL1/CXCR2 signaling
Immunosuppressive signaling axis
• Impair immune balance in TME of HCC.
• Facilitate immune escape via increasing MDSCs recruitment and repressing infiltration of IFNγ+CD8+ T cells.
[121]
CXCL5
C-X-C motif chemokine
• Recruits more TANs infiltration and contributes to TANs-induced HCC immune evasion.
[55]
CCL15
Immunosuppressive signaling
• Serves as an independent factor for HCC prognosis and survival.
• Recruit CCR1 + CD14+ monocytes infiltration, accelerate tumor proliferation and metastasis by activating STAT1/erk1/2 signaling.
• Upregulate immune checkpoints (e.g. PD-L1, Tim3) and immune tolerogenic enzymes (e.g. IDO, ARG)
[122]
Notes: HCC hepatocellular carcinoma, IL- interleukin-, OS overall survival, EMT epithelial-mesenchymal transition, HIF-1α hypoxia inducible factor-1, IFN-γ interferon-γ, NKs natural killer cells, Tregs regulatory T cells, DCs dendritic cells, MDSCs myeloid-derived suppressor cells, PD-1 programmed cell death protein 1, PD-L1 programmed death-ligand 1, LAG3 lymphocyte-activation gene 3, TAA tumor associated antigen, TILs tumor infiltrating lymphocytes, CTLA-4 cytotoxic T-lymphocyte-associated protein 4, IDO indoleamine 2,3-dioxygenase, Tim3 T cell immunoglobulin mucin, CTLs cytotoxic T lymphocytes, VEGF vascular endothelial growth factor, PDGF platelet-derived growth factor, HGF hepatocyte growth factor, TME tumor microenvironment, TAFs tumor-associated-fibroblasts, HSCs hepatic stellate cells, CAFs cancer associated fibroblasts, SDF-1α stromal cell derived factor 1α, CXCR4 chemokine (C-X-C motif) receptor 4, CXCL17 chemokine (C-X-C motif) ligand 17, CCL2 chemokine (C-C motif) ligand 2, MCP-1 monocyte chemotactic protein 1, TANs tumor-associated neutrophils, CXCL1 chemokine (C-X-C motif) ligand 1, CXCR2 chemokine (C-X-C motif) receptor 2, CXCL5 chemokine (C-X-C motif) ligand 5, CCL15 chemokine (C-C motif) ligand 15, CCR1 chemokine (C-C motif) receptor 1, ARG Arginase

Current immunotherapeutic strategies for HCC

As an inflammation-associated cancer, HCC represents a promising target for immune based therapeutics. Clinically, the success of immune oncology in many types of cancer has encouraged implementation of immunotherapeutics in HCC. Recent studies have suggested that tumor antigen-specific immunotherapy and other approaches modulating immunogenicity have become attractive strategies for HCC treatment. Generally, these immunotherapeutic approaches for HCC could be mainly categorized into immune-checkpoint blockade (ICB), cell-based (mainly refers to DCs) /non-cell based vaccines, adoptive cell transfer (ACT), cytokine/antibody based immune regimens, and combination of immunotherapeutic agents with other drugs (Fig. 3.). Here, we collect some representative data on preclinical and clinical trials on immune based strategies of HCC, and discuss our current knowledge on their action mechanisms, rationale and application prospects for HCC treatment in the foreseeable future.

Immune checkpoint inhibitors

Immune checkpoints are a specific sub-type of membrane-bound molecules that act as pivotal regulators of immune escape in cancers. The most studied immune checkpoints in HCC includes cytotoxic T lymphocyte protein 4 (CTLA-4), programmed cell death protein-1 and its ligand (PD-1, PD-L1), lymphocyte activation gene 3 protein (LAG-3) and mucin domain-containing molecule-3 (Tim-3).

Programmed cell death protein-1 and its ligand (PD-1, PD-L1)

PD-1, a regulator immunoglobulin expressed on activated CD4+, CD8+ T cells, B cells and NK cells, plays an important role in maintaining immune tolerance and repressing cytotoxicity of T lymphocytes [123]. Co-inhibitory signals in lymphocytes are mediated by binding of PD-1 to its ligands PD-L1 (B7-H1) and PD-L2 (B7-DC) [124]. In HCC, it’s clear that an increase in the number of both circulating and intratumoral PD-1+ CD8+ T cells predict high postoperative recurrences and poorer prognosis. It is also known that up-regulation of PD-L1 on HCC cells, which is induced by various cytokines, particularly IFN-γ, in turn contributes to impairing anti-tumor immunity and promotes CD8+ T cells apoptosis [110]. New in vitro and in vivo discoveries indicate that PD-1 overexpressed myeloid cells, such as DCs, suppress T cell responses in HCC. CD8+ T cells can be more potently activated to secrete IL-2 and IFN-γ via adoptive transfer of PD-1-deficient DCs [111].
Clinically, a representative phase 1/2 dose escalation and expansion trial on PD-1 immune checkpoint inhibitors Nivolumab (CheckMate 040 study) showed a promising role for immunotherapy in the treatment of advanced HCC, and relevant results were presented at the 2017 ASCO annual meeting [125]. In dose-escalation phase (enrolled number = 48), the objective response rate (ORR) was 15%, the disease control rate (DCR) was 58% and the median time to progression was 3.4 months. In dose-expansion phase (total number = 214; in 4 cohorts), generally, the ORR was reported as 20%, the DCR was 64%, the median time to progression was 4.1 months, and the 6-month and 9-month progression-free survival rates were 83 and 74% respectively. A subsequent CheckMate-040 based analysis compared the ORR and survival between intent-to-treat (ITT) overall population and Asian cohort. It suggested that Nivolumab showed similar mOS and manageable safety profile both in ITT population and Asian patients [126].
The efficacy of another anti-PD-1 monoclonal antibody, Pembrolizumab, was assessed in a phase II, open-label trial (KEYNOTE-224). In this study, Pembrolizumab was proven to be effective and well-tolerated in Sorafenib-experienced patients with advanced HCC, and PD-L1 expression level may act as a useful predictive bio-marker in selecting interested HCC patients. A total of 104 enrolled patients in this study represented 8-month median duration of response (mDOR), with median time to response of 2 months [127].
In addition, another phase 3 randomised clinical trial of nivolumab mono-therapy compared with sorafenib in the first-line setting is ongoing (NCT02576509). Moreover, combination therapies of anti-PD-L1 antibody (Duvalumab) with anti-CTL4–4 antibody (Tremelimumab) for unresectable HCC are under study as well (NCT02519348).
Other reported combination immunotherapy studies are encouraging and really open new avenues for HCC treatment [128130], however, additional strategies are needed to uncover more sensitive predictive biomarkers besides PD-1/PD-L1 axis, optimize treatment selection, and improve HCC patients immune response. (More data of terminated or ongoing clinical trials are available in Table 3.).
Table 3
Representative ongoing immune checkpoint blockade(ICB) based immunotherapy clinical trails in HCC
Regimen
Disease
Mechanism of action
Estimated/Actual enrollment
NCT number
Anti-CTLA-4 antibody based monotherapy/combination therapy
 Tremelimumab+TACE
Liver cancer
Anti-CTLA-4 antibody; chemoembolization
61
NCT01853618
 Tremelimumab
Advanced HCC
Anti-CTLA-4 antibody
20
NCT01008358
 Ipilimumab +Nivolumab/ Nivolumab alone following SBRT
Unresectable HCC
Anti-PD-1 antibody, anti-CTLA-4 antibody
50
NCT03203304
Anti-PD-1 antibody based monotherapy/combination therapy
 Nivolumab+Y90 Radioembolization
HCC
Liver-localized radioembolization, PD-1 blockade
40
NCT03033446
 Nivolumab+cabozantinib
Advanced HCC
Neoadjuvant therapy, PD-1 blockade
15
NCT03299946
 Nivolumab+Pexa Vec
HCC
Oncolytic Immunotherapy, PD-1 blockade
30
NCT03071094
 Nivolumab+Ipilimumab
HCC (Resectable and potentially resectable)
CTLA-4 blocade, PD-1 blockade
45
NCT03222076
 Nivolumab following selective internal radiation therapy (SIRT)
HCC (unresectable)
PD-1 blockade, radiation therapy
40
NCT03380130
 Nivolumab following complete resection
HCC
PD-1 blockade
530
NCT03383458
 Nivolumab+Galunisertib
NSCLC
HCC
TGF-β receptor I kinase inhibitor, PD-1 blockade
75
NCT02423343
 Nivolumab+Lenvatinib
HCC
TKI + PD-1 blockade
26
NCT03418922
 Nivolumab+Y90
HCC
PD-1 blockade+Radioembolization
35
NCT02837029
 Nivolumab+Sorafenib
HCC
PD-1 blockade+chemotherapy
40
NCT03439891
 Nivolumab+CC-122 (Avadomide)
HCC (unresectable)
PD-1 blockade+immunomodulator (targeting protein cereblon)
50
NCT02859324
 Nivolumab+deb-TACE
Advanced HCC
PD-1 blockade+transarterial chemoembolization
14
NCT03143270
 Nivolumab+Mogamulizumab
HCC other solid tumors
PD-1 blockade+anti-CCR4 antibody
188
NCT02705105
 TATE followed by Nivolumab or Pembrolizumab
HCC; mCRC
PD-1 blockade+TACE
40
NCT03259867
 Nivolumab
Advanced HCC (with or without viral infections)
PD-1 blockade
262
NCT01658878
 Nivolumab (vs. Sorafenib)
Advanced HCC
PD-1 blockade
726
NCT02576509
Anti-PD-L1 antibody based monotherapy/combined therapy
 Durvalumab+tremelimumab
Unresectable HCC
Anti-PD-L1 antibody, anti-CTLA-4 antibody
440
NCT02519348
 Durvalumab monotherapy; Durvalumab+Tremelimumab vs. Sorafenib
Unresectable HCC
Anti-PD-L1 antibody, anti-CTLA-4 antibody
1200
NCT03298451
 Durvalumab+Guadecitabine (SGI-110)
Liver cancer; pancreatic cancer; bile duct cancer; gallbladder cancer
Anti-PD-L1 antibody, small molecule DNA methyltransferase 1 (DNMT1) inhibitor
90
NCT03257761
 Durvalumab+Tremelimumab+ablative therapies
Advanced HCC and BTC
Anti-PD-L1 antibody, anti-CTLA-4 antibody
90
NCT02821754
 Durvalumab+Ramucirumab (LY3009806)
GEJ adenocarcinoma;
NSCLC;
HCC
Anti-PD-L1 antibody, anti-VEGFR2 antibody
114
NCT02572687
Anti-LAG-3 antibody in combination with anti-PD-1 blockade
 Relatlimab+Nivolumab
Different types of solid tumor (including HCC)
Anti-LAG-3 antibody,anti-PD-1 antibody
1000
NCT01968109
Notes: Y90 yttrium Y 90 glass microspheres, deb-TACE drug eluting bead transarterial chemoembolization, TATE transarterial tirapazamine embolization, mCRC metastatic colorectal cancer, BTC biliary tract carcinomas, GEJ gastroesophageal junction, SBRT stereotactic body radiotherapy

Cytotoxic T lymphocyte protein 4 (CTLA-4)

Cytotoxic T lymphocyte protein 4 (CTLA-4), an inhibitory co-receptor that is expressed by activated T cells and is constitutively present on Tregs, has great affinity for competing with CD28 by binding to its ligands, CD80 and CD86, on antigen presenting cells (APCs). CTLA-4 plays a critical part in controlling CD4+ T cells function. In HCC and many other types of cancer, CTLA-4 suppresses the proliferation of T cells that have undergone TAA recognition and differentiation [131]. Additionally, inside HCC tissues, CTLA-4 further mediates immunosuppression by inducing Tregs activity and IDO and IL-10 productions in DCs [75].
Many clinical trials of antibodies targeting CTLA-4 are ongoing. A pilot clinical investigation testing anti-tumor and anti-viral effects of Tremelimumab in patients with HCC and HCV infection showed strong signs of anti-tumour efficacy (NCT01008358). The treatment presents a reliable safety profile, as no immune-related adverse events occurred. The median time to progression (TTP) was 6.48 months, and median overall survival (OS) was 8.2 months. The partial response rate was observed as 17.6%, and had a remarkable disease control rate of 76.4%. Moreover, 36% of the patients with AFP levels > 100 ng/ml showed more than 50% drop after Tremelimumab therapies [132]. In another phase 1 clinical study that tests the safety and effectiveness of Tremelimumab with radiofrequency (RFA) (NCT01853618), the median TTP and median OS were respectively 7.4 months (95% CI 4.7 to 19.4 months) and 12.3 months (95% CI 9.3 to 15.4 months). The 6-week HCC biopsies showed a clear increase in CD8+ T cells infiltration demonstrating that the combination of Tremelimumab with RFA in advanced HCC is feasible and results in stronger anti-tumor immunity [133].

Mucin domain-containing molecule-3 (Tim-3) and lymphocyte activation gene 3 protein (LAG-3)

Mucin domain-containing molecule-3 (Tim-3) is a transmembrane protein that is expressed on IFN-γ-secreting Th1 cells, NK cells and CTLs [113]. Tim-3 interacts with its soluble ligand galectin-9, and then negatively regulates T cell responses [91]. The expression of Tim-3 is increased in T cells infiltrating in chronic HBV infection [134], and the Tim-3/galectin-9 pathway consistently predicts poor prognosis in patients with HBV-associated HCC [76].
Lymphocyte activation gene 3 protein (LAG-3), a member of the immunoglobulin super-family proteins, which often binds MHC class II molecules with high affinity, represses the co-stimulatory functions of T cells [135]. Clinically, dual blockade of LAG-3 with anti-PD-1 therapy is being tested in a Phase I trial (NCT01968109) (Table 3.).
The immunosuppressive roles of both Tim-3 and LAG-3 in chronic viral hepatitis and HCC have been uncovered recently. However, their clinical values need to be further elucidated.

Adoptive cell transfer (ACT) based therapy in HCC

Besides the immune-checkpoint blockade (ICB), other effective immunotherapeutic options for HCC are urgently needed. In recent years, the exploration and development of cell-based immunotherapies in treating solid tumors have received considerable attentions. Adoptive cell transfer (ACT) offers robust and more durable anti-tumor immunity in cancer treatment. Recent translational research and clinical cases reported the success of engineered autologous HBV-specific T cell receptor (TCR) redirected therapeutics in treating HBV-associated HCC [136, 137], which broadens the immunotherapeutic approaches and might be used to treat a wider population of patients [138]. Based on the cell types, ACT used in HCC pre-clinical/clinical researches can be mainly classified as: (1) cytokine-induced killer (CIK) cells treatment, and (2) genetically modified NK cells or T cells (CAR-T).
CIK cells are a mixture of T lymphocytes, which are ex vivo expanded in the presence of cytokines (such as IL-1, IL-2, IFN-γ), comprising activated NKT cells, CD3/CD56+ NK cells, and CD3+ /CD56 cytotoxic T cells [139]. CIK cells can be obtained in great numbers from peripheral blood mononuclear cells (PBMCs) and are very easily cultured. More importantly, it has been clarified that the absence of MHC restrictions favors CIK cells’ more potent anti-tumor efficacy compared with traditional CTLs [58, 139]. Previous studies suggested that CIK cells prevent HCC from progression, and effectively kill cancer stem cells (CSCs) mainly through NKG2d-ligands recognition [140]. A retrospective study demonstrated a significant correlation between high number of PD-1+ TILs and favorable outcome in CIK cells treated HCC group, which suggested that PD-1+ TILs may be utilized to predict the efficacy of CIK treatment in post-operative HCC patients [141]. A randomized, phase 3 clinical trial of the efficacy and safety of CIK cells treatment in 230 patients with HCC (NCT00699816) indicates that for post-curative treatment in HCC patients, adjuvant immunotherapy with CIK cells can prolong recurrence-free survival and OS (median RFS:44.0 months in treatment group, 30.0 months in the control group). Additionally, the proportion of patients with serious adverse events did not differ significantly between the treatment and control groups [142]. Several trials of CIK cells treatment in combination with other therapies, such as RFA, arterial chemoembolization and epitope-pulsed DCs, have been reported [143145]. The evidence obtained from a growing body of literature confirms that CIK cells is a very promising adoptive immunotherapy that can be exploited for treatment and prevention of recurrence in HCC. However, a small fraction of patients undergoing standard therapies suffer from ‘immune fatigue’ status, and lack adequate leukocytes [139], a major obstacle for CIK cells treatment that needs to be overcome urgently. (Ongoing CIK-based clinical trials are available in Table 4.)
Table 4
Clinical trials based on CIKs and genetically modified T cells under study for the treatment of HCC
Regimen
Population
Design
Estimated/Actual enrollment
NCT number
CIKs mono-therapy for HCC
 CIKs
• Hepatocellular carcinoma
• Phase 3 clinical trial
• CIK treatments within 3 months after liver resection
200
NCT00769106
 CIKs
• Hepatocellular carcinoma
• Renal cell carcinoma
• Lung cancer
• Phase 1 clinical trial
• CIK treatments following radical resection
40
NCT01914263
 CIKs
• Hepatocellular carcinoma
• Phase 3 clinical trial
• CIK treatments following radical resection
200
NCT01749865
 DC-CIKs
• Hepatocellular carcinoma
• Phase 2 clinical trial
• Dendritic and CIKs used to treat HCC patients who got CR or PR after complete resection/ TACE
100
NCT01821482
CIKs in combination with other therapies for HCC
 CIKs+ anti PD-1 antibodies
• Hepatocellular carcinoma
• Renal cell carcinoma
• Bladder cancer
• Colorectal cancer
• Non-small-cell lung cancer
• Breast cancer
• Phase 2 clinical trial
• Combination therapy
50
NCT02886897
 CIKs+ TACE
• Hepatocellular carcinoma
• Digestive system neoplasms
• Phase 3 clinical trial
• Combination therapy
60
NCT02487017
 CIKs+ RFA
• Hepatocellular carcinoma
• Phase 3 clinical trial
• RFA + Highly-purified CTL vs. RFA Alone for Recurrent HCC after partial hepatectomy
210
NCT02678013
CAR-T trials for HCC treatment
 Anti-GPC3 CAR-T
• Hepatocellular carcinoma (GPC3 + advanced HCC)
• Phase 1/2 clinical trial
20
NCT03084380
 Anti-GPC3 CAR-T
• Hepatocellular carcinoma (GPC3 + advanced HCC)
• Phase 1/2 clinical trial
60
NCT02723942
 Autologous anti-AFP (ET1402L1)-CAR-T
• AFP expressing hepatocellular carcinoma
• Phase 1 clinical trial
• The second generation CAR-T treatment
18
NCT03349255
 Anti-GPC3 CAR-T
• Advanced hepatocellular carcinoma
• Phase 1 clinical trial
13
NCT02395250
 Anti-GPC3 CAR-T
• Advanced hepatocellular carcinoma
• Phase 1 clinical trial
30
NCT03198546
 TAI-GPC3-CAR-T
• Hepatocellular carcinoma
• Phase 1/2 clinical trial
• GPC3-CAR-Ttreatment mediated by the method of transcatheter arterial infusion (TAI)
30
NCT02715362
 Anti-GPC3 CAR-T
• Advanced hepatocellular carcinoma
• Phase 1/2 clinical trial
• GPC3-CAR-Ttreatment by intratumor injection
10
NCT03130712
 Anti-Mucin1 (MUC1) CAR-T
• Hepatocellular carcinoma
• Non-small cell lung cancer
• Pancreatic carcinoma
• Triple-negative invasive breast carcinoma
• Phase 1/2 clinical trial
• Patients with MUC1+ advanced refractory solid tumor
20
NCT02587689
 Anti-GPC3 CAR-T
• Relapsed or refractory hepatocellular carcinoma
• A single arm, open-label pilot study
• GPC3+ hepatocellular carcinoma
20
NCT03146234
 Anti-EpCAM CAR-T
• Colon cancer
• Esophageal carcinoma
• Pancreatic cancer
• Prostate cancer
• Gastric cancer
• Hepatic carcinoma
• Phase 1/2 clinical trial
• Targeting patients with EpCAM+ cancer
60
NCT03013712
 CAR-T targeting TAAs
• Hepatocellular carcinoma
• Pancreatic cancer
• Colorectal cancer
• Phase 1/2 clinical trial
• CAR-T targets: GPC3 for hepatocellular carcinoma
• Mesothelin for pancreatic cancer
• CEA for colorectal cancer
20
NCT02959151
Notes: TACE transcatheter arterial chemoembolization, RFA radiofrequency ablation, DC-CIKs dendritic and cytokine-induced killer cells, CR complete remission, CAR-T cells chimeric antigen receptor-T cells, TAI transcatheter arterial infusion
Another adoptive cell immunotherapy, which uses chimeric antigen receptor-modified T cells (CAR-T) or genetically modified NK cells, has been shown to be a promising strategy for the treatment of HCC. CAR-T can specifically recognize tumor-associated antigens and effectively eliminate tumor cells in a non-MHC restricted manner. Furthermore, additional genes could be used to enable resistance to immune suppression [146]. Successful use of CAR-T cells in treating haematological malignances includes ACT using CD19-specific CAR-T cells and the third-generation of CD20-specific CAR with CD28 and 4-1BB co-stimulatory domains. CAR-T cell treatment is well tolerated and can induce great remission in B-cell lymphoma [147, 148] (NCT00924326, NCT00621452), however, despite this, the achievements of CAR-T cells therapies against solid tumors are still modest. In HCC, recent studies revealed that glypican-3 (GPC-3), an oncofetal proteoglycan anchored to HCC cell membrane that functions to promote HCC progression and is associated with poor prognosis, provides a novel prognostic molecule therapeutic target in HCC immunotherapy [149]. Previous in vitro and orthotopic xenograft models of human HCC experiments both indicated that cytotoxic activities of T cells redirected to GPC-3 seemed to be positively correlated with GPC-3 expression levels in the targeting cell. This suggests that GPC-3-targeted CAR-T cells may offer a promising immune therapeutic intervention for GPC-3+ HCC [150, 151]. Moreover, a series of clinical studies conducted to test the safety and efficacy of CAR-T cells redirected to GPC-3 in HCC treatment are underway (Table 4.). Similarly, CAR-modified NKs represents a newly emerging immunotherapeutic modality for HCC therapy. Potent anti-tumor responses of GPC-3-specific NKs based therapeutics were observed in HCC xenografts with both high and low GPC-3 expression, which extends treatment options for patients with GPC-3+ HCC [152]. Another gene-modified NKs candidate, human interleukin-15 (hIL-15) gene-modified NKL cells (NKL-IL15), has been demonstrated to express high levels of cytolysis-related molecules (TNF-α, IFN-γ, NKp80 and so forth), which induces higher NKG2D ligand expression on target cells and results in increased susceptibility of HCC to NKs-mediated cytolysis [153].
Collectively, genetically modified NK cells and CAR-T cells based treatment provide new avenues for immunotherapies against HCC. Nevertheless, before being widely used as therapeutics in the clinic, their clinical efficacy and on-target-off tumor toxicity still require further assessments in more randomized trials.

Non-cell based vaccines and oncolytic viruses based immunotherapy in HCC

With the identification of a growing number of tumor-associated antigens (TAAs), and as a result, vaccines targeting HCC TAAs have been investigated and developed. A number of tumor antigens, such as human alpha-fetoprotein (AFP), GPC-3 and telomerase-reverse transcriptase (hTERT) have been identified as vaccine-based immunotherapeutic targets for HCC [154]. (Table 5.). Although increased efforts are being made to advance TAAs-based vaccines, the early clinical trials witnessed a mixed history of success and failure [155157, 160]. The first HCC AFP-vaccine clinical trial was completed with only transient immunological responses detected, partially due to the limited number of antigens used or deficient CD4+ helper T cell support [157, 160, 161]. A vaccine with a single 16 amino acids sequence, hTERT-derived peptide (GV1001), and binds multiple HLA class II molecules, results in little clinical activity and no detected absolute antigen-specific CTLs responses [155]. On the other hand, partial clinical data on GPC-3 based vaccines demonstrated that the vaccine could induce measurable anti-tumor responses and are associated with prolonged OS of HCC patients [156, 162].
Table 5
Several representative clinical trials of non-cell based vaccines and oncolytic virus (OVs) based immunotherapy in HCC
Trial (the 1st author/ responsible party)
Agent
Design
Population
Status/Relevant results
Registration no.& Reference order
Non-cell based vaccines
Greten et al. (2010)
GV1001: a telomerase derived peptide vaccine
• A phase 2 open-label trial; 4-week injections with GM-CSF + GV1001 vaccinations
• P:tumor response
• S:TTP, TTSP, PFS, OS, safety and immune responses
40 patients with advanced HCC
Status: terminated
Results: no relevant toxicity, median OS: 11.5 months, median PFS: 57 days, median TTP: 57 days, TTSP: 11.7 months
[155]
NCT00444782
Sawada et al. (2012)
GPC-3-derived peptide vaccine
• A phase 1 Trial
• P: safety
• S:TTP, OS, immune responses (measured by IFN-γ ELISPOT assay)
33 patients with advanced HCC
Status: terminated
Results: well-tolerated, 91% patients were successfully induced with CTLs-mediated responses, median OS: 9.0 months, median TTP: 3.4 months, GPC-3-specific CTL frequency after vaccination correlated with OS
[156]
UMIN-CTR000001395
Butterfield et al. (2003)
AFP peptide vaccine
• A pilot Phase 1 clinical trial
• In vivo studies testing AFP peptide- vaccine reactive T cells responses
6 patients with HCC
Status: terminated
Results: all of the patients generated T-cell responses to most or all of the peptides as measured by direct IFN –γ ELISPOT and MHC class I tetramer assays
[157]
Immunitor LLC et al. (2018)
An oral therapeutic vaccine: hepcortespenlisimut-L (Hepko-V5)
• A phase 3, randomized, placebo-controlled, double-blinded trial
• P:changes in serum AFP levels, tumor burden, OS
Estimated enrollment:120 patients with advanced HCC
Status: recruiting
Results: none
NCT02232490
Roswell Park Cancer Institute (2016)
Vaccine therapy in treating NY-ESO-1 expressing solid tumors
• A phase 1 clinical trial determines the safety of DC205-NY-ESO-1 vaccine
18 patients with NY-ESO-1 solid tumors, including HCC
Status: completed
Results: none
NCT01522820
Butterfield et al. (2013)
AFP+ GM-CSF Plasmid Prime and AFP Adenoviral vector Boost
• A phase 1/2 trial
• Testing immunization with AFP + GM CSF plasmid prime and AFP adenoviral vector
Actual enrollment:
2 patients with HCC
Status: terminated (Poor accrual and limited target patient population for future accrual, did not complete the Phase 1 portion of the trial.)
NCT00669136
Oncolytic virus (OVs) based immunotherapy
Byeong et al. (2008)
JX-594
• A phase 1 clinical trial, assessment of intratumoral injection of JX-594 into primary or metastatic liver tumours
• P:safety, MTD
14 patients with primary or metastatic liver tumors
Status: terminated
Results: well-tolerated;
MTD was determined as 1 × 109 pfu; 10 patients were radiographically evaluable for objective responses; responses in 3 HCC patients: 3 serum tumor markers PR (≥50% decrease); 1 response according to PET
[158] (NCT00629759)
Jeong Heo et al. (2013)
JX-594
• A Prospective, randomized clinical trial with high or low dose JX-594
• P: intrahepatic disease control rate
30 patients with unresectable liver tumors
Status: terminated
Results: 11/16 patients showed cytotoxicity against HCC; 31% anorexia in high dose group
RR: 4 PR, 10 SD by RECIST
[159] (NCT00554372)
Jennerex Biotherapeutics (2008–2011)
JX-594 (Pexa-Vec)
• A phase 2b randomized trial
• JX-594 plus best supportive care versus best supportive care in patients with advanced HCC who have failed Sorafenib treatment
129 patients with advanced HCC who have failed sorafenib
Status: completed
Results: none
(No results posted on ClinicalTrials.​gov)
NCT01387555
SillaJen, Inc. (2015)
Vaccinia virus based immunotherapy (Pexa-Vec) + Sorafenib
• A multi-center, randomized, open-label, Phase 3 trial;
• Comparing Vaccinia Virus based Immunotherapy Plus Sorafenib vs Sorafenib alone
600 patients with advanced HCC
Status: recruiting
Results: none
NCT02562755
Notes: HCC hepatocellular carcinoma, P primary endpoint, S secondary endpoint, OS overall survival, TTP time to progression, TTSP time to symptomatic progression, SD stable disease, RR response rate, JX-594 aoncolyticpox virus carrying human GM-CSF genes, MTD maximum-tolerated dose, RECIST response evaluation criteria in solid tumors, PR partial response
Apart from these classical TAAs, another attractive target is cancer-testis antigens (CTAs), which are considered to be novel targets for HCC immunotherapy due to the restricted expression patterns in a variety of tumors and normal tissues [163]. NY-ESO-1, also known as CTAG1, is one of the most immunogenic CTAs. A number of previous studies reported that NY-ESO-1 is highly expressed in many types of solid tumors, and a number of vaccine strategies targeting NY-ESO-1 are being developed [164166]. In vitro investigations suggested that NY-ESO-1 expression is associated with poor tumor outcomes, and DCs loaded with NY-ESO-1 peptide can stimulate specific T cell responses against HCC cells [98, 167]. This implies that NY-ESO-1 has potential to be a valuable target molecule for immunotherapy against HCC. Clinically, vaccines targeting NY-ESO-1 expressing solid tumors (including HCC) are ongoing (NCT01522820).
Current studies demonstrate that DNA encoding epitope-optimized murine AFP and lentivector-mediated genetic immunization could induce potent AFP-specific CD8+ responses to generate a significant anti-tumor effect in autochthonous HCC model [168]. This may provide additional technology and new perspectives to further maximize the vaccines used in HCC therapy.
Oncolytic viruses (OVs) selectively replicate in tumor cells, damaging them, and subsequently spreading the virus in tumor tissue, while not harming normal cells. This characteristic endows OVs an effective platform for cancer immunotherapy [169]. Pre-clinical and clinical research highlights natural and genetically modified viruses, which are armed with immunomodulatory transgenes, that not only induce potent in situ anti-tumor immunity through mediating immunogenic cell death (ICD) and the inflammatory cascade, but also serve as vectors expressing therapeutic genes to improve cancer treatment [170, 171].
As for OVs used in HCC immunotherapy, previous in vitro and xenograft mice model studies indicated that a broad variety of therapeutic genes recombinant oncolytic adeno-associated viruses (AAVs) can exert a strong cytopathic effect on HCC cells (Fig. 4.). A tumor-selective replicating adenovirus expressing IFN-β, and ZD55-IFN-β, shows an elevated level of IFN-β expression, and 100-fold higher anti-tumor cytotoxicity than replicative adenovirus ONYX-015 [172]. The application of another recombinant AAVs model, AAV vectors containing human telomerase reverse transcriptase (hTERT) and tumor necrosis factor alpha related apoptosis inducing ligand (TRAIL) gene, namely AAV-hTERT-TRAIL, targets telomerase activity in HCC cells, and exhibits specific cytotoxicity and apoptosis to suppress the growth of HCC xenograft tumors [173]. An oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor (GM-CSF), Ad5-D24-GMCSF, induces tumor-specific and virus-specific immunity both in the syngeneic hamster model and patients. This suggests that oncolytic virus–mediated antitumor immunity may be a promising immunotherapeutic candidate for further clinical testing in HCC treatment [174].
In early clinical studies, oncolytic poxvirus JX-594 (also known as PexaVec), showed robust virus replication-dependent oncolysis, well-tolerated character, anti-vascular effects and anti-tumor immunity in HCC and other solid tumors [158, 175, 176]. (NCT00629759) (Table 5.). Another randomized trial in advanced HCC demonstrated oncolytic and immunotherapy mechanisms of action (MOA), tumor responses and dose-related survival (high-dose JX-594 was associated with longer OS) in individuals with HCC [159] (NCT00554372).
In general, OVs is a potent therapeutic agent for cancer treatment, and it’s promising to extend immunotherapeutic options for HCC. Importantly, dosing regimens of OVs must be better defined for its clinical use, and in this regard, further results from clinical trials are awaited.

Conclusions

HCC is characterized by immune tolerance and comprises numerous infiltrated immune cells, a great number of suppressive molecules, complex pro-inflammatory/immunoregulatory signaling and intricate interactions between different components. The picture of immune microenvironment in HCC plays a key role in HCC progression and recurrence. Apparently, interactions of HCC tumor cells and various immune components in TME are really complicated and multifaceted, finally determining the plasticity and heterogeneity of its both innate and adaptive immune responses. Transcriptional and epigenetical alterations [177], metabolic reprogramming [178] and lack of co-stimulatory signals partially contribute to exhausted phenotype of TILs. Moreover, importantly, the benefit of current predictive biomarkers (e.g. PD-L1 expression level and tumor mutation burden (TMB)) in HCC patients receiving ICBs are still limited. Despite some impressive basic and translational discoveries, more details about underlying cellular or molecular mechanisms of immune evasion in HCC need to be further clarified. It’s clear that a better understanding of HCC immune landscape and will provide new breakthroughs in its clinical treatment.
Recently, immunotherapy brings great promises and new opportunities for HCC therapeutics. Its success has been evidenced by extensive studies. However, a subset of patients with HCC has little positive clinical responses to this treatment. In addition to the current combination regimens of ICBs with TKIs, or individualized cell therapeutic approaches, more effective ways to reinvigorate anti-tumor responses are urgently warranted. In this regard, combination of PD-1/PD-L1 monoclonal antibody and targeting co-stimulatory receptors (such as 4-1BB, OX40, CD27) with agonistic antibodies seems to be a potential therapeutic option for HCC, which may enhance and reverse functions of exhausted CD8+ TILs [179]. Additionally, strategies to target altered metabolic characteristics (e.g. the Warburg effect, abnormal glutamine metabolism, and urea cycle deficiency (UCD)) or interfere with the “key point molecules” (e.g. Arginase and indoleamine 2,3-dioxygenase (IDO)) that both influence metabolic reprogramming and T cell exhaustion may be a promising exploration clinically [180]. Moreover, some emerging pre-clinical investigations indicate the developing novel therapeutic approaches like epigenetic therapy using histone deacetylase inhibitors (HDACi) combined with CAR-T treatment are helpful for identification of more precise biomarkers and opening new avenues of HCC immunotherapy.
In general, immunotherapy is becoming one of the most promising approaches for HCC treatment, and it is likely to be more powerful in the foreseeable future.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Thimme R, et al. Comprehensive analysis of the alpha-fetoprotein-specific CD8+ T cell responses in patients with hepatocellular carcinoma. Hepatology. 2008;48(6):1821–33.PubMedCrossRef Thimme R, et al. Comprehensive analysis of the alpha-fetoprotein-specific CD8+ T cell responses in patients with hepatocellular carcinoma. Hepatology. 2008;48(6):1821–33.PubMedCrossRef
2.
Zurück zum Zitat McGlynn KA, Petrick JL, London WT. Global epidemiology of hepatocellular carcinoma: an emphasis on demographic and regional variability. Clin Liver Dis. 2015;19(2):223–38.PubMedPubMedCentralCrossRef McGlynn KA, Petrick JL, London WT. Global epidemiology of hepatocellular carcinoma: an emphasis on demographic and regional variability. Clin Liver Dis. 2015;19(2):223–38.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Rimassa L, et al. Management of adverse events associated with tyrosine kinase inhibitors: improving outcomes for patients with hepatocellular carcinoma. Cancer Treat Rev. 2019;77:20–8.PubMedCrossRef Rimassa L, et al. Management of adverse events associated with tyrosine kinase inhibitors: improving outcomes for patients with hepatocellular carcinoma. Cancer Treat Rev. 2019;77:20–8.PubMedCrossRef
4.
Zurück zum Zitat Ardelt MA, et al. Inhibition of cyclin-dependent kinase 5: a strategy to improve Sorafenib response in hepatocellular carcinoma therapy. Hepatology. 2019;69(1):376–93.PubMedCrossRef Ardelt MA, et al. Inhibition of cyclin-dependent kinase 5: a strategy to improve Sorafenib response in hepatocellular carcinoma therapy. Hepatology. 2019;69(1):376–93.PubMedCrossRef
5.
Zurück zum Zitat Bollard J, et al. Palbociclib (PD-0332991), a selective CDK4/6 inhibitor, restricts tumour growth in preclinical models of hepatocellular carcinoma. Gut. 2017;66(7):1286–96.PubMedCrossRef Bollard J, et al. Palbociclib (PD-0332991), a selective CDK4/6 inhibitor, restricts tumour growth in preclinical models of hepatocellular carcinoma. Gut. 2017;66(7):1286–96.PubMedCrossRef
6.
Zurück zum Zitat Ehrlich SM, et al. Targeting cyclin dependent kinase 5 in hepatocellular carcinoma--a novel therapeutic approach. J Hepatol. 2015;63(1):102–13.PubMedCrossRef Ehrlich SM, et al. Targeting cyclin dependent kinase 5 in hepatocellular carcinoma--a novel therapeutic approach. J Hepatol. 2015;63(1):102–13.PubMedCrossRef
7.
Zurück zum Zitat Joshi JJ, et al. H3B-6527 is a potent and selective inhibitor of FGFR4 in FGF19-driven hepatocellular carcinoma. Cancer Res. 2017;77(24):6999–7013.PubMedCrossRef Joshi JJ, et al. H3B-6527 is a potent and selective inhibitor of FGFR4 in FGF19-driven hepatocellular carcinoma. Cancer Res. 2017;77(24):6999–7013.PubMedCrossRef
9.
Zurück zum Zitat Shlomai A, de Jong YP, Rice CM. Virus associated malignancies: the role of viral hepatitis in hepatocellular carcinoma. Semin Cancer Biol. 2014;26:78–88.PubMedCrossRef Shlomai A, de Jong YP, Rice CM. Virus associated malignancies: the role of viral hepatitis in hepatocellular carcinoma. Semin Cancer Biol. 2014;26:78–88.PubMedCrossRef
10.
Zurück zum Zitat Hato T, et al. Immune checkpoint blockade in hepatocellular carcinoma: current progress and future directions. Hepatology. 2014;60(5):1776–82.PubMedCrossRef Hato T, et al. Immune checkpoint blockade in hepatocellular carcinoma: current progress and future directions. Hepatology. 2014;60(5):1776–82.PubMedCrossRef
11.
Zurück zum Zitat Zhang HH, et al. Regulatory T cells in chronic hepatitis B patients affect the immunopathogenesis of hepatocellular carcinoma by suppressing the anti-tumour immune responses. J Viral Hepat. 2010;17(Suppl 1):34–43.PubMedCrossRef Zhang HH, et al. Regulatory T cells in chronic hepatitis B patients affect the immunopathogenesis of hepatocellular carcinoma by suppressing the anti-tumour immune responses. J Viral Hepat. 2010;17(Suppl 1):34–43.PubMedCrossRef
12.
Zurück zum Zitat Chew V, et al. Delineation of an immunosuppressive gradient in hepatocellular carcinoma using high-dimensional proteomic and transcriptomic analyses. Proc Natl Acad Sci U S A. 2017;114(29):E5900–9.PubMedPubMedCentralCrossRef Chew V, et al. Delineation of an immunosuppressive gradient in hepatocellular carcinoma using high-dimensional proteomic and transcriptomic analyses. Proc Natl Acad Sci U S A. 2017;114(29):E5900–9.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Wu Y, et al. Monocyte/macrophage-elicited natural killer cell dysfunction in hepatocellular carcinoma is mediated by CD48/2B4 interactions. Hepatology. 2013;57(3):1107–16.PubMedCrossRef Wu Y, et al. Monocyte/macrophage-elicited natural killer cell dysfunction in hepatocellular carcinoma is mediated by CD48/2B4 interactions. Hepatology. 2013;57(3):1107–16.PubMedCrossRef
15.
Zurück zum Zitat Kapanadze T, et al. Regulation of accumulation and function of myeloid derived suppressor cells in different murine models of hepatocellular carcinoma. J Hepatol. 2013;59(5):1007–13.PubMedPubMedCentralCrossRef Kapanadze T, et al. Regulation of accumulation and function of myeloid derived suppressor cells in different murine models of hepatocellular carcinoma. J Hepatol. 2013;59(5):1007–13.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Mok MT, et al. CCRK is a novel signalling hub exploitable in cancer immunotherapy. Pharmacol Ther. 2018;186:138–51.PubMedCrossRef Mok MT, et al. CCRK is a novel signalling hub exploitable in cancer immunotherapy. Pharmacol Ther. 2018;186:138–51.PubMedCrossRef
18.
Zurück zum Zitat Deng Y, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–101.PubMedCrossRef Deng Y, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–101.PubMedCrossRef
19.
Zurück zum Zitat Chiu DK, et al. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology. 2016;64(3):797–813.PubMedCrossRef Chiu DK, et al. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology. 2016;64(3):797–813.PubMedCrossRef
20.
Zurück zum Zitat Chiu DK, et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat Commun. 2017;8(1):517.PubMedPubMedCentralCrossRef Chiu DK, et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat Commun. 2017;8(1):517.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Kondo Y, Shimosegawa T. Significant roles of regulatory T cells and myeloid derived suppressor cells in hepatitis B virus persistent infection and hepatitis B virus-related HCCs. Int J Mol Sci. 2015;16(2):3307–22.PubMedPubMedCentralCrossRef Kondo Y, Shimosegawa T. Significant roles of regulatory T cells and myeloid derived suppressor cells in hepatitis B virus persistent infection and hepatitis B virus-related HCCs. Int J Mol Sci. 2015;16(2):3307–22.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Hoechst B, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008;135(1):234–43.PubMedCrossRef Hoechst B, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008;135(1):234–43.PubMedCrossRef
25.
Zurück zum Zitat Srivastava MK, et al. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010;70(1):68–77.PubMedCrossRef Srivastava MK, et al. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010;70(1):68–77.PubMedCrossRef
26.
Zurück zum Zitat Zhang H, et al. Critical role of myeloid-derived suppressor cells in tumor-induced liver immune suppression through inhibition of NKT cell function. Front Immunol. 2017;8:129.PubMedPubMedCentral Zhang H, et al. Critical role of myeloid-derived suppressor cells in tumor-induced liver immune suppression through inhibition of NKT cell function. Front Immunol. 2017;8:129.PubMedPubMedCentral
27.
Zurück zum Zitat Dardalhon V, et al. Tim-3/galectin-9 pathway: regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells. J Immunol. 2010;185(3):1383–92.PubMedCrossRef Dardalhon V, et al. Tim-3/galectin-9 pathway: regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells. J Immunol. 2010;185(3):1383–92.PubMedCrossRef
28.
Zurück zum Zitat Hoechst B, 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.PubMedCrossRef Hoechst B, 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.PubMedCrossRef
29.
Zurück zum Zitat Wu CJ, et al. Combination of radiation and interleukin 12 eradicates large orthotopic hepatocellular carcinoma through immunomodulation of tumor microenvironment. Oncoimmunology. 2018;7(9):e1477459.PubMedPubMedCentralCrossRef Wu CJ, et al. Combination of radiation and interleukin 12 eradicates large orthotopic hepatocellular carcinoma through immunomodulation of tumor microenvironment. Oncoimmunology. 2018;7(9):e1477459.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Movahedi K, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70(14):5728–39.PubMedCrossRef Movahedi K, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70(14):5728–39.PubMedCrossRef
32.
Zurück zum Zitat Cai H, et al. Colony-stimulating factor-1-induced AIF1 expression in tumor-associated macrophages enhances the progression of hepatocellular carcinoma. Oncoimmunology. 2017;6(9):e1333213.PubMedPubMedCentralCrossRef Cai H, et al. Colony-stimulating factor-1-induced AIF1 expression in tumor-associated macrophages enhances the progression of hepatocellular carcinoma. Oncoimmunology. 2017;6(9):e1333213.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Huang W, et al. Interleukin-8 Induces Expression of FOXC1 to Promote Transactivation of CXCR1 and CCL2 in Hepatocellular Carcinoma Cell Lines and Formation of Metastases in Mice. Gastroenterology. 2015;149(4):1053–67 e14.PubMedCrossRef Huang W, et al. Interleukin-8 Induces Expression of FOXC1 to Promote Transactivation of CXCR1 and CCL2 in Hepatocellular Carcinoma Cell Lines and Formation of Metastases in Mice. Gastroenterology. 2015;149(4):1053–67 e14.PubMedCrossRef
34.
Zurück zum Zitat Wang TT, et al. CTGF secreted by mesenchymal-like hepatocellular carcinoma cells plays a role in the polarization of macrophages in hepatocellular carcinoma progression. Biomed Pharmacother. 2017;95:111–9.PubMedCrossRef Wang TT, et al. CTGF secreted by mesenchymal-like hepatocellular carcinoma cells plays a role in the polarization of macrophages in hepatocellular carcinoma progression. Biomed Pharmacother. 2017;95:111–9.PubMedCrossRef
35.
Zurück zum Zitat Capece D, et al. The inflammatory microenvironment in hepatocellular carcinoma: a pivotal role for tumor-associated macrophages. Biomed Res Int. 2013;2013:187204.PubMedCrossRef Capece D, et al. The inflammatory microenvironment in hepatocellular carcinoma: a pivotal role for tumor-associated macrophages. Biomed Res Int. 2013;2013:187204.PubMedCrossRef
38.
Zurück zum Zitat Zhou J, et al. Increased intratumoral regulatory T cells are related to intratumoral macrophages and poor prognosis in hepatocellular carcinoma patients. Int J Cancer. 2009;125(7):1640–8.PubMedCrossRef Zhou J, et al. Increased intratumoral regulatory T cells are related to intratumoral macrophages and poor prognosis in hepatocellular carcinoma patients. Int J Cancer. 2009;125(7):1640–8.PubMedCrossRef
39.
Zurück zum Zitat Sharma S, et al. CD4+CD25+CD127(low) regulatory T cells play predominant anti-tumor suppressive role in hepatitis B virus-associated hepatocellular carcinoma. Front Immunol. 2015;6:49.PubMedPubMedCentralCrossRef Sharma S, et al. CD4+CD25+CD127(low) regulatory T cells play predominant anti-tumor suppressive role in hepatitis B virus-associated hepatocellular carcinoma. Front Immunol. 2015;6:49.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Kuang DM, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma promote expansion of memory T helper 17 cells. Hepatology. 2010;51(1):154–64.PubMedCrossRef Kuang DM, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma promote expansion of memory T helper 17 cells. Hepatology. 2010;51(1):154–64.PubMedCrossRef
41.
Zurück zum Zitat Rani B, et al. Role of the tissue microenvironment as a therapeutic target in hepatocellular carcinoma. World J Gastroenterol. 2014;20(15):4128–40.PubMedPubMedCentralCrossRef Rani B, et al. Role of the tissue microenvironment as a therapeutic target in hepatocellular carcinoma. World J Gastroenterol. 2014;20(15):4128–40.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Lanaya H, et al. EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nat Cell Biol. 2014;16(10):972–7.PubMedPubMedCentralCrossRef Lanaya H, et al. EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nat Cell Biol. 2014;16(10):972–7.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Yan W, et al. Tim-3 fosters HCC development by enhancing TGF-beta-mediated alternative activation of macrophages. Gut. 2015;64(10):1593–604.PubMedCrossRef Yan W, et al. Tim-3 fosters HCC development by enhancing TGF-beta-mediated alternative activation of macrophages. Gut. 2015;64(10):1593–604.PubMedCrossRef
44.
Zurück zum Zitat Wan S, et al. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. 2014;147(6):1393–404.PubMedCrossRef Wan S, et al. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. 2014;147(6):1393–404.PubMedCrossRef
46.
Zurück zum Zitat Nicolas-Avila JA, Adrover JM, Hidalgo A. Neutrophils in homeostasis, immunity, and Cancer. Immunity. 2017;46(1):15–28.PubMedCrossRef Nicolas-Avila JA, Adrover JM, Hidalgo A. Neutrophils in homeostasis, immunity, and Cancer. Immunity. 2017;46(1):15–28.PubMedCrossRef
47.
Zurück zum Zitat Ameratunga M, et al. Neutrophil-lymphocyte ratio kinetics in patients with advanced solid tumours on phase I trials of PD-1/PD-L1 inhibitors. Eur J Cancer. 2018;89:56–63.PubMedCrossRef Ameratunga M, et al. Neutrophil-lymphocyte ratio kinetics in patients with advanced solid tumours on phase I trials of PD-1/PD-L1 inhibitors. Eur J Cancer. 2018;89:56–63.PubMedCrossRef
48.
Zurück zum Zitat Jeyakumar G, et al. Neutrophil lymphocyte ratio and duration of prior anti-angiogenic therapy as biomarkers in metastatic RCC receiving immune checkpoint inhibitor therapy. J Immunother Cancer. 2017;5(1):82.PubMedPubMedCentralCrossRef Jeyakumar G, et al. Neutrophil lymphocyte ratio and duration of prior anti-angiogenic therapy as biomarkers in metastatic RCC receiving immune checkpoint inhibitor therapy. J Immunother Cancer. 2017;5(1):82.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Andzinski L, et al. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. 2016;138(8):1982–93.PubMedCrossRef Andzinski L, et al. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. 2016;138(8):1982–93.PubMedCrossRef
50.
Zurück zum Zitat Michaeli J, et al. Tumor-associated neutrophils induce apoptosis of non-activated CD8 T-cells in a TNFalpha and NO-dependent mechanism, promoting a tumor-supportive environment. Oncoimmunology. 2017;6(11):e1356965.PubMedPubMedCentralCrossRef Michaeli J, et al. Tumor-associated neutrophils induce apoptosis of non-activated CD8 T-cells in a TNFalpha and NO-dependent mechanism, promoting a tumor-supportive environment. Oncoimmunology. 2017;6(11):e1356965.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Margetts J, et al. Neutrophils: driving progression and poor prognosis in hepatocellular carcinoma? Br J Cancer. 2018;118(2):248–57.PubMedCrossRef Margetts J, et al. Neutrophils: driving progression and poor prognosis in hepatocellular carcinoma? Br J Cancer. 2018;118(2):248–57.PubMedCrossRef
52.
Zurück zum Zitat Mano Y, et al. Preoperative neutrophil-to-lymphocyte ratio is a predictor of survival after hepatectomy for hepatocellular carcinoma: a retrospective analysis. Ann Surg. 2013;258(2):301–5.PubMedCrossRef Mano Y, et al. Preoperative neutrophil-to-lymphocyte ratio is a predictor of survival after hepatectomy for hepatocellular carcinoma: a retrospective analysis. Ann Surg. 2013;258(2):301–5.PubMedCrossRef
53.
Zurück zum Zitat Koh MY, et al. A new HIF-1alpha/RANTES-driven pathway to hepatocellular carcinoma mediated by germline haploinsufficiency of SART1/HAF in mice. Hepatology. 2016;63(5):1576–91.PubMedCrossRef Koh MY, et al. A new HIF-1alpha/RANTES-driven pathway to hepatocellular carcinoma mediated by germline haploinsufficiency of SART1/HAF in mice. Hepatology. 2016;63(5):1576–91.PubMedCrossRef
54.
Zurück zum Zitat Zhou SL, et al. Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to Sorafenib. Gastroenterology. 2016;150(7):1646–1658.e17.PubMedCrossRef Zhou SL, et al. Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to Sorafenib. Gastroenterology. 2016;150(7):1646–1658.e17.PubMedCrossRef
56.
Zurück zum Zitat Unitt E, et al. Tumour lymphocytic infiltrate and recurrence of hepatocellular carcinoma following liver transplantation. J Hepatol. 2006;45(2):246–53.PubMedCrossRef Unitt E, et al. Tumour lymphocytic infiltrate and recurrence of hepatocellular carcinoma following liver transplantation. J Hepatol. 2006;45(2):246–53.PubMedCrossRef
57.
Zurück zum Zitat Gao Q, et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007;25(18):2586–93.PubMedCrossRef Gao Q, et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007;25(18):2586–93.PubMedCrossRef
58.
Zurück zum Zitat Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015;12(12):681–700.PubMedCrossRef Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015;12(12):681–700.PubMedCrossRef
59.
Zurück zum Zitat Chen KJ, 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.PubMedPubMedCentralCrossRef Chen KJ, 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.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Jiang R, et al. The long noncoding RNA lnc-EGFR stimulates T-regulatory cells differentiation thus promoting hepatocellular carcinoma immune evasion. Nat Commun. 2017;8:15129.PubMedPubMedCentralCrossRef Jiang R, et al. The long noncoding RNA lnc-EGFR stimulates T-regulatory cells differentiation thus promoting hepatocellular carcinoma immune evasion. Nat Commun. 2017;8:15129.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Zaiss DM, et al. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor. Immunity. 2013;38(2):275–84.PubMedPubMedCentralCrossRef Zaiss DM, et al. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor. Immunity. 2013;38(2):275–84.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Kalathil S, et al. Higher frequencies of GARP(+)CTLA-4(+)Foxp3(+) T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res. 2013;73(8):2435–44.PubMedPubMedCentralCrossRef Kalathil S, et al. Higher frequencies of GARP(+)CTLA-4(+)Foxp3(+) T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res. 2013;73(8):2435–44.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Yuan CH, et al. Amphiregulin activates regulatory T lymphocytes and suppresses CD8+ T cell-mediated anti-tumor response in hepatocellular carcinoma cells. Oncotarget. 2015;6(31):32138–53.PubMedPubMedCentralCrossRef Yuan CH, et al. Amphiregulin activates regulatory T lymphocytes and suppresses CD8+ T cell-mediated anti-tumor response in hepatocellular carcinoma cells. Oncotarget. 2015;6(31):32138–53.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Fu J, et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology. 2007;132(7):2328–39.PubMedCrossRef Fu J, et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology. 2007;132(7):2328–39.PubMedCrossRef
65.
Zurück zum Zitat Huang Y, et al. Tumor-infiltrating FoxP3+ Tregs and CD8+ T cells affect the prognosis of hepatocellular carcinoma patients. Digestion. 2012;86(4):329–37.PubMedCrossRef Huang Y, et al. Tumor-infiltrating FoxP3+ Tregs and CD8+ T cells affect the prognosis of hepatocellular carcinoma patients. Digestion. 2012;86(4):329–37.PubMedCrossRef
66.
Zurück zum Zitat Fu YP, et al. Overexpression of interleukin-35 associates with hepatocellular carcinoma aggressiveness and recurrence after curative resection. Br J Cancer. 2016;114(7):767–76.PubMedPubMedCentralCrossRef Fu YP, et al. Overexpression of interleukin-35 associates with hepatocellular carcinoma aggressiveness and recurrence after curative resection. Br J Cancer. 2016;114(7):767–76.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Ye LY, et al. Hypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76(4):818–30.PubMedCrossRef Ye LY, et al. Hypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76(4):818–30.PubMedCrossRef
68.
Zurück zum Zitat Kuang DM, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206(6):1327–37.PubMedPubMedCentralCrossRef Kuang DM, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206(6):1327–37.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Guo CL, et al. Expression of Fas/FasL in CD8+ T and CD3+ Foxp3+ Treg cells--relationship with apoptosis of circulating CD8+ T cells in hepatocellular carcinoma patients. Asian Pac J Cancer Prev. 2014;15(6):2613–8.PubMedCrossRef Guo CL, et al. Expression of Fas/FasL in CD8+ T and CD3+ Foxp3+ Treg cells--relationship with apoptosis of circulating CD8+ T cells in hepatocellular carcinoma patients. Asian Pac J Cancer Prev. 2014;15(6):2613–8.PubMedCrossRef
70.
Zurück zum Zitat Li L, et al. CXCL17 expression predicts poor prognosis and correlates with adverse immune infiltration in hepatocellular carcinoma. PLoS One. 2014;9(10):e110064.PubMedPubMedCentralCrossRef Li L, et al. CXCL17 expression predicts poor prognosis and correlates with adverse immune infiltration in hepatocellular carcinoma. PLoS One. 2014;9(10):e110064.PubMedPubMedCentralCrossRef
71.
72.
Zurück zum Zitat Moreno-Cubero E, Larrubia JR. Specific CD8(+) T cell response immunotherapy for hepatocellular carcinoma and viral hepatitis. World J Gastroenterol. 2016;22(28):6469–83.PubMedPubMedCentralCrossRef Moreno-Cubero E, Larrubia JR. Specific CD8(+) T cell response immunotherapy for hepatocellular carcinoma and viral hepatitis. World J Gastroenterol. 2016;22(28):6469–83.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Flecken T, et al. Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology. 2014;59(4):1415–26.PubMedCrossRef Flecken T, et al. Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology. 2014;59(4):1415–26.PubMedCrossRef
74.
Zurück zum Zitat Guidotti LG, et al. Immunosurveillance of the liver by intravascular effector CD8(+) T cells. Cell. 2015;161(3):486–500.PubMedCrossRef Guidotti LG, et al. Immunosurveillance of the liver by intravascular effector CD8(+) T cells. Cell. 2015;161(3):486–500.PubMedCrossRef
75.
Zurück zum Zitat Han 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):567–79.PubMedCrossRef Han 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):567–79.PubMedCrossRef
76.
Zurück zum Zitat Li H, et al. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology. 2012;56(4):1342–51.PubMedCrossRef Li H, et al. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology. 2012;56(4):1342–51.PubMedCrossRef
77.
Zurück zum Zitat Woo SR, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–27.PubMedCrossRef Woo SR, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–27.PubMedCrossRef
78.
Zurück zum Zitat Kamimura H, et al. Reduced NKG2D ligand expression in hepatocellular carcinoma correlates with early recurrence. J Hepatol. 2012;56(2):381–8.PubMedCrossRef Kamimura H, et al. Reduced NKG2D ligand expression in hepatocellular carcinoma correlates with early recurrence. J Hepatol. 2012;56(2):381–8.PubMedCrossRef
79.
80.
Zurück zum Zitat Yamamoto M, et al. alpha-Fetoprotein impairs activation of natural killer cells by inhibiting the function of dendritic cells. Clin Exp Immunol. 2011;165(2):211–9.PubMedPubMedCentralCrossRef Yamamoto M, et al. alpha-Fetoprotein impairs activation of natural killer cells by inhibiting the function of dendritic cells. Clin Exp Immunol. 2011;165(2):211–9.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Zhang C, et al. Interleukin-12 improves cytotoxicity of natural killer cells via upregulated expression of NKG2D. Hum Immunol. 2008;69(8):490–500.PubMedCrossRef Zhang C, et al. Interleukin-12 improves cytotoxicity of natural killer cells via upregulated expression of NKG2D. Hum Immunol. 2008;69(8):490–500.PubMedCrossRef
82.
Zurück zum Zitat Vujanovic L, et al. Tumor-derived alpha-fetoprotein directly drives human natural killer-cell activation and subsequent cell death. Cancer Immunol Res. 2017;5(6):493–502.PubMedCrossRef Vujanovic L, et al. Tumor-derived alpha-fetoprotein directly drives human natural killer-cell activation and subsequent cell death. Cancer Immunol Res. 2017;5(6):493–502.PubMedCrossRef
83.
Zurück zum Zitat Sun C, et al. NK cell receptor imbalance and NK cell dysfunction in HBV infection and hepatocellular carcinoma. Cell Mol Immunol. 2015;12(3):292–302.PubMedCrossRef Sun C, et al. NK cell receptor imbalance and NK cell dysfunction in HBV infection and hepatocellular carcinoma. Cell Mol Immunol. 2015;12(3):292–302.PubMedCrossRef
84.
Zurück zum Zitat Langhans B, et al. Regulatory CD4+ T cells modulate the interaction between NK cells and hepatic stellate cells by acting on either cell type. J Hepatol. 2015;62(2):398–404.PubMedCrossRef Langhans B, et al. Regulatory CD4+ T cells modulate the interaction between NK cells and hepatic stellate cells by acting on either cell type. J Hepatol. 2015;62(2):398–404.PubMedCrossRef
85.
Zurück zum Zitat Li T, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–61.PubMedCrossRef Li T, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–61.PubMedCrossRef
86.
Zurück zum Zitat Muhanna N, et al. Amelioration of hepatic fibrosis by NK cell activation. Gut. 2011;60(1):90–8.PubMedCrossRef Muhanna N, et al. Amelioration of hepatic fibrosis by NK cell activation. Gut. 2011;60(1):90–8.PubMedCrossRef
87.
Zurück zum Zitat Zhang M, et al. Apoptotic cells attenuate fulminant hepatitis by priming Kupffer cells to produce interleukin-10 through membrane-bound TGF-beta. Hepatology. 2011;53(1):306–16.PubMedCrossRef Zhang M, et al. Apoptotic cells attenuate fulminant hepatitis by priming Kupffer cells to produce interleukin-10 through membrane-bound TGF-beta. Hepatology. 2011;53(1):306–16.PubMedCrossRef
88.
Zurück zum Zitat Lacotte S, et al. Impact of myeloid-derived suppressor cell on Kupffer cells from mouse livers with hepatocellular carcinoma. Oncoimmunology. 2016;5(11):e1234565.PubMedPubMedCentralCrossRef Lacotte S, et al. Impact of myeloid-derived suppressor cell on Kupffer cells from mouse livers with hepatocellular carcinoma. Oncoimmunology. 2016;5(11):e1234565.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Wu J, et al. The proinflammatory myeloid cell receptor TREM-1 controls Kupffer cell activation and development of hepatocellular carcinoma. Cancer Res. 2012;72(16):3977–86.PubMedPubMedCentralCrossRef Wu J, et al. The proinflammatory myeloid cell receptor TREM-1 controls Kupffer cell activation and development of hepatocellular carcinoma. Cancer Res. 2012;72(16):3977–86.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Sun K, et al. Autophagy-deficient Kupffer cells promote tumorigenesis by enhancing mtROS-NF-kappaB-IL1alpha/beta-dependent inflammation and fibrosis during the preneoplastic stage of hepatocarcinogenesis. Cancer Lett. 2017;388:198–207.PubMedCrossRef Sun K, et al. Autophagy-deficient Kupffer cells promote tumorigenesis by enhancing mtROS-NF-kappaB-IL1alpha/beta-dependent inflammation and fibrosis during the preneoplastic stage of hepatocarcinogenesis. Cancer Lett. 2017;388:198–207.PubMedCrossRef
91.
Zurück zum Zitat Mengshol JA, et al. A crucial role for Kupffer cell-derived galectin-9 in regulation of T cell immunity in hepatitis C infection. PLoS One. 2010;5(3):e9504.PubMedPubMedCentralCrossRef Mengshol JA, et al. A crucial role for Kupffer cell-derived galectin-9 in regulation of T cell immunity in hepatitis C infection. PLoS One. 2010;5(3):e9504.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Kim HY, Park JW. Current immunotherapeutic strategies in hepatocellular carcinoma: recent advances and future directions. Ther Adv Gastroenterol. 2017;10(10):805–14.CrossRef Kim HY, Park JW. Current immunotherapeutic strategies in hepatocellular carcinoma: recent advances and future directions. Ther Adv Gastroenterol. 2017;10(10):805–14.CrossRef
93.
Zurück zum Zitat Fujita K, et al. Correlation between serum galectin-9 levels and liver fibrosis. J Gastroenterol Hepatol. 2018;33(2):492–9.PubMedCrossRef Fujita K, et al. Correlation between serum galectin-9 levels and liver fibrosis. J Gastroenterol Hepatol. 2018;33(2):492–9.PubMedCrossRef
94.
Zurück zum Zitat Radford KJ, Tullett KM, Lahoud MH. Dendritic cells and cancer immunotherapy. Curr Opin Immunol. 2014;27:26–32.PubMedCrossRef Radford KJ, Tullett KM, Lahoud MH. Dendritic cells and cancer immunotherapy. Curr Opin Immunol. 2014;27:26–32.PubMedCrossRef
95.
Zurück zum Zitat Rao Q, et al. Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro. Hepatology. 2016;64(2):456–72.PubMedCrossRef Rao Q, et al. Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro. Hepatology. 2016;64(2):456–72.PubMedCrossRef
96.
Zurück zum Zitat Lu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMedCrossRef Lu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMedCrossRef
97.
Zurück zum Zitat Cao DY, et al. alpha-fetoprotein and interleukin-18 gene-modified dendritic cells effectively stimulate specific type-1 CD4- and CD8-mediated T-Cell response from hepatocellular carcinoma patients in Vitro. Hum Immunol. 2007;68(5):334–41.PubMedCrossRef Cao DY, et al. alpha-fetoprotein and interleukin-18 gene-modified dendritic cells effectively stimulate specific type-1 CD4- and CD8-mediated T-Cell response from hepatocellular carcinoma patients in Vitro. Hum Immunol. 2007;68(5):334–41.PubMedCrossRef
98.
Zurück zum Zitat Chen Y, et al. Potential therapeutic value of dendritic cells loaded with NYESO1 protein for the immunotherapy of advanced hepatocellular carcinoma. Int J Mol Med. 2013;32(6):1366–72.PubMedCrossRef Chen Y, et al. Potential therapeutic value of dendritic cells loaded with NYESO1 protein for the immunotherapy of advanced hepatocellular carcinoma. Int J Mol Med. 2013;32(6):1366–72.PubMedCrossRef
99.
Zurück zum Zitat Vogt A, et al. Improving immunotherapy of hepatocellular carcinoma (HCC) using dendritic cells (DC) engineered to express IL-12 in vivo. Liver Int. 2014;34(3):447–61.PubMedCrossRef Vogt A, et al. Improving immunotherapy of hepatocellular carcinoma (HCC) using dendritic cells (DC) engineered to express IL-12 in vivo. Liver Int. 2014;34(3):447–61.PubMedCrossRef
100.
Zurück zum Zitat Gonzalez-Carmona MA, et al. CD40ligand-expressing dendritic cells induce regression of hepatocellular carcinoma by activating innate and acquired immunity in vivo. Hepatology. 2008;48(1):157–68.PubMedCrossRef Gonzalez-Carmona MA, et al. CD40ligand-expressing dendritic cells induce regression of hepatocellular carcinoma by activating innate and acquired immunity in vivo. Hepatology. 2008;48(1):157–68.PubMedCrossRef
101.
Zurück zum Zitat Tada F, et al. Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Int J Oncol. 2012;41(5):1601–9.PubMedPubMedCentralCrossRef Tada F, et al. Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Int J Oncol. 2012;41(5):1601–9.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Lee JH, 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):1666–76.PubMedPubMedCentralCrossRef Lee JH, 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):1666–76.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Palmer DH, et al. A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology. 2009;49(1):124–32.PubMedCrossRef Palmer DH, et al. A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology. 2009;49(1):124–32.PubMedCrossRef
104.
Zurück zum Zitat El AM, et al. Immunotherapy by autologous dendritic cell vaccine in patients with advanced HCC. J Cancer Res Clin Oncol. 2013;139(1):39–48.CrossRef El AM, et al. Immunotherapy by autologous dendritic cell vaccine in patients with advanced HCC. J Cancer Res Clin Oncol. 2013;139(1):39–48.CrossRef
105.
Zurück zum Zitat Butterfield LH, et al. A phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four alpha-fetoprotein peptides. Clin Cancer Res. 2006;12(9):2817–25.PubMedCrossRef Butterfield LH, et al. A phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four alpha-fetoprotein peptides. Clin Cancer Res. 2006;12(9):2817–25.PubMedCrossRef
106.
Zurück zum Zitat Langhans B, et al. Intrahepatic IL-8 producing Foxp3(+)CD4(+) regulatory T cells and fibrogenesis in chronic hepatitis C. J Hepatol. 2013;59(2):229–35.PubMedCrossRef Langhans B, et al. Intrahepatic IL-8 producing Foxp3(+)CD4(+) regulatory T cells and fibrogenesis in chronic hepatitis C. J Hepatol. 2013;59(2):229–35.PubMedCrossRef
107.
Zurück zum Zitat Ouyang FZ, et al. Dendritic cell-elicited B-cell activation fosters immune privilege via IL-10 signals in hepatocellular carcinoma. Nat Commun. 2016;7:13453.PubMedPubMedCentralCrossRef Ouyang FZ, et al. Dendritic cell-elicited B-cell activation fosters immune privilege via IL-10 signals in hepatocellular carcinoma. Nat Commun. 2016;7:13453.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Ambade A, et al. Hepatocellular carcinoma is accelerated by NASH involving M2 macrophage polarization mediated by hif-1alphainduced IL-10. Oncoimmunology. 2016;5(10):e1221557.PubMedPubMedCentralCrossRef Ambade A, et al. Hepatocellular carcinoma is accelerated by NASH involving M2 macrophage polarization mediated by hif-1alphainduced IL-10. Oncoimmunology. 2016;5(10):e1221557.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Cheng JT, et al. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5:e198.PubMedPubMedCentralCrossRef Cheng JT, et al. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5:e198.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Shi F, et al. PD-1 and PD-L1 upregulation promotes CD8(+) T-cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients. Int J Cancer. 2011;128(4):887–96.PubMedCrossRef Shi F, et al. PD-1 and PD-L1 upregulation promotes CD8(+) T-cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients. Int J Cancer. 2011;128(4):887–96.PubMedCrossRef
111.
Zurück zum Zitat Lim TS, et al. PD-1 expression on dendritic cells suppresses CD8(+) T cell function and antitumor immunity. Oncoimmunology. 2016;5(3):e1085146.PubMedCrossRef Lim TS, et al. PD-1 expression on dendritic cells suppresses CD8(+) T cell function and antitumor immunity. Oncoimmunology. 2016;5(3):e1085146.PubMedCrossRef
112.
Zurück zum Zitat Zhou G, et al. Antibodies against immune checkpoint molecules restore functions of tumor-infiltrating T cells in hepatocellular carcinomas. Gastroenterology. 2017;153(4):1107–1119.e10.PubMedCrossRef Zhou G, et al. Antibodies against immune checkpoint molecules restore functions of tumor-infiltrating T cells in hepatocellular carcinomas. Gastroenterology. 2017;153(4):1107–1119.e10.PubMedCrossRef
113.
Zurück zum Zitat Rong YH, et al. Tim-3 expression on peripheral monocytes and CD3+CD16/CD56+natural killer-like T cells in patients with chronic hepatitis B. Tissue Antigens. 2014;83(2):76–81.PubMedCrossRef Rong YH, et al. Tim-3 expression on peripheral monocytes and CD3+CD16/CD56+natural killer-like T cells in patients with chronic hepatitis B. Tissue Antigens. 2014;83(2):76–81.PubMedCrossRef
114.
Zurück zum Zitat Pan K, et al. Expression and prognosis role of indoleamine 2,3-dioxygenase in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2008;134(11):1247–53.PubMedCrossRef Pan K, et al. Expression and prognosis role of indoleamine 2,3-dioxygenase in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2008;134(11):1247–53.PubMedCrossRef
115.
Zurück zum Zitat Chen Y, et al. Differential effects of sorafenib on liver versus tumor fibrosis mediated by stromal-derived factor 1 alpha/C-X-C receptor type 4 axis and myeloid differentiation antigen-positive myeloid cell infiltration in mice. Hepatology. 2014;59(4):1435–47.PubMedCrossRef Chen Y, et al. Differential effects of sorafenib on liver versus tumor fibrosis mediated by stromal-derived factor 1 alpha/C-X-C receptor type 4 axis and myeloid differentiation antigen-positive myeloid cell infiltration in mice. Hepatology. 2014;59(4):1435–47.PubMedCrossRef
116.
Zurück zum Zitat Sung YC, et al. Combined delivery of sorafenib and a MEK inhibitor using CXCR4-targeted nanoparticles reduces hepatic fibrosis and prevents tumor development. Theranostics. 2018;8(4):894–905.PubMedPubMedCentralCrossRef Sung YC, et al. Combined delivery of sorafenib and a MEK inhibitor using CXCR4-targeted nanoparticles reduces hepatic fibrosis and prevents tumor development. Theranostics. 2018;8(4):894–905.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Li X, et al. Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 2017;66(1):157–67.PubMedCrossRef Li X, et al. Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 2017;66(1):157–67.PubMedCrossRef
118.
Zurück zum Zitat Baeck C, et al. Pharmacological inhibition of the chemokine CCL2 (MCP-1) diminishes liver macrophage infiltration and steatohepatitis in chronic hepatic injury. Gut. 2012;61(3):416–26.PubMedCrossRef Baeck C, et al. Pharmacological inhibition of the chemokine CCL2 (MCP-1) diminishes liver macrophage infiltration and steatohepatitis in chronic hepatic injury. Gut. 2012;61(3):416–26.PubMedCrossRef
119.
Zurück zum Zitat Ren L, et al. Hypoxia-induced CCL28 promotes recruitment of regulatory T cells and tumor growth in liver cancer. Oncotarget. 2016;7(46):75763–73.PubMedPubMedCentralCrossRef Ren L, et al. Hypoxia-induced CCL28 promotes recruitment of regulatory T cells and tumor growth in liver cancer. Oncotarget. 2016;7(46):75763–73.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Facciabene A, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T (reg) cells. Nature. 2011;475(7355):226–30.PubMedCrossRef Facciabene A, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T (reg) cells. Nature. 2011;475(7355):226–30.PubMedCrossRef
122.
Zurück zum Zitat Liu LZ, et al. CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology. 2019;69(1):143–59.PubMedCrossRef Liu LZ, et al. CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology. 2019;69(1):143–59.PubMedCrossRef
123.
124.
Zurück zum Zitat Latchman Y, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2(3):261–8.PubMedCrossRef Latchman Y, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2(3):261–8.PubMedCrossRef
125.
Zurück zum Zitat El-Khoueiry AB, 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):2492–502.PubMedCrossRefPubMedCentral El-Khoueiry AB, 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):2492–502.PubMedCrossRefPubMedCentral
127.
Zurück zum Zitat Zhu AX, 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):940–52.PubMedCrossRef Zhu AX, 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):940–52.PubMedCrossRef
128.
Zurück zum Zitat Chowdhury PS, Chamoto K, Honjo T. Combination therapy strategies for improving PD-1 blockade efficacy: a new era in cancer immunotherapy. J Intern Med. 2018;283(2):110–20.PubMedCrossRef Chowdhury PS, Chamoto K, Honjo T. Combination therapy strategies for improving PD-1 blockade efficacy: a new era in cancer immunotherapy. J Intern Med. 2018;283(2):110–20.PubMedCrossRef
129.
Zurück zum Zitat Mocan T, et al. Programmed cell death protein-1 (PD-1)/programmed death-ligand-1 (PD-L1) axis in hepatocellular carcinoma: prognostic and therapeutic perspectives. Clin Transl Oncol. 2019;21(6):702–12.PubMedCrossRef Mocan T, et al. Programmed cell death protein-1 (PD-1)/programmed death-ligand-1 (PD-L1) axis in hepatocellular carcinoma: prognostic and therapeutic perspectives. Clin Transl Oncol. 2019;21(6):702–12.PubMedCrossRef
130.
Zurück zum Zitat Harding JJ. Immune checkpoint blockade in advanced hepatocellular carcinoma: an update and critical review of ongoing clinical trials. Future Oncol. 2018;14(22):2293–302.PubMedCrossRefPubMedCentral Harding JJ. Immune checkpoint blockade in advanced hepatocellular carcinoma: an update and critical review of ongoing clinical trials. Future Oncol. 2018;14(22):2293–302.PubMedCrossRefPubMedCentral
131.
Zurück zum Zitat Kudo M. Immune checkpoint inhibition in hepatocellular carcinoma: basics and ongoing clinical trials. Oncology. 2017;92(Suppl 1):50–62.PubMedCrossRef Kudo M. Immune checkpoint inhibition in hepatocellular carcinoma: basics and ongoing clinical trials. Oncology. 2017;92(Suppl 1):50–62.PubMedCrossRef
132.
Zurück zum Zitat Sangro B, 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):81–8.PubMedCrossRef Sangro B, 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):81–8.PubMedCrossRef
133.
Zurück zum Zitat Duffy AG, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66(3):545–51.PubMedCrossRef Duffy AG, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66(3):545–51.PubMedCrossRef
134.
Zurück zum Zitat Nebbia G, et al. Upregulation of the Tim-3/galectin-9 pathway of T cell exhaustion in chronic hepatitis B virus infection. PLoS One. 2012;7(10):e47648.PubMedPubMedCentralCrossRef Nebbia G, et al. Upregulation of the Tim-3/galectin-9 pathway of T cell exhaustion in chronic hepatitis B virus infection. PLoS One. 2012;7(10):e47648.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Tan AT, et al. Use of expression profiles of HBV-DNA integrated into genomes of hepatocellular carcinoma cells to select T cells for immunotherapy. Gastroenterology. 2019;156(6):1862–1876.e9.PubMedCrossRef Tan AT, et al. Use of expression profiles of HBV-DNA integrated into genomes of hepatocellular carcinoma cells to select T cells for immunotherapy. Gastroenterology. 2019;156(6):1862–1876.e9.PubMedCrossRef
137.
Zurück zum Zitat Qasim W, 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):486–91.PubMedCrossRef Qasim W, 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):486–91.PubMedCrossRef
138.
Zurück zum Zitat Zhang R, et al. Adoptive cell transfer therapy for hepatocellular carcinoma. Front Med. 2019;13(1):3–11.PubMedCrossRef Zhang R, et al. Adoptive cell transfer therapy for hepatocellular carcinoma. Front Med. 2019;13(1):3–11.PubMedCrossRef
139.
Zurück zum Zitat Schmidt TL, Negrin RS, Contag CH. A killer choice for cancer immunotherapy. Immunol Res. 2014;58(2–3):300–6.PubMedCrossRef Schmidt TL, Negrin RS, Contag CH. A killer choice for cancer immunotherapy. Immunol Res. 2014;58(2–3):300–6.PubMedCrossRef
140.
Zurück zum Zitat Rong XX, et al. Recognition and killing of cancer stem-like cell population in hepatocellular carcinoma cells by cytokine-induced killer cells via NKG2d-ligands recognition. Oncoimmunology. 2016;5(3):e1086060.PubMedCrossRef Rong XX, et al. Recognition and killing of cancer stem-like cell population in hepatocellular carcinoma cells by cytokine-induced killer cells via NKG2d-ligands recognition. Oncoimmunology. 2016;5(3):e1086060.PubMedCrossRef
141.
Zurück zum Zitat Chang B, et al. High number of PD-1 positive intratumoural lymphocytes predicts survival benefit of cytokine-induced killer cells for hepatocellular carcinoma patients. Liver Int. 2018;38(8):1449–58.PubMedPubMedCentralCrossRef Chang B, et al. High number of PD-1 positive intratumoural lymphocytes predicts survival benefit of cytokine-induced killer cells for hepatocellular carcinoma patients. Liver Int. 2018;38(8):1449–58.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Lee JH, et al. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology. 2015;148(7):1383–91.e6.PubMedCrossRef Lee JH, et al. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology. 2015;148(7):1383–91.e6.PubMedCrossRef
143.
Zurück zum Zitat Huang ZM, et al. Cytokine-induced killer cells in combination with transcatheter arterial chemoembolization and radiofrequency ablation for hepatocellular carcinoma patients. J Immunother. 2013;36(5):287–93.PubMedCrossRef Huang ZM, et al. Cytokine-induced killer cells in combination with transcatheter arterial chemoembolization and radiofrequency ablation for hepatocellular carcinoma patients. J Immunother. 2013;36(5):287–93.PubMedCrossRef
144.
Zurück zum Zitat Qiu Y, et al. Hepatocellular carcinoma-specific immunotherapy with synthesized alpha1,3- galactosyl epitope-pulsed dendritic cells and cytokine-induced killer cells. World J Gastroenterol. 2011;17(48):5260–6.PubMedPubMedCentralCrossRef Qiu Y, et al. Hepatocellular carcinoma-specific immunotherapy with synthesized alpha1,3- galactosyl epitope-pulsed dendritic cells and cytokine-induced killer cells. World J Gastroenterol. 2011;17(48):5260–6.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Pan K, et al. The efficacy of cytokine-induced killer cell infusion as an adjuvant therapy for postoperative hepatocellular carcinoma patients. Ann Surg Oncol. 2013;20(13):4305–11.PubMedCrossRef Pan K, et al. The efficacy of cytokine-induced killer cell infusion as an adjuvant therapy for postoperative hepatocellular carcinoma patients. Ann Surg Oncol. 2013;20(13):4305–11.PubMedCrossRef
146.
Zurück zum Zitat Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer. 2013;13(8):525–41.PubMedCrossRef Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer. 2013;13(8):525–41.PubMedCrossRef
147.
Zurück zum Zitat Kochenderfer JN, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012;119(12):2709–20.PubMedPubMedCentralCrossRef Kochenderfer JN, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012;119(12):2709–20.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Till BG, et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood. 2012;119(17):3940–50.PubMedPubMedCentralCrossRef Till BG, et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood. 2012;119(17):3940–50.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Gao H, et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. 2014;20(24):6418–28.PubMedCrossRef Gao H, et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. 2014;20(24):6418–28.PubMedCrossRef
151.
Zurück zum Zitat Jiang Z, et al. Anti-GPC3-CAR T cells suppress the growth of tumor cells in patient-derived xenografts of hepatocellular carcinoma. Front Immunol. 2016;7:690.PubMedCrossRef Jiang Z, et al. Anti-GPC3-CAR T cells suppress the growth of tumor cells in patient-derived xenografts of hepatocellular carcinoma. Front Immunol. 2016;7:690.PubMedCrossRef
152.
Zurück zum Zitat Yu M, et al. Development of GPC3-specific chimeric antigen receptor-engineered natural killer cells for the treatment of hepatocellular carcinoma. Mol Ther. 2018;26(2):366–78.PubMedCrossRef Yu M, et al. Development of GPC3-specific chimeric antigen receptor-engineered natural killer cells for the treatment of hepatocellular carcinoma. Mol Ther. 2018;26(2):366–78.PubMedCrossRef
153.
Zurück zum Zitat Jiang W, et al. hIL-15 gene-modified human natural killer cells (NKL-IL15) augments the anti-human hepatocellular carcinoma effect in vivo. Immunobiology. 2014;219(7):547–53.PubMedCrossRef Jiang W, et al. hIL-15 gene-modified human natural killer cells (NKL-IL15) augments the anti-human hepatocellular carcinoma effect in vivo. Immunobiology. 2014;219(7):547–53.PubMedCrossRef
154.
Zurück zum Zitat Greten TF, Manns MP, Korangy F. Immunotherapy of HCC. Rev Recent Clin Trials. 2008;3(1):31–9.PubMedCrossRef Greten TF, Manns MP, Korangy F. Immunotherapy of HCC. Rev Recent Clin Trials. 2008;3(1):31–9.PubMedCrossRef
155.
Zurück zum Zitat Greten TF, et al. A phase II open label trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer. 2010;10:209.PubMedPubMedCentralCrossRef Greten TF, et al. A phase II open label trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer. 2010;10:209.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Sawada Y, et al. Phase I trial of a glypican-3-derived peptide vaccine for advanced hepatocellular carcinoma: immunologic evidence and potential for improving overall survival. Clin Cancer Res. 2012;18(13):3686–96.PubMedCrossRef Sawada Y, et al. Phase I trial of a glypican-3-derived peptide vaccine for advanced hepatocellular carcinoma: immunologic evidence and potential for improving overall survival. Clin Cancer Res. 2012;18(13):3686–96.PubMedCrossRef
157.
Zurück zum Zitat Butterfield LH, et al. T-cell responses to HLA-A*0201 immunodominant peptides derived from alpha-fetoprotein in patients with hepatocellular cancer. Clin Cancer Res. 2003;9(16 Pt 1):5902–8.PubMed Butterfield LH, et al. T-cell responses to HLA-A*0201 immunodominant peptides derived from alpha-fetoprotein in patients with hepatocellular cancer. Clin Cancer Res. 2003;9(16 Pt 1):5902–8.PubMed
158.
Zurück zum Zitat Park BH, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol. 2008;9(6):533–42.PubMedCrossRef Park BH, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol. 2008;9(6):533–42.PubMedCrossRef
159.
Zurück zum Zitat Heo J, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013;19(3):329–36.PubMedPubMedCentralCrossRef Heo J, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013;19(3):329–36.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Makarova-Rusher OV, et al. The yin and yang of evasion and immune activation in HCC. J Hepatol. 2015;62(6):1420–9.PubMedCrossRef Makarova-Rusher OV, et al. The yin and yang of evasion and immune activation in HCC. J Hepatol. 2015;62(6):1420–9.PubMedCrossRef
161.
Zurück zum Zitat Alisa A, et al. Analysis of CD4+ T-cell responses to a novel alpha-fetoprotein-derived epitope in hepatocellular carcinoma patients. Clin Cancer Res. 2005;11(18):6686–94.PubMedCrossRef Alisa A, et al. Analysis of CD4+ T-cell responses to a novel alpha-fetoprotein-derived epitope in hepatocellular carcinoma patients. Clin Cancer Res. 2005;11(18):6686–94.PubMedCrossRef
162.
Zurück zum Zitat Tada Y, et al. Analysis of cytotoxic T lymphocytes from a patient with hepatocellular carcinoma who showed a clinical response to vaccination with a glypican3derived peptide. Int J Oncol. 2013;43(4):1019–26.PubMedPubMedCentralCrossRef Tada Y, et al. Analysis of cytotoxic T lymphocytes from a patient with hepatocellular carcinoma who showed a clinical response to vaccination with a glypican3derived peptide. Int J Oncol. 2013;43(4):1019–26.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci. 2009;100(11):2014–21.PubMedCrossRef Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci. 2009;100(11):2014–21.PubMedCrossRef
164.
Zurück zum Zitat Jager E, et al. Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1-specific immune responses in cancer patients. Proc Natl Acad Sci U S A. 2006;103(39):14453–8.PubMedPubMedCentralCrossRef Jager E, et al. Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1-specific immune responses in cancer patients. Proc Natl Acad Sci U S A. 2006;103(39):14453–8.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Chen JL, et al. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer. 2015;136(6):E590–601.PubMedCrossRef Chen JL, et al. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer. 2015;136(6):E590–601.PubMedCrossRef
166.
Zurück zum Zitat Kim SH, et al. Expression of cancer-testis antigens MAGE-A3/6 and NY-ESO-1 in non-small-cell lung carcinomas and their relationship with immune cell infiltration. Lung. 2009;187(6):401–11.PubMedCrossRef Kim SH, et al. Expression of cancer-testis antigens MAGE-A3/6 and NY-ESO-1 in non-small-cell lung carcinomas and their relationship with immune cell infiltration. Lung. 2009;187(6):401–11.PubMedCrossRef
167.
Zurück zum Zitat Nakamura S, et al. Expression and immunogenicity of NY-ESO-1 in hepatocellular carcinoma. J Gastroenterol Hepatol. 2006;21(8):1281–5.PubMedCrossRef Nakamura S, et al. Expression and immunogenicity of NY-ESO-1 in hepatocellular carcinoma. J Gastroenterol Hepatol. 2006;21(8):1281–5.PubMedCrossRef
168.
Zurück zum Zitat Hong Y, et al. Epitope-optimized alpha-fetoprotein genetic vaccines prevent carcinogen-induced murine autochthonous hepatocellular carcinoma. Hepatology. 2014;59(4):1448–58.PubMedCrossRef Hong Y, et al. Epitope-optimized alpha-fetoprotein genetic vaccines prevent carcinogen-induced murine autochthonous hepatocellular carcinoma. Hepatology. 2014;59(4):1448–58.PubMedCrossRef
169.
Zurück zum Zitat Liu TC, Kirn D. Systemic efficacy with oncolytic virus therapeutics: clinical proof-of-concept and future directions. Cancer Res. 2007;67(2):429–32.PubMedCrossRef Liu TC, Kirn D. Systemic efficacy with oncolytic virus therapeutics: clinical proof-of-concept and future directions. Cancer Res. 2007;67(2):429–32.PubMedCrossRef
171.
Zurück zum Zitat Jiang H, et al. Oncolytic adenovirus and tumor-targeting immune modulatory therapy improve autologous Cancer vaccination. Cancer Res. 2017;77(14):3894–907.PubMedPubMedCentralCrossRef Jiang H, et al. Oncolytic adenovirus and tumor-targeting immune modulatory therapy improve autologous Cancer vaccination. Cancer Res. 2017;77(14):3894–907.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat He LF, et al. Significant antitumor activity of oncolytic adenovirus expressing human interferon-beta for hepatocellular carcinoma. J Gene Med. 2008;10(9):983–92.PubMedCrossRef He LF, et al. Significant antitumor activity of oncolytic adenovirus expressing human interferon-beta for hepatocellular carcinoma. J Gene Med. 2008;10(9):983–92.PubMedCrossRef
173.
Zurück zum Zitat Wang Y, et al. Potent antitumor effect of TRAIL mediated by a novel adeno-associated viral vector targeting to telomerase activity for human hepatocellular carcinoma. J Gene Med. 2008;10(5):518–26.PubMedCrossRef Wang Y, et al. Potent antitumor effect of TRAIL mediated by a novel adeno-associated viral vector targeting to telomerase activity for human hepatocellular carcinoma. J Gene Med. 2008;10(5):518–26.PubMedCrossRef
174.
Zurück zum Zitat Cerullo V, et al. Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res. 2010;70(11):4297–309.PubMedCrossRef Cerullo V, et al. Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res. 2010;70(11):4297–309.PubMedCrossRef
175.
Zurück zum Zitat Heo J, et al. Sequential therapy with JX-594, a targeted oncolytic poxvirus, followed by sorafenib in hepatocellular carcinoma: preclinical and clinical demonstration of combination efficacy. Mol Ther. 2011;19(6):1170–9.PubMedPubMedCentralCrossRef Heo J, et al. Sequential therapy with JX-594, a targeted oncolytic poxvirus, followed by sorafenib in hepatocellular carcinoma: preclinical and clinical demonstration of combination efficacy. Mol Ther. 2011;19(6):1170–9.PubMedPubMedCentralCrossRef
176.
Zurück zum Zitat Hwang TH, et al. A mechanistic proof-of-concept clinical trial with JX-594, a targeted multi-mechanistic oncolytic poxvirus, in patients with metastatic melanoma. Mol Ther. 2011;19(10):1913–22.PubMedPubMedCentralCrossRef Hwang TH, et al. A mechanistic proof-of-concept clinical trial with JX-594, a targeted multi-mechanistic oncolytic poxvirus, in patients with metastatic melanoma. Mol Ther. 2011;19(10):1913–22.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Scharping NE, et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive Intratumoral T cell metabolic insufficiency and dysfunction. Immunity. 2016;45(2):374–88.PubMedPubMedCentralCrossRef Scharping NE, et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive Intratumoral T cell metabolic insufficiency and dysfunction. Immunity. 2016;45(2):374–88.PubMedPubMedCentralCrossRef
180.
Metadaten
Titel
From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma
verfasst von
Yaojie Fu
Shanshan Liu
Shan Zeng
Hong Shen
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2019
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1396-4

Weitere Artikel der Ausgabe 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.