Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Journal of Oncology Research and Treatment - Immunotherapy for Hepatocellular Carcinoma and Its New Development

Journal of Oncology Research and Treatment
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Review Article   
  • J Oncol Res Treat 2019, Vol 4(1): 134

Immunotherapy for Hepatocellular Carcinoma and Its New Development

Heping Li1, Ildiko Gyory2, Xiaoli Zhang3, Mingyan Xu3 and Shiqiu Xiong4*
1Department of Medical Oncology, First affiliated hospital, Sun Yat-sen University, Guangzhou, China
2Department of Molecular and Cell Biology, University of Leicester, UK
3HaploX Biotechnology Co. Ltd, Shenzhen, China
4LGC Limited, Fordham, Cambridgeshire, CB75WW, UK
*Corresponding Author: Shiqiu Xiong, Senior Investigator, LGC Ltd, Molecular Biology, Fordham, Cambridgeshire, CB75WW.UK, UK, Tel: 00447449965484, Email: xiongshiqiu2008@yahoo.com

Received: 18-Jan-2019 / Accepted Date: 11-Feb-2019 / Published Date: 18-Feb-2019

Abstract

Hepatocellular Carcinoma (HCC) is the most common primary liver cancer with high mortality. Because of the limitation on the conventional therapeutic options and immunological characters of this disease, variable immunotherapy strategies for HCC have been designed and investigated. A few of them look promising. In this review, we comprehensively summarized recent and novel immunotherapies for HCC, with a particular focus on clinical trials on the basis of their physiological rationales for these strategies. Insights into these clinical progress and their ideologies are essential to improve efficacy and clinical outcome of these immunotherapies

Keywords: Hepatocellular carcinoma; Immunotherapy; liver tolerance; Clinical trial

Core Tip

This review article aims to connect the physiological characters of liver immunology with the rationales of HCC immunotherapies, provide the updated information about recent clinical progress, and summarize the advantage and disadvantage of each regiment, new vision on the potential research future of immunotherapies.

Introduction

Hepatocellular Carcinoma (HCC) is the most common histologic type of primary liver cancer, accounting for between 85% and 90% of liver cancers. HCC is one of the cancers with highest morbidity worldwide. There are about 750,000 new cases every year, accounting for 5.7% of all new cancer cases, 85% of these new cases reported in sub-Saharan Africa and East Asia. This geographic distribution of HCC overlaps with that of chronic viral hepatitis, according to epidemiological data [1]. It is widely accepted that HCC is developed from viral hepatitis B and C, and subsequent stage liver cirrhosis. The other less frequent causes are chronic exposure to toxins or hereditary liver diseases, such as non-alcoholic fatty liver disease [2].

HCC is the fifth most common malignant tumour, but in mortality it is ranked as third world-wide. Although there are great advances in medical diagnosis technology and surveillance programs for highrisk populations, only 20% of HCC patient can be diagnosed in phase I, and only these patients are amenable to curative therapy (liver transplantation, surgical resection or ablative therapies); most HCC patients with liver dysfunction, are not feasible to these aggressive treatments. Therefore, the only reliable therapeutic option is Sorafenib which yielded a median overall survival of 6.5-10.7 months [3]. Furthermore, HCC tumour recurrence happens in 50%-80% of patients after 5 years’ treatment [4]. A significant unmet medical need remains.

The challenge of immunotherapy in HCC

There is a growing evidence to suggest that HCC may be considered an immunogenic tumour, arising from chronic inflammation in an immunosuppressive environment.

The strong association between spontaneous regression of this disease and systemic inflammatory response sparked greater interest in the immunological pathogenesis in HCC [5]. Further reports on particular types of immunological cells, which associated with favourable prognosis of HCC, introduced novel immunotherapy concepts to the medical management of patients with HCC [6]. The characters of immunotherapy elicit non-toxic, systemic, memorial and specific anti-tumour activity makes it particularly promising in the setting of HCC.

The immunosuppressive microenvironment is an important factor that contributes to tumour initiation, progression and their therapeutic resistance. Immunological cells and molecules are fundamental components of the tumour microenvironment. All biological aspects of HCC are shaped by the immune system.

The rationale of immunotherapy for tumours is to counteract the immunosuppressive mechanisms and/or amplify tumour-specific and memory immunity, which is expected to improve clinical outcomes profoundly by causing long-lasting regression and preventing relapse in cancer patients [7].

Although HCC is assumed as a good disease model for immunotherapy, most current clinical trials on this disease achieved only marginally positive response, which attribute to the inherently tolerogenic character of the liver in both healthy and diseased states [8]. Understanding the characteristics of the immune system in HCC setting is crucial to improve future immunotherapy strategies.

Because of the vast array and constant flux of blood-derived antigens and bacterial molecules from the intestinal microbiota, human liver has evolved to respond with a plethora of immune-regulatory mechanisms to exhibit a distinctive form of immune privilege or tolerance, best exemplified by orthotopic liver transplantation resulting in systemic, donor-specific T-cell tolerance [9].

The liver’s pathway to immune tolerance is multifaceted. Structurally, the liver consists of repetitive hepatic lobules that are perfused by sinusoidal vessels. Hepatocytes are shielded from direct interaction with the bloodstream by densely populated nonparenchymal liver cells: including Liver Sinusoidal Endothelial Cells (LSECs); stellate cells; dendritic cells; Kupffer Cells (KC); natural killer cells; natural killer T cells and others. These non-parenchymal cells serve as a protective barrier between the hepatocytes and pathogens [10].

To protect the liver from autoimmune damage that could result from the continuous stimulation by intestinal antigens, effector T cells in the liver are kept in an induced state of tolerance; Kupffer Cells (KC) constitutively release inhibitory cytokines, such as Interleukin 10 (IL-10) and Transforming Growth Factor beta (TGF-β); hepatocytes, Hepatic Stellate Cells (HSC), KC, LSEC and intrahepatic leukocytes (LC) upregulate immune checkpoints, involving programmed death-ligand 1 or 2 (PD-L1 or PD-L2); regulatory T cells (Treg) highly express Cytotoxic T-lymphocyte Associated Protein 4 (CTLA-4). All these factors directly or indirectly suppress the immune effector cells CTL and NK (Natural Killer) cells [11]. The liver-specific physiological immune tolerance is beneficial in case of harmless antigens, however, it is potentially detrimental under hepatitis settings, and the chronic inflammation amplifies this immune suppression. Without proper immune surveillance, the initial lesions of HCC, which are induced by viral and immunologic factors, escape immune clearance and develop into solid tumours.

The summarized main components of immunological aetiology in HCC are: immunosuppressive tumour environment; negative regulation of PD-1/PDL1 pathway; failure of Tumour Associate Antigen (TAA) processing and presentation; suppression of CD4+ and CD8+ T cells by Treg; insufficient help of CD4+ T cells; direct suppression on effector cells, such as CTL, NK, NKT. Hence, the rationale of immunotherapy in HCC is to interfere these immunosuppressive pathways and recover their immune potency (Figure 1).

advances-oncology-research-treatments

Figure 1: Main factors involve in immunotherapy for HCC.

Antigen-presenting cells (APC, such as DC, Kuffers, macrophage, B cells etc.) capture tumour antigens, process and present them, along with cytokines (including IL-12, IL-15 etc.), to effector CTL cells, to help them maturation and activation. Meanwhile, these APC also convey CTLA-4, PD-1 mediated inhibitory signals to balance CTL activation; activated CTL cells kill target tumour cells directly, or secret cytokine to amplify the anti-tumour response; APC also help NK cells maturation and function by secreted IL-12, IL-15; activated NK cells kill target tumour cells in a similar way of CTL.

On the basis of this rationale, HCC immune therapies were classified into 4 main regimens:

a. Blockade of T cells response checkpoint molecules PD-1and CTLA-4;

b. To perform the adoptive transfer with anti-tumour effector immune cells, such as CAR-T, CTL and NK [12];

c. To generally counteract immunosuppressive mechanisms by administrating cytokines, such as IFN, IL-12, IL-15 etc.;

d. Boosting antigen presentation by tumour antigen-pulsed DC cells;

We are trying to summarize the promising and recent immunotherapy for HCC respectively, mainly focus on clinical trials. Limited to our knowledge, we sincerely apologize to those colleagues whose important works have been missed in this report.

Immune checkpoint blockade

The field of cancer immunotherapy is undergoing a renaissance due to a deeper understanding of the immune system and immunosurveillance. The balance between co-stimulatory signals and immune checkpoints determines T cell activation and the overall intensity of the immune response. Whenever CD80/CD86 from APC cells recognized by CD28 on effector T cells, they deliver co-stimulatory signals to synergize first activation signalling from TCR recognizing antigen/ MHC (Major Histocompatibility Complex) complex; meanwhile, to control the intensity and remission of immune response tightly, T cells also present inhibitory receptors, such as CTLA-4 and PD-1. CTLA- 4 compete CD28 binding to CD80/CD86 and thereby inhibits T-cell activation [13]. PD-1 receptors are expressed on several immune cells, including effector T and NK cells, which associate with PD-L1 from APC or tumour cells. Engaged PD-1 receptors deliver pro-apoptotic signalling to effector immune cells. After astonishing success of clinical trials on CTLA-4 and PD-L1 blockade to treat melanoma patients [14- 16], similar positive results are highly anticipated in HCC patients [17].

In a recent pilot clinical trial, 37 patients with HCC or HCV infection were injected with 15 mg/kg of the CTLA-4-blocking antibody tremelimumab for 90 days, to test the anti-tumour and antiviral effect of this antibody. In addition to demonstrating the safety of such approach in patients, a 17.6% partial response rate and a 76.4% disease control rate was achieved. A significant drop in viral load was also observed [18]. Another phase I clinical trial of tremelimumab with percutaneous Radiofrequency Ablation (RFA) or trans-arterial therapy is ongoing (NCT01853618) [19].

The anti-PD-1 antibody based therapy is even more promising than a CTLA-4 blockade. Nivolumab was the first immune drug approved as a second-line treatment for patients with HCC by FDA in September 2017. In the CheckMate040 study, 48 patients with HCC had received nivolumab 0.1-10 mg/kg intravenously for 2 years. The Objective Remission Rate (ORR) was 15%; median Overall Survival (OS) was 15.1 months and the 9 months OS rate was 67% [20]. In a dose-expansion study of nivolumab, 174 patients with HCC received nivolumab 3 mg/kg intravenously. The ORR was 20% and tumor burden was reduced In 68 patients (39%) [21]. Besides, in 80 HCC patients treated with nivolumab only, the ORR was 23%; Disease Control Rate (DCR) was 63%; and 40% of patients had stable disease over than 6 months. However, in the group of sorafenib-experienced patients that received nivolumab, 91% (166/182) of patients progressed on sorafenib treatment. The ORR was 16%-19% [22]. Most recently, the efficacy and safety of anti-PD1 therapy of advanced HCC was evaluated on 11 cases, no related adverse effects were noted; the disease control rate reached 81.8% comparing to an objective response of 63.6%, this clinical trial proved promising application of PD-1 in HCC [23]. There are more clinical trials on other immune checkpoint blockade (Figure 2). Greten summarised clinical data of the therapies based on PD-1/PD-L1 blockade under study for the treatment of hepatocellular carcinoma [24].

advances-oncology-research-treatments

Figure 2: Molecular rationale of immune checkpoint blockade.

Indeed, the combination of PD-1 and CTLA-4 blockade resulted in an objective response with limited toxicity in 40% of the patients [25]. In a recent clinical trial, 40 HCC patients received combination treatment with the anti-PD-L1 antibody durvalumab and the anti- CTLA-4 antibody tremelimumab to evaluate the efficacy. The ORR was 17.5, DCR at 16 weeks was 57.5%. In the 20 HCC patients without HCV infection, DCR at 16 weeks was 70% [26,27].

In immune checkpoint therapy, effective biomarkers could be used to assess efficacy and predict prognosis to predict survival benefits of patients with HCC. In the tumour microenvironment of HCC, PDL1 is overexpressed, and this associates with CD8+T cell immune responses and tumour aggressiveness [28-32]. In recent studies, PDL1 expression was associated with the therapeutic response of immune checkpoint therapy of HCC and postoperative recurrence [30]. Tumor Mutation Burden (TMB) is an emerging biomarker of sensitivity to immune checkpoint inhibitors, which estimated the total number of mutations in the tumor genome coding region. Tumor cells with high-level TMB are more likely to be identified by the immune system. The efficacy of anti-PD-1 antibody in lung cancer with high TMB was significantly better than that with low TMB, and the PFS was four times [33]. A study of the relationship between TMB and ORR had collected the data in 27 types of cancer. It indicated that there was a positive correlation between TMB and ORR for anti–PD-1 or anti–PDL1 therapy. The ORR was near 20% for HCC on the basis of a median tumor mutational burden of <10 mutations/MB [34]. The application of TMB as a biomarker for immunotherapy in HCC also requires more clinical researches.

The evaluation of efficacy and potency of anti-checkpoint inhibitors in HCC is still in phase I/II trial. Large randomized phase III trials need to be designed and conducted to confirm the promising efficacy.

Given these evolutionary progress in tumour therapy, recent nobel prize rewarding the pioneers in this field is expected, which in turn will heat up these immunotherapy works. However, the resiliency and complexity of immune system and tumor cells may disappoint most scientists in the near future. Combination these agents with other immune regulators or complimentary drugs will shed light on the future in this field.

Antigen Presenting Cells (APC) capture tumour antigens, process and present them to T cells by recognizing TCR, which induced first activation signal. To be fully activated, T cells need co-stimulating signal mediated by CD28 binding with CD80/CD86. CTLA-4 competes CD28 binding to CD80/CD86 and thereby regulate intensity and remission of T-cell activation delicately. Activated T cells recognize and kill target tumour cells by TCR binding peptide/MHC specifically, this process is attenuated by PD-1/PDL-1 signalling. Specific Abs against CTLA-4 or PD-1 can terminate negative signals for T cells, therefore augment T cells mediated immunity.

Adoptive cell transfer

These immune cells are the immune effectors counteracting the tumour cell-mediated negative effects directly, the above-mentioned therapeutic approaches are all indirect and mediated by these ultimate cell types. The suppression of immune effector cells in the liver under physiologic and pathogenic conditions is well-documented. Both regulatory T cells [35-37] and γδ+ T cells [38,39] have been shown to mediate this suppression recently. Adoptive transfer of activated effector cells in vitro may hurdle this problem and trigger host immune response against tumour cells directly.

CAR-T

Chimeric Antigen Receptor T-Cell Immunotherapy (CAR-T) is the new and promising cell immunotherapy by combination of engineering T cells and Chimeric Antigen Receptor (CAR) which redirects T cell specificity and function [40]. CAR is composed of an extracellular antigen recognition, an extracellular spacer/hinge region, a trans-membrane domain, and an intracellular signaling domain. And the extracellular antigen recognition can identify TAAs which are not restricted via HLA. Thus, CAR-T can overcome HLA-mediated tumor escape mechanisms [41]. CAR-T immunotherapy has been a great success in haematological malignancies [42]. But the application to HCC is still in research stage. Glypican-3 (GPC3) is a potential target for CAR-T immunotherapy against HCC. GPC3 is a cell surface oncofetal proteoglycan, which was overexpressed in 75% of HCC [43]. Previous studies showed that anti-GPC3-CAR T cells could against HCC cell lines in vivo [44,45], and it efficiently suppressed tumor growth in the tumor cell in which GPC3 proteins were low expressed and impressively eradicated tumor cells in which GPC3 proteins were overexpressed [46]. Currently, 13 clinical trials using CAR-T immunotherapy are under way. In a recent clinical trial, 13 patients with refractory or relapsed HCC treated with GPC3 CAR-T. All 13 patients tolerated the treatment well. In the five patients who could evaluate the efficacy, 1 was partial response, 2 were stable disease and 2 were progressive disease. And 4 of them are still alive; the median OS was longer than 14 months [47].

The lack of specific antigens in HCC causes the CAR-T cells attack the normal tissues which express the same antigens. It leads to severe toxicity [48]. Dual-targeted CAR-T cells have been proposed as a potential way to reduce on-target, off-tumor toxicity. Chen et al. developed dual-targeted CAR-T cells, which co-express GPC3 and Asialoglycoprotein Receptor 1 (ASGR1). They found that the dualtargeted CAR-T cells might reduce the risk of on-target, off-tumor toxicity while maintaining relatively potent anti-tumor activities on GPC3+ASGR1+ HCC [49]. More clinical trials are required as CAR-T based immunotherapy is a new strategy for HCC treatment.

CTL

Adoptive immunotherapy of HCC is often hampered by the low frequency of tumour-specific T cells, therefore screening and choosing proper TAAs to boost T cells in vitro is critical to the success of CTLbased strategies [50]. Hiroishi and colleagues identified 3 HCC TAAs, namely, glypican-3, NY-ESO-1, and MAGE-1 in their screen. They measured the magnitude of TAA -specific CD8+ T-cell responses against these antigens in 20 HCC patients treated by radiofrequency ablation or Trans-catheter Chemo-embolization (TACE), and found that these responses represented the only significant prognostic factor to indicate a prolonged tumor-free Interval [51]. The responses to these antigens were compared and described in detail in subsequent publications [52]. These investigations highlighted the importance of CTL in HCC immunotherapy. Combining sunitinib, a receptor tyrosine kinase inhibitor, with adoptive transfer of tumour antigen-specific CD8+ T cells led to the elimination of established tumours without recurrence in an orthotopic murine tumour model, highlighting the potential of a synergistic chemo-immunotherapeutic approach to HCC [53]. Qasim W and colleagues designed an experiment on one patient who had undergone liver transplantation for HBV-related HCC. Genetically modified autologous T cells that express an HBsAgspecific T cell receptor was introduced to this patient, with the rationale of HBV-DNA integration frequently occurring in HBV-related hepatocellular carcinoma. These T cells have been shown to mediate a reduction in HBsAg levels without causing an exacerbation of liver inflammation or other toxicity. This work marked the path to a novel adoptive immunotherapy in hepatitis B-associated HCC [54].

NK

Apart from CTL, NK cells are the main effector immune cells, which can eliminate tumours or virus infected cells without specific antigen priming. NK cell activation depends on the signals from a repertoire of activating and inhibitory receptors, the integration of these distinct signals dictates the quality and the intensity of the NK cell response [55]. 25%-40% of liver lymphocytes are NK cells, this proportion is significantly higher than their 5% cellular abundance in peripheral blood [56]. This enrichment of liver NK cells indicates their potential clinical applications.

The therapeutic potential of NK cells has been most extensively explored in haematological malignancies, because NK cells’ killing requires one-to-one target engagement and site-directed release of cytolytic granules. Nevertheless, an increasing number of studies already indicated that NK cell based immunotherapy is a promising therapeutic approach for HCC, Tsuchiyama and colleagues have shown that HSV thymidine kinase and MCP-1 transduced HCC cells can be eliminated by NK cells in a mouse model [57]. In a study of 37 HCC patients undergoing radiofrequency thermal ablation, their peripheral blood NK cells showed a more differentiated phenotype profile with a general increase of pro-activatory functional activities [58]. These may come from their ability to precisely kill cancer stem cells, antibody coated cells, and stressed cells specifically, which makes them appealing therapeutic effectors for all cancer forms, including metastases. Meanwhile activated NK cells produce pro-inflammatory cytokines, which can reverse the anti-inflammatory tumour microenvironment and augment adaptive immune responses [59,60].

In vivo and in vitro studies have been shown the involvement of TLR3 to contribute to the efficacy of NK cell mediated eradication of HCC, although the presence of ligands for this innate receptor in tumour cells has not been demonstrated [61]. Another report, which showed apoptosis of stressed liver cells by TNF-related Apoptosisinducing Ligand (TRAIL) that was induced by activated NK cells. This interaction is mediated by the receptor DR5 which is expressed on these target cells at higher levels [62]. Apart from DR5, UL16-binding Protein (ULBP) 1expression level, which was regulated by proteasome, correlated well with HCC progression and early recurrence, suggested that signalling from NKG2D (ULBP1 receptor) in NK cells played a crucial role in immune response to HCC [63]. Another NKG2D ligand, MICA was proved to be a key molecule in tumour immune surveillance in the setting of HCC [64]. Intriguingly, a novel B7 family member B7-H6, the ligands of another NK activating receptor NKP30, proved to express highly on hepatocellular carcinoma samples [65]. IL-37 is a member of the interleukin 1 cytokine family. A new report implied that their expression level associated with the prognosis of HCC patients via stimulating the migration of the activated memory CD57+ NK cells [66].

In a recent clinical trial of 62 HCC patients, Injection of IL-2 and CD16 antibody induced NK cells helped to prevent the recurrence of HCC after radiofrequency ablation treatment [67]. Human allogeneic suicide gene-modified killer cells (aSGMKCs) exhibit a high, rapid, interleukin-2–dependent, and non–major histocompatibility complex class I-restricted in vitro and in vivo cytotoxicity to HCC. This, mainly NK and NK-like T cell mediated strategy opened a new perspective for immunotherapy of HCC [68]. Another promising NK cell mediated immunotherapy is the combination of adoptive transfer of NK cells with pharmacological STAT3 inhibitors as shown by two new reports demonstrating synergistic anti-tumour effects [69,70].

Moreover, two ongoing clinical trials are designed to assess NK cell therapy combined with liver resection (NCT02008929) or liver transplantation for HCC (NCT01147380).

Cytokine-induced killer cells

Cytokine-induced Killer (CIK) cells are mixed T and NK cell, which generated from human Peripheral Blood Mononuclear Cells (PBMC) activated with IFN-gamma, anti-CD3 antibody and IL-2. In murine tumour models, these CIK cells demonstrated significant antitumor effects against a number of hematopoietic and solid tumours, including HCC. The safety and antitumor effect of CIK cell-based therapy for patients also has been proved, it induced minimal toxicity and no graft-vs-host disease [71, 72]. In a recent multi-centre, randomized, open-label, phase III trial, IL-2 and CD3 antibody-induced killer (CIK) cells were injected into 230 HCC patients after conventional treatment. This immunotherapy resulted in increased recurrence-free (44.0 months vs 30.0 months, p=0.01) and overall survival [73]. Recent meta-analysis showed that immunotherapy based on CIK cells induces a significant increase in both Overall Survival (OS) and Progressionfree Survival (PFS) of HCC patients [74,75]. PD-L1 expression could be as a predictive biomarker for CIK cell immunotherapy in patients with HCC. Previous studies showed that patients who treated with surgery and CIK with 5% PD-L1 expression had better OS and Recurrence-free Survival (RFS) than patients with <5% PD-L1 expression. However, PD-L1 expression did not show a direct impact on the survival of patients in the surgery alone group. High number of PD-1 positive intra-tumoral lymphocytes also could predict survival benefit of CIK cells for HCC patients [76].

Because of the small HCC patient numbers in above clinical trials, further validation of the efficacy of CIK immunotherapy for HCC is needed in the near future, by increasing the number of evaluating HCC patients.

Interferon

Interferons (IFN) are cytokines, named after their ability to “interfere” with viral replication and thereby protecting cells from viral infections. They are widely applied in clinical anti-viral therapy, including HBV and HCV infections. It is proven that IFNs also have various immune regulatory functions, such as activation of natural killer cells and macrophages, up-regulation of antigen presentation by increasing the expression of Major Histocompatibility Complex (MHC) [77].

In a clinical trial enrolled a large cohort of post-operative HCC patients (236 patients in total), IFN-α treatment had been applied in long-term, randomized and observation-controlled settings. In these patients, Sun and his colleagues reported significant beneficial effect of IFN-α treatment, represented as improved overall survival (63.8 vs. 38.8, p=0.0003) and longer cancer-free survival period (31.2 vs. 17.7 months) [78]. However, other clinical trials did not confirm this significant improvement [79-83]. Most of these clinical trials concluded with just a marginal beneficial effect of IFN-α or a combination of IFN-α and IFN2β on overall survival and/or partial response, at the expense of severe side effects such as flu-like symptoms (chills, fever, headache, myalgia, malaise), myelosuppression, depression, and hepatotoxicity. Interestingly, a recent clinical trial applying PEGIFN а-2β/ribavirin (RBV) to a cohort of 54 randomized patients with HCV-associated Stage I/II HCC showed very promising outcome. The 3 year overall survival rate in the experimental group was 90.2% contrast to 61.2% in the control group [84]. Because systemic immune-stimulation by IFN is usually bad tolerated, cytokine treatment in combination with other antiviral drugs or immunotherapy could be a future direction.

IL-12

The other cytokine, besides interferons, which has been clinically investigated is IL-12. Produced by DCs and macrophages, this cytokine activates immune effector cells, such as CTLs and NKs. To avoid the severe side effects that were previously reported in mouse models, Sangro B and his colleagues injected an adenoviral vector encoding IL-12 directly into gastrointestinal carcinomas, including primary liver tumours. This vector introduced the exogenous Il-12 gene directly to the autologous DC cells of the patients. Adenoviral vector delivered Il-12 alone exerts only mild antitumor effects; this approach elicited only partial response in only 1 patient out of 9 [85,86]. Kayashima H and colleagues achieved more promising results using combined IL- 12 gene and dendritic cell therapy in a mouse model, however, this combination is yet to be introduced to patients [87]. Although it’s direct boost immune effector cells, the strong side effect of IL-12 limited its application in HCC.

IL-15

IL-15 is a cytokine produced by monocytes, macrophages and DCs. Il-15 is firstly recognized by IL-15Rα and upon association of the ligand with this receptor subunit, it is trans-presented to the cells expressing the β and γ subunits of the IL-15R. IL-15 regulates development, proliferation, maintenance and activation of T and NK cells [88]. IL-15 is widely anticipated to be the most promising therapeutic cytokine in HCC on the basis of its safety [89]. In rhesus macaques, sub-optimal IL-15 administration was associated with increased numbers of circulating NK and CD8+ central and effector-memory T cells [90]. In a recent clinical trial, IL-15 has been safely administered to patients with metastatic malignancy. This cytokine markedly altered the homeostasis of the patients’ peripheral blood lymphocyte subsets, with NK cells and γδ+ T cells most dramatically affected, followed by CD8+ memory T cells. Five out of eight patients showed a 10% to 30% volume-decrease in their marker lesions, while two of them experienced clearing of the lesion [91]. Scientists are quite optimistic in IL-15 for HCC immunotherapy.

Tumour antigen pulsed DC vaccine

DCs are the most potent antigen-presenting cells, with the capacity to take up, process, and present tumour antigens to T cells and stimulate an immune response, thus providing a rational platform for vaccine development. Failure of Tumour Associate Antigen (TAA) processing and presentation by DCs is one of the main pathogenic factors in HCC [92,93], rendering the development of tumour antigen-pulsed DC vaccines to be a good immunotherapeutic strategy [94].

On an autochthonous hepatocellular carcinoma mouse model, a plasmid, expressing Alpha-foetoprotein (AFP), induced AFP-specific response and this led to a significant (65%) reduction in tumour size [95]. In a pilot phase I clinical trial, Butterfield and colleagues showed that 4 immuno-dominant peptides, derived from alpha-fetoprotein, along with incomplete Freund’s adjuvant induced specific T-cell responses in 6 HCC patients [96]. Furthermore, these peptide-pulsed DC cells induced significant antigen-specific T cell responses in a follow-up phase I/II trial in hepatocellular carcinoma patients [97].

Encouraged by these results, a series of clinical trials on HCC patients with tumour antigen-pulsed DC vaccines were carried out. Of 31 patients with advanced HCC, autologous tumour lysate-pulsed dendritic cells have increased the 1-year survival rates readily (63.3% against 10.7%; P < 0.001) [98]. In the follow-up phase II clinical study, thirty-five patients with advanced HCC were vaccinated intravenously with mature autologous Dendritic Cells (DCs) pulsed ex vivo with a liver tumour cell line lysate (HepG2). In this study, 28% of the disease control rate was observed without obvious toxicity and autoimmunity [99]. To augment the interaction between DCs and antigen-specific T cells, Gonzalez et al. transduced TAA-pulsed DCs with a CD40 L-encoding adenovirus, which increased the stimulatory capacity of DCs significantly. Intra-tumoral injection of DCs activated both acquired and innate immunity in mouse models, elicited complete regression of established tumours and long-term immunity was established against tumour recurrence [100]. DCs from HCC patients have been proven to strongly inhibit liver NK and NKT cells via an NK activating receptor NKP30 to boost these effector cells, α1, 3-galactosyl epitope-pulsed dendritic cells were injected into 9 HCC patients in a new clinical trial. Comparing to the 9 patients in the control group, this therapy significantly prolonged the survival of treated patients (17.1 ± 2.01 month vs 10.1 ± 4.5 month, p=0.00121) [101]. A new phase I clinical trial to evaluate the safety of an allogeneic dendritic cell vaccine COMBIG-DC in HCC patients is now recruiting participants (NCT01974661).

Because of the potent antigen-presenting capacity and feasibility of cells culture and expansion of autologous DCs, DC vaccines are one the most promising candidates for HCC immunotherapy. Frustratingly, current clinical trials haven’t demonstrated expected therapeutic efficacy. Since the efficacy of DC-vaccines critically depends on techniques for the priming antigen loading onto DCs efficiently and their ability to migrate to the draining Lymph Nodes (LNs). Therefore, improving DC-vaccines homing to draining LNs and increasing the antigen-loading efficacy appears as critical next steps in this field [102].

Future Perspectives

(1) CAR-T and checkpoint blockage antibodies are stepping into the centre of immunotherapy stage, both strategies gained great success in treating other tumours. There are good reasons to keep optimistic of their efficacy in HCC.

(2) Adoptive transfer in vitro modified TIL, NK or DC cells showed efficacy in several cases. They still have a lot to learn from CAR-T before they reach clinical satisfaction.

(3) Administrating cytokines in management HCC is still hugely challenging. Combination with chemotherapy or other therapy may be their future.

(4) The combination of immunotherapy with local ablative therapies such as RFA or cryoablation, or the combination of two different immunotherapy modalities seems to be particularly promising approaches.

(5) Development of additional animal models that reflect characteristics of different HCC subtypes would be necessary; magnificent genetic information from NGS helps better understanding and improving immunotherapy of HCC; Investigation of biology-driven biomarkers in response to different treatment strategies will help to follow treatment efficacy.

Acknowledgement

This work was sponsored by Nature Science Foundation of China (No. 81602701), Nature Science Foundation of Guangdong Province (No. 2017A030313547), and Nature Science Foundation of Guangdong Province (No. 2018A030313176).

References

  1. Lafaro KJ, Demirjian AN, Pawlik TM (2015) Epidemiology of hepatocellular carcinoma. Surg Oncol Clin N Am 24: 1.
  2. Breous E, Thimme R (2011) Potential of immunotherapy for hepatocellular carcinoma. J Hepatol 54: 830.
  3. Siegel AB, Olsen SK, Magun A, Brown RS, Jr. Sorafenib (2010) where do we go from here? Hepatology 52: 360.
  4. Osaki Y, Nishikawa H (2015) Treatment for hepatocellular carcinoma in Japan over the last three decades: Our experience and published work review. Hepatol Res 45: 59.
  5. Makarova-Rusher OV, Medina-Echeverz J, Duffy AG, Greten TF (2015) The yin and yang of evasion and immune activation in HCC. J Hepatol.
  6. Greten TF, Wang XW, Korangy F (2015) Current concepts of immune based treatments for patients with HCC: from basic science to novel treatment approaches. Gut 64: 842.
  7. Joyce JA, Fearon DT (2015) T cell exclusion, immune privilege, and the tumor microenvironment. Science 348: 74.
  8. Knolle PA, Thimme R (2014) Hepatic immune regulation and its involvement in viral hepatitis infection. Gastroenterology 146: 1193.
  9. Adams DH, Sanchez-Fueyo A, Samuel D (2015) From immunosuppression to tolerance. J Hepatol 62: S170.
  10. Jenne CN, Kubes P (2013) Immune surveillance by the liver. Nat Immunol 14: 996.
  11. Crispe IN, Giannandrea M, Klein I, John B, Sampson B, et al. (2006) Cellular and molecular mechanisms of liver tolerance. Immunol Rev 213: 101.
  12. Sharma P, Allison JP (2015) The future of immune checkpoint therapy. Science 348: 56.
  13. Schneider H, Downey J, Smith A, Zinselmeyer BH, Rush C, et al. (2006) Reversal of the TCR stop signal by CTLA-4. Science 313: 1972.
  14. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363: 711.
  15. Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, et al. (2011) Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364: 2517.
  16. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, et al. (2013) Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369: 134.
  17. Sharma P, Allison JP (2015) Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161: 205.
  18. Sangro B, Gomez-Martin C, de la Mata M, Inarrairaegui M, Garralda E, et al. (2013) A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol 59: 81.
  19. Hato T, Goyal L, Greten TF, Duda DG, Zhu AX (2014) Immune checkpoint blockade in hepatocellular carcinoma: current progress and future directions. Hepatology 60: 1776.
  20. Sangro B, Melero I, Yau TC, Hsu C, Kudo M, et al. Safety and antitumor activity of nivolumab (nivo) in patients (pts) with advanced hepatocellular carcinoma (HCC): Interim analysis of dose-expansion cohorts from the phase 1/2 CheckMate-040 study. J Clin Oncol 34.
  21. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, et al. (2017) Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 389: 2492.
  22. Crocenzi TS, El-Khoueiry AB, Yau TC, Melero I, Sangro B, et al. (2017) Nivolumab (nivo) in sorafenib (sor)-naive and -experienced pts with advanced hepatocellular carcinoma (HCC): CheckMate 040 study. J Clin Oncol 35.
  23. Feng D, Hui X, Shi-Chun L, Yan-Hua B, Li C, et al. (2017) Initial experience of anti-PD1 therapy with nivolumab in advanced hepatocellular carcinoma. Oncotarget 8: 96649.
  24. Greten TF, Sangro B (2017) Targets for immunotherapy of liver cancer. J Hepatol.
  25. Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, et al. (2008) Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res 14: 3044.
  26. Abou-Alfa GK, Sangro B, Morse M, Zhu AX, Kim RD, et al. (2016) Phase 1/2 study of durvalumab and tremelimumab as monotherapy and in combination in patients with unresectable hepatocellular carcinoma (HCC). J Clin Oncol 34.
  27. Kelley RK, Abou-Alfa GK, Bendel JC, Kim TY, Borad MJ, et al. (2017) Phase I/II study of durvalumab and tremelimumab in patients with unresectable hepatocellular carcinoma (HCC): Phase I safety and efficacy analyses. J Clin Oncol 35.
  28. Gao Q, Wang XY, Qiu SJ, Yamato I, Sho M, et al. (2009) Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res 15: 971.
  29. Umemoto Y, Okano S, Matsumoto Y, Nakagawara H, Matono R, et al. (2015) Prognostic impact of programmed cell death 1 ligand 1 expression in human leukocyte antigen class I-positive hepatocellular carcinoma after curative hepatectomy. J Gastroenterol 50: 65.
  30. Han Y, Chen Z, Yang Y, Jiang Z, Gu Y, et al. (2014) 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 59: 567.
  31. Calderaro J, Rousseau B, Amaddeo G, Mercey M, Charpy C, et al. (2016) PD-L1 expression in hepatocellular carcinoma: relationship with clinical and pathological features. Hepatology 64: 624a.
  32. Huang CY, Wang Y, Luo GY, Han F, Li YQ, et al. (2017) Relationship Between PD-L1 Expression and CD8(+) T-cell Immune Responses in Hepatocellular Carcinoma. J Immunother 40: 323.
  33. Davis AA, Chae YK, Agte S, Pan A, Iams WT, et al. (2017) Association of circulating tumor DNA (ctDNA) tumor mutational burden (TMB) with DNA repair mutations and response to anti-AD 1/PD-L1 therapy in non-small cell lung cancer (NSCLC). J Clin Oncol 35.
  34. Yarchoan M, Hopkins A, Jaffee EM (2017) Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N Engl J Med 377: 2500.
  35. Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Kruger C, et al. (2008) A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology 135: 234.
  36. Kalathil S, Lugade AA, Miller A, Iyer R, Thanavala Y (2013) 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 73:2435.
  37. Pedroza-Gonzalez A, Verhoef C, Ijzermans JN, Peppelenbosch MP, Kwekkeboom J, et al. (2013) Activated tumor-infiltrating CD4+ regulatory T cells restrain antitumor immunity in patients with primary or metastatic liver cancer. Hepatology 57: 183.
  38. Yi Y, He HW, Wang JX, Cai XY, Li YW, et al. (2013) The functional impairment of HCC-infiltrating gammadelta T cells, partially mediated by regulatory T cells in a TGFbeta- and IL-10-dependent manner. J Hepatol 58: 977.
  39. Ma S, Cheng Q, Cai Y, Gong H, Wu Y, et al. (2014) IL-17A produced by gammadelta T cells promotes tumor growth in hepatocellular carcinoma. Cancer Res 74: 1969.
  40. Curran KJ, Pegram HJ, Brentjens RJ (2012) Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. J Gene Med 14:405.
  41. Srivastava S, Riddell SR (2015) Engineering CAR-T cells: Design concepts. Trends Immunol 36: 494
  42. Dotti G (2016) CAR-T cells for hematological malignancies: exploring alternative targets. Human Gene Therapy 27: A14.
  43. Dargel C, Bassani-Sternberg M, Hasreiter J, Zani F, Bockmann JH, et al. (2015) T Cells Engineered to Express a T-Cell Receptor Specific for Glypican-3 to Recognize and Kill Hepatoma Cells In Vitro and in Mice. Gastroenterology 149: 1042.
  44. Gao HP, Li KS, Tu H, Pan XR, Jiang H, et al. (2014) Development of T Cells Redirected to Glypican-3 for the Treatment of Hepatocellular Carcinoma. Clin Cancer Res 20: 6418
  45. Li WP, Guo LJ, Rathi P, Marinova E, Gao XH, et al. (2017) Redirecting T Cells to Glypican-3 with 4-1BB Zeta Chimeric Antigen Receptors Results in Th1 Polarization and Potent Antitumor Activity. Human Gene Therapy 28: 437.
  46. Jiang ZW, Jiang XF, Chen SM, Lai YX, Wei XR, et al. (2017) Anti-GPEC3-CAR T Cells Suppress the Growth of Tumor Cells in Patient-Derived Xenogratfts of Hepatocelluar Carcinoma. Front Immunol 7.
  47. Zhai B, Shi DH, Gao HP, Qi XX, Jiang H, et al. (2017) A phase I study of anti-GPC3 chimeric antigen receptor modified T cells (GPC3 CAR-T) in Chinese patients with refractory or relapsed GPC3+hepatocellular carcinoma (r/r GPC3+HCC). J Clin Oncol 35.
  48. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, et al. (2010) Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen Receptor Recognizing ERBB2. Mol Ther 18: 843.
  49. Chen C, Li KS, Jiang H, Song F, Gao HP, et al. (2017) Development of T cells carrying two complementary chimeric antigen receptors against glypican-3 and asialoglycoprotein receptor 1 for the treatment of hepatocellular carcinoma. Cancer Immunol Immunother 66: 475.
  50. Schumacher TN, Schreiber RD (2015) Neoantigens in cancer immunotherapy. Science 348: 69.
  51. Hiroishi K, Eguchi J, Baba T, Shimazaki T, Ishii S, et al. (2010) Strong CD8(+) T-cell responses against tumor-associated antigens prolong the recurrence-free interval after tumor treatment in patients with hepatocellular carcinoma. J Gastroenterol 45: 451.
  52. Mizukoshi E, Nakamoto Y, Arai K, Yamashita T, Sakai A, et al. (2011) Comparative analysis of various tumor-associated antigen-specific t-cell responses in patients with hepatocellular carcinoma. Hepatology 53: 1206.
  53. Avella DM, Li G, Schell TD, Liu D, Zhang SS, et al. (2012) Regression of established hepatocellular carcinoma is induced by chemoimmunotherapy in an orthotopic murine model. Hepatology 55: 141.
  54. Qasim W, Brunetto M, Gehring AJ, Xue SA, Schurich A, et al. (2015) Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. J Hepatol 62: 486.
  55. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, et al. (2011) Innate or adaptive immunity? The example of natural killer cells. Science 331: 44.
  56. Shi FD, Ljunggren HG, La Cava A, Van Kaer L (2011) Organ-specific features of natural killer cells. Nat Rev Immunol 11: 658.
  57. Tsuchiyama T, Nakamoto Y, Sakai Y, Marukawa Y, Kitahara M, et al. (2007) Prolonged, NK cell-mediated antitumor effects of suicide gene therapy combined with monocyte chemoattractant protein-1 against hepatocellular carcinoma. J Immunol 178: 574.
  58. Zerbini A, Pilli M, Laccabue D, Pelosi G, Molinari A, et al. (2010) Radiofrequency thermal ablation for hepatocellular carcinoma stimulates autologous NK-cell response. Gastroenterology 138: 1931.
  59. Sun C, Sun H, Zhang C, Tian Z (2015) NK cell receptor imbalance and NK cell dysfunction in HBV infection and hepatocellular carcinoma. Cell Mol Immunol 12: 292.
  60. Gras Navarro A, Bjorklund AT, Chekenya M (2015) Therapeutic potential and challenges of natural killer cells in treatment of solid tumors. Front Immunol 6: 202.
  61. Chew V, Tow C, Huang C, Bard-Chapeau E, Copeland NG, et al. (2012) Toll-like receptor 3 expressing tumor parenchyma and infiltrating natural killer cells in hepatocellular carcinoma patients. J Natl Cancer Inst 104: 1796.
  62. Beraza N, Malato Y, Sander LE, Al-Masaoudi M, Freimuth J, et al. (2009) Hepatocyte-specific NEMO deletion promotes NK/NKT cell- and TRAIL-dependent liver damage. J Exp Med 206: 1727.
  63. Kamimura H, Yamagiwa S, Tsuchiya A, Takamura M, Matsuda Y, et al. (2012) Reduced NKG2D ligand expression in hepatocellular carcinoma correlates with early recurrence. J Hepatol 56: 381.
  64. Goto K, Kato N (2015) MICA SNPs and the NKG2D system in virus-induced HCC. J Gastroenterol 50: 261.
  65. Fiegler N, Textor S, Arnold A, Rolle A, Oehme I, et al. (2013) Downregulation of the activating NKp30 ligand B7-H6 by HDAC inhibitors impairs tumor cell recognition by NK cells. Blood 122: 684.
  66. Zhao JJ, Pan QZ, Pan K, Weng DS, Wang QJ, et al. (2014) Interleukin-37 mediates the antitumor activity in hepatocellular carcinoma: role for CD57+ NK cells. Sci Rep 4: 5177.
  67. Cui J, Wang N, Zhao H, Jin H, Wang G, et al. (2014) Combination of radiofrequency ablation and sequential cellular immunotherapy improves progression-free survival for patients with hepatocellular carcinoma. Int J Cancer 134: 342.
  68. Leboeuf C, Mailly L, Wu T, Bour G, Durand S, et al. (2014) In vivo proof of concept of adoptive immunotherapy for hepatocellular carcinoma using allogeneic suicide gene-modified killer cells. Mol Ther 22: 634.
  69. Sun X, Sui Q, Zhang C, Tian Z, Zhang J (2013) Targeting blockage of STAT3 in hepatocellular carcinoma cells augments NK cell functions via reverse hepatocellular carcinoma-induced immune suppression. Mol Cancer Ther 12: 2885.
  70. Sui Q, Zhang J, Sun X, Zhang C, Han Q, et al. (2014) NK cells are the crucial antitumor mediators when STAT3-mediated immunosuppression is blocked in hepatocellular carcinoma. J Immunol 193: 2016.
  71. Ma Y, Xu YC, Tang L, Zhang Z, Wang J, et al. (2012) Cytokine-induced killer (CIK) cell therapy for patients with hepatocellular carcinoma: efficacy and safety. Exp Hematol Oncol 1: 11.
  72. Nishimura R, Baker J, Beilhack A, Zeiser R, Olson JA, et al. (2008) In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood 112: 2563.
  73. Lee JH, Lee JH, Lim YS, Yeon JE, Song TJ, et al. (2015) Adjuvant Immunotherapy With Autologous Cytokine-Induced Killer Cells for Hepatocellular Carcinoma. Gastroenterology.
  74. Su Y, Yang Y, Ma Y, Zhang Y, Rao W, et al. (2016) The Efficacy and Safety of Dendritic Cells Co-Cultured with Cytokine-Induced Killer Cell Therapy in Combination with TACE-Predominant Minimally-Invasive Treatment for Hepatocellular Carcinoma: a Meta-Analysis. Clin Lab 62: 599.
  75. Li X, Dai D, Song X, Liu J, Zhu L, et al. (2014) A meta-analysis of cytokine-induced killer cells therapy in combination with minimally invasive treatment for hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 38: 583.
  76. Chen CL, Pan QZ, Zhao JJ, Wang Y, Li YQ, et al. (2016) PD-L1 expression as a predictive biomarker for cytokine-induced killer cell immunotherapy in patients with hepatocellular carcinoma. Oncoimmunology 5.
  77. Platanias LC (2005) Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol 5: 375.
  78. Sun HC, Tang ZY, Wang L, Qin LX, Ma ZC, et al. (2006) Postoperative interferon alpha treatment postponed recurrence and improved overall survival in patients after curative resection of HBV-related hepatocellular carcinoma: a randomized clinical trial. J Cancer Res Clin Oncol 132: 458.
  79. Llovet JM, Sala M, Castells L, Suarez Y, Vilana R, et al. (2000) Randomized controlled trial of interferon treatment for advanced hepatocellular carcinoma. Hepatology 31: 54.
  80. Mazzaferro V, Romito R, Schiavo M, Mariani L, Camerini T,  et al. (2006) Prevention of hepatocellular carcinoma recurrence with alpha-interferon after liver resection in HCV cirrhosis. Hepatology 44: 1543.
  81. Shiratori Y, Shiina S, Teratani T, Imamura M, Obi S, et al. (2003) Interferon therapy after tumor ablation improves prognosis in patients with hepatocellular carcinoma associated with hepatitis C virus. Ann Intern Med 138: 299.
  82. Lo CM, Liu CL, Chan SC, Lam CM, Poon RT, et al. (2007) A randomized, controlled trial of postoperative adjuvant interferon therapy after resection of hepatocellular carcinoma. Ann Surg 245: 831.
  83. Chen LT, Chen MF, Li LA, Lee PH, Jeng LB, et al. (2012) Long-term results of a randomized, observation-controlled, phase III trial of adjuvant interferon Alfa-2b in hepatocellular carcinoma after curative resection. Ann Surg 255: 8.
  84. Ishikawa T, Higuchi K, Kubota T, Seki K, Honma T, et al. (2012) Combination PEG-IFN a-2b/ribavirin therapy following treatment of hepatitis C virus-associated hepatocellular carcinoma is capable of improving hepatic functional reserve and survival. Hepatogastroenterology 59: 529.
  85. Sangro B, Mazzolini G, Ruiz J, Herraiz M, Quiroga J, et al. (2004) Phase I trial of intratumoral injection of an adenovirus encoding interleukin-12 for advanced digestive tumors. J Clin Oncol 22: 1389.
  86. Mazzolini G, Alfaro C, Sangro B, Feijoo E, Ruiz J, et al. (2005) Intratumoral injection of dendritic cells engineered to secrete interleukin-12 by recombinant adenovirus in patients with metastatic gastrointestinal carcinomas. J Clin Oncol 23: 999.
  87. Kayashima H, Toshima T, Okano S, Taketomi A, Harada N, et al. (2010) Intratumoral neoadjuvant immunotherapy using IL-12 and dendritic cells is an effective strategy to control recurrence of murine hepatocellular carcinoma in immunosuppressed mice. J Immunol 185: 698.
  88. Mishra A, Sullivan L, Caligiuri MA (2014) Molecular pathways: interleukin-15 signaling in health and in cancer. Clin Cancer Res 20: 2044.
  89. Waldmann TA (2015) The shared and contrasting roles of IL2 and IL15 in the life and death of normal and neoplastic lymphocytes: implications for cancer therapy. Cancer Immunol Res 3: 219.
  90. Waldmann TA, Lugli E, Roederer M, Perera LP, Smedley JV, et al. (2011) Safety (toxicity), pharmacokinetics, immunogenicity, and impact on elements of the normal immune system of recombinant human IL-15 in rhesus macaques. Blood 117: 4787.
  91. Conlon KC, Lugli E, Welles HC, Rosenberg SA, Fojo AT, et al. (2015) Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J Clin Oncol 33: 74.
  92. Ormandy LA, Farber A, Cantz T, Petrykowska S, Wedemeyer H, et al. (2006) Direct ex vivo analysis of dendritic cells in patients with hepatocellular carcinoma. World J Gastroenterol 12: 3275.
  93. Strauss O, Dunbar PR, Bartlett A, Phillips A (2015) The immunophenotype of antigen presenting cells of the mononuclear phagocyte system in normal human liver--a systematic review. J Hepatol 62: 458.
  94. Thomson AW, Knolle PA (2010) Antigen-presenting cell function in the tolerogenic liver environment. Nat Rev Immunol 10: 753.
  95. Cany J, Barteau B, Tran L, Gauttier V, Archambeaud I, et al. (2011) AFP-specific immunotherapy impairs growth of autochthonous hepatocellular carcinoma in mice. J Hepatol 54: 115.
  96. Butterfield LH, Ribas A, Meng WS, Dissette VB, Amarnani S, et al. (2003) T-cell responses to HLA-A*0201 immunodominant peptides derived from alpha-fetoprotein in patients with hepatocellular cancer. Clin Cancer Res 9: 5902.
  97. Butterfield LH, Ribas A, Dissette VB, Lee Y, Yang JQ, et al. (2006) A phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four alpha-fetoprotein peptides. Clin Cancer Res 12: 2817.
  98. Lee WC, Wang HC, Hung CF, Huang PF, Lia CR, et al. (2005) Vaccination of advanced hepatocellular carcinoma patients with tumor lysate-pulsed dendritic cells: a clinical trial. J Immunother 28: 496.
  99. Palmer DH, Midgley RS, Mirza N, Torr EE, Ahmed F, et al. (2009) A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology 49: 124.
  100. Gonzalez-Carmona MA, Lukacs-Kornek V, Timmerman A, Shabani S, Kornek M, et al. (2008) CD40ligand-expressing dendritic cells induce regression of hepatocellular carcinoma by activating innate and acquired immunity in vivo. Hepatology 48: 157.
  101. Qiu Y, Xu MB, Yun MM, Wang YZ, Zhang RM, et al. (2011) Hepatocellular carcinoma-specific immunotherapy with synthesized alpha1,3- galactosyl epitope-pulsed dendritic cells and cytokine-induced killer cells. World J Gastroenterol 17: 5260.
  102. Shang N, Figini M, Shangguan J, Wang B, Sun C, et al. (2017) Dendritic cells based immunotherapy. Am J Cancer Res 7: 2091.

Citation: Li H, Gyory I, Zhang X, Xu M, Xiong S (2019) Immunotherapy for Hepatocellular Carcinoma and Its New Development. J Oncol Res Treat 4: 134.

Copyright: © 2019 Li H, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top