376

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells Peter R. Galle, MD, PhD1

1 First Medical Department, University Medical Center,

Johannes Gutenberg University, Mainz, Germany Semin Liver Dis 2014;34:376–388.

Abstract

Keywords

► hepatocellular carcinoma ► macrophages ► myeloid suppressor cells ► tumor environment ► immune surveillance

Address for correspondence Peter R. Galle, MD, PhD, First Medical Department, University Medical Center, Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany (e-mail: [email protected]).

Immune control of hepatocellular carcinoma (HCC) is executed by effector immune cells, which efficiently eliminate malignant transformed cells. However, progression of HCC clearly documents failure of tumor immune control, which led to the concept of immune subversion by the tumor environment. Particularly tumor-associated stromal cells cooperate within an inflammatory network, which is responsible for immune privilege. The stromal cell composition matures during tumor growth and is derived from surrounding noncancerous tissue or from circulating cells recruited to the tumor site. Therefore, immunosuppressive stromal cells represent heterogeneous cell lineages, including myeloid cells, lymphocytes, endothelial cells, and fibroblasts, which interact by direct cell contact, secretion of soluble factors, or production of extracellular matrix. As the stromal cells determine tumor immune control and clinical outcome of HCC, they represent a promising target for cancer immunotherapy.

Epidemiological data clearly support inflammation driven development of hepatocellular carcinoma (HCC), as chronic viral hepatitis B and C as well as nutritive-toxic hepatitis are the most prominent causes of HCC. 1 In these heterogeneous entities, sequence of liver fibrosis, cirrhosis and HCC is clearly related to the inflammatory activity in the liver, 2–4 which involves cell types from the innate and adaptive immune system. Several experimental tumor models confirm that chronic inflammation initiates early hepatocarcinogenesis and eventually drives tumor progression. 5–11 Throughout this process, HCC is exposed to an ambivalent inflammatory environment, as chronic unresolved inflammation promotes cancer development, whereas profound immune control is able to prevent tumor progression. In this context, protective immune surveillance is generally executed by tumor-directed natural killer (NK) cells and lymphocytes, which efficiently recognize and delete malignant cells. However, tumor growth clearly documents failure of tumor

Issue Theme Hepatocellular Carcinoma: Current Evidence and Future Research; Guest Editor, Jordi Bruix, MD

immune control suggesting inhibition of anticancer immune responses. Stromal cells associated with HCC mirror the complex inflammatory network responsible for the immune-privileged tumor milieu. The stromal cell composition evolves during an inflammatory adaption, which starts with tumor induction and matures during tumor growth. This process requires selective recruitment, differentiation, and activation of immune modulatory cells. Stromal cells hereby interact with the malignant tissue by direct cell contact, secretion of soluble factors, or production of extracellular matrix. This takes into account that stromal components may also be located outside the primary HCC in adjacent noncancerous liver tissue. Following these considerations we will outline the prime mechanisms of anticancer immune control and highlight the HCC-associated stromal cell components affecting tumor immune surveillance. Particularly, as HCC-associated immune-modulatory cells are potential targets of anticancer immune therapy.

Copyright © 2014 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

DOI http://dx.doi.org/ 10.1055/s-0034-1394138. ISSN 0272-8087.

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Martin F. Sprinzl, MD1

Immune Control during HCC Initiation and Progression Innate Immune Control of HCC Innate immune responses are capable to eliminate malignant cells and belong to the first-line defense to control tumor initiation and progression. Particularly, NK cells (CD3-/ CD56þ) reside in the liver and represent approximately 30% to 50% of the hepatic lymphocytes in humans.12–14 Natural killer cells, therefore, represent a relevant effector cell population contributing to tumor surveillance within the liver. Tumor control by NK-dependent elimination of senescent tumor cells was conclusively shown in a murine hepatoma model.15 Natural killer cells also control tumor progression of transplanted tumors of different etiology in mice.16 The balanced activation of NK cells is tightly regulated by activating and inhibiting receptors,17 which facilitate selective tumor cell elimination by granzyme-B, TRAIL and FAS-ligand(FasL-) dependent cytotoxicity.18–21 The NK cell receptor NKG2D and its corresponding ligands in mice (H60 and Rae1) and humans (MICA/B) are well-characterized activators of anticancer NK cell response.16,22 In this context, NKG2D ligands are predominantly expressed by transformed cells, which selectively promote tumor-directed NK cell cytotoxicity.16,23,24 However, regulation of NK cell activation also includes inhibitory NK receptors, which bind to major histocompatibility complex (MHC) class I molecules and belong to the inhibitory killer Ig-like receptor (KIR) family.17 Downregulation of MHC-class I surface expression on malignant cells restrains NK cell inhibition and promotes tumor-directed responses. A part of direct cell-interaction NK cells is incorporated in a cytokine network of IL2, IL12, IL15, and type I interferons, which increase their viability, proliferation, and cytotoxicity.25,26 In return, activated NK cells release granulocyte-macrophage colony-stimulating factor (GMCSF) and interferon-γ (IFN-γ),27,28 which eventually support maturation of antigen-presenting cells and drive Th1 responses to control the tumor. Eventually, NK cells expressing Fc receptor (CD16) also execute anticancer B cell responses as they facilitate antibody-dependent cellular cytotoxicity of opsonized tumor cells.29 The relevant contribution of NK cells to HCC control in humans is indicated by prolonged recurrence-free survival after resection of HCC in presence of increased cytotoxic NK cell activity or tumor NKG2D-ligand (UL16-binding protein 1) expression.30,31 Corresponding histological analysis also correlated proliferating NK cells (CD56þ/Ki67þ) in HCC with tumor cell apoptosis, which linked NK cell activation and target cell killing.32 High TRAIL-receptor (DR4, DR5) expression and low MHC-I expression in HCC further emphasizes the sensitivity of HCC against NK cell responses.33,34 Indirect evidence for NK-cell-driven HCC control is provided by the prognostic value of peritumoral IL2 and IL15 expression, which represent potent NK cell activators.33,35 Natural killer T (NKT) cells, another component of innate tumor defense, represent approximately 5% to 10% of the entire hepatic NK cell population. NKT cells close a link between innate and adaptive immunity as they express NK

Sprinzl, Galle

cell markers beside invariant T cell receptors to detect pathogen-associated molecular patterns.12 Natural killer T cells provide cytotoxic effector functions and important regulatory functions during immune response priming. Their contribution in antitumor immune control has emerged in recent years as NKT cells are capable to eliminate tumors upon specific stimulation with α-galactosylceramide, which is recognized by the T-cell receptor (TCR) repertoire expressed on NKT cells.36,37 In addition, CD1 knockout mice as well as IL12 receptor deficient mice, lacking efficient NKT cell responses, show impaired tumor control.38,39 The activated NKT cells either execute direct lytic activity or prime secondary-tumor-directed immune responses via secretion of IFN-γ.40–42 In patients with chronic hepatitis C, the responsiveness of hepatic NK and NKT cells is reduced during cirrhosis progression, resulting in low-lytic activity and IFN-γ release upon tumor cell contact.43 The progressive NK/NKT dysfunction is associated with an increased HCC risk and suggests hampered innate tumor surveillance in cirrhosis.43 Accumulation of NKT cells in HCC tissue provides further evidence for innate tumor-directed immune responses, particularly if NKT cell infiltrations were combined with elevated IFN-γ expression.44 However, HCC progression along with increasing NKT tumor infiltration indicate impaired anticancer effects in the tumor environment45 some findings even suggest NKT-driven tumor immune escape.46

Adaptive Immune Control of Hepatocellular Carcinoma Adaptive tumor surveillance develops along with innate immune responses, which eventually target tumor-associated antigens (TAA). Therefore, specific elimination of transformed cells by adaptive immune responses protects from tumor manifestation and progression. Adaptive immunity involves CD4þ T lymphocytes, which represent different helper lineages (including Th1, Th2, Th17), defined by specific cytokine secretion profiles. The fundamental role of CD4 T cells in early tumor prevention via senescent cell elimination has been confirmed in a murine HCC model.47 The mechanism of CD4 T cell dependent tumor surveillance is based on Th1-polarized responses, which eventually drive macrophages to deplete senescent cells. The cytokine milieu associated with T-cell polarization guides immune responses responsible for tumor control. Particularly, local Th1-polarized T cell responses in human HCC support favorable prognosis.48 Thus, Th1-cytokines (IL1A, IL1B, IL2, and IFN-γ) are prominently expressed in localized HCC, whereas Th2-cytokines (IL4, IL5, IL8, and IL10) are present in HCC with vascular invasion and metastasis.48 Furthermore, expression of T-cell-promoting cytokines (IL2 and IL15) in peritumoral tissue is associated with a low HCC recurrence rate following resection.35 Despite these facts, TAA-specific CD4 T lymphocytes were not observed in HCC patients under normal circumstances so far, indicating impaired T-cell help in established HCC.49,50 However, detection of tumor-specific antibodies in HCC patients provides indirect evidence for Th2-skewed help, which orchestrates Bcell response and antibody production.51 As circulating antibodies recognize different HCC-associated antigens, they may Seminars in Liver Disease

Vol. 34

No. 4/2014

377

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells be relevant for HCC immune surveillance.51–53 Yet no clear correlation between tumor-reactive antibodies and clinical outcome has been established. Th17-polarized immune cells represent another distinct Tcell lineage, which differs from Th1/Th2-lymphocytes by secretion of IL17.54 Th17 lymphocytes are promoters of autoimmune disease and typically contribute to pathogen defense.55 Th17 cell involvement in tumor control has been confirmed in murine tumor models56–58 and Th17 cells are associated with different human cancer entities.59–61 Albeit Th17 lymphocytes show diminished cytotoxic effector function, they are capable to prime cytotoxic T cells involved in tumor control.56,58,62 In HCC patients, activation of Th17polarized responses following chemoembolization is associated with improved outcome and supports their contribution to tumor surveillance.63 However, conflicting data correlate Th17 lymphocyte infiltrations in HCC with poor prognosis,61 which may reflect the activation of Th17 responses by tumorpromoting cytokines.5,64–66 Cytotoxic CD8 T cells (CTL) are the basis of immunological tumor surveillance, as they efficiently eliminate transformed cells. CTL effector functions are unleashed, following the TCR binding to the corresponding MHC/antigen complex expressed on tumor cells. Eventually CTL killing is mediated by FasL, perforin, and granzyme B expressed or secreted during activation.67 First data showing therapeutic tumor control by CTL in humans was provided by TAA-specific responses, which maintained remission of chronic myeloid leukemia following bone marrow transplantation.68 Evidence for their impact on solid tumor progression has eventually stretched out on HCC, which is regarded as potential target for cytotoxic T-cell-based interventions.69 CTL infiltrations in HCC tissue are a prognostic factor for prolonged diseasefree survival and overall survival after HCC resection.70 Corresponding tumor-directed cytotoxic CD8 T cell responses

Sprinzl, Galle

are present in patients with established HCC (►Table 1). These T cells recognize TAA expressed in the HCC tissue, such as α-fetoprotein, glypicane-3, NY-ESO-1, and MAGE.51,71–75 Their prognostic relevance has been confirmed in patients receiving local ablative therapy, where tumordirected T cells indicate better tumor response.76,77 However, effective T-cell responses are limited by low T-cell frequencies and impaired T-cell function. The lack of function is largely attributed to central tolerance caused by thymic deletion of autoreactive T cells, leading to a low TCR affinity against TAA compared with the naïve T-cell repertoire.78 Expression of the checkpoint molecule programmed cell death protein-1 (PD-1) by HCC-infiltrating lymphocytes indicates additional T-cell exhaustion with functional impairment.75 The mechanism of T-cell exhaustion has been conclusively confirmed in a murine model of SV40 large T-antigen (TAg-) inducible HCC.79 In this model, the initial elimination of nearly all TAgþ hepatocytes was followed by development of TAgþ tumors along with hepatic accumulation of TAg-specific PD-1þ CD8 T cells. Local tolerance was concluded, as TAgþ cancer cells expressing a luciferase reporter were rejected at a subcutaneous injection site, whereas the same cancer cells established tumors in the permissive HCC environment.79

HCC Associated Stromal Cells and Immune Control HCC-Associated Stromal Cells from the Local Environment Hepatocellular carcinoma progression in the presence of innate and adaptive immune effector mechanisms documents inhibition of tumor immune surveillance. Stromal cells associated with HCC have entered research focus in recent years, as they are key inducers of local immune tolerance. The

Table 1 Hepatocellular carcinoma directed cytotoxic T-cell responses T cell target antigen

Source

Method

MAGE-A1

PBMC, HCC

ELISPOT, Tetramer

MAGE-A2

HCC

ELISPOT

MAGE-C2

HCC

ELISPOT

SSX-2

HCC

ELISPOT, ICS

Outcomea

Reference

Local ablation

Increased RFS, PFS

72,75,76,180

NR

NR

75

NR

NR

75

NR

NR

75

Clinical setting

GPC-3

PBMC, HCC

ELISPOT

Local ablation

Increased RFS, PFS

72,76

NY-ESO-1

PBMC, HCC

ELISPOT, ICS, CSA

Local ablation

Increased PFS

51,75,76

hTERT

PBMC, HCC

ELISPOT, ICS; CrR

Local ablation

Increased PFS

71,75,77

AFP

PBMC, HCC

ELISPOT, ICS; CrR

Local ablation / NR

Increased PFS

72,74,77

p53

PBMC

ELISPOT, ICS; CrR

NR

NR

71

SART-3

PBMC

ELISPOT, ICS; CrR

Local ablation

Increased PFS

71,77

MRP-3

PBMC

ELISPOT

Local ablation

Increased PFS

71,77

Abbreviations: AFP, α-fetoprotein; CSA, cytokine secretions assay; CrR, chromium release assay; ELISPOT, enzyme-linked immunospot; GPC-3, glypicane-3; hTERT, human reverse transcriptase telomerase; ICS, intracellular stain; MAGE, melanoma-associated genes; MRP-3, multidrug resistanceassociated protein-3; NR, not rated; RFS, recurrence-free survival; SART-3, squamous cell carcinoma antigen recognized by T cells 3; SSX-2, synovial sarcoma X chromosome breakpoint. a Clinical data related to outcome are provided in references 72,76,77. Seminars in Liver Disease

Vol. 34

No. 4/2014

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

378

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

Liver Sinusoidal Endothelial Cells Liver sinusoidal endothelial cells (LSECs) line the sinusoidal space and are directly exposed to the portal blood flow, which contains nutrient-derived antigens and lipopolysaccharides (LPSs) from intestinal bacteria. Therefore, LSECs shield the liver from incoming antigens and are the prime checkpoint to shape local immune tolerance. Liver sinusoidal endothelial cells efficiently clear LPS from the portal system, which leads to a reduced MHC-dependent antigen presentation on LSEC. LPS exposure further prevents lysosomal antigen processing and downmodulates costimulatory CD80 and CD86 expression in LSECs.80 Induction of programmed death ligand-1 (PD-L1, B7-H1) on LSEC also promotes coinhibitory PD-1 signaling, which induces CD8 T cell tolerance.81 PD-L1 hereby binds to PD-1, which is an immunoglobulin superfamily member related to CD28 and CTLA-4, incapable to transduce activating signals to T cells. LSEC lack IL12 expression and secrete Th2-polarizing factors upon stimulation, which prime Th0 (IL4, IL10, IFN-γ) T cells, but fail induction of Th1 responses.82 Interestingly, the robust Th0-polarized T-cell phenotype is not affected by stimulatory cytokines (IL1, IL12, and IL18) and is only inhibited by neutralizing anti-IL4 antibodies during LSEC priming.80 Activation of LSECs also induces regulatory CD4 T-cell (Treg) populations of CD25low FoxP3- phenotype and induce classical CD25high FoxP3þ Tregs in a TGF-β dependent

379

manner.83,84 The role of LSECs in early HCC progression is supported by histological findings, which confirm endothelial cells (positive for Stabilin-1/2, LYVE-1, and CD32b) in close vicinity to malignant cells and in the peritumoral tissue.85 Throughout tumor progression, the LSECs are displaced by the expanding tumor supporting their hepatic origin. Nevertheless, presence of LSEC markers in the HCC environment is associated with impaired outcome.85,86

Hepatic Tissue Macrophages (Kupffer Cells) Kupffer cells (KCs) are the most prominent hepatic tissue macrophages, which could contribute to local tolerance. KCs are found close to the sinusoids and have access to the portal blood circulation. They share functional features of alternatively polarized macrophages, as they produce TGF-β and IL10 upon LPS stimulation via Toll–like receptor (TLR-) 4.84,87,88 Kupffer cells derived TGF-β induces CD25high FoxP3þ Tregs,84,89 whereas IL10 suppresses NK-cell function90 and Th1 responses.87,91 Corresponding mouse data indicate that KCs are essential to establish Treg-driven hepatic tolerance against antigen challenge in vivo.92 KCs also produce prostaglandin E2 (PGE-2) upon stimulation,93,94 which regulates activation of myeloid-derived suppressor cells (MDSC).95 Recruitment of regulatory cell types link KCs function to cancer immune evasion, as Treg and MDSC are associated with HCC progression.96,97 Inhibitory PD-L1 expressed on KCs directly transduces tolerizing signals to cytotoxic T cells via PD-1. This mechanism eventually drives T-cell inhibition in context of the tumor milieu, as IL10 secreted by hepatoma cells increases PD-L1 expression on KCs.98 Tolerogenic capacity of KC has also been attributed to their surface expression of galectin-9, which is upregulated by IFN-γ during inflammation in HBV-associated HCC.99 Galectin-9 induces cell death in TIM-3 expressing lymphocytes and tempers Th1 responses.100 Coherent data show that galectin-9 expressed by KCs induces senescence of TIM-3þ T cells in HCC, which accumulate in HCC and determine outcome.99 The finding is

Fig. 1 Stromal cell architecture in hepatocellular carcinoma (HCC). (A) Early HCC lesions develop in the immune-privileged niche of hepatic trabeculae. Liver sinusoidal endothelial cells (LSECs) separate hepatocytes and tumor cells from the sinusoidal lumen, which drains the hepatic blood from the portal system to the central vein. LSECs and Kupffer cells sense circulating immune stimulators (e.g., nutrient antigens, LPS), secret anti-inflammatory cytokines (IL10, TGF-β) and prevent efficient T-cell priming. Chronic inflammation activates hepatic stellate cells, which shape the inhibitory environment by production of cytokines and extracellular matrix. (B) Established HCC replaces the hepatic micro-architecture by pauci-immune tumor tissue. Hepatic tissue collapse during HCC expansion and myo-/fibroblast activation results in a peritumoral accumulation of extracellular matrix. HCC typically accumulates stromal cells at the invasion front or at the margin of necrotic areas. The stromal cells are either derived from the neighboring noncancerous tissue or from the circulating pool of immune cells.

Seminars in Liver Disease

Vol. 34

No. 4/2014

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

stromal cells associated with HCC are initially derived from the hepatic tissue, which provides an immune-privileged niche for early tumor lesions and shapes the surrounding noncancerous tumor environment (►Fig. 1). Hepatic tolerance, which copes with continuous immune stimulation by immunogenic components absorbed in the gastrointestinal system, provides an ideal foundation for tumor progression. Liver cirrhosis, which forms the background of most HCC, further accentuates the immune-modulatory functions of the hepatic environment.

Sprinzl, Galle

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells in line with the controversial hepatic “graveyard theory,” which suggests tolerance induction by deletion of activated T cells in the liver.101 However, the effect of KCs on tumor progression remains unresolved, particularly as depletion of KCs in a model of hepatic metastasis promotes cancer growth.102 These contrasting results may reflect the functional plasticity of KC depending on the underlying inflammatory stimulus. Thus, toll-like receptor (TLR-) 3 engagement by Poly I:C on KCs induces secretion of IL12 and IL18, which may promote effector cell responses with antitumor activity.90

Cirrhotic Liver Environment Hepatocellular carcinoma develops during chronic liver damage accompanied with destructive liver fibrosis. The typical noncancerous tissue harboring the HCC therefore shows a cirrhotic liver architecture in approximately 80% of all cases.103 Cirrhotic liver anatomy increases portal pressure, which promotes translocation of intestinal bacteria into the portal circulation. Consecutive increase of LPS activates LSECs and KCs, which drive hepatic immune privilege. LPS, reactive oxygen species (ROS) from activated KCs, or stromal-derived factor 1α (SDF-1α) secreted by LSECs further induce hepatic stellate cells (HSCs), which enhance tumor progression, produce collagen and are the main storage of retinoic acid.104–107 Again, activated HSCs induce regulatory T cells via secreted Th2-type cytokines or retinoic acid,108,109 which has a major impact on immune control of HCCs.110 HSCs have also been identified as inducers of MDSC, which may inhibit alloreactive T-cell responses111,112 and hamper tumor response during systemic HCC therapy.113 Additional crosstalk between activated HSCs and T cells engages PD-1/PD-L1 interaction leading to T-cell exhaustion.114,115 Evidence also supports that HCSs transdifferentiate to myofibroblasts or fibroblasts and shape the HCC environment. This particularly accounts for the characteristic peritumoral fibrotic capsule, which evolves from myofibroblasts expressing smooth muscle actin-α (SMA-α).116 Initial data attributed accelerated hepatoma cell growth and HCC progression to the growth factor release by activated HSCs or fibroblasts.117,118 However, similar SMA-α positive fibroblasts promote tumor invasion of desmoplastic carcinomas via SDF-1/CXCL12 secretion119 and recruit macrophages to the tumor site.120 Therapeutic fibroblast elimination even reduces macrophage (F4/80) tumor infiltrations in a murine mamma carcinoma model and restored intratumoral cytotoxic (granzyme-Bþ) CD8 T cell frequencies.121 These data indicate either direct T-cell inhibitory effects of fibroblasts or indirect immune modulation by recruited macrophages. Direct NK-cell inhibition was recently confirmed in coculture experiments with HCC-associated fibroblasts. Specific inhibitors of fibroblast signaling restored NK-cell cytotoxicity after target cell contact and identified PGE-2 and indoleamine 2,3dioxygenase (IDO) as inhibitory factors secreted by fibroblasts.122 However, the complete impact of the cirrhotic HCC environment on immune surveillance has just started to reveal. Seminars in Liver Disease

Vol. 34

No. 4/2014

Sprinzl, Galle

This includes the role of platelet stimulation during portal hypertension. This hypothesis is supported by elevated sCD40L serum concentrations in cirrhotic patients, as sCD40L is released from activated platelets into the circulation of cirrhotic patients.123 Analysis of TAg-induced HCC shows early thrombocyte aggregation and sequential macrophage recruitment within the dysplastic stroma, supporting the role of thrombocytes in HCC formation.124 Along with platelet activation tumor cells may be shielded against cytotoxic NK cells, which might affect innate tumor control.125 Activated thrombocytes also release vascular endothelial growth factor (VEGF), which could induce regulatory T cells.126 However, more research is required to unravel the interaction between HCC stroma, coagulation, and immune control in liver cirrhosis.

HCC-Associated Stromal Cells from the Circulation Throughout HCC progression, selective recruitment of circulating immune modulatory cells determines the stromal cell composition (►Fig. 1). The privileged milieu is eventually maintained by tumor-derived factors, which differentiate and activate immunosuppressive stromal cells. Hence, tumorassociated stroma cells do not delineate entirely from the local tissue context and are regularly recruited to the tumor site from the blood circulation (►Fig. 2). Circulating stromal cell precursors are typically derived from myeloid or lymphocytic cell lineages. The coordinated expression of chemokines on endothelial cells and malignant cells finally navigates the immune cells to the tumor stroma.127 Local chemokines determine the composition of effector or regulatory immune cells, which express cell specific chemokine receptors families (CCR, CXCR, CX3CR, and XCR).128 Lymphocyte recruitment to HCC tissue is mediated by CXCR3 or CCR5 and their corresponding chemokine ligands. This is confirmed by monoclonal antiCXCR3 and anti-CCR5 antibodies inhibiting tumor lymphocyte infiltration in tumor bearing mice.127 The increased CXCR3 expression in HCC-infiltrating lymphocytes compared with their circulating counterparts supports CXCR3selective lymphocyte recruitment to the tumor.129 Corresponding data correlate CXCL10, CCL5, and CCL2 expression in resected HCC with protective Th1-skewed lymphocyte infiltration associated with improved outcome.130 However, downregulation of CXCR3 in HCC patients compared with healthy controls could also indicate disrupted T-cell recruitment into cancer.131 In turn, CCL22-dependent mechanisms recruit regulatory CD4 T cells (Treg) to the tumor environment, which express CCR4 and represent the immunosuppressive counterpart of effector lymphocytes.132 CCL22dependent Treg-recruitment signaling has been confirmed in HCC and is driven by chronic inflammation and TGF-β release during chronic hepatitis B.133 The CXCL12-CXCR4 axis is also relevant for stromal cell composition as it recruits myeloid-derived cell subsets to the tumor environment.113 CXCR4 has regularly been confirmed in HCC tissue and its high expression may result from impaired TGF-β inhibition or from the rigid HCC matrix.134–136 Endothelial cells and HSCs associated with HCC

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

380

Sprinzl, Galle

Fig. 2 Crosstalk between effector immune responses and stromal cells in the tumor. (1) Chemokines originating from the tumor environment recruit stromal cells and effector immune cells. (2) Effector immune cells specifically recognize and eliminate malignant tissue. (3) Factors derived from the tumor polarize anti-inflammatory (M2) tumor-associated macrophages, which stimulate inhibitory regulatory T cells (Treg), myeloidderived suppressor cells (MDSC), and myofibroblasts. (4) M2 macrophages directly inhibit cytotoxic T cells and induce Th2-skewed responses, which in turn stimulate M2-macrophages via IL4. (5,6) Stimulated Tregs and MDSC eventually inhibit tumor specific T cell and NK/NKT cell responses, (7) whereas stimulated fibroblasts shape the immune milieu by production of extracellular matrix and soluble cytokines.

are potential sources of CXCL12 (SDF-1α) to drive CXCR4dependent myeloid cell attraction. Conditional myeloid (MxCre) CXCR4-ko mice show reduced myeloid (Gr1þ) cell recruitment into lung cancer, which results in impaired tumor growth in combination with disrupted VEGF signaling.137 Inhibition of CXCR4 signaling with AMD3100 reduces myeloid (Gr1þ) cell frequencies in HCC as well.113 Chemokines may also drive recruitment of macrophages to HCC, given that myeloid precursors may differentiate into macrophages.138 However, the recruitment of macrophages to the HCC environment remains controversial, as resident KCs are mainly involved in HCC induction.10 Presumably local (KCs) and distant macrophage populations contribute to the accumulation of tumor-associated macrophages (TAMs) in HCC.

Dendritic Cells Activated macrophages recruit dendritic cells (DCs) from the sinusoidal circulation, which home in the liver by CC chemokine receptor (CCR)-5/7 dependent trafficking.139,140 This observation has relevant impact on antitumor immunity as DCs could efficiently prime antigen-specific immune responses. Immune-stimulatory DCs typically express MHC class I/II along with costimulatory molecules (CD40, CD80, CD83, CD86) and release Th1-cytokines (IL2, IL12, IL23, and type I interferons).101,141 However, hepatic DCs include lineage subsets that only cause partial T-cell activation and may lead to local tolerance. Hepatic DCs hereby represent a mixture of activating and dysfunctional DC subtypes, which show very distinct activation competency if analyzed separately.142,143 Minimal T-cell activation by a mixture of hepatic DCs indicates that the relative abundance of dysfunctional DC subtypes compared with activating DCs determines local tolerance.144,145 IL10 is capable to prevent DC maturation and increases with progressive tumor burden in the serum of

HCC patients. This may explain how immature DCs accumulate in the context of HCC.146 High frequencies of circulating immature DCs in HCC patients147 and lack of HCC-infiltrating activated DCs with CD83 expression148 support the concept of inhibitory DC composition in context of cancer.

Tumor-Associated Macrophages Tumor-associated macrophages are the most-versatile HCCassociated stromal cell population to have a major impact on anticancer immune responses. Their functional plasticity ranges from proinflammatory (classical) M1 macrophages to anti-inflammatory (alternative) M2 macrophages.149 M1 macrophages prime immune responses via IL6, IL12, and TNF-α secretion and present antigens via MHC molecules. In turn, M2 macrophages lack sufficient MHC expression and release anti-inflammatory factors, such as IL10, IL23, PGE-2, and TGF-β. During inflammatory resolution M2 macrophages provide growth factors (EGF, IGF, and VEGF) and matrix metalloproteinases (MMP), which promote tissue regeneration or remodeling. Along with functional macrophage polarization their phenotypic marker composition changes (M1: CD68, HLA-DRhigh vs. M2: CD163, CD203, and HLA-DRlow).149 Tumor-associated macrophages are generally regarded as negative prognostic marker for HCC due to their immunemodulatory and growth promoting properties. Hence, CD68þ macrophage densities in resected HCC are inversely correlated with overall survival.150 However, high CD68þ macrophage densities in conjunction with tumor-infiltrating memory CD45ROþ lymphocytes indicated a better overall survival after HCC resection.151 In addition, macrophages are capable to eliminate opsonized tumor cells and execute senescent cell deletion.47,152,153 These conflicting data highlight the importance of alternative TAM polarization, which Seminars in Liver Disease

Vol. 34

No. 4/2014

381

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells determines HCC prognosis. Assessment of CD163þ TAMs densities eventually confirmed the predominant M2 polarization in resected HCCs, which was associated with reduced overall survival.154 Increased shedding of soluble CD163 (sCD163) into the bloodstream of HCC patients during tumor progression supports the polarization theory.155 The M2 polarization of TAMs is maintained by colony stimulating factor-1 (CSF-1) and IL4, which are both highly expressed in HCC tissue.48,156 CSF-1 functions as central macrophage maturation factor, whereas IL4 augments M2 polarization.157 Recently, lactate has also been identified as a fundamental M2 polarization stimulus in hypoxic tumor tissue,158 which likely affects TAM polarization in HCC showing critical perfusion. Along with polarization, TAMs are activated by LPS and apoptotic bodies present in the HCC environment, which induces secretion of immune-modulatory factors.159 Tumor-associated macrophages located in murine HCCs, for example, inhibit tumor-directed T cells by secretion of IL10, which induces PD-L1 expression on HCC-associated monocytes.160 Alternative macrophages further produce anti-inflammatory TGF-β and PGE-2 upon stimulation.161 Macrophages are capable to produce arginase and IDO to suppress T-cell functions162,163 and release VEGF in context of inflammatory stimuli, which activates regulatory T cells via VEGF-receptor 2.126 In a murine ovarial cancer model, TAMs recruit regulatory T cells to the tumor via CCL22,132 a chemokine axis also activated in hepatitis B associated HCC.133 MMPs expressed by TAMs show additional immune modulatory activity. Elevated sCD163 serum concentrations in HCC patients indicates involvement of the proteinase ADAM17, which is responsible for sCD163 shedding during macrophage activation.164 ADAM17 activation potentially affects HCC and inhibits NK-cell induction by cleavage of the activating NKp30 ligand B7-H6. 165 In addition, HCC-associated TAMs express MMP9, 166 an enzyme able to shed NK-cell-inhibiting MICA fragments from tumor cells.167

Myeloid-Derived Suppressor Cells Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous cell population with monocytic or granulocytic morphology, which share immune-suppressive functions. MDSCs represent a stromal component of different tumor entities and suppress T-cell function by arginase-1, nitric oxide, and ROS production.138 Human MDSCs typically express myeloid-lineage markers (CD33, CD11b) and lack relevant MHC class II HLA-DR expression. In mice, MDSCs are characterized by myeloid GR1 and CD11b expression. The intermediate maturation state of MDSCs leads to phenotypic plasticity and allows maturation into DCs and macrophages in monocytic subsets.138 MDSCs with a CD14þ/HLA-DR/low phenotype have been isolated from resected HCC tissue.168 These monocytic MDSCs inhibit T-cell responses directly by production of arginase-1 or via regulatory T-cell activation. Interestingly, CD14þ/HLA-DR/low MDSC did not secrete immune-modulatory TGF-β or IL10 ex vivo and were resistant to DC maturation stimuli, indicating a stable phenotype. CorreSeminars in Liver Disease

Vol. 34

No. 4/2014

Sprinzl, Galle

lation of circulating MDSC frequencies with reduced recurrence-free survival after local HCC ablation supports the prognostic impact of this MDSC subtype.97 Another report describes a correlation between HCC stage an circulating MDSCs, which were defined by a Lin/CD33þ/HLA-DR/low phenotype.169 Reconstitution of T-cell function after combined Treg and MDSC depletion in PBMC obtained from HCC patients suggests a related immune modulation.170 However, nonselective depletion in this report did not allow assessment of specific MDSC function.

Regulatory T Cells Regulatory CD4 T lymphocytes (Treg) are the prototype of immune-suppressive cells, which inhibit antigen-specific T-cell responses. Tregs express activation markers (CD25, CD62L, and CD127), which are commonly found in chronically stimulated lymphocyte subsets without immunosuppressive function. The more-specific surface marker GARP and the transcription factor FoxP3 eventually define immunosuppressive Treg subtypes.171,172 Tregs activation involves specific antigen recognition by TCRs and cooperative IL10 and TGF-β signaling. Activated FoxP3þ Treg block effector T lymphocytes by direct cell-cell contact via inhibitory molecules, such as CTLA-4, Lag-3, and membranebound TGF-β. 173 Release of soluble IL10 and TGF-β contribute to inhibitory Treg function, but are not essential for effector T-cell inhibition in vivo. Interestingly, constitutive expression of CD25 (IL2 receptor) by Tregs deprives IL2 from the circulation, which impairs effector T-cell proliferation.173 These effects eventually suppress T-cell maturation, priming, and effector functions required for tumor control. First reports described CD4þ/CD25þ Treg accumulation in HCC, which inversely correlated with HCC infiltrating CD8þ T lymphocyte functions.174,175 Other studies identified elevated Treg frequencies in the blood circulation of HCC patients.96,170 As expected, Treg isolated from HCC patients broadly inhibited T-cell activation upon stimulation with anti-CD3/CD28 antibodies.96,174,175 Specific inhibition of tumor-directed T cells was confirmed by Treg depletion, which reconstituted T-cell responses against HCC-related AFP.50 The potent immunosuppressive Treg function translates into decreased overall survival of HCC patients with elevated circulating Treg frequencies96 and accounts for reduced overall survival after HCC resection.70 Several studies confirmed the impact of Tregs on HCC prognosis, which has been summarized by a recent meta-analysis.110

Perspective The summarized mechanisms of immune control and the corresponding stromal cell components clearly determine the clinical outcome of HCC (►Table 2). Therefore, reconstitution of local immune surveillance may provide a potent approach for HCC therapy.69 Following this concept, reconstitution of anticancer immune responses via vaccination has been pursued for a long time. However, vaccination approaches were hampered by impaired immune cell priming of tumor-directed effector T cells. Alternative introduction of tumor-directed

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

382

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

Sprinzl, Galle

383

Table 2 Impact of immune-regulatory stromal cells on hepatocellular carcinoma prognosis Cell lineage

Source

Clinical setting

Outcome

Reference

HCC

Resection

Increased OS

181,182

PBMC

NR

HCC Stage

146

Dendritic cells S100þ -

Lin CD11c

high

HLA-DR

þ

CD4þ CD25þ

PBMC

NR

Reduced OS

96

FoxP3

þ

HCC

Resection

Reduced DFS, OS

70,183,184

FoxP3

þ

HCC

Resection

Reduced OS

185

FoxP3þ

HCC

Resection

Reduced DFS

186

CD4þCD25þFoxP3þ

PBMC

Resection

Reduced DFS, OS

184

Macrophages, monocytes, and MDSCs CD68þ

HCC

Resection

Increased DFS, OS

151

CD68þ

HCC

Resection

Reduced DFS, OS

156,187

HCC

Resection

Reduced RFS, OS

154

þ

CD68 CD163 þ

CD68 PD-L1

þ

þ

HCC

Resection

Reduced DFS, OS

98,160

CD14þ TIE2þ

PBMC

Resection / local ablation

Reduced RFS

188

CD14þ HLA-DR-/low

PBMC

Local ablation

Reduced RFS

97

Abbreviations: DFS, disease-free survival; MDSCs, myeloid-derived suppressor cells; NR, not rated; OS, overall survival, PBMC, periphery blood mononuclear cells; RFS, recurrence-free survival.

T cells, which were specifically engineered to recognize and eliminate malignant cells also face some unresolved obstacles. This includes the basic choice of the optimal TAA target to avoid side effects. The narrow target spectrum of selected antigen-specific T cells bears the risk of tumor resistance by TAA downregulation or epitope evasion. Furthermore, the limited tumor infiltration of therapeutic T cells and the restricted T-cell expansion requires additional optimization. Particularly, the functional stability of therapeutic T cells is challenging, as activated T cells may undergo apoptosis or face functional inhibition/exhaustion in the tumor environment as outlined above. Modulation of inhibitory stromal cells provides an alternative approach for anticancer immunotherapy, which could support specific approaches by a broader mode of action. Reconstitution of suppressed tumor-directed immune responses is promising as T cells specific for several HCCassociated antigens have been confirmed.69 This could be achieved by immune-stimulatory cytokines or TLR-ligands, shifting the Th1/Th2 balance toward a local Th1 response. Early experiments of complete TAM depletion have been successfully introduced to improve treatment response in murine HCC models.176 More specific targeting of TAM polarization via nuclear-factor kappa B inhibition or CSF-1receptor blockage, were successfully tested in murine cancer models.177,178 Particularly, therapeutic CSF-1-receptor inhibition seems promising for the treatment of CSF-1 expressing HCC.48,156 Monoclonal antibodies targeting lymphocytic checkpoint molecules are currently the most promising approach for tumor immune therapy. Inhibitory PD-1/PD-L1 antibodies,

for example, activate antitumor T-cell responses in HCCbearing mice79 and reconstitute function of HCC-specific T cells isolated from HCC patients.75 CTLA-4 blocking antibodies showed similar immune-stimulatory activity71 and were tested in HCC patients with a good safety profile.179 Despite initial insights into the biology of HCC-associated stromal cells the immune-modulatory armamentarium for an effective tumor treatment is still limited. Hence, further research about the environmental tumor network is required to develop innovative HCC-directed immunotherapies.

References 1 El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiolo-

2

3

4

5

6

gy and molecular carcinogenesis. Gastroenterology 2007;132(7): 2557–2576 Villeneuve JP, Desrochers M, Infante-Rivard C, et al. A long-term follow-up study of asymptomatic hepatitis B surface antigenpositive carriers in Montreal. Gastroenterology 1994;106(4): 1000–1005 Liaw YF, Tai DI, Chu CM, et al. Early detection of hepatocellular carcinoma in patients with chronic type B hepatitis. A prospective study. Gastroenterology 1986;90(2):263–267 Ikeda K, Saitoh S, Suzuki Y, et al. Disease progression and hepatocellular carcinogenesis in patients with chronic viral hepatitis: a prospective observation of 2215 patients. J Hepatol 1998; 28(6):930–938 Naugler WE, Sakurai T, Kim S, et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 2007;317(5834):121–124 Pikarsky E, Porat RM, Stein I, et al. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004;431(7007):461–466 Seminars in Liver Disease

Vol. 34

No. 4/2014

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Regulatory T cells

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

Sprinzl, Galle

7 Haybaeck J, Zeller N, Wolf MJ, et al. A lymphotoxin-driven

27 Subleski JJ, Hall VL, Back TC, Ortaldo JR, Wiltrout RH. Enhanced

pathway to hepatocellular carcinoma. Cancer Cell 2009;16(4): 295–308 Jiang R, Tan Z, Deng L, et al. Interleukin-22 promotes human hepatocellular carcinoma by activation of STAT3. Hepatology 2011;54(3):900–909 Greten FR, Karin M. The IKK/NF-kappaB activation pathway-a target for prevention and treatment of cancer. Cancer Lett 2004; 206(2):193–199 Maeda S, Kamata H, Luo JL, Leffert H, Karin M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 2005;121(7): 977–990 Luedde T, Beraza N, Kotsikoris V, et al. Deletion of NEMO/ IKKgamma in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer Cell 2007;11(2):119–132 Gao B, Jeong WI, Tian Z. Liver: An organ with predominant innate immunity. Hepatology 2008;47(2):729–736 Doherty DG, Norris S, Madrigal-Estebas L, et al. The human liver contains multiple populations of NK cells, T cells, and CD3þCD56þ natural T cells with distinct cytotoxic activities and Th1, Th2, and Th0 cytokine secretion patterns. J Immunol 1999;163(4):2314–2321 Kita H, Naidenko OV, Kronenberg M, et al. Quantitation and phenotypic analysis of natural killer T cells in primary biliary cirrhosis using a human CD1d tetramer. Gastroenterology 2002; 123(4):1031–1043 Iannello A, Thompson TW, Ardolino M, Lowe SW, Raulet DH. p53dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells. J Exp Med 2013;210(10):2057–2069 Diefenbach A, Jensen ER, Jamieson AM, Raulet DH. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature 2001;413(6852):165–171 Thielens A, Vivier E, Romagné F. NK cell MHC class I specific receptors (KIR): from biology to clinical intervention. Curr Opin Immunol 2012;24(2):239–245 Kägi D, Ledermann B, Bürki K, et al. Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforindeficient mice. Nature 1994;369(6475):31–37 van den Broek ME, Kägi D, Ossendorp F, et al. Decreased tumor surveillance in perforin-deficient mice. J Exp Med 1996;184(5): 1781–1790 Takeda K, Smyth MJ, Cretney E, et al. Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development. J Exp Med 2002; 195(2):161–169 Screpanti V, Wallin RP, Ljunggren HG, Grandien A. A central role for death receptor-mediated apoptosis in the rejection of tumors by NK cells. J Immunol 2001;167(4):2068–2073 Bauer S, Groh V, Wu J, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999; 285(5428):727–729 Guerra N, Tan YX, Joncker NT, et al. NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 2008;28(4):571–580 Cerwenka A, Baron JL, Lanier LL. Ectopic expression of retinoic acid early inducible-1 gene (RAE-1) permits natural killer cellmediated rejection of a MHC class I-bearing tumor in vivo. Proc Natl Acad Sci U S A 2001;98(20):11521–11526 Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP. Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol 1999;17:189–220 Nguyen KB, Salazar-Mather TP, Dalod MY, et al. Coordinated and distinct roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J Immunol 2002;169(8): 4279–4287

antitumor response by divergent modulation of natural killer and natural killer T cells in the liver. Cancer Res 2006;66(22): 11005–11012 Cuturi MC, Anegón I, Sherman F, et al. Production of hematopoietic colony-stimulating factors by human natural killer cells. J Exp Med 1989;169(2):569–583 Tong AW, Lee JC, Wang RM, Ordonez G, Stone MJ. Augmentation of lymphokine-activated killer cell cytotoxicity by monoclonal antibodies against human small cell lung carcinoma. Cancer Res 1989;49(15):4103–4108 Kamimura H, Yamagiwa S, Tsuchiya A, et al. Reduced NKG2D ligand expression in hepatocellular carcinoma correlates with early recurrence. J Hepatol 2012;56(2):381–388 Taketomi A, Shimada M, Shirabe K, Kajiyama K, Gion T, Sugimachi K. Natural killer cell activity in patients with hepatocellular carcinoma: a new prognostic indicator after hepatectomy. Cancer 1998;83(1):58–63 Chew V, Tow C, Teo M, et al. Inflammatory tumour microenvironment is associated with superior survival in hepatocellular carcinoma patients. J Hepatol 2010;52(3):370–379 Ishiyama K, Ohdan H, Ohira M, Mitsuta H, Arihiro K, Asahara T. Difference in cytotoxicity against hepatocellular carcinoma between liver and periphery natural killer cells in humans. Hepatology 2006;43(2):362–372 Kurokohchi K, Carrington M, Mann DL, et al. Expression of HLA class I molecules and the transporter associated with antigen processing in hepatocellular carcinoma. Hepatology 1996;23(5): 1181–1188 Zhou H, Huang H, Shi J, et al. Prognostic value of interleukin 2 and interleukin 15 in peritumoral hepatic tissues for patients with hepatitis B-related hepatocellular carcinoma after curative resection. Gut 2010;59(12):1699–1708 Kawano T, Cui J, Koezuka Y, et al. CD1d-restricted and TCRmediated activation of valpha14 NKT cells by glycosylceramides. Science 1997;278(5343):1626–1629 Kobayashi E, Motoki K, Uchida T, Fukushima H, Koezuka Y. KRN7000, a novel immunomodulator, and its antitumor activities. Oncol Res 1995;7(10-11):529–534 Smyth MJ, Thia KY, Street SE, et al. Differential tumor surveillance by natural killer (NK) and NKT cells. J Exp Med 2000;191(4): 661–668 Swann JB, Uldrich AP, van Dommelen S, et al. Type I natural killer T cells suppress tumors caused by p53 loss in mice. Blood 2009; 113(25):6382–6385 Arase H, Arase N, Nakagawa K, Good RA, Onoé K. NK1.1þ CD4þ CD8- thymocytes with specific lymphokine secretion. Eur J Immunol 1993;23(1):307–310 Dao T, Mehal WZ, Crispe IN. IL-18 augments perforin-dependent cytotoxicity of liver NK-T cells. J Immunol 1998;161(5): 2217–2222 Arase H, Arase N, Kobayashi Y, Nishimura Y, Yonehara S, Onoé K. Cytotoxicity of fresh NK1.1þ T cell receptor alpha/betaþ thymocytes against a CD4þ8þ thymocyte population associated with intact Fas antigen expression on the target. J Exp Med 1994; 180(2):423–432 Kawarabayashi N, Seki S, Hatsuse K, et al. Decrease of CD56(þ)T cells and natural killer cells in cirrhotic livers with hepatitis C may be involved in their susceptibility to hepatocellular carcinoma. Hepatology 2000;32(5):962–969 Xiao YS, Gao Q, Xu XN, et al. Combination of intratumoral invariant natural killer T cells and interferon-gamma is associated with prognosis of hepatocellular carcinoma after curative resection. PLoS ONE 2013;8(8):e70345 Bricard G, Cesson V, Devevre E, et al. Enrichment of human CD4þ V(alpha)24/Vbeta11 invariant NKT cells in intrahepatic malignant tumors. J Immunol 2009;182(8):5140–5151

8

9

10

11

12 13

14

15

16

17

18

19

20

21

22

23

24

25

26

Seminars in Liver Disease

Vol. 34

No. 4/2014

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

384

Sprinzl, Galle

46 Terabe M, Matsui S, Noben-Trauth N, et al. NKT cell-mediated

66 Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a

repression of tumor immunosurveillance by IL-13 and the IL-4RSTAT6 pathway. Nat Immunol 2000;1(6):515–520 Kang TW, Yevsa T, Woller N, et al. Senescence surveillance of premalignant hepatocytes limits liver cancer development. Nature 2011;479(7374):547–551 Budhu A, Forgues M, Ye QH, et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell 2006;10(2):99–111 Witkowski M, Spangenberg HC, Neumann-Haefelin C, et al. Lack of ex vivo peripheral and intrahepatic α-fetoprotein-specific CD4þ responses in hepatocellular carcinoma. Int J Cancer 2011; 129(9):2171–2182 Greten TF, Ormandy LA, Fikuart A, et al. Low-dose cyclophosphamide treatment impairs regulatory T cells and unmasks AFPspecific CD4þ T-cell responses in patients with advanced HCC. J Immunother 2010;33(2):211–218 Korangy F, Ormandy LA, Bleck JS, et al. Spontaneous tumorspecific humoral and cellular immune responses to NY-ESO-1 in hepatocellular carcinoma. Clin Cancer Res 2004;10(13): 4332–4341 Sahin U, Türeci O, Schmitt H, et al. Human neoplasms elicit multiple specific immune responses in the autologous host. Proc Natl Acad Sci U S A 1995;92(25):11810–11813 Stockert E, Jäger E, Chen YT, et al. A survey of the humoral immune response of cancer patients to a panel of human tumor antigens. J Exp Med 1998;187(8):1349–1354 Aarvak T, Chabaud M, Miossec P, Natvig JB. IL-17 is produced by some proinflammatory Th1/Th0 cells but not by Th2 cells. J Immunol 1999;162(3):1246–1251 Bettelli E, Korn T, Oukka M, Kuchroo VK. Induction and effector functions of T(H)17 cells. Nature 2008;453(7198):1051–1057 Benchetrit F, Ciree A, Vives V, et al. Interleukin-17 inhibits tumor cell growth by means of a T-cell-dependent mechanism. Blood 2002;99(6):2114–2121 Muranski P, Boni A, Antony PA, et al. Tumor-specific Th17polarized cells eradicate large established melanoma. Blood 2008;112(2):362–373 Martin-Orozco N, Muranski P, Chung Y, et al. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity 2009;31(5):787–798 Kryczek I, Banerjee M, Cheng P, et al. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood 2009;114(6): 1141–1149 Miyahara Y, Odunsi K, Chen W, Peng G, Matsuzaki J, Wang RF. Generation and regulation of human CD4þ IL-17-producing T cells in ovarian cancer. Proc Natl Acad Sci U S A 2008;105(40): 15505–15510 Zhang JP, Yan J, Xu J, et al. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. J Hepatol 2009;50(5):980–989 Hinrichs CS, Kaiser A, Paulos CM, et al. Type 17 CD8þ T cells display enhanced antitumor immunity. Blood 2009;114(3): 596–599 Liao Y, Wang B, Huang ZL, et al. Increased circulating Th17 cells after transarterial chemoembolization correlate with improved survival in stage III hepatocellular carcinoma: a prospective study. PLoS ONE 2013;8(4):e60444 Kortylewski M, Xin H, Kujawski M, et al. Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. Cancer Cell 2009;15(2):114–123 Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol 2007;8(9):942–949

pathogenic T cell population that induces autoimmune inflammation. J Exp Med 2005;201(2):233–240 Russell JH, Ley TJ. Lymphocyte-mediated cytotoxicity. Annu Rev Immunol 2002;20:323–370 Molldrem JJ, Lee PP, Wang C, et al. Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nat Med 2000;6(9):1018–1023 Schmidt N, Neumann-Haefelin C, Thimme R. Cellular immune responses to hepatocellular carcinoma: lessons for immunotherapy. Dig Dis 2012;30(5):483–491 Gao Q, Qiu SJ, Fan J, 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): 2586–2593 Mizukoshi E, Nakamoto Y, Arai K, et al. Comparative analysis of various tumor-associated antigen-specific t-cell responses in patients with hepatocellular carcinoma. Hepatology 2011; 53(4):1206–1216 Flecken T, Schmidt N, Hild S, et al. Immunodominance and functional alterations of tumor-associated antigen-specific CD8þ T-cell responses in hepatocellular carcinoma. Hepatology 2014;59(4):1415–1426 Shang XY, Chen HS, Zhang HG, et al. The spontaneous CD8þ T-cell response to HLA-A2-restricted NY-ESO-1b peptide in hepatocellular carcinoma patients. Clin Cancer Res 2004;10(20): 6946–6955 Thimme R, Neagu M, Boettler T, et al. Comprehensive analysis of the alpha-fetoprotein-specific CD8þ T cell responses in patients with hepatocellular carcinoma. Hepatology 2008;48(6): 1821–1833 Gehring AJ, Ho ZZ, Tan AT, et al. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology 2009;137(2):682–690 Hiroishi K, Eguchi J, Baba T, et al. Strong CD8(þ) T-cell responses against tumor-associated antigens prolong the recurrence-free interval after tumor treatment in patients with hepatocellular carcinoma. J Gastroenterol 2010;45(4):451–458 Mizukoshi E, Yamashita T, Arai K, et al. Enhancement of tumorassociated antigen-specific T cell responses by radiofrequency ablation of hepatocellular carcinoma. Hepatology 2013;57(4): 1448–1457 Kumari S, Wälchli S, Fallang LE, et al. Alloreactive cytotoxic T cells provide means to decipher the immunopeptidome and reveal a plethora of tumor-associated self-epitopes. Proc Natl Acad Sci U S A 2014;111(1):403–408 Willimsky G, Schmidt K, Loddenkemper C, Gellermann J, Blankenstein T. Virus-induced hepatocellular carcinomas cause antigen-specific local tolerance. J Clin Invest 2013;123(3):1032–1043 Knolle PA, Germann T, Treichel U, et al. Endotoxin down-regulates T cell activation by antigen-presenting liver sinusoidal endothelial cells. J Immunol 1999;162(3):1401–1407 Diehl L, Schurich A, Grochtmann R, Hegenbarth S, Chen L, Knolle PA. Tolerogenic maturation of liver sinusoidal endothelial cells promotes B7-homolog 1-dependent CD8þ T cell tolerance. Hepatology 2008;47(1):296–305 Knolle PA, Schmitt E, Jin S, et al. Induction of cytokine production in naive CD4(þ) T cells by antigen-presenting murine liver sinusoidal endothelial cells but failure to induce differentiation toward Th1 cells. Gastroenterology 1999;116(6):1428–1440 Kruse N, Neumann K, Schrage A, et al. Priming of CD4þ T cells by liver sinusoidal endothelial cells induces CD25low forkhead box protein 3- regulatory T cells suppressing autoimmune hepatitis. Hepatology 2009;50(6):1904–1913 Carambia A, Freund B, Schwinge D, et al. TGF-β-dependent induction of CD4(þ)CD25(þ)Foxp3(þ) Tregs by liver sinusoidal endothelial cells. J Hepatol 2014;61(3):594–599

47

48

49

50

51

52

53

54

55 56

57

58

59

60

61

62

63

64

65

67 68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

Seminars in Liver Disease

Vol. 34

No. 4/2014

385

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells 85 Géraud C, Mogler C, Runge A, et al. Endothelial transdifferentia-

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

tion in hepatocellular carcinoma: loss of Stabilin-2 expression in peri-tumourous liver correlates with increased survival. Liver Int 2013;33(9):1428–1440 Bergé M, Allanic D, Bonnin P, et al. Neuropilin-1 is upregulated in hepatocellular carcinoma and contributes to tumour growth and vascular remodelling. J Hepatol 2011;55(4):866–875 Knolle P, Schlaak J, Uhrig A, Kempf P, Meyer zum Büschenfelde KH, Gerken G. Human Kupffer cells secrete IL-10 in response to lipopolysaccharide (LPS) challenge. J Hepatol 1995;22(2): 226–229 de Waal Malefyt R, Abrams J, Bennett B, Figdor CG, de Vries JE. Interleukin 10(IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10 produced by monocytes. J Exp Med 1991;174(5):1209–1220 Wiegard C, Frenzel C, Herkel J, Kallen KJ, Schmitt E, Lohse AW. Murine liver antigen presenting cells control suppressor activity of CD4þCD25þ regulatory T cells. Hepatology 2005;42(1):193–199 Tu Z, Bozorgzadeh A, Pierce RH, Kurtis J, Crispe IN, Orloff MS. TLRdependent cross talk between human Kupffer cells and NK cells. J Exp Med 2008;205(1):233–244 Fiorentino DF, Bond MW, Mosmann TR. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med 1989;170(6):2081–2095 Breous E, Somanathan S, Vandenberghe LH, Wilson JM. Hepatic regulatory T cells and Kupffer cells are crucial mediators of systemic T cell tolerance to antigens targeting murine liver. Hepatology 2009;50(2):612–621 Tzung SP, Gaines KC, Lance P, Ehrke MJ, Cohen SA. Suppression of hepatic lymphokine-activated killer cell induction by murine Kupffer cells and hepatocytes. Hepatology 1990;12(4 Pt 1): 644–652 Billiar TR, Lysz TW, Curran RD, Bentz BG, Machiedo GW, Simmons RL. Hepatocyte modulation of Kupffer cell prostaglandin E2 production in vitro. J Leukoc Biol 1990;47(4):305–311 Rodriguez PC, Hernandez CP, Quiceno D, et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med 2005;202(7):931–939 Fu J, Xu D, Liu Z, et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 2007;132(7):2328–2339 Arihara F, Mizukoshi E, Kitahara M, et al. Increase in CD14þHLADR -/low myeloid-derived suppressor cells in hepatocellular carcinoma patients and its impact on prognosis. Cancer Immunol Immunother 2013;62(8):1421–1430 Wu K, Kryczek I, Chen L, Zou W, Welling TH. Kupffer cell suppression of CD8þ T cells in human hepatocellular carcinoma is mediated by B7-H1/programmed death-1 interactions. Cancer Res 2009;69(20):8067–8075 Li H, Wu K, Tao K, 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):1342–1351 Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 2005;6(12):1245–1252 Crispe IN, Giannandrea M, Klein I, John B, Sampson B, Wuensch S. Cellular and molecular mechanisms of liver tolerance. Immunol Rev 2006;213:101–118 Heuff G, Oldenburg HS, Boutkan H, et al. Enhanced tumour growth in the rat liver after selective elimination of Kupffer cells. Cancer Immunol Immunother 1993;37(2):125–130 Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 2004;127(5, Suppl 1):S35–S50 Paik YH, Schwabe RF, Bataller R, Russo MP, Jobin C, Brenner DA. Toll-like receptor 4 mediates inflammatory signaling by bacterial

Seminars in Liver Disease

Vol. 34

No. 4/2014

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

Sprinzl, Galle

lipopolysaccharide in human hepatic stellate cells. Hepatology 2003;37(5):1043–1055 Dapito DH, Mencin A, Gwak GY, et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell 2012;21(4):504–516 Svegliati Baroni G, D’Ambrosio L, Ferretti G, et al. Fibrogenic effect of oxidative stress on rat hepatic stellate cells. Hepatology 1998; 27(3):720–726 Hong F, Tuyama A, Lee TF, et al. Hepatic stellate cells express functional CXCR4: role in stromal cell-derived factor-1alphamediated stellate cell activation. Hepatology 2009;49(6): 2055–2067 Dunham RM, Thapa M, Velazquez VM, et al. Hepatic stellate cells preferentially induce Foxp3þ regulatory T cells by production of retinoic acid. J Immunol 2013;190(5):2009–2016 Dangi A, Sumpter TL, Kimura S, et al. Selective expansion of allogeneic regulatory T cells by hepatic stellate cells: role of endotoxin and implications for allograft tolerance. J Immunol 2012;188(8):3667–3677 Zhao HQ, Li WM, Lu ZQ, Yao YM. Roles of Tregs in development of hepatocellular carcinoma: a meta-analysis. World J Gastroenterol 2014;20(24):7971–7978 Chou HS, Hsieh CC, Yang HR, et al. Hepatic stellate cells regulate immune response by way of induction of myeloid suppressor cells in mice. Hepatology 2011;53(3):1007–1019 Höchst B, Schildberg FA, Sauerborn P, et al. Activated human hepatic stellate cells induce myeloid derived suppressor cells from peripheral blood monocytes in a CD44-dependent fashion. J Hepatol 2013;59(3):528–535 Chen Y, Huang Y, Reiberger T, 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):1435–1447 Yu MC, Chen CH, Liang X, et al. Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology 2004;40(6):1312–1321 Chen CH, Kuo LM, Chang Y, et al. In vivo immune modulatory activity of hepatic stellate cells in mice. Hepatology 2006;44(5): 1171–1181 Ooi LP, Crawford DH, Gotley DC, et al. Evidence that “myofibroblast-like” cells are the cellular source of capsular collagen in hepatocellular carcinoma. J Hepatol 1997;26(4):798–807 Neaud V, Faouzi S, Guirouilh J, et al. Human hepatic myofibroblasts increase invasiveness of hepatocellular carcinoma cells: evidence for a role of hepatocyte growth factor. Hepatology 1997; 26(6):1458–1466 Amann T, Bataille F, Spruss T, et al. Activated hepatic stellate cells promote tumorigenicity of hepatocellular carcinoma. Cancer Sci 2009;100(4):646–653 Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005;121(3):335–348 Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell 2010;17(2):135–147 Liao D, Luo Y, Markowitz D, Xiang R, Reisfeld RA. Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS ONE 2009;4(11):e7965 Li T, Yang Y, Hua X, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett 2012;318(2):154–161 Basili S, Raparelli V, Riggio O, et al; CALC Group. NADPH oxidasemediated platelet isoprostane over-production in cirrhotic

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

386

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

125

126

127

128 129

130

131

132

133

134

135

136

137

138

139

140

141 142

387

143 Jomantaite I, Dikopoulos N, Kröger A, et al. Hepatic dendritic cell

subsets in the mouse. Eur J Immunol 2004;34(2):355–365 144 De Creus A, Abe M, Lau AH, Hackstein H, Raimondi G, Thomson

145

146

147

148

149 150

151

152

153

154

155

156

157

158

159

160

161

AW. Low TLR4 expression by liver dendritic cells correlates with reduced capacity to activate allogeneic T cells in response to endotoxin. J Immunol 2005;174(4):2037–2045 Pillarisetty VG, Shah AB, Miller G, Bleier JI, DeMatteo RP. Liver dendritic cells are less immunogenic than spleen dendritic cells because of differences in subtype composition. J Immunol 2004; 172(2):1009–1017 Beckebaum S, Zhang X, Chen X, et al. Increased levels of interleukin-10 in serum from patients with hepatocellular carcinoma correlate with profound numerical deficiencies and immature phenotype of circulating dendritic cell subsets. Clin Cancer Res 2004;10(21):7260–7269 Ninomiya T, Akbar SM, Masumoto T, Horiike N, Onji M. Dendritic cells with immature phenotype and defective function in the peripheral blood from patients with hepatocellular carcinoma. J Hepatol 1999;31(2):323–331 Chen S, Akbar SM, Tanimoto K, et al. Absence of CD83-positive mature and activated dendritic cells at cancer nodules from patients with hepatocellular carcinoma: relevance to hepatocarcinogenesis. Cancer Lett 2000;148(1):49–57 Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141(1):39–51 Ding T, Xu J, Wang F, et al. High tumor-infiltrating macrophage density predicts poor prognosis in patients with primary hepatocellular carcinoma after resection. Hum Pathol 2009;40(3): 381–389 Li YW, Qiu SJ, Fan J, et al. Tumor-infiltrating macrophages can predict favorable prognosis in hepatocellular carcinoma after resection. J Cancer Res Clin Oncol 2009;135(3):439–449 Herlyn D, Koprowski H. IgG2a monoclonal antibodies inhibit human tumor growth through interaction with effector cells. Proc Natl Acad Sci U S A 1982;79(15):4761–4765 Adams DO, Hall T, Steplewski Z, Koprowski H. Tumors undergoing rejection induced by monoclonal antibodies of the IgG2a isotype contain increased numbers of macrophages activated for a distinctive form of antibody-dependent cytolysis. Proc Natl Acad Sci U S A 1984;81(11):3506–3510 Kong LQ, Zhu XD, Xu HX, et al. The clinical significance of the CD163þ and CD68þ macrophages in patients with hepatocellular carcinoma. PLoS ONE 2013;8(3):e59771 Waidmann O, Köberle V, Bettinger D, et al. Diagnostic and prognostic significance of cell death and macrophage activation markers in patients with hepatocellular carcinoma. J Hepatol 2013;59(4):769–779 Zhu XD, Zhang JB, Zhuang PY, et al. High expression of macrophage colony-stimulating factor in peritumoral liver tissue is associated with poor survival after curative resection of hepatocellular carcinoma. J Clin Oncol 2008;26(16):2707–2716 Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol 2006;177(10):7303–7311 Colegio OR, Chu NQ, Szabo AL, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014 [Epub ahead of print] Sprinzl MF, Reisinger F, Puschnik A, et al. Sorafenib perpetuates cellular anticancer effector functions by modulating the crosstalk between macrophages and natural killer cells. Hepatology 2013; 57(6):2358–2368 Kuang DM, Zhao Q, Peng C, 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):1327–1337 Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro Seminars in Liver Disease

Vol. 34

No. 4/2014

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

124

patients: implication for platelet activation. Liver Int 2011; 31(10):1533–1540 Dupuy E, Hainaud P, Villemain A, et al. Tumoral angiogenesis and tissue factor expression during hepatocellular carcinoma progression in a transgenic mouse model. J Hepatol 2003;38(6): 793–802 Palumbo JS, Talmage KE, Massari JV, et al. Platelets and fibrin (ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 2005;105(1): 178–185 Terme M, Pernot S, Marcheteau E, et al. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res 2013;73(2):539–549 Yoong KF, Afford SC, Jones R, et al. Expression and function of CXC and CC chemokines in human malignant liver tumors: a role for human monokine induced by gamma-interferon in lymphocyte recruitment to hepatocellular carcinoma. Hepatology 1999; 30(1):100–111 Oo YH, Adams DH. The role of chemokines in the recruitment of lymphocytes to the liver. J Autoimmun 2010;34(1):45–54 Liu Y, Poon RT, Feng X, Yu WC, Luk JM, Fan ST. Reduced expression of chemokine receptors on peripheral blood lymphocytes in patients with hepatocellular carcinoma. Am J Gastroenterol 2004;99(6):1111–1121 Chew V, Chen J, Lee D, et al. Chemokine-driven lymphocyte infiltration: an early intratumoural event determining longterm survival in resectable hepatocellular carcinoma. Gut 2012; 61(3):427–438 Liu YQ, Poon RT, Hughes J, Li QY, Yu WC, Fan ST. Desensitization of T lymphocyte function by CXCR3 ligands in human hepatocellular carcinoma. World J Gastroenterol 2005;11(2):164–170 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004;10(9):942–949 Yang P, Li QJ, Feng Y, et al. TGF-β-miR-34a-CCL22 signalinginduced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell 2012;22(3): 291–303 Schimanski CC, Bahre R, Gockel I, et al. Dissemination of hepatocellular carcinoma is mediated via chemokine receptor CXCR4. Br J Cancer 2006;95(2):210–217 Schrader J, Gordon-Walker TT, Aucott RL, et al. Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology 2011;53(4): 1192–1205 Ding ZY, Jin GN, Wang W, et al. Reduced expression of TIF1γ promotes metastasis and indicates poor prognosis of hepatocellular carcinoma. Hepatology 2014 [Epub ahead of print] PubMed Hiratsuka S, Duda DG, Huang Y, et al. C-X-C receptor type 4 promotes metastasis by activating p38 mitogen-activated protein kinase in myeloid differentiation antigen (Gr-1)-positive cells. Proc Natl Acad Sci U S A 2011;108(1):302–307 Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9(3): 162–174 Yoneyama H, Matsuno K, Zhang Y, et al. Regulation by chemokines of circulating dendritic cell precursors, and the formation of portal tract-associated lymphoid tissue, in a granulomatous liver disease. J Exp Med 2001;193(1):35–49 Kudo S, Matsuno K, Ezaki T, Ogawa M. A novel migration pathway for rat dendritic cells from the blood: hepatic sinusoids-lymph translocation. J Exp Med 1997;185(4):777–784 Thomson AW, O’Connell PJ, Steptoe RJ, Lu L. Immunobiology of liver dendritic cells. Immunol Cell Biol 2002;80(1):65–73 Bosma BM, Metselaar HJ, Mancham S, et al. Characterization of human liver dendritic cells in liver grafts and perfusates. Liver Transpl 2006;12(3):384–393

Sprinzl, Galle

Immune Control in Hepatocellular Carcinoma Development and Progression: Role of Stromal Cells

162

163

164

165

166

167

168

169

170

171

172 173

174

inhibit proinflammatory cytokine production through autocrine/ paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest 1998;101(4):890–898 Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med 1999;189(9):1363–1372 Rodriguez PC, Quiceno DG, Zabaleta J, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res 2004;64(16):5839–5849 Etzerodt A, Maniecki MB, Møller K, Møller HJ, Moestrup SK. Tumor necrosis factor α-converting enzyme (TACE/ADAM17) mediates ectodomain shedding of the scavenger receptor CD163. J Leukoc Biol 2010;88(6):1201–1205 Schlecker E, Fiegler N, Arnold A, et al. Metalloprotease-mediated tumor cell shedding of B7-H6, the ligand of the natural killer cellactivating receptor NKp30. Cancer Res 2014;74(13):3429–3440 Roderfeld M, Rath T, Lammert F, Dierkes C, Graf J, Roeb E. Innovative immunohistochemistry identifies MMP-9 expressing macrophages at the invasive front of murine HCC. World J Hepatol 2010;2(5):175–179 Sun D, Wang X, Zhang H, Deng L, Zhang Y. MMP9 mediates MICA shedding in human osteosarcomas. Cell Biol Int 2011;35(6): 569–574 Hoechst B, Ormandy LA, Ballmaier M, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(þ)CD25(þ)Foxp3(þ) T cells. Gastroenterology 2008;135(1):234–243 Shen P, Wang A, He M, Wang Q, Zheng S. Increased circulating Lin (-/low) CD33(þ) HLA-DR(-) myeloid-derived suppressor cells in hepatocellular carcinoma patients. Hepatol Res 2014;44(6): 639–650 Kalathil S, Lugade AA, Miller A, Iyer R, Thanavala Y. Higher frequencies of GARP(þ)CTLA-4(þ)Foxp3(þ) T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res 2013;73(8):2435–2444 Wang R, Kozhaya L, Mercer F, Khaitan A, Fujii H, Unutmaz D. Expression of GARP selectively identifies activated human FOXP3þ regulatory T cells. Proc Natl Acad Sci U S A 2009; 106(32):13439–13444 Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008;133(5):775–787 Sojka DK, Huang YH, Fowell DJ. Mechanisms of regulatory T-cell suppression - a diverse arsenal for a moving target. Immunology 2008;124(1):13–22 Yang XH, Yamagiwa S, Ichida T, et al. Increase of CD4þ CD25þ regulatory T-cells in the liver of patients with hepatocellular carcinoma. J Hepatol 2006;45(2):254–262

Seminars in Liver Disease

Vol. 34

No. 4/2014

Sprinzl, Galle

175 Unitt E, Rushbrook SM, Marshall A, et al. Compromised lympho-

176

177

178

179

180

181

182

183

184

185

186

187

188

cytes infiltrate hepatocellular carcinoma: the role of T-regulatory cells. Hepatology 2005;41(4):722–730 Zhang W, Zhu XD, Sun HC, et al. Depletion of tumor-associated macrophages enhances the effect of sorafenib in metastatic liver cancer models by antimetastatic and antiangiogenic effects. Clin Cancer Res 2010;16(13):3420–3430 Hagemann T, Lawrence T, McNeish I, et al. “Re-educating” tumorassociated macrophages by targeting NF-kappaB. J Exp Med 2008;205(6):1261–1268 Pyonteck SM, Akkari L, Schuhmacher AJ, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med 2013;19(10):1264–1272 Sangro B, Gomez-Martin C, de la Mata M, et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol 2013;59(1): 81–88 Zerbini A, Pilli M, Soliani P, et al. Ex vivo characterization of tumor-derived melanoma antigen encoding gene-specific CD8þcells in patients with hepatocellular carcinoma. J Hepatol 2004;40(1):102–109 Cai XY, Gao Q, Qiu SJ, et al. Dendritic cell infiltration and prognosis of human hepatocellular carcinoma. J Cancer Res Clin Oncol 2006; 132(5):293–301 Yin XY, Lu MD, Lai YR, Liang LJ, Huang JF. Prognostic significances of tumor-infiltrating S-100 positive dendritic cells and lymphocytes in patients with hepatocellular carcinoma. Hepatogastroenterology 2003;50(53):1281–1284 Huang Y, Wang FM, Wang T, et al. Tumor-infiltrating FoxP3þ Tregs and CD8þ T cells affect the prognosis of hepatocellular carcinoma patients. Digestion 2012;86(4):329–337 Wang F, Jing X, Li G, et al. Foxp3þ regulatory T cells are associated with the natural history of chronic hepatitis B and poor prognosis of hepatocellular carcinoma. Liver Int 2012;32(4):644–655 Huang Y, Wang F, Wang Y, et al. Intrahepatic interleukin-17þ T cells and FoxP3þ regulatory T cells cooperate to promote development and affect the prognosis of hepatocellular carcinoma. J Gastroenterol Hepatol 2014;29(4):851–859 Sasaki A, Tanaka F, Mimori K, et al. Prognostic value of tumorinfiltrating FOXP3þ regulatory T cells in patients with hepatocellular carcinoma. Eur J Surg Oncol 2008;34(2):173–179 Fan QM, Jing YY, Yu GF, et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett 2014;352(2): 160–168 Matsubara T, Kanto T, Kuroda S, et al. TIE2-expressing monocytes as a diagnostic marker for hepatocellular carcinoma correlates with angiogenesis. Hepatology 2013;57(4):1416–1425

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

388

Copyright of Seminars in Liver Disease is the property of Thieme Medical Publishing Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Immune control in hepatocellular carcinoma development and progression: role of stromal cells.

Immune control of hepatocellular carcinoma (HCC) is executed by effector immune cells, which efficiently eliminate malignant transformed cells. Howeve...
331KB Sizes 0 Downloads 5 Views