Next Article in Journal
Asymmetric Presynaptic Depletion of Dopamine Neurons in a Drosophila Model of Parkinson’s Disease
Next Article in Special Issue
Predicting Outcomes of Atezolizumab and Bevacizumab Treatment in Patients with Hepatocellular Carcinoma
Previous Article in Journal
The Role of Krüppel-like Factors in Pancreatic Physiology and Pathophysiology
Previous Article in Special Issue
Synergistic Nanomedicine: Photodynamic, Photothermal and Photoimmune Therapy in Hepatocellular Carcinoma: Fulfilling the Myth of Prometheus?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Synergistic Effect of Interventional Locoregional Treatments and Immunotherapy for the Treatment of Hepatocellular Carcinoma

Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(10), 8598; https://doi.org/10.3390/ijms24108598
Submission received: 18 April 2023 / Revised: 5 May 2023 / Accepted: 8 May 2023 / Published: 11 May 2023

Abstract

:
Immunotherapy has remarkably revolutionized the management of advanced HCC and prompted clinical trials, with therapeutic agents being used to selectively target immune cells rather than cancer cells. Currently, there is great interest in the possibility of combining locoregional treatments with immunotherapy for HCC, as this combination is emerging as an effective and synergistic tool for enhancing immunity. On the one hand, immunotherapy could amplify and prolong the antitumoral immune response of locoregional treatments, improving patients’ outcomes and reducing recurrence rates. On the other hand, locoregional therapies have been shown to positively alter the tumor immune microenvironment and could therefore enhance the efficacy of immunotherapy. Despite the encouraging results, many unanswered questions still remain, including which immunotherapy and locoregional treatment can guarantee the best survival and clinical outcomes; the most effective timing and sequence to obtain the most effective therapeutic response; and which biological and/or genetic biomarkers can be used to identify patients likely to benefit from this combined approach. Based on the current reported evidence and ongoing trials, the present review summarizes the current application of immunotherapy in combination with locoregional therapies for the treatment of HCC, and provides a critical evaluation of the current status and future directions.

1. The New Era in Hepatocellular Carcinoma Treatment: The Breakthrough of Immunotherapy

Hepatocellular carcinoma (HCC) is the sixth most commonly occurring cancer worldwide, and due to its constantly increasing incidence, it has become the third leading cause of cancer-related death among general populations. Moreover, it represents the most common cause of death in patients with cirrhosis [1,2]. Multiple classification schemes are available to stratify HCC patients in an effort to determine which therapies they can undergo to increase their overall survival. The Barcelona Clinic Liver Cancer Staging (BCLC) system is one of the most widely used, and takes into account hepatic function, the extent of tumor involvement, and performance status [3].
The definitive therapies for HCC remain surgical resection and liver transplantation that can be performed only in patients at very early (0) and early (A) stages. However, given the similar survival benefit paired with the less invasiveness and lower costs compared to surgical resection, percutaneous ablative therapies such as radiofrequency ablation (RFA) and microwave ablation (MWA) are now considered the first treatment approach in both very early and early stages, especially in patients with small HCC (≤3 cm) [4,5]. Despite inducing an effective local antitumor effect, the responses to ablation techniques are relatively weak and might not completely control the tumor, as testified to by the high local recurrence rates. In particular, the size, number and location of tumors can be responsible for incomplete treatment response [6]; in addition, by promoting angiogenesis of residual cancer cells through both transcriptional and epigenetic regulations, insufficient ablation could lead to the recurrence of HCC with a more aggressive phenotype [7]. Therefore, novel techniques to improve ablation efficacy are currently being investigated.
Despite the improvement in screening and surveillance programs, most patients with HCC (about 65–70%) are still diagnosed in the intermediate (B) or advanced (C) tumoral stages, and are thus ineligible for radical therapies [8,9,10]; therefore, patients with intermediate and/or advanced HCCs are considered for transarterial therapies or systemic therapies [11,12] which, albeit effective, are deemed non-curative or “palliative” and still yield a lower 5-year survival rate [13,14]. According to BCLC tumor staging and management [3], transarterial chemoembolization (TACE) is recommended as first-line therapy for unresectable intermediate-stage HCC (stage B). Therefore, it is not surprising that this treatment was the most widely used first line treatment for the treatment of HCC across the world. More interestingly, instead, TACE emerged as the most frequently used first-line treatment for early and advanced stages, thus making it the most frequent treatment for HCC overall [8]. In fact, TACE is potentially suitable and safe for selected patients in the advanced stage with tumor vein thrombosis [15,16,17], or in combination with systemic therapies, without safety concerns [18,19]. Additionally, TACE can be safely and effectively performed in patients at very early and early stages that are partial responders to surgery or ablation, or that are unfit for these curative therapies due to contraindications [20], or prior to liver transplantation to downstage the tumor burden [21]. Despite evidence of beneficial short-term outcomes with locoregional treatments, recurrence and distant metastasis continue to have a significant effect on the overall survival of patients with HCC, especially in intermediate and advanced stages. This may be partly explained by the hypoxic environment created by the TACE procedure, which can induce neoangiogenesis by stimulating vascular endothelial growth factor (VEGF) and other angiogenic pathways, promoting revascularisation and growth of residual viable tumors or even new lesions [22]. Moreover, when it comes to transarterial therapies, one important consideration is that the blockade of hepatic arteries, especially if repeated several times, can compromise liver function and lead to collateral vessel formation, thus limiting the ability to repeat embolization by conventional hepatic vasculature [23,24]. In an effort to address this problem, many studies have been conducted combining TACE with systemic anti-angiogenic agents, most commonly sorafenib, with the aim to counteract this paradoxical effect and thus extend the clinical benefit derived from TACE. However, although most of these studies report the safety of the combination [18], a large number of clinical trials have failed to demonstrate any significant improvement in clinically relevant outcomes for patients with intermediate-stage HCC [25,26]. Even the more recent TACTICS trial, despite being the first study to demonstrate a longer progression-free survival (PFS) in patients receiving sorafenib plus TACE than in those receiving TACE plus placebo [27], it did not significantly extend overall survival (OS) in its final post-hoc analysis [28]. Therefore, better strategies to improve the outcomes for HCC patients treated with TACE are being developed.
Along with TACE, the role of other radiological locoregional therapies has expanded in recent years. For example, transarterial radioembolization (TARE) with yttrium-90 has been suggested as a safe and effective alternative treatment option for HCC patients with a liver-dominant disease who cannot tolerate systemic therapies [29,30,31], even with a significant cost advantage [32]. Moreover, the recent availability of new microspheres with a different radioisotope (such as 166-holmium) and the new technological developments will probably contribute to further reinforce the role of this option in HCC treatment and expand its clinical indication even in early and intermediate stages [33,34]. Nonetheless, due to the current lack of evidence demonstrating its superiority and non-inferiority to sorafenib, TARE is now recommended only in single HCCs ≤ 8 cm [3,35], and its role behind this indication remains uncertain.
Therefore, despite current limitations, the role of interventional radiology in the treatment of HCC is continuing to grow at each stage of the disease, especially at centers of excellence with multidisciplinary tumor boards, whether it is performed with curative, downstaging, bridging, debulking or palliative intent (Figure 1) [36,37]. Moreover, its expansion is expected to further progress as technical and clinical innovation continue to outpace large randomized controlled trials, with 50–60% of HCC patients that are expected to receive these treatments in their lifespan, globally [38].
In recent years, immunotherapy has led to a major shift in the treatment of HCC and prompted clinical trials, with therapeutic agents being used to selectively target immune cells rather than cancer cells [39]. In particular, the combination of atezolizumab and bevacizumab is now regarded as the standard first-line treatment for patients with advanced HCC due to the significant and clinically meaningful improvements in terms of OS, PFS, objective response rate (ORR) and complete response rate (CRR) compared with sorafenib monotherapy [3,40,41]. More recently, the combination of tremelimumab and durvalumab has been reported to be superior to sorafenib in patients with advanced or unresectable HCC, adding another first-line treatment option [42]. The impressive benefit provided by immunotherapy in patients with advanced HCC has led to the question if there is a rationale to support the combination of these new drugs with locoregional therapies in an adjuvant or neoadjuvant setting even in the early and/or intermediate stages [43]. In fact, it has now been demonstrated that locoregional treatments can positively alter the immune microenvironment of HCC and, theoretically, have a synergistic effect, further enhancing antitumor immune responses and thus improving patient survival [44]. In addition, novel immunotherapies, including new target antibodies, bispecific antibodies, combination regimens, engineered cytokines, adoptive T-cell therapy, tumor vaccines, and oncolytic viruses might be available to treat all stages of HCC in the near future. Based on the current reported evidence and ongoing trials, immunotherapy, especially in combination with other therapies, has the potential to act as a significant approach to the treatment of HCC.

2. The Immunogenic Proprieties of Hepatocellular Carcinoma

HCC arises almost exclusively in the setting of chronic liver diseases, and chronic inflammation is now regarded as one of the main triggers of hepatocarcinogenesis [45,46,47]. Since the background of chronic inflammation promotes immune suppression, there is a tightly interwoven, exceedingly complex relationship between HCC and the anti-tumor immune response in the liver. Due to the presence of an immune-suppressed microenvironment, HCC is indeed considered an immunogenic tumor [48].
First of all, chronic inflammation plays a key role in the initiation, evolution, and progression of neoplasms by creating a microenvironment that supports the malignant transformation of hepatocytes through hepatocellular DNA damage and genetic and epigenetic aberrations [49]. When liver damage occurs, thanks to the liver’s unique considerable ability to repair itself, differentiated hepatocytes can re-enter the cell cycle and serve as their own main source of replacement [50]. However, the chronic activation of non-parenchymal cells induces altered survival and proliferation signals, resulting in cellular stress, epigenetic modifications, mitochondrial alterations, DNA damage, senescence, and chromosomal aberrations. This leads to continual cell death, compensatory regeneration and liver fibrosis, which collectively induce tumorigenesis [51]. Moreover, the increased production of pro-inflammatory cytokines occurring in the setting of chronic inflammation promotes the expression of pro-oncogenic transcription factors (such as STAT3 and NF-κB), further contributing to HCC development [52].
Secondly, chronic inflammation can boost tumor immunogenicity, creating an immunosuppressive surrounding and allowing cancer cells to escape the host immune surveillance and progress [53]. One of the main functions of the liver is to continuously remove a large and diverse spectrum of pathogen components [i.e., pathogen-associated molecular patterns (PAMP)] and endogen molecules derived from damaged or necrotized cells [i.e., damage-associated molecular patterns (DAMPs)] from the circulation, thus ensuring organ protection by maintaining immunotolerance [54]. In chronic liver diseases, however, this tightly controlled immunological network is deregulated, thus leading to the failure of efficient detection and the elimination of transformed cells and causing the breakdown of proper tolerance [53]. Once HCC has developed, an intra-tumor infiltration by lymphocytes occurs, in an attempt by the host to mediate an anti-tumor reaction [55]. Under normal circumstances, tumor antigens would be internalized by the host antigen-presenting cells (APCs) and then, after being processed, be bonded to Major Histocompatibility Complex II (MHC-II) molecules. Subsequently, if properly stimulated, dendritic cells would present these tumor antigens to T cells located in the lymphatic organs, thus promoting their activation and the stimulation of effector cells, including CD8+ T cells and Natural Killer (NK) cells. Once activated, tumor-specific effector cells would migrate from lymph nodes to the tumor location, where they would exert their cytotoxic effect on neoplastic cells. Unfortunately, these cellular responses can be dysfunctional and unable to efficiently eliminate cancer cells, thus leading to HCC progression [56].
Tumoral cells can indeed promote an elevated production of immunosuppressor cytokines (such as IL-10 and TGFβ1) that downregulate the anti-tumor response at different levels. The number of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) increases in the HCC microenvironment, which directly inhibits the tumor killing effect of NK cells and CD8+ T cells through overexpression of multiple factors [57]. In addition, MCH II is often functionally depleted in HCC, thus being unable to induce the activation of CD8+ T cells and leading to tumor immune escape [58]. Furthermore, tumoral cells inhibit the activation of APCs and promote the M2 polarization of macrophages, thus further impairing the effector functions of CD8+ T cells and NK cells [59,60]. Lastly, there is an abnormal expression and function of immune checkpoint molecules that, rather than preventing the excessive immune response from injuring normal hepatocytes as it happens in normal conditions, inhibit the host immune function and thus promote the growth of tumor cells. In particular, the most studied of them are programmed cell death protein 1 (PD-1) and its ligand (PD-L1), which leads to the T-cell exhaustion status, and cytotoxic T-lymphocyte protein 4 (CTLA-4), which inhibits the activation of T cells [61,62].
The current combined strategy of immunotherapy and locoregional treatments essentially aims to enhance the effects of immune checkpoint inhibitors (ICIs) that selectively target these immune checkpoints (PD-1/PD-L1 and CTLA-4); therefore, rather than stimulating new or different immune responses, ICIs can restore and unleash a preexisting immune reactivity to cancer which is being held in check by tumoral microenvironmental factors (Figure 2) [63,64].

2.1. PD-1 and PD-L1 Inibithors

PD-1 plays a key role in the regulation and maintenance of the balance between T cell activation and immune tolerance, especially in peripheral tissues. It is widely expressed on human cells but is mainly detected in activated T cells, NK cells and APCs [65].
PD-1 has two ligands, PD-L1 and PD-L2. PD-L1 is expressed on a variety of cells, both hematopoietic and non-hematopoietic, whereas PD-L2 is selectively induced on fewer cells post-activation, especially APCs, and has a higher affinity to PD1 than PD-L1; however, recent studies have shown that PD-L2 can be found also on other immune cells and even on tumor cells under microenvironment stimulation [66]. When PD-1 binds to its ligands on T cells, it leads to dephosphorylation of T cell receptors and blockage of CD28 signaling with subsequent reduction of T cell proliferation, adhesion, cytolytic function and cytokine production [67,68]. Reportedly, it also promotes the differentiation, maintenance and function of Tregs, further enhancing tumor immune escape [69].
In the HCC microenvironment, PD-L1 is highly expressed by intra-tumoral inflammatory cells, especially Kupffer cells and other APCs, which thereby prevent the activation of anti-tumor T cells [70,71]. Additionally, tumor cells can turn this immune checkpoint signaling to their own advantage through the expression of PD-L1 or PD-L2 on their surface, thus favoring the escape of immune surveillance [72].
PD-1 inhibitors can prevent the interaction of PD-1 with its ligands PD-L1 on tumor cells and inflammatory cells by binding to PD-1, leading to the restoration of antitumor activity of functionally depleted T cells. The first anti-PD-1 drug to be used for HCC was nivolumab, a fully human IgG4 monoclonal antibody that blocks PD-1 interactions with PD-L1 and PD-L2; now, several other PD-1 inhibitors are available for the treatment of HCC and are currently undergoing clinical trials, including pembrolizumab, tislelizumab, toripalimab and camrelizumab, which are all humanized IgG4 antibodies, and sintiliamab, a fully human IgG4 monoclonal antibody [73,74]. Different from PD-1 inhibitors, anti-PD-L1 drugs exert their anti-tumor efficacy by binding directly to the PD-L1 receptor on the surface of cancer cells rather than to PD-1 [75]. Durvalumab, a fully human IgG1 antibody, and atezolizumab, a humanized IgG1 antibody, are currently the most relevant anti-PD-L1 agents investigated in the field of HCC [73]; however, recently, avelumab, another IgG1 human antibody, has also demonstrated encouraging results in clinical trials with HCC patients [76]. Since the knowledge about the PD-L2 regulatory network is relatively ambiguous, there are no clinical trials about immunotherapy regimens against PD-L2 so far.
In theory, anti-PD-1 antibodies can block the binding of PD-1 to both its ligands (PD-L1 and PD-L2), whereas PD-L1 antibodies can only inhibit the binding of PD-1 to PD-L1 and, therefore, could be less effective. One meta-analysis of 19 randomized clinical trials involving more than 11.000 patients with cancer revealed a statistically significantly greater OS for patients treated with PD-1 inhibitors compared with patients treated with anti-PD-L1 drugs [77]. However, no patients with HCC were included in the analysis, thus further studies are required to confirm whether anti-PD-1 antibodies are associated with better outcomes compared to PD-L1 inhibitors also in HCC patients.

2.2. CTLA-4 Inibithors

CTLA-4 is an inhibitory co-receptor that is inducibly expressed on activated T cells and constitutively expressed on Tregs [78]. Due to its higher affinity, CTLA-4 competes with its homologous CD28 and binds to CD80/CD86 on the surface of APCs, transmitting an inhibitory signal that downregulates the function of T cells [79]. Additionally, CTLA-4 has been shown to lower levels of CD80/CD86 costimulatory molecules available on APCs through CTLA-4-dependent sequestration via transendocytosis [80]. Therefore, when the T cells are activated and the number of Tregs increases, as it occurs in the HCC microenvironment, the expression of CTLA-4 is up-regulated and the degree of T cell inflammatory response is reduced [81,82]. Contrary to PD-1/PD-L1 activity, however, the downregulation of T cells’ immune response occurs mainly in lymphatic tissues [75].
Any drugs that block CTLA-4 activity can counteract its immunosuppressive mechanism in the process of T cell activation, thus up-regulating the immune system and increasing its ability to recognize and destroy neoplastic cells [75]. The first CTLA-4 inhibitor investigated in the field of HCC was tremelimumab, an IgG2 human antibody; now, ipilimumab, an IgG1 human antibody, is also available for the treatment of HCC [83].

3. The Immune Modulation Effect of Locoregional Therapies

In several animal and human studies, locoregional treatments have been shown to induce immune responses in HCC patients, positively altering their tumor microenvironment [84,85]. The release of tumor antigens due to cell death and subsequent recruitment and activation of APCs and effector immune cells are the main processes responsible for the changes in anti-tumor immune responses after locoregional treatments [84].
Immunogenic cell death involves the translocation of calreticulin on the cell surface, the secretion of ATP, and the release of the non-histone chromatin protein high-mobility group box 1 (HMGB1) and other immunostimulatory molecules that collectively facilitate the recruitment and activation of APCs into the tumor microenvironment, the engulfment of tumor antigens from dying tumor cells and, finally, the optimal antigen presentation to T cells [85,86,87,88,89,90]. Locoregional treatments can induce both apoptosis and necrosis of tumor cells. Necrosis is a form of cell death characterized by loss of plasma membrane integrity, culminating in the escape of cell contents into the extracellular space, including tumor specific antigens, thus is known to be immunogenic; conversely, apoptosis is a programmed cell death in which the plasma membrane is not disrupted and cellular contents are packaged and then released into apoptotic bodies, thus it is regarded as immunologically “silent” [91,92]; nevertheless, previous reports have also implicated that certain types of apoptosis could be immunogenic and therefore favor the immune response against the tumor [93,94].
A plethora of cytokines, chemokines, and inflammatory/cell stress molecular markers have been described following the execution of the majority of locoregional treatments for HCC, supporting the immune modulation effect of these techniques. The effect of MWA as a single therapy was one of the first to be investigated, demonstrating the activation of Tregs, CD4+ and CD8+ T cell and NK cells, as well as the release of IL-12 [95,96]. The evidence that ablative therapy can cause tumor-specific immune responses was observed also in patients who underwent RFA, which can increase the number of tumor-associated antigen-derived peptides in peripheral blood [97], induce APCs activation and proliferation [98] and stimulate the secretion of Th1 cytokines (such as IL-2, TNF-α and IFN-γ) that promote CD8+ T activity [99]. Similarly, also TACE was reported to promote immunogenic cell death, as testified to by the increased serum levels of immunogenic cell death biomarkers following the procedure [100]; moreover, TACE can also promote Th17 and CD8+ activation and reduce the number of Tregs [101,102]. More recently, infiltration of CD8+ T cells and NK cells and an increase in cytokines levels (especially IL-1, IL-6 and IL-8) was found after TARE with yttrium-90 [103,104,105].
Locoregional therapy can promote systemic immune response by releasing neoantigens into blood circulation, but their effect alone might be too modest to prevent tumor recurrence and metastasis, even after successful treatments. Moreover, especially when incomplete, locoregional treatments can also induce immunosuppressive factors (such as IL-6, VEGF, HIF-1α, TGF-β, PD-1 and PD-L1), stimulate the accumulation of Tregs in the tumor and cause lymphopenia, leading to tumor progression in the end [106,107,108,109,110]. Incomplete T cell restoration despite antigen clearance and immune-tolerant liver environment might also affect the attenuation of immune surveillance. Additionally, their immunological effects appear limited in time. Indeed, as demonstrated by a previous study, the memory phenotype and lifetime of tumor-specific T cells were not sufficient to prevent HCC recurrence completely after RFA [97]. For all these reasons, the efficacy of locoregional treatments could be enhanced by their combination with immunotherapeutic drugs, which would guarantee the achievement of an immunologically more favorable tumor microenvironment [111,112]; at the same time, through a mutually beneficial and synergistic mechanism, the positive alteration of the tumor microenvironment derived from locoregional treatments may enhance ICI therapy efficacy (Figure 3) [38].
To date, there is no direct comparison between the different ablation or intra-arterial techniques, therefore it is not known whether one technique is superior to the others in inducing tumor-specific immune response [113]. In a previous study, it was demonstrated that serum levels of Glypican-3, a carcinoembryonic antigen inducing tumor-specific activation of cytotoxic T cells, were increased in 55% of patients with HCC after RFA and in 44% of patients after TACE, although these results were non-significant [114]. Interestingly, more recent evidence seems to suggest that TACE may have a greater immunogenic role than other locoregional treatments, possibly due to the potential immunogenic cell death induced by doxorubicin [115]. Doxorubicin is the most used chemotherapeutic agent for TACE and, despite the absence of a proven superiority compared to other drugs (such as cisplatin, epirubicin and mitomycin), is the only one to have demonstrated to possess immunogenic properties and thus trigger a significant tumor-specific immunological response [116]. In particular, anthracyclines such as doxorubicin seem to cause the post-transcriptional translocation of calreticulin from the endoplasmic reticulum, where it is involved in the maintenance of Ca2+ homeostasis, to the plasma membrane of tumor cells; surface-exposed calreticulin then acts as an “eat me” signal for phagocytosis by neighboring APCs, which is required for subsequent antigen cross-presentation to cytotoxic T cells [117]. Because chemotherapy is an integral part of TACE, these studies indicate that not only the immunogenic effects of embolization must be considered, but also the immune effects of the chemotherapy of choice. Therefore, if TACE is combined with immunotherapy, doxorubicin likely would lead to better outcomes compared to other chemotherapeutic agents.

4. The Current Evidence from Clinical Trials

The high risk of local and distant recurrence after locoregional treatments indicates the need for efficient adjuvant strategies to improve cure rates, even at very early and early stages. Features, such as large tumors, multinodularity, and vascular invasion (macroscopic or microscopic), are significantly related to higher recurrence rates in both ablative and intra-arterial therapies [118,119,120]. With this perspective, the addition of immunotherapy after locoregional treatments could amplify the effect of these treatments against micro-metastatic residual disease, especially in patients with a high risk of recurrence or those who would present clinical or hepatic deterioration after treatment. Similarly, there is a rationale to integrate immunotherapy in the neoadjuvant setting as well, especially in intermediate and advanced stages. The pre-treatment administration of ICIs can indeed leverage the higher levels of tumor antigens and thus promote the expansion of tumor-specific T cells, increasing the chance of cure following locoregional treatments [121,122].
One of the first trials that investigated the role of ICIs in combination with locoregional treatments in HCC patients evaluated the safety and efficacy of tremelimumab plus subtotal conventional TACE, RFA or cryoablation in patients who were non-responders to sorafenib. In particular, the protocol was shown to be safe and feasible, with no clear trends in adverse events or dose-limited toxicity; moreover, this therapeutic combination resulted in objective tumor responses even outside of the ablated or embolized zone, indicating that the systemic effects brought by locoregional therapies indeed exist [123]. The combination of tremelimumab plus ablation (RFA or cryoablation) or drug-eluting beads TACE (DEB-TACE) was also assessed in another study with HCC patients progressed on sorafenib therapy, proving the safety and efficacy of the protocol; in particular, the primary lesion kept shrinking and almost disappeared at 6 months and the untreated other intrahepatic lesions reduced in size gradually [84]. The enhanced efficacy of anti-PD-1 and ablative combined therapy was later confirmed in another retrospective study, where patients who underwent RFA plus camrelizumab or sintilimab demonstrated a longer OS and a higher recurrence-free survival (RFS) compared to those treated with RFA alone (32.5% vs. 10.0% and 51 weeks vs. 47.6 weeks, respectively) [124]. Similarly, a proof-of-concept clinical trial enrolling 50 patients with advanced HCC after sorafenib failure reported that additional RFA or MWA to anti-PD-1 therapy (nivolumab or pembrolizumab) increased the response rate from 10% to 24%. This latter study, moreover, documented that repeated ablations were also proved feasible and safe, reporting only common ablation-related complications that were easily managed as per the standard of care [125].
Three different studies [126,127,128] indicated that anti-PD-1 therapy (camrelizumab) plus TACE regimen is effective and safe, with effective tumor control, improved survival and manageable ICI-related adverse effects, leading to better outcomes than treatment with anti-PD-1 inhibitors alone; moreover, a longer interval between camrelizumab administration and TACE was related to the unsatisfying OS, whereas the timing of administration (before or after TACE) did not significantly influence the results. However, another study reported similar efficacy of TACE combined with camrelizumab compared to TACE alone, although the protocol was safe and tolerable [129]. Among the most common adverse events, itching was the most common, and is often associated with dermatitis and increased liver transaminases; whereas the appearance of colitis, thyroiditis and pneumonia is rarer. An interesting study compared the efficacy and safety of conventional TACE + camrelizumab with DEB-TACE + camrelizumab with the aim of determining which technique was superior. Despite both protocols being safe and well-tolerated, DEB-TACE produced better tumor response and PFS (70.4% vs. 40.7% and 10 vs. 3 months, respectively); however, these results could have been influenced by the inclusion of patients with large and multiple HCCs, who are theoretically more susceptible to this type of intra-arterial procedure; thus, further studies are needed [130].
Similar to TACE, even TARE in combination with nivolumab was demonstrated as a safe and effective treatment for HCC patients, showing a higher objective response rate (ORR) compared to both TARE alone and anti-PD-1 agents alone (30.6% vs. 20% vs. 15–23%, respectively) [131]; of note, the ORR in patients without extrahepatic spread was 43.5%, suggesting that TARE followed by nivolumab should be further evaluated in patients with BCLC B or BCLC C with no extrahepatic spread. One small retrospective trial examined patients with advanced HCC but preserved liver function who had received TARE and nivolumab with or without ipilimumab, documenting the safety of this association; moreover, there were no differences in toxicities between patients who received both therapies within 30 days of each other and those who received both therapies within 30–90 days [132]. The safety and efficacy of TARE plus anti-PD-1 therapy were also confirmed in other studies [133,134].
Despite this encouraging evidence, larger and comparative studies are needed to confirm the efficacy of immunotherapy combined with locoregional treatments in HCC patients. Currently, several other trials are exploring the role of ICIs in combination with locoregional treatments in HCC patients, with or without other drugs (such as tyrosine-kinase inhibitors), but participants are still being recruited or are receiving intervention, or data have yet to be analyzed. The role of numerous immunotherapeutic drugs is being tested in the adjuvant setting of patients who underwent ablative therapies, including nivolumab (the CheckMate 9DX trial, NCT03383458), atezolizumab plus bevacizumab (the IMbrave050 trial, NCT04102098) and pembrolizumab (the KEYNOTE-937 trial, NCT03867084); similarly, the use of nivolumab in both adjuvant and neoadjuvant settings after electroporation is currently being investigated (the NIVOLEP trial, NCT03630640). The number of studies that are evaluating the combination of immunotherapy with TACE and TARE in intermediate and advanced patients is even larger. The results of the LEAP-012 trial exploring TACE plus pembrolizumab plus lenvatinib in advanced HCC patients are eagerly awaited (NCT04246177), as are those of ongoing trials evaluating TACE plus atezolizumab plus bevacizumab (NCT04712643), TACE plus durvalumab plus bevacizumab (the EMERALD-1 trial, NCT03778957), TACE plus durvalumab plus bevacizumab plus tremelimumab (the EMERALD-3 trial, NCT05301842), TACE plus nivolumab (the TACE-3 trial, NCT04268888) and TACE plus nivolumab plus ipilimumab (the CheckMate 74W trial, NCT04340193). Similarly, the results that will emerge from trials combining TARE with yttrium-90 plus nivolumab (NCT03033446), pembrolizumab (NCT03099564) or durvalumab plus tremelimumab (the MEDI4736 trial, NCT04522544) are highly anticipated.

5. Current Challenges and Limitations of Combined Immunotherapy and Locoregional Therapies

The greatest challenge in investigating this combination approach still lies in the design of the clinical trials, specifically in the selection of an appropriate target population, proper control arms and adequate primary endpoints [135]. Moreover, the heterogeneity of both population and tumor burden should be considered before randomization since it can potentially limit the results [136]. For example, the different outcomes between virus-related and non-virus-related HCC observed in other trials of immunotherapies seem to suggest that this element should be incorporated as a stratification factor in addition to the geographical region [137]. Furthermore, the optimal regimens of locoregional treatment (dose/fraction of radiation therapy, types of chemotherapeutic agents, etc.) that will best induce immunogenic cell death and the timing and sequence of both locoregional treatments and immunotherapy should also be addressed [138]. Finally, there are still methodologic differences in how these trials assess treatment response since they combine agents that require iRECIST with therapeutical procedures requiring mRECIST, adding a complexity that remains to be determined.
Besides these “theoretical” challenges, however, there are also several “practical” issues in implementing this combination therapy in clinical practice. For example, the Italian Liver Cancer (ITA.LI.CA) group has shown that in real-life practice, due to the numerous restricted selection parameters, only 10–20% of HCC patients are eligible for first-line ICI therapy and this percentage is reduced to <10% in the second-line treatment. Therefore, considering that about 30–40% of them do not respond to these agents, only a small number of HCC patients could actually benefit from immunotherapy [139]. Moreover, the contraindications and the feasibility of locoregional treatments in these patients should also be acknowledged, since they are not negligible [140].
Therefore, there is an urgent need for effective predictive serological and/or tissue biomarkers to identify patients likely to benefit from immunotherapy and thus dictate patient-specific therapy choices and reduce the economic burdens on healthcare systems; in addition, it would be possible to avoid ICI-associated adverse events in those patients identified as non-responders. PD-L1 expression is widely used today for the selection of anti-PD-1 therapy in patients with non-small cell lung cancer and melanoma [141,142]; as for HCC, this association has not yet been sufficiently investigated and PD-L1 expression cannot be considered a binary marker to help decide which patients should receive anti- PD-1 therapy. In addition, a study revealed a significant correlation between tumor mutation burden (TMB) and clinical outcomes after PD-1 inhibitors, implying that tumors with high TMB would present a greater number of tumoral neoantigens and thereby would have a greater chance of being recognized by tumor-specific T cells [143]; however, since HCC proved to be less immunogenic compared to other tumors and showed a low TMB (approximately 5 Mut/Mb), the role of this biomarker in these patients remains limited [144]. More recently, overexpression of TIM-3 and LAG-3 as well as Wnt/β-catenin mutations have emerged as possible biomarkers of response in patients receiving anti-PD-1 therapies; nonetheless, data are still preliminary and further confirmations are necessary before drawing firm conclusions [145,146,147]. Therefore, to date, no validated molecular and/or genetic biomarkers predicting response to ICIs in patients with HCC have been identified.
Finally, there is a unique and challenging subset of patients that deserves special attention, i.e., those in whom HCC recurs after liver transplantation. In fact, despite its proven curative efficacy, the recurrence rate of HCC following transplantation is still 15–20%. [148] However, organ transplantation has typically been an exclusion criterion in every clinical trial testing ICIs since immunotherapy, through the activation of T cells, can cause allograft rejection and, subsequently, lead to end-stage organ failure in a high percentage of subjects (37.5% and 75%, respectively) [149]. Moreover, due to the immunosuppressive status, the efficacy of immunotherapy could be reduced because ICIs require competent T-cell populations to exert their antitumor effects [150]. Therefore, until further experience is provided, the use of ICI should be avoided in the neoadjuvant setting for patients on the liver transplant waiting list as well as in the post-transplant setting, and sorafenib or lenvatinib should remain the first-line treatment for this subgroup of HCC patients.

6. Future Perspectives and Promises of Combined Immunotherapy and Locoregional Therapies

Despite that the therapeutic combination of ICIs and locoregional treatments appears promising, its antitumor efficacy may be impaired by the hypoxic mechanisms secondary to locoregional approaches, which might increase the level of pro-angiogenetic cytokines (such as VEGF-1, VEGF-2, TGFβ) and, therefore, promote tumor angiogenesis and metastasis development [38]. Moreover, hypoxia can reactivate APCs, promote the activation, infiltration and migration of lymphocytes and reduce the recruitment of inhibitory cells such as Tregs and MDSCs [151]; furthermore, PD-L1 expression is strongly dependent on transcriptional regulation of hypoxia-inducible factor 1 alpha (HIF1α) [152]. Based on these premises and the approval of the combination of atezolizumab and bevacizumab as first-line treatment for unresectable HCC [41], several studies are currently investigating the synergistic effects of locoregional therapies with immunotherapy and antiangiogenic agents with the hope to extend the clinical benefit of this combination. Therefore, VEGF/VEGFR inhibitors could have a double effect on cancer cells, counteracting the paradoxical angiogenic effect of locoregional therapies and boosting immunity through parallel and distinct mechanisms, thus further priming tumors for immunotherapy and leading to a stronger immune response. Some studies have shown the effectiveness and safety of TACE combined with antiangiogenic therapy and immunotherapy in advanced HCC [153,154,155]. In addition, TACE combined with antiangiogenic therapy and immunotherapy has been demonstrated to remarkably improve OS and PFS over both the combined antiangiogenic and immunotherapy or the combined TACE and antiangiogenic therapy in unresectable HCC patients [156,157,158,159]. Similar results have also been observed with other locoregional approaches, including DEB-TACE [160] and MWA [161].
Although the preliminary outcomes of these therapeutic combinations are encouraging, countless other treatment possibilities can still be explored. For example, radioimmunotherapy has recently emerged as a valid therapeutic option for HCC. In particular, this technique offers a selective internal radiation therapy approach using radionuclides conjugated with tumor-specific monoclonal antibodies, thus combining immunotherapy and radiotherapy. In particular, by binding to the cancer cell surface, these radioimmunoconjugates enable a targeted concentration of radiation in tumor tissue, leading to DNA damage and finally causing tumor cell death. Following this principle, iodine-131 labeled metuximab (also commercially known as Licartin) gained approval for clinical therapy of primary HCC from the China State Food and Drug Administration [162]. This radioimmunoconjugate directly targets HAb18G/CD147, a cell antigen with multiple functions highly expressed on HCC cells. In addition, to allow for the concentration of radionuclides in HCC tissues, iodine-131 metuximab can directly impair the adhesion and motion of tumor cells; furthermore, it can block metalloproteinases production and VEGFR-2 phosphorylation, thus effectively inhibiting HCC growth and metastasis [163,164]. A combination of iodine-131 metuximab with TACE has recently demonstrated an improved clinical efficacy in HCC therapy compared to TACE alone, in terms of both tumor response and OS, with a similar tolerability profile [165,166]. Theoretically, the arterial embolization activity of TACE can indeed enhance the anti-tumor effects of iodine-131 metuximab by reducing tumoral blood flow and resulting in tumor retention of the radioimmunoconjugate; moreover, retention of the anti-cancer drug in the tumor may have a radiosensitizing effect and iodine-131 metuximab can eliminate residual cancer cells after TACE for its continuous radiation. Interestingly, one randomized trial reported that also the combination of iodine-131 metuximab with RFA resulted in improved outcomes for patients at very early, early and intermediate stages, with greater anti-recurrence benefit than RFA alone; however, this significant improvement was not detected in the CD147-negative subpopulation, thus further analysis is still needed [167]. Therefore, radioimmunotherapy in combination with TACE or RFA and, hypothetically, even with systemic therapies (including sorafenib and other VEGF/VEGFR inhibitors) could certainly represent a promising field of investigation in the near future.
Adoptive cell therapies represent a novel approach to HCC therapy. In this approach, autologous immune cells are extracted from the patient, activated and expanded ex vivo, then reinfused into the patient; these can include cytokine-induced killer (CIK) cells, γδ T cells, dendritic cells, NK cells, lymphokine-activated killer (LAK) cells and tumour-infiltrating lymphocyte (TILs). Moreover, genetically modified immune cells, including chimeric antigen receptor T cells (CAR-T) directed against glypican-3 (GPC3), are currently in development [168,169]. Despite that it is too early for robust results in the HCC setting, it is worth mentioning that the few trials available in the literature seem to confirm that the combination of adoptive cell therapies with local ablation or TACE can be exploited to augment therapeutic efficacy and prevent tumor recurrence [170,171]. Recent data suggest that CIK cells transfusion therapy combined with TACE and/or RFA treatment is associated with longer OS and PFS compared to TACE and/or RFA treatment alone [172]; similarly, even the combination of locoregional treatments with NK cells, γδ T cells and CAR-T cells was reported to be safe and showed encouraging clinical efficacy [173,174,175]. With the identification of a growing number of HCC-associated antigens, tumor vaccines which increase specific immune responses to tumor antigens as well as oncolytic viruses—that can selectively replicate in tumor cells and damage them without harming normal cells—are being investigated and developed [176]. Unfortunately, clinical trials based on adoptive cellular therapies, tumor vaccines and oncolytic vaccines are relatively few compared to those studying ICIs, probably due to the limitations of in-house cell therapy facilities and the obvious higher costs [63,177].
Finally, multiple studies have revealed that qualitative and quantitative alterations of the gut microbiota play an important role in HCC pathogenesis. Chronic liver diseases, indeed, are associated with an imbalance in bacterial composition and metabolic activities (dysbiosis) as well as with changes in the intestinal barrier leakiness, which altogether lead to hepatic exposure to bacterial metabolites and microbiota-associated molecular patterns (MAMPs); additionally, in patients with chronically injured liver, hepatic detoxification, degradation, and clearance are compromised, thus hepatic exposure to this gut-derived microbial toxicity is further enhanced. Both bacterial metabolites and MAMPs promote hepatocarcinogenesis via multiple mechanisms, supporting a chronic inflammatory state, favoring fibrosis development and inducing senescence of hepatic stellate cells [178,179]. The gut microbiome seems to also have a notable impact on responses to immunotherapy in HCC patients. For example, an interesting study showed that fecal samples from patients responding to PD-1 inhibitors presented higher taxa richness compared to fecal samples of non-responders [180]; similarly, a significant microbial dissimilarity was observed in fecal bacteria between patients with unresectable HCC who responded to immunotherapy and patients who did not respond to therapy [181]. Therefore, considering the potential modulatory role of human microbiota on antitumoral immune response, the additional administration of specific probiotics, as well as the more complex fecal microbiota transplantation in non-responder patients are currently being investigated, with rather promising preliminary results. Preclinical studies have demonstrated that supplementation of certain commensal bacterial genera (such as Akkermansia muciniphila) can restore responsiveness to immunotherapy in mice with melanoma [182]. More recently, fecal microbiota transplantation from responder donors in patients with metastatic melanoma, refractory to anti-PD-L1 therapy, led to a clinical response and tumor regression in 3 out of 10 subjects; moreover, all 3 participants that responded to immunotherapy following transplantation received samples from the same donor, indicating the choice of donor stool may be critical in inducing sensitivity to immunotherapy [183]. To date, however, there are no available studies that have analyzed the associations between microbiota alterations and locoregional treatments. In the future, with the increasing possibility of acting on the gut bacterial composition, targeted studies investigating this theme should be considered.

7. Conclusions

Locoregional treatments have been shown to positively alter the tumor immune microenvironment of HCC; therefore, the association of locoregional treatments and immunotherapy could contribute to increasing their efficacy through a synergistic mechanism, thus improving the survival of HCC. Despite the encouraging results, however, many unanswered questions still remain, including which immunotherapy and locoregional treatment can guarantee the best survival and clinical outcomes, the most effective timing and sequence to obtain the most effective therapeutic response and which biological and/or genetic biomarkers can be used to identify patients likely to benefit from these combined approaches. Therefore, when planning new clinical studies, it is essential to homogenize, as far as possible, the trial designs to objectively evaluate the clinical contribution of this association. Finally, to maximize the results of the combined anti-tumor strategy, it is pivotal to enhance the multidisciplinary cooperation between hepatologists, radiologists, nuclear doctors and oncologists.

Author Contributions

Conceptualization, N.B. and M.R.; methodology, N.B. and M.R.; software, N.B. and M.R.; validation, N.B. and M.R.; formal analysis, N.B. and M.R.; investigation, N.B. and M.R.; resources, N.B. and M.R.; data curation, N.B. and M.R.; writing—original draft preparation, N.B. and M.R.; writing—review and editing, N.B. and M.R.; visualization, N.B. and M.R.; supervision, N.B. and M.R.; project administration, N.B. and M.R.; funding acquisition, N.B. and M.R. All authors have read and agreed to the published version of the manuscript.

Funding

The work reported in this publication was funded by the Italian Ministry of Health, RC-2022-2773478 project.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Liver Factsheet. Globocan. Available online: https://gco.iarc.fr/today/data/factsheets/cancers/11-Liver-fact-sheet.pdf (accessed on 20 March 2023).
  2. Dasgupta, P.; Henshaw, C.; Youlden, D.R.; Clark, P.J.; Aitken, J.F.; Baade, P.D. Global Trends in Incidence Rates of Primary Adult Liver Cancers: A Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 171. [Google Scholar] [CrossRef] [PubMed]
  3. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update. J. Hepatol. 2022, 76, 681–693. [Google Scholar] [CrossRef] [PubMed]
  4. Takayama, T.; Hasegawa, K.; Izumi, N.; Kudo, M.; Shimada, M.; Yamanaka, N.; Inomata, M.; Kaneko, S.; Nakayama, H.; Kawaguchi, Y.; et al. Surgery versus Radiofrequency Ablation for Small Hepatocellular Carcinoma: A Randomized Controlled Trial (SURF Trial). Liver Cancer 2021, 11, 209–218. [Google Scholar] [CrossRef] [PubMed]
  5. Cucchetti, A.; Piscaglia, F.; Cescon, M.; Colecchia, A.; Ercolani, G.; Bolondi, L.; Pinna, A.D. Cost-effectiveness of hepatic resection versus percutaneous radiofrequency ablation for early hepatocellular carcinoma. J. Hepatol. 2013, 59, 300–307. [Google Scholar] [CrossRef] [PubMed]
  6. Tiong, L.; Maddern, G.J. Systematic review and meta-analysis of survival and disease recurrence after radiofrequency ablation for hepatocellular carcinoma. Br. J. Surg. 2011, 98, 1210–1224. [Google Scholar] [CrossRef]
  7. Kong, J.; Kong, L.; Kong, J.; Ke, S.; Gao, J.; Ding, X.; Zheng, L.; Sun, H.; Sun, W. After insufficient radiofrequency ablation, tumor-associated endothelial cells exhibit enhanced angiogenesis and promote invasiveness of residual hepatocellular carcinoma. J. Transl. Med. 2012, 10, 230. [Google Scholar] [CrossRef]
  8. Park, J.; Chen, M.; Colombo, M.; Roberts, L.; Schwartz, M.; Chen, P.-J.; Kudo, M.; Johnson, P.; Wagner, S.; Orsini, L.S.; et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: The BRIDGE Study. Liver Int. 2015, 35, 2155–2166. [Google Scholar] [CrossRef]
  9. Golfieri, R.; Garzillo, G.; Ascanio, S.; Renzulli, M. Focal Lesions in the Cirrhotic Liver: Their Pivotal Role in Gadoxetic Acid-Enhanced MRI and Recognition by the Western Guidelines. Dig. Dis. 2014, 32, 696–704. [Google Scholar] [CrossRef]
  10. Renzulli, M.; Golfieri, R. Bologna Liver Oncology Group (BLOG) Proposal of a new diagnostic algorithm for hepatocellular carcinoma based on the Japanese guidelines but adapted to the Western world for patients under surveillance for chronic liver disease. J. Gastroenterol. Hepatol. 2016, 31, 69–80. [Google Scholar] [CrossRef]
  11. Facciorusso, A.; Licinio, R.; Muscatiello, N.; Di Leo, A.; Barone, M. Transarterial chemoembolization: Evidences from the literature and applications in hepatocellular carcinoma patients. World J. Hepatol. 2015, 7, 2009–2019. [Google Scholar] [CrossRef]
  12. Renzulli, M.; Peta, G.; Vasuri, F.; Marasco, G.; Caretti, D.; Bartalena, L.; Spinelli, D.; Giampalma, E.; D’errico, A.; Golfieri, R. Standardization of conventional chemoembolization for hepatocellular carcinoma. Ann. Hepatol. 2021, 22, 100278. [Google Scholar] [CrossRef] [PubMed]
  13. Guarino, M.; Viganò, L.; Ponziani, F.R.; Giannini, E.G.; Lai, Q.; Morisco, F. Special Interest Group on Hepatocellular carcinoma and new anti-HCV therapies” of the Italian Association for the Study of the Liver. Recurrence of hepatocellular carcinoma after direct acting antiviral treatment for hepatitis C virus infection: Literature review and risk analysis. Dig. Liver Dis. 2018, 50, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
  14. Renzulli, M.; Pecorelli, A.; Brandi, N.; Brocchi, S.; Tovoli, F.; Granito, A.; Carrafiello, G.; Ierardi, A.M.; Golfieri, R. The Feasibility of Liver Biopsy for Undefined Nodules in Patients under Surveillance for Hepatocellular Carcinoma: Is Biopsy Really a Useful Tool? J. Clin. Med. 2022, 11, 4399. [Google Scholar] [CrossRef]
  15. Xue, T.-C.; Xie, X.-Y.; Zhang, L.; Yin, X.; Zhang, B.-H.; Ren, Z.-G. Transarterial chemoembolization for hepatocellular carcinoma with portal vein tumor thrombus: A meta-analysis. BMC Gastroenterol. 2013, 13, 60. [Google Scholar] [CrossRef] [PubMed]
  16. Deng, J.; Liao, Z.; Gao, J. Efficacy of Transarterial Chemoembolization Combined with Tyrosine Kinase Inhibitors for Hepatocellular Carcinoma Patients with Portal Vein Tumor Thrombus: A Systematic Review and Meta-Analysis. Curr. Oncol. 2023, 30, 1243–1254. [Google Scholar] [CrossRef] [PubMed]
  17. Guo, L.; Wei, X.; Feng, S.; Zhai, J.; Guo, W.; Shi, J.; Lau, W.Y.; Meng, Y.; Cheng, S. Radiotherapy prior to or after transcatheter arterial chemoembolization for the treatment of hepatocellular carcinoma with portal vein tumor thrombus: A randomized controlled trial. Hepatol. Int. 2022, 16, 1368–1378. [Google Scholar] [CrossRef]
  18. Geschwind, J.-F.; Kudo, M.; Marrero, J.A.; Venook, A.P.; Chen, X.-P.; Bronowicki, J.-P.; Dagher, L.; Furuse, J.; De Guevara, L.L.; Papandreou, C.; et al. TACE Treatment in Patients with Sorafenib-treated Unresectable Hepatocellular Carcinoma in Clinical Practice: Final Analysis of GIDEON. Radiology 2016, 279, 630–640. [Google Scholar] [CrossRef]
  19. Wu, F.-X.; Chen, J.; Bai, T.; Zhu, S.-L.; Yang, T.-B.; Qi, L.-N.; Zou, L.; Li, Z.-H.; Ye, J.-Z.; Li, L.-Q. The safety and efficacy of transarterial chemoembolization combined with sorafenib and sorafenib mono-therapy in patients with BCLC stage B/C hepatocellular carcinoma. BMC Cancer 2017, 17, 645. [Google Scholar] [CrossRef]
  20. Granito, A.; Facciorusso, A.; Sacco, R.; Bartalena, L.; Mosconi, C.; Cea, U.V.; Cappelli, A.; Antonino, M.; Modestino, F.; Brandi, N.; et al. TRANS-TACE: Prognostic Role of the Transient Hypertransaminasemia after Conventional Chemoembolization for Hepatocellular Carcinoma. J. Pers. Med. 2021, 11, 1041. [Google Scholar] [CrossRef]
  21. Pommergaard, H.-C.; Rostved, A.A.; Adam, R.; Thygesen, L.C.; Salizzoni, M.; Bravo, M.A.G.; Cherqui, D.; De Simone, P.; Boudjema, K.; Mazzaferro, V.; et al. Locoregional treatments before liver transplantation for hepatocellular carcinoma: A study from the European Liver Transplant Registry. Transpl. Int. 2018, 31, 531–539. [Google Scholar] [CrossRef]
  22. Wang, B.; Xu, H.; Gao, Z.Q.; Ning, H.F.; Sun, Y.Q.; Cao, G.W. Increased expression of vascular endothelial growth factor in hepatocellular carcinoma after transcatheter arterial chemoembolization. Acta Radiol. 2008, 49, 523–529. [Google Scholar] [CrossRef]
  23. Kudo, M.; Arizumi, T.; Ueshima, K.; Sakurai, T.; Kitano, M.; Nishida, N. Subclassification of BCLC B Stage Hepatocellular Carcinoma and Treatment Strategies: Proposal of Modified Bolondi’s Subclassification (Kinki Criteria). Dig. Dis. 2015, 33, 751–758. [Google Scholar] [CrossRef] [PubMed]
  24. Miyayama, S.; Matsui, O.; Taki, K.; Minami, T.; Ryu, Y.; Ito, C.; Nakamura, K.; Inoue, D.; Notsumata, K.; Toya, D.; et al. Extrahepatic Blood Supply to Hepatocellular Carcinoma: Angiographic Demonstration and Transcatheter Arterial Chemoembolization. Cardiovasc. Interv. Radiol. 2006, 29, 39–48. [Google Scholar] [CrossRef] [PubMed]
  25. Lencioni, R.; Llovet, J.M.; Han, G.; Tak, W.Y.; Yang, J.; Guglielmi, A.; Paik, S.W.; Reig, M.; Kim, D.Y.; Chau, G.-Y.; et al. Sorafenib or placebo plus TACE with doxorubicin-eluting beads for intermediate stage HCC: The SPACE trial. J. Hepatol. 2016, 64, 1090–1098. [Google Scholar] [CrossRef] [PubMed]
  26. Meyer, T.; Fox, R.; Ma, Y.T.; Ross, P.J.; James, M.W.; Sturgess, R.; Stubbs, C.; Stocken, D.D.; Wall, L.; Watkinson, A.; et al. Sorafenib in combination with transarterial chemoembolisation in patients with unresectable hepatocellular carcinoma (TACE 2): A randomised placebo-controlled, double-blind, phase 3 trial. Lancet Gastroenterol. Hepatol. 2017, 2, 565–575, Erratum in Lancet Gastroenterol. Hepatol. 2017, 2, e6. [Google Scholar] [CrossRef] [PubMed]
  27. Kudo, M.; Ueshima, K.; Ikeda, M.; Torimura, T.; Tanabe, N.; Aikata, H.; Izumi, N.; Yamasaki, T.; Nojiri, S.; Hino, K.; et al. Randomised, multicentre prospective trial of transarterial chemoembolisation (TACE) plus sorafenib as compared with TACE alone in patients with hepatocellular carcinoma: TACTICS trial. Gut 2020, 69, 1492–1501. [Google Scholar] [CrossRef]
  28. Kudo, M.; Ueshima, K.; Ikeda, M.; Torimura, T.; Tanabe, N.; Aikata, H.; Izumi, N.; Yamasaki, T.; Nojiri, S.; Hino, K.; et al. Final Results of TACTICS: A Randomized, Prospective Trial Comparing Transarterial Chemoembolization Plus Sorafenib to Transarterial Chemoembolization Alone in Patients with Unresectable Hepatocellular Carcinoma. Liver Cancer 2022, 11, 354–367. [Google Scholar] [CrossRef]
  29. Vilgrain, V.; Pereira, H.; Assenat, E.; Guiu, B.; Ilonca, A.D.; Pageaux, G.-P.; Sibert, A.; Bouattour, M.; Lebtahi, R.; Allaham, W.; et al. Efficacy and safety of selective internal radiotherapy with yttrium-90 resin microspheres compared with sorafenib in locally advanced and inoperable hepatocellular carcinoma (SARAH): An open-label randomised controlled phase 3 trial. Lancet Oncol. 2017, 18, 1624–1636. [Google Scholar] [CrossRef]
  30. Sangro, B.; Carpanese, L.; Cianni, R.; Golfieri, R.; Gasparini, D.; Ezziddin, S.; Paprottka, P.M.; Fiore, F.; Van Buskirk, M.; Bilbao, J.I.; et al. Survival after yttrium-90 resin microsphere radioembolization of hepatocellular carcinoma across Barcelona clinic liver cancer stages: A European evaluation. Hepatology 2011, 54, 868–878. [Google Scholar] [CrossRef]
  31. Chow, P.K.; Gandhi, M.; Tan, S.-B.; Khin, M.W.; Khasbazar, A.; Ong, J.; Choo, S.P.; Cheow, P.C.; Chotipanich, C.; Lim, K.; et al. SIRveNIB: Selective Internal Radiation Therapy Versus Sorafenib in Asia-Pacific Patients with Hepatocellular Carcinoma. J. Clin. Oncol. 2018, 36, 1913–1921. [Google Scholar] [CrossRef]
  32. Alonso, J.C.; Casans, I.; González, F.M.; Fuster, D.; Rodríguez, A.; Sánchez, N.; Oyagüez, I.; Burgos, R.; Williams, A.O.; Espinoza, N. Economic evaluations of radioembolization with Itrium-90 microspheres in hepatocellular carcinoma: A systematic review. BMC Gastroenterol. 2022, 22, 6. [Google Scholar] [CrossRef]
  33. Radosa, C.G.; Radosa, J.C.; Grosche-Schlee, S.; Zöphel, K.; Plodeck, V.; Kühn, J.P.; Kotzerke, J.; Hoffmann, R.-T. Holmium-166 Radioembolization in Hepatocellular Carcinoma: Feasibility and Safety of a New Treatment Option in Clinical Practice. Cardiovasc. Interv. Radiol. 2019, 42, 405–412. [Google Scholar] [CrossRef] [PubMed]
  34. Guiu, B.; Garin, E.; Allimant, C.; Edeline, J.; Salem, R. TARE in Hepatocellular Carcinoma: From the Right to the Left of BCLC. Cardiovasc. Interv. Radiol. 2022, 45, 1599–1607. [Google Scholar] [CrossRef] [PubMed]
  35. Salem, R.; Johnson, G.E.; Kim, E.; Riaz, A.; Bishay, V.; Boucher, E.; Fowers, K.; Lewandowski, R.; Padia, S.A. Yttrium-90 Radioembolization for the Treatment of Solitary, Unresectable HCC: The LEGACY Study. Hepatology 2021, 74, 2342–2352. [Google Scholar] [CrossRef] [PubMed]
  36. Holzwanger, D.J.; Madoff, D.C. Role of interventional radiology in the management of hepatocellular carcinoma: Current status. Chin. Clin. Oncol. 2018, 7, 49. [Google Scholar] [CrossRef]
  37. Aubé, C.; Bouvier, A.; Lebigot, J.; Vervueren, L.; Cartier, V.; Oberti, F. Radiological treatment of HCC: Interventional radiology at the heart of management. Diagn. Interv. Imaging 2015, 96, 625–636. [Google Scholar] [CrossRef]
  38. Llovet, J.M.; De Baere, T.; Kulik, L.; Haber, P.K.; Greten, T.F.; Meyer, T.; Lencioni, R. Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 293–313. [Google Scholar] [CrossRef]
  39. Granito, A.; Muratori, L.; Lalanne, C.; Quarneti, C.; Ferri, S.; Guidi, M.; Lenzi, M.; Muratori, P. Hepatocellular carcinoma in viral and autoimmune liver diseases: Role of CD4+ CD25+ Foxp3+ regulatory T cells in the immune microenvironment. World J. Gastroenterol. 2021, 27, 2994–3009. [Google Scholar] [CrossRef]
  40. Cheng, A.-L.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Lim, H.Y.; Kudo, M.; Breder, V.; Merle, P.; et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J. Hepatol. 2022, 76, 862–873. [Google Scholar] [CrossRef]
  41. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  42. Kelley, R.K.; Sangro, B.; Harris, W.; Ikeda, M.; Okusaka, T.; Kang, Y.-K.; Qin, S.; Tai, D.W.-M.; Lim, H.Y.; Yau, T.; et al. Safety, Efficacy, and Pharmacodynamics of Tremelimumab Plus Durvalumab for Patients with Unresectable Hepatocellular Carcinoma: Randomized Expansion of a Phase I/II Study. J. Clin. Oncol. 2021, 39, 2991–3001. [Google Scholar] [CrossRef] [PubMed]
  43. Singh, P.; Toom, S.; Avula, A.; Kumar, V.; Rahma, O.E. The Immune Modulation Effect of Locoregional Therapies and Its Potential Synergy with Immunotherapy in Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2020, 7, 11–17. [Google Scholar] [CrossRef] [PubMed]
  44. Mattos, A.Z.; Debes, J.D.; Boonstra, A.; Vogel, A.; Mattos, A.A. Immune aspects of hepatocellular carcinoma: From immune markers for early detection to immunotherapy. World J. Gastrointest. Oncol. 2021, 13, 1132–1143. [Google Scholar] [CrossRef] [PubMed]
  45. Ringelhan, M.; Pfister, D.; O’connor, T.; Pikarsky, E.; Heikenwalder, M. The immunology of hepatocellular carcinoma. Nat. Immunol. 2018, 19, 222–232. [Google Scholar] [CrossRef]
  46. Cariani, E.; Missale, G. Immune landscape of hepatocellular carcinoma microenvironment: Implications for prognosis and therapeutic applications. Liver Int. 2019, 39, 1608–1621. [Google Scholar] [CrossRef] [PubMed]
  47. Prieto, J.; Melero, I.; Sangro, B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 681–700. [Google Scholar] [CrossRef]
  48. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer Immunoediting: Integrating Immunity’s Roles in Cancer Suppression and Promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef]
  49. Refolo, M.G.; Messa, C.; Guerra, V.; Carr, B.I.; D’alessandro, R. Inflammatory Mechanisms of HCC Development. Cancers 2020, 12, 641. [Google Scholar] [CrossRef]
  50. Jörs, S.; Jeliazkova, P.; Ringelhan, M.; Thalhammer, J.; Dürl, S.; Ferrer, J.; Sander, M.; Heikenwalder, M.; Schmid, R.M.; Siveke, J.T.; et al. Lineage fate of ductular reactions in liver injury and carcinogenesis. J. Clin. Investig. 2015, 125, 2445–2457. [Google Scholar] [CrossRef]
  51. Hernandez–Gea, V.; Toffanin, S.; Friedman, S.L.; Llovet, J.M. Role of the Microenvironment in the Pathogenesis and Treatment of Hepatocellular Carcinoma. Gastroenterology 2013, 144, 512–527. [Google Scholar] [CrossRef]
  52. Subramaniam, A.; Shanmugam, M.K.; Perumal, E.; Li, F.; Nachiyappan, A.; Dai, X.; Swamy, S.N.; Ahn, K.S.; Kumar, A.P.; Tan, B.K.; et al. Potential role of signal transducer and activator of transcription (STAT)3 signaling pathway in inflammation, survival, proliferation and invasion of hepatocellular carcinoma. Biochim. Biophys. Acta 2013, 1835, 46–60. [Google Scholar] [CrossRef] [PubMed]
  53. Jenne, C.N.; Kubes, P. Immune surveillance by the liver. Nat. Immunol. 2013, 14, 996–1006. [Google Scholar] [CrossRef] [PubMed]
  54. Crispe, I.N. The Liver as a Lymphoid Organ. Annu. Rev. Immunol. 2009, 27, 147–163. [Google Scholar] [CrossRef] [PubMed]
  55. Qin, L.-X. Inflammatory Immune Responses in Tumor Microenvironment and Metastasis of Hepatocellular Carcinoma. Cancer Microenviron. 2012, 5, 203–209. [Google Scholar] [CrossRef]
  56. Fu, J.; Xu, D.; Liu, Z.; Shi, M.; Zhao, P.; Fu, B.; Zhang, Z.; Yang, H.; Zhang, H.; Zhou, C.; et al. Increased Regulatory T Cells Correlate with CD8 T-Cell Impairment and Poor Survival in Hepatocellular Carcinoma Patients. Gastroenterology 2007, 132, 2328–2339. [Google Scholar] [CrossRef]
  57. Liu, M.; Zhou, J.; Liu, X.; Feng, Y.; Yang, W.; Wu, F.; Cheung, O.K.-W.; Sun, H.; Zeng, X.; Tang, W.; et al. Targeting monocyte-intrinsic enhancer reprogramming improves immunotherapy efficacy in hepatocellular carcinoma. Gut 2020, 69, 365–379. [Google Scholar] [CrossRef]
  58. Greten, T.F.; Wang, X.W.; Korangy, F. Current concepts of immune based treatments for patients with HCC: From basic science to novel treatment approaches. Gut 2015, 64, 842–848. [Google Scholar] [CrossRef]
  59. Okumoto, K.; Hattori, E.; Tamura, K.; Kiso, S.; Watanabe, H.; Saito, K.; Saito, T.; Togashi, H.; Kawata, S. Possible contribution of circulating transforming growth factor-beta1 to immunity and prognosis in unresectable hepatocellular carcinoma. Liver Int. 2004, 24, 21–28. [Google Scholar] [CrossRef]
  60. Zhang, F.; Wang, H.; Wang, X.; Jiang, G.; Liu, H.; Zhang, G.; Wang, H.; Fang, R.; Bu, X.; Cai, S.; et al. TGF-β induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget 2016, 7, 52294–52306. [Google Scholar] [CrossRef]
  61. Gordon, S.R.; Maute, R.L.; Dulken, B.W.; Hutter, G.; George, B.M.; McCracken, M.N.; Gupta, R.; Tsai, J.M.; Sinha, R.; Corey, D.; et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017, 545, 495–499. [Google Scholar] [CrossRef]
  62. Jain, N.; Nguyen, H.; Chambers, C.; Kang, J. Dual function of CTLA-4 in regulatory T cells and conventional T cells to prevent multiorgan autoimmunity. Proc. Natl. Acad. Sci. USA 2010, 107, 1524–1528. [Google Scholar] [CrossRef] [PubMed]
  63. Johnston, M.P.; Khakoo, S.I. Immunotherapy for hepatocellular carcinoma: Current and future. World J. Gastroenterol. 2019, 25, 2977–2989. [Google Scholar] [CrossRef] [PubMed]
  64. Donisi, C.; Puzzoni, M.; Ziranu, P.; Lai, E.; Mariani, S.; Saba, G.; Impera, V.; Dubois, M.; Persano, M.; Migliari, M.; et al. Immune Checkpoint Inhibitors in the Treatment of HCC. Front. Oncol. 2021, 10, 601240. [Google Scholar] [CrossRef]
  65. Nikolova, M.; Lelievre, J.-D.; 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. [Google Scholar] [CrossRef] [PubMed]
  66. Tanegashima, T.; Togashi, Y.; Azuma, K.; Kawahara, A.; Ideguchi, K.; Sugiyama, D.; Kinoshita, F.; Akiba, J.; Kashiwagi, E.; Takeuchi, A.; et al. Immune Suppression by PD-L2 against Spontaneous and Treatment-Related Antitumor Immunity. Clin. Cancer Res. 2019, 25, 4808–4819. [Google Scholar] [CrossRef] [PubMed]
  67. Gou, Q.; Dong, C.; Xu, H.; Khan, B.; Jin, J.; Liu, Q.; Shi, J.; Hou, Y. PD-L1 degradation pathway and immunotherapy for cancer. Cell Death Dis. 2020, 11, 955. [Google Scholar] [CrossRef]
  68. Wu, X.; Gu, Z.; Chen, Y.; Chen, B.; Chen, W.; Weng, L.; Liu, X. Application of PD-1 Blockade in Cancer Immunotherapy. Comput. Struct. Biotechnol. J. 2019, 17, 661–674. [Google Scholar] [CrossRef] [PubMed]
  69. Li, B.; Yan, C.; Zhu, J.; Chen, X.; Fu, Q.; Zhang, H.; Tong, Z.; Liu, L.; Zheng, Y.; Zhao, P.; et al. Anti–PD-1/PD-L1 Blockade Immunotherapy Employed in Treating Hepatitis B Virus Infection–Related Advanced Hepatocellular Carcinoma: A Literature Review. Front. Immunol. 2020, 11, 1037. [Google Scholar] [CrossRef]
  70. Wu, K.; Kryczek, I.; Chen, L.; Zou, W.; Welling, T.H. Kupffer Cell Suppression of CD8+ T Cells in Human Hepatocellular Carcinoma Is Mediated by B7-H1/Programmed Death-1 Interactions. Cancer Res. 2009, 69, 8067–8075. [Google Scholar] [CrossRef]
  71. Simon, S.; Labarriere, N. PD-1 expression on tumor-specific T cells: Friend or foe for immunotherapy? Oncoimmunology 2017, 7, e1364828. [Google Scholar] [CrossRef]
  72. 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. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297. [Google Scholar] [CrossRef] [PubMed]
  73. Li, Q.; Han, J.; Yang, Y.; Chen, Y. PD-1/PD-L1 checkpoint inhibitors in advanced hepatocellular carcinoma immunotherapy. Front. Immunol. 2022, 13, 1070961. [Google Scholar] [CrossRef] [PubMed]
  74. Harkus, U.; Wankell, M.; Palamuthusingam, P.; McFarlane, C.; Hebbard, L. Immune checkpoint inhibitors in HCC: Cellular, molecular and systemic data. Semin. Cancer Biol. 2022, 86 Pt 3, 799–815. [Google Scholar] [CrossRef]
  75. Anagnostou, V.K.; Brahmer, J.R. Cancer Immunotherapy: A Future Paradigm Shift in the Treatment of Non–Small Cell Lung Cancer. Clin. Cancer Res. 2015, 21, 976–984. [Google Scholar] [CrossRef]
  76. Kudo, M.; Motomura, K.; Wada, Y.; Inaba, Y.; Sakamoto, Y.; Kurosaki, M.; Umeyama, Y.; Kamei, Y.; Yoshimitsu, J.; Fujii, Y.; et al. Avelumab in Combination with Axitinib as First-Line Treatment in Patients with Advanced Hepatocellular Carcinoma: Results from the Phase 1b VEGF Liver 100 Trial. Liver Cancer 2021, 10, 249–259. [Google Scholar] [CrossRef] [PubMed]
  77. Duan, J.; Cui, L.; Zhao, X.; Bai, H.; Cai, S.; Wang, G.; Zhao, Z.; Zhao, J.; Chen, S.; Song, J.; et al. Use of Immunotherapy with Programmed Cell Death 1 vs Programmed Cell Death Ligand 1 Inhibitors in Patients with Cancer. JAMA Oncol. 2020, 6, 375–384. [Google Scholar] [CrossRef]
  78. Callahan, M.K.; Wolchok, J.D.; Allison, J.P. Anti–CTLA-4 Antibody Therapy: Immune Monitoring During Clinical Development of a Novel Immunotherapy. Semin. Oncol. 2010, 37, 473–484. [Google Scholar] [CrossRef]
  79. Manzotti, C.N.; Liu, M.K.P.; Burke, F.; Dussably, L.; Zheng, Y.; Sansom, D.M. Integration of CD28 and CTLA-4 function results in differential responses of T cells to CD80 and CD86. Eur. J. Immunol. 2006, 36, 1413–1422. [Google Scholar] [CrossRef]
  80. Qureshi, O.S.; Zheng, Y.; Nakamura, K.; Attridge, K.; Manzotti, C.; Schmidt, E.M.; Baker, J.; Jeffery, L.E.; Kaur, S.; Briggs, Z.; et al. Trans-Endocytosis of CD80 and CD86: A Molecular Basis for the Cell-Extrinsic Function of CTLA-4. Science 2011, 332, 600–603. [Google Scholar] [CrossRef]
  81. Hosseini, A.; Gharibi, T.; Marofi, F.; Babaloo, Z.; Baradaran, B. CTLA-4: From mechanism to autoimmune therapy. Int. Immunopharmacol. 2020, 80, 106221. [Google Scholar] [CrossRef]
  82. Han, Y.; Chen, Z.; Yang, Y.; Jiang, Z.; Gu, Y.; Liu, Y.; Lin, C.; Pan, Z.; Yu, Y.; Jiang, M.; et al. Human CD14+CTLA-4+regulatory dendritic cells suppress T-cell response by cytotoxic T-lymphocyte antigen-4-dependent IL-10 and indoleamine-2,3-dioxygenase production in hepatocellular carcinoma. Hepatology 2014, 59, 567–579. [Google Scholar] [CrossRef] [PubMed]
  83. Mandlik, D.S.; Mandlik, S.K.; Choudhary, H.B. Immunotherapy for hepatocellular carcinoma: Current status and future perspectives. World J. Gastroenterol. 2023, 29, 1054–1075. [Google Scholar] [CrossRef] [PubMed]
  84. Greten, T.F.; Mauda-Havakuk, M.; Heinrich, B.; Korangy, F.; Wood, B.J. Combined locoregional-immunotherapy for liver cancer. J. Hepatol. 2019, 70, 999–1007. [Google Scholar] [CrossRef] [PubMed]
  85. Greten, T.F.; Duffy, A.G.; Korangy, F. Hepatocellular Carcinoma from an Immunologic Perspective. Clin. Cancer Res. 2013, 19, 6678–6685. [Google Scholar] [CrossRef] [PubMed]
  86. Pinato, D.J.; Murray, S.M.; Forner, A.; Kaneko, T.; Fessas, P.; Toniutto, P.; Mínguez, B.; Cacciato, V.; Avellini, C.; Diaz, A.; et al. Trans-arterial chemoembolization as a loco-regional inducer of immunogenic cell death in hepatocellular carcinoma: Implications for immunotherapy. J. Immunother. Cancer 2021, 9, e003311. [Google Scholar] [CrossRef]
  87. Barker, H.E.; Paget, J.T.E.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425, Erratum in Nat. Rev. Cancer 2015, 15, 509. [Google Scholar] [CrossRef]
  88. Rubner, Y.; Wunderlich, R.; Rühle, P.-F.; Kulzer, L.; Werthmöller, N.; Frey, B.; Weiss, E.-M.; Keilholz, L.; Fietkau, R.; Gaipl, U.S. How Does Ionizing Irradiation Contribute to the Induction of Anti-Tumor Immunity? Front. Oncol. 2012, 2, 75. [Google Scholar] [CrossRef]
  89. Casares, N.; Pequignot, M.O.; Tesniere, A.; Ghiringhelli, F.; Roux, S.; Chaput, N.; Schmitt, E.; Hamai, A.; Hervas-Stubbs, S.; Obeid, M.; et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J. Exp. Med. 2005, 202, 1691–1701. [Google Scholar] [CrossRef]
  90. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
  91. Tonnus, W.; Meyer, C.; Paliege, A.; Belavgeni, A.; Von Mässenhausen, A.; Bornstein, S.R.; Hugo, C.; Becker, J.U.; Linkermann, A. The pathological features of regulated necrosis. J. Pathol. 2019, 247, 697–707. [Google Scholar] [CrossRef]
  92. Asadzadeh, Z.; Safarzadeh, E.; Safaei, S.; Baradaran, A.; Mohammadi, A.; Hajiasgharzadeh, K.; Derakhshani, A.; Argentiero, A.; Silvestris, N.; Baradaran, B. Current Approaches for Combination Therapy of Cancer: The Role of Immunogenic Cell Death. Cancers 2020, 12, 1047. [Google Scholar] [CrossRef] [PubMed]
  93. Birmpilis, A.I.; Paschalis, A.; Mourkakis, A.; Christodoulou, P.; Kostopoulos, I.V.; Antimissari, E.; Terzoudi, G.; Georgakilas, A.G.; Armpilia, C.; Papageorgis, P.; et al. Immunogenic Cell Death, DAMPs and Prothymosin α as a Putative Anticancer Immune Response Biomarker. Cells 2022, 11, 1415. [Google Scholar] [CrossRef] [PubMed]
  94. Kroemer, G.; Galassi, C.; Zitvogel, L.; Galluzzi, L. Immunogenic cell stress and death. Nat. Immunol. 2022, 23, 487–500. [Google Scholar] [CrossRef] [PubMed]
  95. Jing, X.; Zhou, Y.; Xu, X.; Ding, J.; Wang, F.; Wang, Y.; Wang, P. Dynamic changes of T-cell subsets and their relation with tumor recurrence after microwave ablation in patients with hepatocellular carcinoma. J. Cancer Res. Ther. 2018, 14, 40–45. [Google Scholar] [CrossRef]
  96. Zhang, H.; Hou, X.; Cai, H.; Zhuang, X. Effects of microwave ablation on T-cell subsets and cytokines of patients with hepatocellular carcinoma. Minim. Invasive Ther. Allied Technol. 2017, 26, 207–211. [Google Scholar] [CrossRef]
  97. Mizukoshi, E.; Yamashita, T.; Arai, K.; Sunagozaka, H.; Ueda, T.; Arihara, F.; Kagaya, T.; Yamashita, T.; Fushimi, K.; Kaneko, S. Enhancement of tumor-associated antigen-specific T cell responses by radiofrequency ablation of hepatocellular carcinoma. Hepatology 2013, 57, 1448–1457. [Google Scholar] [CrossRef]
  98. Zerbini, A.; Pilli, M.; Fagnoni, F.; Pelosi, G.; Pizzi, M.G.; Schivazappa, S.; Laccabue, D.; Cavallo, C.; Schianchi, C.; Ferrari, C.; et al. Increased Immunostimulatory Activity Conferred to Antigen-presenting Cells by Exposure to Antigen Extract from Hepatocellular Carcinoma After Radiofrequency Thermal Ablation. J. Immunother. 2008, 31, 271–282. [Google Scholar] [CrossRef]
  99. Ji, L.; Gu, J.; Chen, L.; Miao, D. Changes of Th1/Th2 cytokines in patients with primary hepatocellular carcinoma after ultrasound-guided ablation. Int. J. Clin. Exp. Pathol. 2017, 10, 8715–8720. [Google Scholar]
  100. Kohles, N.; Nagel, D.; Jüngst, D.; Stieber, P.; Holdenrieder, S. Predictive value of immunogenic cell death biomarkers HMGB1, sRAGE, and DNase in liver cancer patients receiving transarterial chemoembolization therapy. Tumor Biol. 2012, 33, 2401–2409. [Google Scholar] [CrossRef]
  101. Liao, J.; Xiao, J.; Zhou, Y.; Liu, Z.; Wang, C. Effect of transcatheter arterial chemoembolization on cellular immune function and regulatory T cells in patients with hepatocellular carcinoma. Mol. Med. Rep. 2015, 12, 6065–6071. [Google Scholar] [CrossRef]
  102. Liao, Y.; Wang, B.; Huang, Z.-L.; Shi, M.; Yu, X.-J.; Zheng, L.; Li, S.; Li, L. Increased Circulating Th17 Cells after Transarterial Chemoembolization Correlate with Improved Survival in Stage III Hepatocellular Carcinoma: A Prospective Study. PLoS ONE 2013, 8, e60444. [Google Scholar] [CrossRef] [PubMed]
  103. Chew, V.; Lee, Y.H.; Pan, L.; Nasir, N.J.M.; Lim, C.J.; Chua, C.; Lai, L.; Hazirah, S.N.; Lim, T.K.H.; Goh, B.K.P.; et al. Immune activation underlies a sustained clinical response to Yttrium-90 radioembolisation in hepatocellular carcinoma. Gut 2019, 68, 335–346. [Google Scholar] [CrossRef]
  104. Fernandez-Ros, N.; Iñarrairaegui, M.; Paramo, J.A.; Berasain, C.; Avila, M.A.; Chopitea, A.; Varo, N.; Sarobe, P.; Bilbao, J.I.; Dominguez, I.; et al. Radioembolization of hepatocellular carcinoma activates liver regeneration, induces inflammation and endothelial stress and activates coagulation. Liver Int. 2015, 35, 1590–1596. [Google Scholar] [CrossRef] [PubMed]
  105. Seidensticker, M.; Powerski, M.; Seidensticker, R.; Damm, R.; Mohnike, K.; Garlipp, B.; Klopffleisch, M.; Amthauer, H.; Ricke, J.; Pech, M. Cytokines and 90Y-Radioembolization: Relation to Liver Function and Overall Survival. Cardiovasc. Interv. Radiol. 2017, 40, 1185–1195. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, J.; Li, H.; Gao, D.; Zhang, B.; Zheng, M.; Lun, M.; Wei, M.; Duan, R.; Guo, M.; Hua, J.; et al. A prognosis and impact factor analysis of DC-CIK cell therapy for patients with hepatocellular carcinoma undergoing postoperative TACE. Cancer Biol. Ther. 2018, 19, 475–483. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, H.L.; Jang, J.W.; Lee, S.W.; Yoo, S.H.; Kwon, J.H.; Nam, S.W.; Bae, S.H.; Choi, J.Y.; Han, N.I.; Yoon, S.K. Inflammatory cytokines and change of Th1/Th2 balance as prognostic indicators for hepatocellular carcinoma in patients treated with transarterial chemoembolization. Sci. Rep. 2019, 9, 3260. [Google Scholar] [CrossRef]
  108. Chen, M.-F.; Chen, P.-T.; Chen, W.-C.; Lu, M.-S.; Lin, P.-Y.; Lee, K.-D. The role of PD-L1 in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression. Oncotarget 2016, 7, 7913–7924. [Google Scholar] [CrossRef]
  109. Duan, X.-H.; Li, H.; Han, X.-W.; Ren, J.-Z.; Li, F.-Y.; Ju, S.-G.; Chen, P.-F.; Kuang, D.-L. Upregulation of IL-6 is involved in moderate hyperthermia induced proliferation and invasion of hepatocellular carcinoma cells. Eur. J. Pharmacol. 2018, 833, 230–236. [Google Scholar] [CrossRef]
  110. Domouchtsidou, A.; Barsegian, V.; Mueller, S.P.; Best, J.; Ertle, J.; Bedreli, S.; Horn, P.A.; Bockisch, A.; Lindemann, M. Impaired lymphocyte function in patients with hepatic malignancies after selective internal radiotherapy. Cancer Immunol. Immunother. 2018, 67, 843–853. [Google Scholar] [CrossRef]
  111. Han, J.-W.; Yoon, S.-K. Immune Responses Following Locoregional Treatment for Hepatocellular Carcinoma: Possible Roles of Adjuvant Immunotherapy. Pharmaceutics 2021, 13, 1387. [Google Scholar] [CrossRef]
  112. Zeng, P.; Shen, D.; Zeng, C.-H.; Chang, X.-F.; Teng, G.-J. Emerging Opportunities for Combining Locoregional Therapy with Immune Checkpoint Inhibitors in Hepatocellular Carcinoma. Curr. Oncol. Rep. 2020, 22, 76. [Google Scholar] [CrossRef]
  113. Gravante, G.; Sconocchia, G.; Ong, S.L.; Dennison, A.; Lloyd, D.M. Immunoregulatory effects of liver ablation therapies for the treatment of primary and metastatic liver malignancies. Liver Int. 2009, 29, 18–24. [Google Scholar] [CrossRef] [PubMed]
  114. Nakatsura, T.; Nobuoka, D.; Motomura, Y.; Shirakawa, H.; Yoshikawa, T.; Kuronuma, T.; Takahashi, M.; Nakachi, K.; Ishii, H.; Furuse, J.; et al. Radiofrequency ablation for hepatocellular carcinoma induces glypican-3 peptide-specific cytotoxic T lymphocytes. Int. J. Oncol. 2012, 40, 63–70. [Google Scholar] [CrossRef]
  115. Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015, 28, 690–714. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, Y.-J.; Fletcher, R.; Yu, J.; Zhang, L. Immunogenic effects of chemotherapy-induced tumor cell death. Genes Dis. 2018, 5, 194–203. [Google Scholar] [CrossRef] [PubMed]
  117. Apetoh, L.; Mignot, G.; Panaretakis, T.; Kroemer, G.; Zitvogel, L. Immunogenicity of anthracyclines: Moving towards more personalized medicine. Trends Mol. Med. 2008, 14, 141–151. [Google Scholar] [CrossRef] [PubMed]
  118. Kloeckner, R.; Galle, P.R.; Bruix, J. Local and Regional Therapies for Hepatocellular Carcinoma. Hepatology 2021, 73 (Suppl. S1), 137–149. [Google Scholar] [CrossRef]
  119. Cho, Y.K.; Kim, J.K.; Kim, M.Y.; Rhim, H.; Han, J.K. Systematic review of randomized trials for hepatocellular carcinoma treated with percutaneous ablation therapies. Hepatology 2008, 49, 453–459. [Google Scholar] [CrossRef]
  120. Lencioni, R. Loco-regional treatment of hepatocellular carcinoma. Hepatology 2010, 52, 762–773. [Google Scholar] [CrossRef]
  121. Foerster, F.; Gairing, S.J.; Ilyas, S.I.; Galle, P.R. Emerging immunotherapy for HCC: A guide for hepatologists. Hepatology 2022, 75, 1604–1626. [Google Scholar] [CrossRef]
  122. Marzi, L.; Mega, A.; Gitto, S.; Pelizzaro, F.; Seeber, A.; Spizzo, G. Impact and Novel Perspective of Immune Checkpoint Inhibitors in Patients with Early and Intermediate Stage HCC. Cancers 2022, 14, 3332. [Google Scholar] [CrossRef] [PubMed]
  123. Duffy, A.G.; Ulahannan, S.V.; Makorova-Rusher, O.; Rahma, O.; Wedemeyer, H.; Pratt, D.; Davis, J.L.; Hughes, M.S.; Heller, T.; ElGindi, M.; et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J. Hepatol. 2017, 66, 545–551. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, X.; Liu, G.; Chen, S.; Bi, H.; Xia, F.; Feng, K.; Ma, K.; Ni, B. Combination therapy with PD-1 blockade and radiofrequency ablation for recurrent hepatocellular carcinoma: A propensity score matching analysis. Int. J. Hyperth. 2021, 38, 1519–1528. [Google Scholar] [CrossRef]
  125. Lyu, N.; Kong, Y.; Li, X.; Mu, L.; Deng, H.; Chen, H.; He, M.; Lai, J.; Li, J.; Tang, H.; et al. Ablation Reboots the Response in Advanced Hepatocellular Carcinoma with Stable or Atypical Response During PD-1 Therapy: A Proof-of-Concept Study. Front. Oncol. 2020, 10, 580241. [Google Scholar] [CrossRef]
  126. Zhang, J.-X.; Chen, P.; Liu, S.; Zu, Q.-Q.; Shi, H.-B.; Zhou, C.-G. Safety and Efficacy of Transarterial Chemoembolization and Immune Checkpoint Inhibition with Camrelizumab for Treatment of Unresectable Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2022, 9, 265–272. [Google Scholar] [CrossRef]
  127. You, R.; Xu, Q.; Wang, Q.; Zhang, Q.; Zhou, W.; Cao, C.; Huang, X.; Ji, H.; Lv, P.; Jiang, H.; et al. Efficacy and safety of camrelizumab plus transarterial chemoembolization in intermediate to advanced hepatocellular carcinoma patients: A prospective, multi-center, real-world study. Front. Oncol. 2022, 12, 816198. [Google Scholar] [CrossRef] [PubMed]
  128. Ren, Y.; Liu, Z.; Makamure, J.; Kan, X.; Song, S.; Liu, Y.; Qian, K.; Zheng, C.; Liang, B. Addition of Camrelizumab to Transarterial Chemoembolization in Hepatocellular Carcinoma with Untreatable Progression. Technol. Cancer Res. Treat. 2022, 21, 15330338221131385. [Google Scholar] [CrossRef]
  129. Guo, Y.; Ren, Y.; Chen, L.; Sun, T.; Zhang, W.; Sun, B.; Zhu, L.; Xiong, F.; Zheng, C. Transarterial chemoembolization combined with camrelizumab for recurrent hepatocellular carcinoma. BMC Cancer 2022, 22, 270. [Google Scholar] [CrossRef]
  130. Ren, Y.; Guo, Y.; Chen, L.; Sun, T.; Zhang, W.; Sun, B.; Zhu, L.; Xiong, F.; Zheng, C. Efficacy of Drug-Eluting Beads Transarterial Chemoembolization Plus Camrelizumab Compared with Conventional Transarterial Chemoembolization Plus Camrelizumab for Unresectable Hepatocellular Carcinoma. Cancer Control 2022, 29, 10732748221076806. [Google Scholar] [CrossRef]
  131. Tai, D.; Loke, K.; Gogna, A.; Kaya, N.A.; Tan, S.H.; Hennedige, T.; Ng, D.; Irani, F.; Lee, J.; Lim, J.Q.; et al. Radioembolisation with Y90-resin microspheres followed by nivolumab for advanced hepatocellular carcinoma (CA 209-678): A single arm, single centre, phase 2 trial. Lancet Gastroenterol. Hepatol. 2021, 6, 1025–1035. [Google Scholar] [CrossRef]
  132. Zhan, C.; Ruohoniemi, D.; Shanbhogue, K.P.; Wei, J.; Welling, T.H.; Gu, P.; Park, J.S.; Dagher, N.N.; Taslakian, B.; Hickey, R.M. Safety of Combined Yttrium-90 Radioembolization and Immune Checkpoint Inhibitor Immunotherapy for Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2020, 31, 25–34. [Google Scholar] [CrossRef]
  133. De la Torre-Aláez, M.; Matilla, A.; Varela, M.; Iñarrairaegui, M.; Reig, M.; Lledó, J.L.; Arenas, J.I.; Lorente, S.; Testillano, M.; Márquez, L.; et al. Nivolumab after selective internal radiation therapy for the treatment of hepatocellular carcinoma: A phase 2, single-arm study. J. Immunother. Cancer 2023, 10, e005457, Erratum in J. Immunother. Cancer 2023, 11, e005457corr1. [Google Scholar] [CrossRef] [PubMed]
  134. Marinelli, B.; Cedillo, M.; Pasik, S.D.; Charles, D.; Murthy, S.; Patel, R.S.; Fischman, A.; Ranade, M.; Bishay, V.; Nowakowski, S.; et al. Safety and Efficacy of Locoregional Treatment during Immunotherapy with Nivolumab for Hepatocellular Carcinoma: A Retrospective Study of 41 Interventions in 29 Patients. J. Vasc. Interv. Radiol. 2020, 31, 1729–1738.e1. [Google Scholar] [CrossRef] [PubMed]
  135. Llovet, J.M.; Villanueva, A.; Marrero, J.A.; Schwartz, M.; Meyer, T.; Galle, P.R.; Lencioni, R.; Greten, T.F.; Kudo, M.; Mandrekar, S.J.; et al. Trial Design and Endpoints in Hepatocellular Carcinoma: AASLD Consensus Conference. Hepatology 2021, 73 (Suppl. S1), 158–191. [Google Scholar] [CrossRef]
  136. Biondetti, P.; Saggiante, L.; Ierardi, A.M.; Iavarone, M.; Sangiovanni, A.; Pesapane, F.; Fumarola, E.M.; Lampertico, P.; Carrafiello, G. Interventional Radiology Image-Guided Locoregional Therapies (LRTs) and Immunotherapy for the Treatment of HCC. Cancers 2021, 13, 5797. [Google Scholar] [CrossRef] [PubMed]
  137. Haber, P.K.; Puigvehí, M.; Castet, F.; Lourdusamy, V.; Montal, R.; Tabrizian, P.; Buckstein, M.; Kim, E.; Villanueva, A.; Schwartz, M.; et al. Evidence-Based Management of Hepatocellular Carcinoma: Systematic Review and Meta-analysis of Randomized Controlled Trials (2002–2020). Gastroenterology 2021, 161, 879–898. [Google Scholar] [CrossRef] [PubMed]
  138. Cheng, A.-L.; Hsu, C.; Chan, S.L.; Choo, S.-P.; Kudo, M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J. Hepatol. 2020, 72, 307–319. [Google Scholar] [CrossRef]
  139. Giannini, E.G.; Aglitti, A.; Borzio, M.; Gambato, M.; Guarino, M.; Iavarone, M.; Lai, Q.; Sandri, G.B.L.; Melandro, F.; Morisco, F.; et al. Overview of Immune Checkpoint Inhibitors Therapy for Hepatocellular Carcinoma, and The ITA.LI.CA Cohort Derived Estimate of Amenability Rate to Immune Checkpoint Inhibitors in Clinical Practice. Cancers 2019, 11, 1689. [Google Scholar] [CrossRef] [PubMed]
  140. Renzulli, M.; Ramai, D.; Singh, J.; Sinha, S.; Brandi, N.; Ierardi, A.M.; Albertini, E.; Sacco, R.; Facciorusso, A.; Golfieri, R. Locoregional Treatments in Cholangiocarcinoma and Combined Hepatocellular Cholangiocarcinoma. Cancers 2021, 13, 3336. [Google Scholar] [CrossRef]
  141. Lantuejoul, S.; Tsao, M.; Cooper, W.A.; Girard, N.; Hirsch, F.R.; Roden, A.C.; Lopez-Rios, F.; Jain, D.; Chou, T.-Y.; Motoi, N.; et al. PD-L1 Testing for Lung Cancer in 2019: Perspective from the IASLC Pathology Committee. J. Thorac. Oncol. 2020, 15, 499–519. [Google Scholar] [CrossRef]
  142. Jung, H.I.; Jeong, D.; Ji, S.; Ahn, T.S.; Bae, S.H.; Chin, S.; Chung, J.C.; Kim, H.C.; Lee, M.S.; Baek, M.-J. Overexpression of PD-L1 and PD-L2 Is Associated with Poor Prognosis in Patients with Hepatocellular Carcinoma. Cancer Res. Treat. 2017, 49, 246–254. [Google Scholar] [CrossRef] [PubMed]
  143. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
  144. Totoki, Y.; Tatsuno, K.; Covington, K.R.; Ueda, H.; Creighton, C.J.; Kato, M.; Tsuji, S.; Donehower, L.A.; Slagle, B.L.; Nakamura, H.; et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 2014, 46, 1267–1273. [Google Scholar] [CrossRef]
  145. Zucman-Rossi, J.; Villanueva, A.; Nault, J.-C.; Llovet, J.M. Genetic Landscape and Biomarkers of Hepatocellular Carcinoma. Gastroenterology 2015, 149, 1226–1239.e4. [Google Scholar] [CrossRef] [PubMed]
  146. Harding, J.J.; Nandakumar, S.; Armenia, J.; Khalil, D.N.; Albano, M.; Ly, M.; Shia, J.; Hechtman, J.F.; Kundra, R.; El Dika, I.; et al. Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies. Clin. Cancer Res. 2019, 25, 2116–2126. [Google Scholar] [CrossRef]
  147. Rizzo, A.; Ricci, A.D.; Di Federico, A.; Frega, G.; Palloni, A.; Tavolari, S.; Brandi, G. Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy in Hepatocellular Carcinoma: Where Do We Stand? Front. Oncol. 2021, 11, 803133. [Google Scholar] [CrossRef]
  148. Filgueira, N.A. Hepatocellular carcinoma recurrence after liver transplantation: Risk factors, screening and clinical presentation. World J. Hepatol. 2019, 11, 261–272. [Google Scholar] [CrossRef] [PubMed]
  149. D’izarny-Gargas, T.; Durrbach, A.; Zaidan, M. Efficacy and tolerance of immune checkpoint inhibitors in transplant patients with cancer: A systematic review. Am. J. Transplant. 2020, 20, 2457–2465. [Google Scholar] [CrossRef]
  150. Bartlett, A.S.; McCall, J.L.; Ameratunga, R.; Howden, B.; Yeong, M.; Benjamin, C.D.; Hess, D.; Peach, R.; Munn, S.R. Costimulatory blockade prevents early rejection, promotes lymphocyte apoptosis, and inhibits the upregulation of intragraft interleukin-6 in an orthotopic liver transplant model in the rat. Liver Transplant. 2002, 8, 458–468. [Google Scholar] [CrossRef]
  151. Pinato, D.J.; Fessas, P.; Cortellini, A.; Rimassa, L. Combined PD-1/VEGFR Blockade: A New Era of Treatment for Hepatocellular Cancer. Clin. Cancer Res. 2021, 27, 908–910. [Google Scholar] [CrossRef]
  152. Chen, J.; Jiang, C.; Jin, L.; Zhang, X. Regulation of PD-L1: A novel role of pro-survival signalling in cancer. Ann. Oncol. 2016, 27, 409–416. [Google Scholar] [CrossRef]
  153. Yang, F.; Yang, J.; Xiang, W.; Zhong, B.-Y.; Li, W.-C.; Shen, J.; Zhang, S.; Yin, Y.; Sun, H.-P.; Wang, W.-S.; et al. Safety and Efficacy of Transarterial Chemoembolization Combined with Immune Checkpoint Inhibitors and Tyrosine Kinase Inhibitors for Hepatocellular Carcinoma. Front. Oncol. 2022, 11, 657512. [Google Scholar] [CrossRef] [PubMed]
  154. Cao, F.; Yang, Y.; Si, T.; Luo, J.; Zeng, H.; Zhang, Z.; Feng, D.; Chen, Y.; Zheng, J. The Efficacy of TACE Combined with Lenvatinib Plus Sintilimab in Unresectable Hepatocellular Carcinoma: A Multicenter Retrospective Study. Front. Oncol. 2021, 11, 783480. [Google Scholar] [CrossRef] [PubMed]
  155. Teng, Y.; Ding, X.; Li, W.; Sun, W.; Chen, J. A Retrospective Study on Therapeutic Efficacy of Transarterial Chemoembolization Combined with Immune Checkpoint Inhibitors Plus Lenvatinib in Patients with Unresectable Hepatocellular Carcinoma. Technol. Cancer Res. Treat. 2022, 21, 15330338221075174. [Google Scholar] [CrossRef] [PubMed]
  156. Zheng, L.; Fang, S.; Wu, F.; Chen, W.; Chen, M.; Weng, Q.; Wu, X.; Song, J.; Zhao, Z.; Ji, J. Efficacy and Safety of TACE Combined with Sorafenib Plus Immune Checkpoint Inhibitors for the Treatment of Intermediate and Advanced TACE-Refractory Hepatocellular Carcinoma: A Retrospective Study. Front. Mol. Biosci. 2021, 7, 609322. [Google Scholar] [CrossRef] [PubMed]
  157. Ju, S.; Zhou, C.; Yang, C.; Wang, C.; Liu, J.; Wang, Y.; Huang, S.; Li, T.; Chen, Y.; Bai, Y.; et al. Apatinib Plus Camrelizumab with/without Chemoembolization for Hepatocellular Carcinoma: A Real-World Experience of a Single Center. Front. Oncol. 2022, 11, 835889. [Google Scholar] [CrossRef]
  158. Chen, S.; Wu, Z.; Shi, F.; Mai, Q.; Wang, L.; Wang, F.; Zhuang, W.; Chen, X.; Chen, H.; Xu, B.; et al. Lenvatinib plus TACE with or without pembrolizumab for the treatment of initially unresectable hepatocellular carcinoma harbouring PD-L1 expression: A retrospective study. J. Cancer Res. Clin. Oncol. 2022, 148, 2115–2125. [Google Scholar] [CrossRef] [PubMed]
  159. Qin, J.; Huang, Y.; Zhou, H.; Yi, S. Efficacy of Sorafenib Combined with Immunotherapy Following Transarterial Chemoembolization for Advanced Hepatocellular Carcinoma: A Propensity Score Analysis. Front. Oncol. 2022, 12, 807102. [Google Scholar] [CrossRef]
  160. Wang, M.; Sun, L.; Han, X.; Ren, J.; Li, H.; Wang, W.; Xu, W.; Liang, C.; Duan, X. The addition of camrelizumab is effective and safe among unresectable hepatocellular carcinoma patients who progress after drug-eluting bead transarterial chemoembolization plus apatinib therapy. Clin. Res. Hepatol. Gastroenterol. 2023, 47, 102060. [Google Scholar] [CrossRef] [PubMed]
  161. Li, X.; Zhang, Q.; Lu, Q.; Cheng, Z.; Liu, F.; Han, Z.; Yu, X.; Yu, J.; Liang, P. Microwave ablation combined with apatinib and camrelizumab in patients with advanced hepatocellular carcinoma: A single-arm, preliminary study. Front. Immunol. 2022, 13, 1023983. [Google Scholar] [CrossRef]
  162. Chen, Z.-N.; Mi, L.; Xu, J.; Song, F.; Zhang, Q.; Zhang, Z.; Xing, J.-L.; Bian, H.-J.; Jiang, J.-L.; Wang, X.-H.; et al. Targeting radioimmunotherapy of hepatocellular carcinoma with iodine (131I) metuximab injection: Clinical Phase I/II trials. Int. J. Radiat. Oncol. 2006, 65, 435–444. [Google Scholar] [CrossRef]
  163. Zhang, Q.; Zhou, J.; Ku, X.-M.; Chen, X.-G.; Zhang, L.; Xu, J.; Chen, G.-S.; Li, Q.; Qian, F.; Tian, R.; et al. Expression of CD147 as a significantly unfavorable prognostic factor in hepatocellular carcinoma. Eur. J. Cancer Prev. 2007, 16, 196–202. [Google Scholar] [CrossRef]
  164. He, Q.; Lu, W.S.; Liu, Y.; Guan, Y.S. Kuang AR131I-labeled metuximab combined with chemoembolization for unresectable hepatocellular carcinoma. World J. Gastroenterol. 2013, 19, 9104–9110. [Google Scholar] [CrossRef]
  165. Zhu, Z.-X.; Liao, M.-H.; Wang, X.-X.; Huang, J.-W. Transcatheter Arterial Chemoembolization Plus131I-Labelled Metuximab versus Transcatheter Arterial Chemoembolization Alone in Intermediate/Advanced Stage Hepatocellular Carcinoma: A Systematic Review and Meta-Analysis. Korean J. Radiol. 2016, 17, 882–892. [Google Scholar] [CrossRef]
  166. Chen, H.; Nan, G.; Wei, D.; Zhai, R.-Y.; Huang, M.; Yang, W.-W.; Xing, B.-C.; Zhu, X.; Xu, H.-F.; Wang, X.-D.; et al. Hepatic Artery Injection of 131I-Metuximab Combined with Transcatheter Arterial Chemoembolization for Unresectable Hepatocellular Carcinoma: A Prospective Nonrandomized, Multicenter Clinical Trial. J. Nucl. Med. 2022, 63, 556–559. [Google Scholar] [CrossRef]
  167. Bian, H.; Zheng, J.-S.; Nan, G.; Li, R.; Chen, C.; Hu, C.-X.; Zhang, Y.; Sun, B.; Wang, X.-L.; Cui, S.-C.; et al. Randomized Trial of [131I] Metuximab in Treatment of Hepatocellular Carcinoma After Percutaneous Radiofrequency Ablation. J. Natl. Cancer Inst. 2014, 106, dju239. [Google Scholar] [CrossRef] [PubMed]
  168. Rochigneux, P.; Chanez, B.; De Rauglaudre, B.; Mitry, E.; Chabannon, C.; Gilabert, M. Adoptive Cell Therapy in Hepatocellular Carcinoma: Biological Rationale and First Results in Early Phase Clinical Trials. Cancers 2021, 13, 271. [Google Scholar] [CrossRef]
  169. Dendy, M.S.; Ludwig, J.M.; Stein, S.M.; Kim, H.S. Locoregional Therapy, Immunotherapy and the Combination in Hepatocellular Carcinoma: Future Directions. Liver Cancer 2019, 8, 326–340. [Google Scholar] [CrossRef] [PubMed]
  170. Ding, M.; Wang, Y.; Chi, J.; Wang, T.; Tang, X.; Cui, D.; Qian, Q.; Zhai, B. Is Adjuvant Cellular Immunotherapy Essential after TACE-Predominant Minimally-Invasive Treatment for Hepatocellular Carcinoma? A Systematic Meta-Analysis of Studies Including 1774 Patients. PLoS ONE 2016, 11, e0168798. [Google Scholar] [CrossRef] [PubMed]
  171. Li, Y.-C.; Zhao, L.; Wu, J.-P.; Qu, C.-X.; Song, Q.-K.; Wang, R.-B. Cytokine-induced killer cell infusion combined with conventional treatments produced better prognosis for hepatocellular carcinoma patients with barcelona clinic liver cancer B or earlier stage: A systematic review and meta-analysis. Cytotherapy 2016, 18, 1525–1531. [Google Scholar] [CrossRef]
  172. Li, X.; Dai, D.; Song, X.; Liu, J.; Zhu, L.; Xu, W. A meta-analysis of cytokine-induced killer cells therapy in combination with minimally invasive treatment for hepatocellular carcinoma. Clin. Res. Hepatol. Gastroenterol. 2014, 38, 583–591. [Google Scholar] [CrossRef] [PubMed]
  173. Cui, J.; Wang, N.; Zhao, H.; Jin, H.; Wang, G.; Niu, C.; Terunuma, H.; He, H.; Li, W. Combination of radiofrequency ablation and sequential cellular immunotherapy improves progression-free survival for patients with hepatocellular carcinoma. Int. J. Cancer 2014, 134, 342–351. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, T.; Chen, J.; Niu, L.; Liu, Y.; Ye, G.; Jiang, M.; Qi, Z. Clinical Safety and Efficacy of Locoregional Therapy Combined with Adoptive Transfer of Allogeneic γδ T Cells for Advanced Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. J. Vasc. Interv. Radiol. 2021, 33, 19–27.e3. [Google Scholar] [CrossRef] [PubMed]
  175. Qiu, Y.; Xu, M.-B.; Yun, M.M.; Wang, Y.-Z.; Zhang, R.-M.; Meng, X.-K.; Ou-Yang, X.-H.; Yun, S. Hepatocellular carcinoma-specific immunotherapy with synthesized α1,3- galactosyl epitope-pulsed dendritic cells and cytokine-induced killer cells. World J. Gastroenterol. 2011, 17, 5260–5266. [Google Scholar] [CrossRef] [PubMed]
  176. Greten, T.F.; Manns, M.; Korangy, F. Immunotherapy of HCC. Rev. Recent Clin. Trials 2008, 3, 31–39. [Google Scholar] [CrossRef]
  177. Sangro, B.; Sarobe, P.; Hervás-Stubbs, S.; Melero, I. Advances in immunotherapy for hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 525–543. [Google Scholar] [CrossRef]
  178. Schwabe, R.F.; Greten, T.F. Gut microbiome in HCC—Mechanisms, diagnosis and therapy. J. Hepatol. 2020, 72, 230–238. [Google Scholar] [CrossRef]
  179. Dapito, D.H.; Mencin, A.; Gwak, G.-Y.; Pradere, J.-P.; Jang, M.-K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef]
  180. Zheng, Y.; Wang, T.; Tu, X.; Huang, Y.; Zhang, H.; Tan, D.; Jiang, W.; Cai, S.; Zhao, P.; Song, R.; et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. Immunother. Cancer 2019, 7, 193. [Google Scholar] [CrossRef]
  181. Lee, P.-C.; Wu, C.-J.; Hung, Y.-W.; Lee, C.J.; Chi, C.-T.; Lee, I.-C.; Yu-Lun, K.; Chou, S.-H.; Luo, J.-C.; Hou, M.-C.; et al. Gut microbiota and metabolites associate with outcomes of immune checkpoint inhibitor–treated unresectable hepatocellular carcinoma. J. Immunother. Cancer 2022, 10, e004779. [Google Scholar] [CrossRef]
  182. Routy, B.; le Chatelier, E.; DeRosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [PubMed]
  183. Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021, 371, 602–609. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The main locoregional techniques for the treatment of hepatocellular carcinoma.
Figure 1. The main locoregional techniques for the treatment of hepatocellular carcinoma.
Ijms 24 08598 g001
Figure 2. Immune checkpoint inhibitors in hepatocellular carcinoma. PD-1 binding its ligand PD-L1/PD-L2 prevents TCR signaling, blocks T cell proliferation, and induces the exhaustion of T cells. CTLA-4 binds CD80/CD86 and blocks the activation of the T cells. The inhibition of these immune checkpoints with PD-1/PD-L1 inhibitors and CTLA-4 inhibitors promote T cell activation and up-regulate the immune system, thus reactivating the anticancer immune response.
Figure 2. Immune checkpoint inhibitors in hepatocellular carcinoma. PD-1 binding its ligand PD-L1/PD-L2 prevents TCR signaling, blocks T cell proliferation, and induces the exhaustion of T cells. CTLA-4 binds CD80/CD86 and blocks the activation of the T cells. The inhibition of these immune checkpoints with PD-1/PD-L1 inhibitors and CTLA-4 inhibitors promote T cell activation and up-regulate the immune system, thus reactivating the anticancer immune response.
Ijms 24 08598 g002
Figure 3. The rationale behind combining locoregional therapies and immunotherapy. Locoregional therapies, especially when incomplete, can increase the level of pro-angiogenetic cytokines and thus promote neoangiogenesis of residual cancer cells and metastasis development; at the same time, however, they promote systemic immune response by releasing neoantigens into blood circulation, although this immunogenic effect might be too modest. The immunological efficacy of locoregional treatments could be enhanced by their combination with immunotherapeutic drugs, which would promote immune cell activation and proliferation, positively influencing the tumor microenvironment.
Figure 3. The rationale behind combining locoregional therapies and immunotherapy. Locoregional therapies, especially when incomplete, can increase the level of pro-angiogenetic cytokines and thus promote neoangiogenesis of residual cancer cells and metastasis development; at the same time, however, they promote systemic immune response by releasing neoantigens into blood circulation, although this immunogenic effect might be too modest. The immunological efficacy of locoregional treatments could be enhanced by their combination with immunotherapeutic drugs, which would promote immune cell activation and proliferation, positively influencing the tumor microenvironment.
Ijms 24 08598 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brandi, N.; Renzulli, M. The Synergistic Effect of Interventional Locoregional Treatments and Immunotherapy for the Treatment of Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 8598. https://doi.org/10.3390/ijms24108598

AMA Style

Brandi N, Renzulli M. The Synergistic Effect of Interventional Locoregional Treatments and Immunotherapy for the Treatment of Hepatocellular Carcinoma. International Journal of Molecular Sciences. 2023; 24(10):8598. https://doi.org/10.3390/ijms24108598

Chicago/Turabian Style

Brandi, Nicolò, and Matteo Renzulli. 2023. "The Synergistic Effect of Interventional Locoregional Treatments and Immunotherapy for the Treatment of Hepatocellular Carcinoma" International Journal of Molecular Sciences 24, no. 10: 8598. https://doi.org/10.3390/ijms24108598

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop