Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Nov 1.
Published in final edited form as: Hepatology. 2014 Sep 26;60(5):1776–1782. doi: 10.1002/hep.27246

Immune checkpoint blockade in hepatocellular carcinoma: Current progress and future directions

Tai Hato 1, Lipika Goyal 2, Tim F Greten 3, Dan G Duda 1,*, Andrew X Zhu 2,*
PMCID: PMC4211962  NIHMSID: NIHMS604607  PMID: 24912948

Abstract

Immune checkpoint blockade has recently emerged as a promising therapeutic approach for various malignancies including hepatocellular carcinoma (HCC). Preclinical and clinical studies have shown the potential benefit of modulating immunogenicity of HCC. In addition, recent advances in tumor immunology have broadened our understanding of the complex mechanism of immune evasion. In this review, we summarize the current knowledge on HCC immunology, and discuss the potential of immune checkpoint blockade as a novel HCC therapy from the basic, translational, and clinical perspectives.

Introduction

The failure of the immune system to prevent hepatocellular carcinoma (HCC) and to halt its progression is closely linked with pathogenesis and the survival of patients with HCC. Thus, immunotherapy aimed at boosting HCC-specific immune responses is considered a promising treatment approach. In this review, we will summarize the current knowledge of immune responses in HCC—with the focus on immune checkpoints—and the current status of translational clinical research in this field.

Why use immunotherapy for HCC patients?

The success of immune checkpoint blockade with anti-cytotoxic T lymphocyte associated antigen 4 (CTLA-4) antibodies in advanced melanoma patients has brought renewed hope for immunotherapy in cancer. In addition, immunotherapy is particularly attractive for HCC for several reasons. HCC is typically an inflammation-associated cancer and can be immunogenic. Indeed, cases of spontaneous regression of HCC have been reported and many of these cases were related to systemic inflammatory responses [1]. In addition, the majority of HCC patients suffer from cirrhosis of viral etiology, alcoholism, or nonalcoholic steatohepatitis. Since immunotherapeutic drugs are not metabolized in the liver, they may have predictable pharmacokinetic profiles in cirrhotic patients. Indeed, preliminary clinical data with antibody-based therapy has not shown any severe hepatoxicity [2]. Nevertheless, the successful application of immunotherapy in HCC will have to take into account the liver cancer-specific immune microenvironment and responses.

How do HCCs evade anti-tumor immunity?

Spontaneous anti-tumor responses have been detected in HCC patients. Activation of immune response and T cell infiltration has been reported after percutaneous ethanol injection and radiofrequency ablation (RFA) [3,4]. In addition, tumor-associated antigen (TAA) specific CD8+ T cell immune responses have been described. Among the most studied antigens in HCC are alpha-fetoprotein (AFP), glypican-3 (GPC-3), NY-ESO-1, SSX-2, melanoma antigen gene-A (MAGE-A) and human telomerase-reverse transcriptase (hTERT). One report estimated that more than 50% of HCC patients develop spontaneous cellular or humoral immune response against NY-ESO-1 [5]. Another study reported that HCC-infiltrating TAA-specific CD8+ T cells were detectable in more than 50% of patients, and these cell numbers correlated with progression-free survival [6].

The immune microenvironment of the liver plays a major role in anti-tumor immunity. Liver is generally “tolerogenic” to prevent undesirable immune response to antigens absorbed from the gut. The tolerability is maintained by direct activation of naïve T cells in liver through antigen presentation by liver sinusoidal endothelial cells, Kuppfer cells, dendritic cells (DCs) and hepatocytes [710]. In addition, intricate immunosuppressive mechanisms become activated in the HCC microenvironment and further interfere with the development of meaningful anti-tumor immune responses. Multiple such mechanisms have been proposed, including defective antigen presentation, recruitment of immunosuppressive myeloid and lymphoid cell populations, suppression of natural killer (NK) cells, impaired CD4+ T cell functions, and up-regulation of immune checkpoint pathways [1123].

Among immunosuppressive cell populations, T regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSCs) are thought to play key roles in cancer evasion from immunosurveillance. In HCC, the number of Tregs is increased both in the blood circulation and inside the tumor [11]. Intratumoral Treg accumulation correlates with disease progression and poor prognosis [12, 13]. MDSCs are immature/progenitor myeloid cells with immunosuppressive and pro-angiogenic activity. MDSC accumulation is found not only within the tumors but also in blood circulation, spleen, bone marrow and liver [14]. The MDSCs inhibit the function of effector T cells, and decrease NK cell cytotoxicity and cytokine production [15, 16]. The frequency of MDSCs has been shown to correlate with recurrence-free survival of HCC patients who have undergone RFA [17]. It has also been suggested that MDSCs interact with Kuppfer cells to induce PD-L1 expression, which in turn inhibits antigen presentation [14]. MDSCs may also help expand Treg population. Depletion of Tregs or MDSCs could prompt spontaneous immune responses against AFP, suggesting the potential of immune reactivation [15, 23]. Recently, Han et al. have identified new subset of immune suppressive cells in HCC patients called regulatory DCs [24]. These regulatory DCs can suppress T cell activation through interleukin (IL)-10 and indoleamine 2,3-dioxygenase (IDO) production.

Exhaustion of CD4+ T cells has also been reported as a mechanism of immune evasion in HCC. While infrequent AFP-specific CD4+ T cells are detectable in early disease, they became exhausted and fail to execute their immune supportive function once the disease has advanced [18, 19].

Finally, while the immune response to specific antigen is recognized by major histocompatibility receptors, co-stimulatory and co-inhibitory molecules regulate the intensity of response. Immune checkpoints are co-inhibitory molecules that are physiologically expressed for the maintenance of self-tolerance. In the tumor microenvironment, immune checkpoint molecules such as CTLA-4 and PD-L1 are often overexpressed and participate in the evasive mechanism as discussed above [2529]. In the next section, we will discuss the exciting potential of blocking the immune checkpoint for HCC therapy.

Immune checkpoint blockade: Potential for cancer immunotherapy

The balance between co-stimulatory signals and immune checkpoints determines the cytotoxic T cell activation and intensity of immune response [30]. The immune checkpoints are often activated in the tumor tissue, and this promotes tumor evasion from host immunity. The most studied immune checkpoint receptors are CTLA-4, PD-1, TIM-3, BTLA, VISTA, LAG-3 and OX40 (Fig.1 &Suppl. Table S1). Inhibitors of CTLA-4 and PD-1 have already been FDA-approved for the treatment of melanoma and are currently in development in HCC. Our review focuses primarily on the potential of these two pathways as targets in HCC.

Figure 1. Role of immune checkpoint molecules in anti-tumor immune responses against hepatocellular carcinoma.

Figure 1

(a) Cancer cells enhance the intratumoral recruitment and expansion of MDSCs and Tregs. Tregs and MDSCs suppress innate immune response of NK cells through activation of immune checkpoint pathways such as PD-L1/PD-1. MDSC or Treg activity is regulated by the inflammatory tumor microenvironment, helper T cells or dendritic cell activities. (b) The acquired immune responses are suppressed through multiple mechanisms by immune checkpoint molecules. Cytotoxic T cell function is not only subdued by the immune suppressive cells such as Tregs and MDSCs but also repressed by the co-stimulation of CTLA-4 with antigen presentation from DCs. The tolerogenic liver microenvironment also inhibits cytotoxic T cell activation.

CTLA-4: Mechanism of action

Although the mechanism of action of CTLA-4 (also known as CD152) is not fully elucidated, it is clear that this immune checkpoint acts as a “break” for immune responses. CTLA-4 is expressed on activated T cells and Tregs, and, at low levels, may also be expressed on naïve T cells [3133]. Upon stimulation via the T cell receptor, CTLA-4 localizes to the plasma membrane. CTLA-4 can bind to CD80 and CD86 with much higher affinity than CD28 [34]. In doing so, CTLA-4 antagonizes the CD28 binding to CD80 and CD86 and inhibits T cell activation. CTLA-4 is known to inhibit the binding of antigen presentation by antigen-presenting cell (APC) [35]. Reverse signaling through CD80 or CD86 activates IDO in APC, which leads to tryptophan degradation and suppression of T cell-mediated anti-tumor immune responses [36]. CTLA-4 signaling may stimulate the expression of immune regulatory cytokines such as TGF-beta [37]. In addition, inhibition of CD28 interaction with CD80 or CD86 binding in APCs results in reduced T cell activation. Knockout of CTLA-4 is embryonically lethal in mice due to autoimmune response and excessive CD4+ T cell proliferation, suggesting that CTLA-4 function is primarily important in CD4+ T cells [38]. Although the exact mechanism is not known, CTLA-4 is also thought to have an effect on the differentiation and activation of Tregs, which constitutively express this receptor. Treg-specific knockout or blockade of CTLA-4 inhibits their ability to regulate both autoimmunity and anti-cancer immunity [39, 40].

Despite this understanding, and the promising results obtained so far with CTLA-4 inhibitors in clinical trials, mechanistic data on CTLA-4 blockade from preclinical models of HCC are limited. CTLA-4 blockade combined with microwave ablation and local GM-CSF administration showed promising efficacy in subcutaneous models of HCC [20]. Re-challenging the mice by re-implantation of the cancer cells resulted in tumor rejection in 90% of the cases. Anti-tumor responses by CD4+ and CD8+ T cells and NK cells in the circulating blood were also observed, suggesting successful immunization by this strategy. Future studies in more clinically relevant models of HCC should provide mechanistic insights to guide clinical development.

Clinical studies of CTLA-4 blockade

Two CTLA-4 blocking antibodies (ipilimumab and tremelimumab) are currently in advanced stages of clinical development (Table 1). The US FDA approved ipilimumab for the treatment of melanoma in 2011 based on randomized phase III trial showing improved overall survival [41, 42]. Recently, tremelimumab showed early evidence of antitumor activity in a single arm phase II trial in malignant mesothelioma [43]. CTLA-4 blockade has also been tested in other malignancies, including breast, colon, lung, prostate and brain cancers, melanoma, lymphoma and sarcomas. Two observations are consistent in the published studies [44]. Firstly, the efficacy of CTLA-4 blockade may correlate with the immunogenicity of the tumor. Secondly, the immunotherapy may be more effective against smaller tumors.

Table 1.

Immune checkpoint blocking antibodies and status of clinical development for HCC

Target Antibody Trial ID Phase Treatment Status Results
CTLA-4 Tremelimumab(formerly referred to asticilimumab, CP-675,206, MedImmune, USA & Pfizer, USA) NCT01008358 I Monotherapy Completed Well tolerated Disease control rate of 76.4% Median OS 8.2 month [95%CI: 4.64,21.34]
NCT01853618 I In combination with RFA or TACE Ongoing N/A
Ipilimumab(MDX-010, Bristol-Myers Squibb, USA) N/A N/A N/A N/A N/A

PD-1 Nivolumab (BMS-936558, Bristol-Myers Squibb, USA) NCT01658878 I Monotherapy Ongoing N/A
CT-011 (CureTech, Israel) NCT00966251 I/II Monotherapy Terminated Stopped due to slow accrual
Lambrolizumab (MK-3475, Merck, USA) N/A N/A N/A N/A N/A
AMP-224 (Amplimmune, USA) N/A N/A N/A N/A N/A

PD-L1 MPDL3280A/RG7446 (Genentech, USA &Roche, Switzerland) N/A N/A N/A N/A N/A
MEDI4376 (MedImmune, USA &AstraZeneca, UK) N/A N/A N/A N/A N/A

A phase I trial of tremelimumab in HCC patients has recently been reported (NCT01008358) [43]. The study enrolled 21 patients with chronic hepatitis C with Child-Pugh A or B cirrhosis and advanced HCC not amenable to percutaneous ablation or transarterial embolization. Tremelimumab was well tolerated, and there were no treatment related deaths. Almost half of the patients experienced a grade 3/4 rise in AST but this was not associated with a parallel decline in liver dysfunction. Partial responses were seen in 17.6% of the cases, and 45% of the patients had stable disease for more than 6 months. Interestingly, the patients with stable levels of interferon (IFN)-γ during the treatment showed better treatment response compared to those who showed a decrease, suggestive of more active anti-tumor immunity. A phase I clinical trial of tremelimumab with radiofrequency ablation or transarterial therapy is ongoing [NCT01853618].

PD-1: Mechanism of action

PD-1 is another CD28 superfamily member that conveys co-inhibitory signals for TCR receptor. PD-1 binds to its ligands PD-L1 (CD274) or PD-L2 (CD273) [45, 46]. PD-1 is primarily expressed on CD8+ T cells, but can also be detected on Tregs and MDSCs [47]. PD-1 mediates the differentiation and proliferation of Tregs [48]. PD-1 also regulates peripheral tolerance and autoimmunity. Chronic exposure to antigens leads to the overexpression of PD-1 in T cells, which induces anergy or cell exhaustion [49]. By chronic antigen stimulation, IFN-γ induces IRF9 binding to Pdcd-1 promoter and PD-1 transcription in T cells. When PD-1 binds to PD-L1 or PD-L2, T cell proliferation and cytokine release are inhibited through SHP2, which inactivates ZAP70, a major TCR signaling integrator [50]. T cell function is differentially affected by the level of PD-1 activity [51]. Cancer cells can highjack PD-L1/PD-1 signaling by expressing PD-L1 or PD-L2 to activate PD-1 in tumor-infiltrating lymphocytes and evade immune surveillance [52, 53].

While the mechanisms of immune tolerance to viral hepatitis are well described, limited data are available for HCC [54, 55]. Two mouse models showed the potential relevance of PD-1/PD-L1-induced immune tolerance in HCC. In a genetic model of c-Myc-induced HCC and doxycycline-induced expression of IL-12 in hepatocytes, doxycycline treatment induced IFN-γ expression but only a partial regression of the tumors [21]. Treatment resistance was associated with an increase in Treg numbers and upregulation of several immune checkpoint molecules (including PD-L1/PD-1). In another mouse model of HCC induced by adenovirus-mediated inducible SV40 large T antigen expression in hepatocytes, T cell infiltration into the tumor was found to be decreased in the advanced lesions [22].

Translational clinical studies also support the potential role of PD-1/PD-L1 pathway-induced immune tolerance in HCC. PD-L1 was found to be expressed by cancer cells and PD-1 by CD8+ T cells in HCC tissue specimens [25]. Moreover, correlative studies have shown that both PD-L1 expression and PD-1 expression in tumors were significantly correlated with HCC stage, local recurrence rate and poor prognosis [26, 5658]. Similarly, the frequency of intratumoral or circulating PD-1+CD8+ T cells correlated with HCC progression and postoperative recurrence [27].

Even in cases when CD8+ T lymphocytes were activated and recognized the tumor (as evidenced by HLA-1 expression), high PD-L1 expression negatively impacted prognosis in resected HCC patients [56]. Furthermore, PD-L1 and PD-1 expression in circulating cells correlated with poor prognosis in HBV-positive HCC patients who underwent cryoablation [28]. Finally, simultaneous depletion of Tregs, MDSC and PD-1+ T cells from the peripheral blood of advanced HCC patients could restore CD8+ T cell activation ex vivo [29].

Clinical studies of PD-1 blockade

Five anti-PD-1 antibodies and 3 anti-PD-L1 antibodies are currently under development, emphasizing the growing interest in this immune checkpoint pathway as a target for cancer therapy (Table 1). Lambrolizumab induced tumor regression in advanced melanoma patients and showed a favorable safety profile [59]. Interestingly, lambrolizumab was effective even in patients who failed ipilimumab treatment, which suggests a differential mechanism of action for PD-1 inhibition versus CTLA-4 blockade. Indeed, combination of nivolumab with ipilimumab achieved objective response in 40% of the patients with limited toxicity [60]. CT-011 and MPDL3280A/RG7446 were tested in phase I trials and showed with favorable safety profiles [61]. MEDI4376 targets PD-L1, and phase I trial is ongoing. AMP-224 is a recombinant B7-DC-Fc fusion protein, and a phase I trial of this agent is also underway. In HCC, a phase I/II trial of CT-011 in advanced HCC was initiated but stopped due to slow accrual. A phase I trial of nivolumab is currently ongoing for patients with advanced HCC (NCT01658878). This study plans to enroll three cohorts of patients stratified by viral etiology—HCV, HBV, no viral infection—with a target enrollment of 90 patients.

Future perspectives

Immunotherapy using immune checkpoint blockers has already shown great promise in intractable cancers such as advanced melanoma. This approach has recently begun clinical testing in advanced HCC patients. The limited evidence from pre-clinical studies in animal models further supports the development of CTLA-4 and PD-L1/PD-1 inhibitors in this disease. Successful development of this strategy will have to address several aspects specific to HCC. Firstly, it will need to address the specific issues of immunotolerance and chronic liver inflammation (cirrhosis) and hypoxia that underlie HCC development and response to treatment [62]. Secondly, the strategy will be most likely to succeed if the development will be guided by biology-driven biomarkers of response to such agents. Thirdly, therapy with immune checkpoint blockers is most likely to succeed in combination with other surgical, cytotoxic, immune or targeted therapies. Combination of immune checkpoint inhibitors with local ablative therapies such as RFA or cryoablation—which may induce tumor antigen release—would particularly promising approaches [63]. Furthermore, integration of immunotherapy with systemic treatments (e.g., sorafenib) will require mechanistic understanding of treatment interactions. Finally, the integration of immune checkpoint blockers—alone or in combination with other agents—will have to address the safety concerns specific to this population of patients such as hepatotoxicity. The potential risk of fueling acute exacerbation of viral hepatitis in HBV/HCV positive patients will need to be elucidated and carefully observed in the clinical setting. Addressing these issues will greatly help the field bring to fruition the great promise of these novel immune-therapeutics in this intractable disease.

Supplementary Material

Supp TableS1
Supplementary Material

Acknowledgments

The research of the authors is supported by National Institutes of Health grants P01-CA080124, R01-CA159258, and R21-CA139168, a National Cancer Institute/Proton Beam Federal Share Program award, and the American Cancer Society grant 120733-RSG-11-073-01-TBG (to DGD); and a Postdoctoral Fellowship from Astellas Foundation for Research on Metabolic Disorders, Japan (to TH).

References

  • 1.Huz JI, Melis M, Sarpel U. Spontaneous regression of hepatocellular carcinoma is most often associated with tumour hypoxia or a systemic inflammatory response. HPB. 2012;14:500–5. doi: 10.1111/j.1477-2574.2012.00478.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sangro B, Gomez-Martin C, de la Mata M, Iñarrairaegui M, Garralda E, Barrera P, Riezu-Boj JI, Larrea E, Alfaro C, Sarobe P, Lasarte JJ, Pérez-Gracia JL, Melero I, Prieto J. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol. 2013;59:81–88. doi: 10.1016/j.jhep.2013.02.022. [DOI] [PubMed] [Google Scholar]
  • 3.Ohnishi K, Yoshioka H, Ito S, Fujiwara K. Prospective randomized controlled trial comparing percutaneous acetic acid injection and percutaneous ethanol injection for small hepatocellular carcinoma. Hepatology. 1998;27:67–72. doi: 10.1002/hep.510270112. [DOI] [PubMed] [Google Scholar]
  • 4.Shiina S, Tagawa K, Unuma T, Takanashi R, Yoshiura K, Komatsu Y, Hata Y, Niwa Y, Shiratori Y, Terano A, et al. Percutaneous ethanol injection therapy for hepatocellular carcinoma. A histopathologic study. Cancer. 1991;68:1524–1530. doi: 10.1002/1097-0142(19911001)68:7<1524::aid-cncr2820680711>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 5.Korangy F, Ormandy LA, Bleck JS, Klempnauer J, Wilkens L, Manns MP. Greten TFSpontaneous tumor-specific humoral and cellular immune responses to NY-ESO-1 in hepatocellular carcinoma. Clin Cancer Res. 2004;10:4332–41. doi: 10.1158/1078-0432.CCR-04-0181. [DOI] [PubMed] [Google Scholar]
  • 6.Flecken T, Schmidt N, Hild S, Gostick E, Drognitz O, Zeiser R, Schemmer P, Bruns H, Eiermann T, Price DA, Blum HE, Neumann-Haefelin C, Thimme R. Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology. 2014;59(4):1415–26. doi: 10.1002/hep.26731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Schurich A, Berg M, Stabenow D, Böttcher J, Kern M, Schild HJ, Kurts C, Schuette V, Burgdorf S, Diehl L, Limmer A, Knolle PA. Dynamic regulation of CD8 T cell tolerance induction by liver sinusoidal endothelial cells. J Immunol. 2010;184:4107–14. doi: 10.4049/jimmunol.0902580. [DOI] [PubMed] [Google Scholar]
  • 8.Kuniyasu Y, Marfani SM, Inayat IB, Sheikh SZ, Mehal WZ. Kupffer cells required for high affinity peptide-induced deletion, not retention, of activated CD8+ T cells by mouse liver. Hepatology. 2004;39:1017–27. doi: 10.1002/hep.20153. [DOI] [PubMed] [Google Scholar]
  • 9.Thomson AW, Knolle PA. Antigen-presenting cell function in the tolerogenic liver environment. Nat Rev Immunol. 2010;10:753–66. doi: 10.1038/nri2858. [DOI] [PubMed] [Google Scholar]
  • 10.Holz LE, Bowen DG, Bertolino P. Mechanisms of T cell death in the liver: to Bim or not to Bim? Dig Dis. 2010;28:14–24. doi: 10.1159/000282060. [DOI] [PubMed] [Google Scholar]
  • 11.Ormandy LA, Hillemann T, Wedemeyer H, Manns MP, Greten TF, Korangy F. Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res. 2005;65:2457–2464. doi: 10.1158/0008-5472.CAN-04-3232. [DOI] [PubMed] [Google Scholar]
  • 12.Chen KJ, Lin SZ, Zhou L, Xie HY, Zhou WH, Taki-Eldin A, Zheng SS. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS One. 2011;6:e24671. doi: 10.1371/journal.pone.0024671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, Xu Y, Li YW, Tang ZY. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007;25:2586–2593. doi: 10.1200/JCO.2006.09.4565. [DOI] [PubMed] [Google Scholar]
  • 14.Ilkovitch D, Lopez DM. The liver is a site for tumor-induced myeloid-derived suppressor cell accumulation and immunosuppression. Cancer Res. 2009;69:5514–21. doi: 10.1158/0008-5472.CAN-08-4625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, Greten TF, Korangy F. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008;135:234–243. doi: 10.1053/j.gastro.2008.03.020. [DOI] [PubMed] [Google Scholar]
  • 16.Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, Lehner F, Manns MP, Greten TF, Korangy F. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50:799–807. doi: 10.1002/hep.23054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Arihara F, Mizukoshi E, Kitahara M, Takata Y, Arai K, Yamashita T, Nakamoto Y, Kaneko S. Increase in CD14+HLA-DR -/low myeloid-derived suppressor cells in hepatocellular carcinoma patients and its impact on prognosis. Cancer Immunol Immunother. 2013;62:1421–30. doi: 10.1007/s00262-013-1447-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Alias A, Ives A, Pathan AA, Navarrete CV, Williams R, Bertoletti A, Behboudi S. Analysis of CD4+ T-Cell responses to a novel alpha-fetoprotein-derived epitope in hepatocellular carcinoma patients. Clin Can Res. 2005;11:6686–6694. doi: 10.1158/1078-0432.CCR-05-0382. [DOI] [PubMed] [Google Scholar]
  • 19.Witkowski M, Spangenberg HC, Neumann-Haefelin C, Büttner N, Breous E, Kersting N, Drognitz O, Hopt UT, Blum HE, Semmo N, Thimme R. Lack of ex vivo peripheral and intrahepatic α-fetoprotein-specific CD4+ responses in hepatocellular carcinoma. Int J Cancer. 2011;129:2171–2182. doi: 10.1002/ijc.25866. [DOI] [PubMed] [Google Scholar]
  • 20.Chen Z, Shen S, Peng B, Tao J. Intratumoural GM-CSF microspheres and CTLA-4 blockade enhance the antitumour immunity induced by thermal ablation in a subcutaneous murine hepatoma model. Int J Hyperthermia. 2009;25:374–382. doi: 10.1080/02656730902976807. [DOI] [PubMed] [Google Scholar]
  • 21.Zabala M, Lasarte JJ, Perret C, Sola J, Berraondo P, Alfaro M, Larrea E, Prieto J, Kramer MG. Induction of immunosuppressive molecules and regulatory T cells counteracts the antitumor effect of interleukin-12-based gene therapy in a transgenic mouse model of liver cancer. J Hepatol. 2007;47(6):807–15. doi: 10.1016/j.jhep.2007.07.025. [DOI] [PubMed] [Google Scholar]
  • 22.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):1032–43. doi: 10.1172/JCI64742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Greten TF, Ormandy LA, Fikuart A, Höchst B, Henschen S, Hörning M, Manns MP, Korangy F. Low-dose cyclophosphamide treatment impairs regulatory T cells and unmasks AFP-specific CD4+ T-cell responses in patients with advanced HCC. J Immunother. 33:211–8. doi: 10.1097/CJI.0b013e3181bb499f. [DOI] [PubMed] [Google Scholar]
  • 24.Han Y, Chen Z, Yang Y, Jiang Z, Gu Y, Liu Y, Lin C, Pan Z, Yu Y, Jiang M, Zhou W, Cao X. 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;59:567–579. doi: 10.1002/hep.26694. [DOI] [PubMed] [Google Scholar]
  • 25.Wang BJ, Bao JJ, Wang JZ, Wang Y, Jiang M, Xing MY, Zhang WG, Qi JY, Roggendorf M, Lu MJ, Yang DL. Immunostaining of PD-1/PD-Ls in liver tissues of patients with hepatitis and hepatocellular carcinoma. World J Gatroenterol. 2011;17:3322–3329. doi: 10.3748/wjg.v17.i28.3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gao Q, Wang XY, Qiu SJ, Yamato I, Sho M, Nakajima Y, Zhou J, Li BZ, Shi YH, Xiao YS, Xu Y, Fan J. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Can Res. 2009;15:971–979. doi: 10.1158/1078-0432.CCR-08-1608. [DOI] [PubMed] [Google Scholar]
  • 27.Shi F, Shi M, Zeng Z, Qi RZ, Liu ZW, Zhang JY, Yang YP, Tien P, Wang FS. Int J PD-1 and PD-L1 upregulation promotes CD8(+) T-cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients. Cancer. 2011;128:887–896. doi: 10.1002/ijc.25397. [DOI] [PubMed] [Google Scholar]
  • 28.Zeng Z, Shi F, Zhou L, Zhang MN, Chen Y, Chang XJ, Lu YY, Bai WL, Qu JH, Wang CP, Wang H, Lou M, Wang FS, Lv JY, Yang YP. Upregulation of circulating PD-L1/PD-1 is associated with poor post-cryoablation prognosis in patients with HBV-related hepatocellular carcinoma. PLoS One. 2011;6:e23621. doi: 10.1371/journal.pone.0023621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kalathil S, Lugade AA, Miller A, Iyer R, Thanavala Y. 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;73:2435–44. doi: 10.1158/0008-5472.CAN-12-3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13:227–242. doi: 10.1038/nri3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Manzotti CN, Liu MK, Burke F, Dussably L, Zheng Y, Sansom DM. Integration of CD28 and CTLA-4 function results in differential responses of T cells to CD80 and CD86. Eur J Immnol. 2006;36:1413–1422. doi: 10.1002/eji.200535170. [DOI] [PubMed] [Google Scholar]
  • 32.Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan AD, Stroud JC, Bates DL, Guo L, Han A, Ziegler SF, Mathis D, Benoist C, Chen L, Rao A. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell. 2006;126:375–387. doi: 10.1016/j.cell.2006.05.042. [DOI] [PubMed] [Google Scholar]
  • 33.Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, Rudensky AY. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445:771–775. doi: 10.1038/nature05543. [DOI] [PubMed] [Google Scholar]
  • 34.Collins AV, Brodie DW, Gilbert RJ, Iaboni A, Manso-Sancho R, Walse B, Stuart DI, van der Merwe PA, Davis SJ. The interaction properties of costimulatory molecules revisited. Immunity. 2002;17:201–210. doi: 10.1016/s1074-7613(02)00362-x. [DOI] [PubMed] [Google Scholar]
  • 35.Schneider H, Downey J, Smith A, Zinselmeyer BH, Rush C, Brewer JM, Wei B, Hogg N, Garside P, Rudd CE. Reversal of the TCR stop signal by CTLA-4. Science. 2006;313:1972–1975. doi: 10.1126/science.1131078. [DOI] [PubMed] [Google Scholar]
  • 36.Grohmann U, Orabona C, Fallarino F, Vacca C, Calcinaro F, Falorni A, Candeloro P, Belladonna ML, Bianchi R, Fioretti MC, Puccetti P. CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat Immunol. 2002;3:1097–1101. doi: 10.1038/ni846. [DOI] [PubMed] [Google Scholar]
  • 37.Chen W, Jin W, Wahl SM. Engagement of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) induces transforming growth factor beta (TGF-beta) production by murine CD4(+) T cells. J Exp Med. 1998;188:1849–1857. doi: 10.1084/jem.188.10.1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tivol EA, Boyd SD, McKeon S, Borriello F, Nickerson P, Strom TB, Sharpe AH. CTLA-4-Ig prevents lymphoproliferation and fatal multiorgan tissue destruction in CTLA-4-deficient mice. J Immunol. 1997;158:5091–5094. [PubMed] [Google Scholar]
  • 39.Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, Nomura T, Sakaguchi S. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322:271–275. doi: 10.1126/science.1160062. [DOI] [PubMed] [Google Scholar]
  • 40.Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–1725. doi: 10.1084/jem.20082492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Robert C, Thomas L, Bondarenko I, O’Day S, MD JW, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, Davidson N, Richards J, Maio M, Hauschild A, Miller WH, Jr, Gascon P, Lotem M, Harmankaya K, Ibrahim R, Francis S, Chen TT, Humphrey R, Hoos A, Wolchok JD. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
  • 43.Calabro L, Morra A, Fonsatti E, Cutaia O, Amato G, Giannarelli D, Di Giacomo AM, Danielli R, Altomonte M, Mutti L, Maio M. Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial. Lancet Oncol. 2013;14:1104–11. doi: 10.1016/S1470-2045(13)70381-4. [DOI] [PubMed] [Google Scholar]
  • 44.Grosso JF, Jure-Kunker MN. CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immunity. 2013;13:5–19. [PMC free article] [PubMed] [Google Scholar]
  • 45.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R, Greenfield EA, Bourque K, Boussiotis VA, Carter LL, Carreno BM, Malenkovich N, Nishimura H, Okazaki T, Honjo T, Sharpe AH, Freeman GJ. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–268. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
  • 47.Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, Sharpe AH. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206:3015–3029. doi: 10.1084/jem.20090847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nikolova M, Lelievre JD, Carriere M, Bensussan A, Lévy Y. Regulatory T cells differentially modulate the maturation and apoptosis of human CD8+ T-cell subsets. Blood. 2009;113:4556–4565. doi: 10.1182/blood-2008-04-151407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–687. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
  • 50.Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. ProcNatlAcadSci U S A. 2001;98:13866–13871. doi: 10.1073/pnas.231486598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wei F, Zhong S, Ma Z, Kong H, Medvec A, Ahmed R, Freeman GJ, Krogsgaard M, Riley JL. Strength of PD-1 signaling differentially affects T-cell effector functions. ProcNatlAcadSci U S A. 2013;110:2480–2489. doi: 10.1073/pnas.1305394110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. ProcNatlAcadSci U S A. 2002;99:12293–12297. doi: 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8:467–477. doi: 10.1038/nri2326. [DOI] [PubMed] [Google Scholar]
  • 54.Iwai Y, Terawaki S, Ikegawa M, Okazaki T, Honjo T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J Exp Med. 2003;198:39–50. doi: 10.1084/jem.20022235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Maier H, Isogawa M, Freeman GJ, Chisari FV. PD-1:PD-L1 interactions contribute to the functional suppression of virus-specific CD8+ T lymphocytes in the liver. J Immunol. 2007;178:2714–2720. doi: 10.4049/jimmunol.178.5.2714. [DOI] [PubMed] [Google Scholar]
  • 56.Umemoto Y, Okano S, Matsumoto Y, Nakagawara H, Matono R, Yoshiya S, Yamashita YI, Yoshizumi T, Ikegami T, Soejima Y, Harada M, Aishima S, Oda Y, Shirabe K, Maehara Y. Prognostic impact of programmed cell death 1 ligand 1 expression in human leukocyte antigen class I-positive hepatocellular carcinoma after curative hepatectomy. J Gastroenterol. 2014 doi: 10.1007/s00535-014-0933-3. [epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 57.Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC, Lim SG, Bertoletti A. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gstroenterology. 2009;137:682–690. doi: 10.1053/j.gastro.2009.04.045. [DOI] [PubMed] [Google Scholar]
  • 59.Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A, Mateus C, Boasberg P, Tumeh PC, Chmielowski B, Ebbinghaus SW, Li XN, Kang SP, Ribas A. N Engl J Med. 2013;369:134–44. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I, Inzunza HD, Feely W, Horak CE, Hong Q, Korman AJ, Wigginton JM, Gupta A, Sznol M. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–133. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, Koren-Michowitz M, Shimoni A, Nagler A. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Can Res. 2008;14:3044–3051. doi: 10.1158/1078-0432.CCR-07-4079. [DOI] [PubMed] [Google Scholar]
  • 62.Chen Y, Huang Y, Reiberger T, Duyverman AM, Huang P, Samuel R, Hiddingh L, Roberge S, Koppel C, Lauwers GY, Zhu AX, Jain RK, Duda DG. 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. doi: 10.1002/hep.26790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Greten TF, Duffy AG, Korangy F. Hepatocellular carcinoma from an immunologic perspective. Clin Cancer Res. 2013;19:6678–85. doi: 10.1158/1078-0432.CCR-13-1721. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp TableS1
Supplementary Material

RESOURCES