Next Article in Journal
Determining the Effectiveness of Fibrin Sealants in Reducing Complications in Patients Undergoing Lateral Neck Dissection (DEFeND): A Randomised External Pilot Trial
Next Article in Special Issue
Dose Consideration of Lenvatinib’s Anti-Cancer Effect on Hepatocellular Carcinoma and the Potential Benefit of Combined Colchicine Therapy
Previous Article in Journal
Compromised Blood-Brain Barrier Junctions Enhance Melanoma Cell Intercalation and Extravasation
Previous Article in Special Issue
Association of Hepatobiliary Phase of Gadoxetic-Acid-Enhanced MRI Imaging with Immune Microenvironment and Response to Atezolizumab Plus Bevacizumab Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Combination Therapy of Immune Checkpoint Inhibitors with Locoregional Therapy for Hepatocellular Carcinoma

1
Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan
2
Department of Hepatobiliary Pancreatic and Transplant Surgery, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan
*
Author to whom correspondence should be addressed.
Submission received: 29 September 2023 / Revised: 17 October 2023 / Accepted: 18 October 2023 / Published: 20 October 2023
(This article belongs to the Special Issue Systemic Therapy for Hepatocellular Carcinoma)

Abstract

:

Simple Summary

Immune checkpoint inhibitor (ICI) therapy has recently become the standard treatment for advanced hepatocellular carcinoma (HCC); however, clinical outcomes remain unsatisfactory. Locoregional therapies, such as ablation, transarterial embolization, and radiotherapy, which are usually used for local treatment of HCC at an earlier stage, have been actively explored to enhance ICI efficacy. This review focuses on the rationale and clinical trials of combination therapy with ICIs and locoregional therapy for HCC.

Abstract

Hepatocellular carcinoma (HCC) is estimated to be the fourth leading cause of cancer-related deaths globally, and its overall prognosis is dismal because most cases are diagnosed at a late stage and are unamenable to curative treatment. The emergence of immune checkpoint inhibitors (ICIs) has dramatically improved the therapeutic efficacy for advanced hepatocellular carcinoma; however, their response rates remain unsatisfactory, partly because >50% of HCC exhibit an ICI-nonresponsive tumor microenvironment characterized by a paucity of cytotoxic T cells (immune-cold), as well as difficulty in their infiltration into tumor sites (immune excluded). To overcome this limitation, combination therapies with locoregional therapies, including ablation, transarterial embolization, and radiotherapy, which are usually used for early stage HCCs, have been actively explored to enhance ICI efficacy by promoting the release of tumor-associated antigens and cytokines, and eventually accelerating the so-called cancer–immunity cycle. Various combination therapies have been investigated in early- to late-phase clinical trials, and some have shown promising results. This comprehensive article provides an overview of the immune landscape for HCC to understand ICI efficacy and its limitations and, subsequently, reviews the status of combinatorial therapies of ICIs with locoregional therapy for HCC.

1. Introduction

Hepatocellular carcinoma (HCC), representing >80% of primary liver cancers, is estimated to be the fourth leading cause of cancer-related deaths worldwide [1]. While the early detection of HCC enables curative treatments, such as liver transplantation, surgical resection, and ablation therapies, that lead to prolonged survival [2,3,4,5], the most common HCC stage at diagnosis is an advanced stage that is not amenable to curative treatments in North America, Europe, China, and South Korea, resulting in poor prognosis (5-year survival < 15%) [6]. In addition, as evidenced by the observation that the major cause of death in patients with early-stage HCC is HCC itself due to highly frequent metachronous and multicentric recurrences in chronically injured livers [7], the majority of HCC can eventually progress to an advanced stage even when the primary tumor is diagnosed at an early stage and is curatively treated. Therefore, more efficacious treatments for advanced HCC are required to improve the overall prognosis for HCC patients.
Advancements in immune checkpoint inhibitors (ICIs) have revolutionized systemic antitumor therapy for a variety of malignancies, including HCC. Anti-programmed death ligand-1 (PD-L1) atezolizumab plus anti-vascular endothelial growth factor (VEGF) bevacizumab and anti-programmed cell death protein 1 (PD-1) camrelizumab plus anti-VEGF receptor 2 (VEGFR2) rivoceranib demonstrated prolonged overall survival and progression-free survival compared with sorafenib that had been the only therapeutic option for unresectable HCC since 2008 [8,9,10]. A combination regimen of anti-PD-L1 durvalumab and anti-cytotoxic T-lymphocyte antigen 4 (CTLA4) tremelimumab also demonstrated significantly improved overall survival compared with sorafenib, with a 3-year survival rate of 30.7% [11]. Consequently, multiple expert societies consistently recommend ICIs as first-line systemic therapy for unresectable HCC, unless contraindications are identified [12]. Furthermore, the use of ICI in adjuvant or neoadjuvant settings has also been intensively investigated. Numerous clinical trials are ongoing to test antibodies targeting other immune checkpoints, such as lymphocyte activation gene 3 (LAG3), T cell immunoglobulin mucin-3 (TIM3), and T cell immunoreceptor with lg and ITIM domains (TIGIT) [13,14,15]. Thus, ICIs will continue to be the primary therapeutic option for HCC over the next decade.
Nevertheless, their current response rates are limited, with an objective response rate of 20–30% in clinical trials and real-world studies [8,11,16,17]. On the other hand, it should also be emphasized that the survival curves for ICI therapy frequently exhibit a plateau at the tail end that has never been observed in those with traditional cytotoxic chemotherapies or molecular target agents, suggesting that ICIs can enable almost complete remission in a subset of patients with HCC and a long cancer-free status [18]. Therefore, maximizing the potential antitumor ability of ICIs in combination with other modalities/therapies may enable further improvement in patients with advanced HCCs. Recently, locoregional therapies, such as ablation, transarterial embolization, and radiotherapy, which are usually used for HCC at an earlier stage, have been actively explored to bolster ICI efficacy. In this article, we first provide an overview of the immune landscape for HCC to understand ICI efficacy and its limitations, and subsequently review the status of combinatorial therapies of ICI with locoregional therapies for HCC and their future prospects.

2. Tumor Immune Microenvironment for HCC and ICI Efficacy

Most carcinomas, including HCC, exhibit a heterogeneous pattern of immune cells within the tumor site, according to type, density, and localization. The widely accepted concept of immune heterogeneity associated with ICI efficacy classifies tumors into “hot”, “excluded”, and “cold” based on the T cell landscape (Figure 1). While immune-hot tumors have abundant T cells that exert antitumor activity and are, therefore, expected to be more susceptible to ICIs, immune-cold tumors are characterized by a paucity of T cells, likely resulting in a poor response. Immune-excluded tumors are expected to show an intermediate response to ICIs, because T cells are mainly observed at the edge of the tumor without being capable of infiltrating themselves. Indeed, this simplified but powerful concept predicted the treatment outcomes of ICIs for various malignancies [19,20,21]. Bagaev et al. developed conserved pan-cancer microenvironment subtypes from >10,000 transcriptome profiles and classified cancers into four types: immune-enriched/non-fibrotic (equivalent to hot), immune-enriched/fibrotic (equivalent to excluded), fibrotic, and immune-depleted (equivalent to cold) tumors [22]. Notably, only the immune-enriched/non-fibrotic subtype was associated with a favorable response to ICIs, suggesting that physical fibrotic barriers, that is, excluded status, can diminish ICI efficacy despite the accumulation of T cells. Based on this classification, one-fourth of HCCs were classified as the ICI-responsive immune-enriched/non-fibrotic subtype. Comprehensive immunohistochemistry-based assessment of HCC, including 919 regions from 158 HCC nodules, revealed that HCC can be classified into three subtypes based on the abundance of the immune cells (immune-high, -mid, and -low) and coexisting cell types. Immune-mid can be further subdivided into mid1 and 2, the former of which is characterized by enrichment of the granular formation and infiltration of mast cells and neutrophils, and the latter shows a more homogeneous distribution of various immune cell types. The immune-low subtype can also be subdivided into two distinct patterns, characterized by higher and lower regulatory T cell (Treg)/CD4 ratios. According to this histological classification, approximately 10% of HCC cases were classified as immune-high, which was significantly associated with a lower recurrence rate after surgical resection.
The underlying mechanisms and biomarkers of ICI effectiveness in HCC have been explored. Zhu et al. performed integrated molecular analyses of HCC samples (n = 358) from patients enrolled in the phase 1b or IMbrave150 phase 3 clinical trial of atezolizumab and bevacizumab to identify potential biomarkers for predicting clinical outcomes. Pre-existing immunity characterized by intratumoral CD8 T cell density, high expression of CD274 encoding PD-L1, and T-effector signature was favorably associated with the outcome, whereas a high regulatory T cell to effector T cell ratio and high expression of oncofetal genes (GPC3 and AFP) were associated with reduced benefit from the combination therapy. A variety of underlying mechanisms that promote T cell accumulation within tumors have been revealed in animal models and comprehensive human HCC analyses [23]. Ruiz de Galarreta et al. showed that β-catenin activation promotes immune escape by defecting dendritic cell recruitment and impairing T cell activity [24]. In steatotic HCC, palmitate-induced lipid accumulation upregulates PD-L1 expression and promotes immunosuppressive phenotypes of co-cultured macrophages and fibroblasts [25]. To explore the molecular correlates of response to ICIs among immune-hot HCCs, Magen et al. analyzed resected HCC samples from a neoadjuvant anti-PD-1 trial [26]. First, the authors confirmed that none of the immune-excluded or immune-cold HCCs responded to the ICI therapy. Simultaneously, the authors found that half of the immune-hot HCCs were nonresponsive. Multilayer omics analyses, such as single-cell RNA sequencing and spatial profiling, revealed that clonal expansion of a subset of T cells, more specifically intratumoral C-X-C chemokine ligand 13 (CXCL13)+ cholesterol 25-hydroxylase (CH25H)+ interleukin (IL)-21+ PD-1+ CD4 T helper cells named “CXCL13+ TH” and granzyme K (GZMK)+ PD-1+ CD8 effector-like T cells, was observed in responders, while terminally exhausted T cells were more enriched in non-responders. Interestingly, the potential antitumor CD8 effector-like T cells clonally expanded and differentiated from progenitor CD8 T cells in micro-niches formed by maturation regulatory dendritic cells and CXCL13+ TH in response to ICI treatment. This was not observed in non-responders, indicating that it not only increases in CD8 T cells but also the creation of a favorable microenvironment before ICI initiation plays a pivotal role in ICI responsiveness.

Rationale for Combination of ICI and Locoregional Therapies

As mentioned above, the efficient delivery and infiltration of specific T cells that exert antitumor activity at the tumor site are crucial for maximizing ICI efficacy. The rationales for multimodal approaches can be conceptualized by the so-called cancer–immunity cycle that illustrates the step-by-step anticancer immune responses and optimizes the combination therapies with ICIs accordingly (Figure 2) [27]. Briefly, the cancer–immunity cycle can be summarized as follows: (i) the release of cancer cell antigens, dying cancer cells release antigens (tumor-associated antigens (TAAs) and tumor-specific antigens aka neoantigen) that antigen-presenting cells (APCs), such as dendritic cells, recognize and engulf, as well as damage-associated molecular patterns (DAMPs) and cytokines; (ii) antigen presentation, APCs present tumor antigens on their surfaces; (iii) T cell priming and activation, T cells recognize tumor antigens presented by APCs through T cell receptors on their surface and are activated by recognition in the lymph node; (iv) T cell trafficking, activated T cells leave the lymph node towards the tumor site and infiltrate the tumor; (v) cancer cell recognition, T cells recognize and bind to cancer cells through interactions between the T cell receptors and cancer cell antigens present on the cancer cell surface; (vi) killing cancer cells, T cells release cytotoxic molecules, such as perforin and granzymes, to directly kill cancer cells, as well as cytokines and chemokines to activate other immune cells, such as macrophages. According to this concept, the anti-CTLA-4 antibody inhibits the immunosuppressive interaction between CTLA4 on T cells and APCs (step iii). Targeting VEGF with bevacizumab and rivoceranib is expected to increase cytotoxic immune cell infiltration into tumors by normalizing aberrant tumor vascularity, as well as other pathways (step iv). It is also important to evaluate how the combined therapy modifies the entire tumor microenvironment. We previously reported that anti-VEGF lenvatinib effectively recruits GZMK+ CD8 T cells to the tumor site through CXCL9 released from tumor-associated macrophages; however, it also enriched intratumoral stroma by upregulating fibrosis-related pathways [28].
In this scenario, dying cancer cells after locoregional therapies can release various immunogenic substances, such as TAAs and DAMPs, which may enhance local and systemic immune responses to ICIs. Zerbini et al. demonstrated that HCC-specific T cell responses were activated by coculturing patient monocytes in vitro together with tumor debris generated by radiofrequency ablation (RFA) and granulocyte macrophage colony-stimulating factor [29]. Importantly, apoptotic and necrotic cell death can induce different immune responses [30]. Antigen release by locoregional treatment can also expect the “abscopal effect” that leads to tumor shrinkage outside of tumors treated with locoregional treatment via the activated systemic immune response against tumors [31]. As overviewed below and illustrated in Figure 2, locoregional therapies mainly accelerate steps (i), (ii), (v), and (vi), as well as systemic inflammation. Given that the current ICI, with or without tyrosine kinase inhibitors (TKIs), primarily targets steps (iii) to (vi), combination therapies of locoregional therapies, ICI, and TKI are likely to activate the cancer–immunity cycle more efficiently. Indeed, the phase 3 EMERALD-3 trial is conducted to evaluate the efficacy and safety of durvalumab + tremelimumab + transarterial chemoembolization (TACE), with or without lenvatinib, compared with TACE alone (NCT05301842) [32].

3. Cancer–Immunity Cycle Acceleration by Locoregional Therapies

3.1. Ablation Therapy

Thermal ablation induces irreversible cell injury, tumor apoptosis, and coagulative necrosis through the local application of extremely high or low temperatures from electrodes that are directly inserted into tumors under image guidance. For early-stage HCCs, ablative techniques have been used for decades for the curative treatment of HCC. A multicenter randomized controlled study in Japan recently showed that percutaneous RFA is similarly effective for improving the prognosis of early-stage HCC compared to surgical resection [33]. Generally, ablation therapy can be classified into hyperthermal techniques, including RFA and microwave ablation (MWA), and cryoablation. RFA generates frictional heating (60–100 °C) through a high-frequency alternating current in the electrodes, whereas MWA generates heat using electromagnetic waves from an intratumorally placed antenna that forces the rotation of the molecules and increases their kinetic energy, thereby elevating the temperature within the tumor [34]. In contrast, cryoablation uses liquefied gas, such as argon, to reach a lethal cold temperature between −20 and −40 °C. These technical differences may lead to distinct immunogenicity, which can affect the effectiveness of combination therapy with ICIs [35]. Wang et al. performed in vitro experiments to compare the effects of different ablation temperatures (−80 °C, −40 °C, 0 °C, 37 °C, and 60 °C) on immunogenic cell death-related substances in multiple cell lines and found that the release of ATP, high mobility group box 1 (HMGB-1), and CXCL10 in HCC cell lines was significantly increased after both cryoablation and thermal ablation, whereas the expression levels of calreticulin, one of the DAMPs, were significantly different between high- and low-temperature ablation [36].

3.2. RFA

In a mouse HCC model, tumor ablation significantly increased and activated antigen-loaded dendritic cells that primed T cell activation in draining lymph nodes and, subsequently, activated tumor-specific T cells [37,38]. Mizukoshi et al. revealed that some peptides from TAAs, including lymphocyte-specific protein tyrosine kinases, p53, and human telomerase reverse transcriptase (TERT), are recognized by T cells only after locoregional treatment of human HCC [39]. Furthermore, the addition of the anti-CTLA4 antibody might enhance the immune response by modulating the cytokine and chemokine profiles of peripheral mononuclear cells. RFA may also induce pyroptosis, inflammasome-related programmed cell death, leading to the cleavage of gasdermin D and activation of IL-18 and IL-1β. Yang et al. showed that thermal sub-ablation of endothelial cells and hemangiomas induces HMGB-1-induced pyroptosis [40].
RFA also affects the systemic inflammatory status of patients with HCC. Myeloid-derived immunosuppressive cells (MDSCs) are a dominant component of the immunosuppressive network [41]. A pharmacological inhibitor in their differentiation from early-stage myeloid progenitors augmented the antitumor activity of ICIs in mouse models of mammary cancer [42]. The proportion of circulating MDSC was significantly decreased after RFA and was inversely associated with survival in patients with HCC [43]. RFA also triggers a systemic immune response, mainly involving innate immune cells, such as dendritic cells and natural killer (NK) cells, even after ablation of small HCC nodules in patients with cirrhosis [44]. An increase in activated NKp30+ NK cells 24 h after RFA was associated with reduced HCC recurrence, suggesting the importance of innate immunity in suppressing residual but clinically invisible malignant cells. RFA stimulated NK cell cytotoxicity and NK-mediated antibody-dependent cellular cytotoxicity. Furthermore, the production of nitric oxide (NO) and active oxygen species is activated after RFA [45]. NO induced the repolarization of tumor-associated macrophages to the tumoricidal form and the recruitment of NK cells and may complement ICI efficacy [46]. It is also known that sublethal heat treatment by RFA transforms HCC to the progenitor-like, more proliferative phenotype via epithelial–mesenchymal transition and promotes metastases associated with poor prognoses, supporting the importance of complete ablation for overall prognosis improvement [47,48].

3.3. MWA

In addition to heat-induced apoptosis and necrosis, Yu et al. speculated that MWA might induce ferroptosis, a newly proposed type of cell death, through the induction of reactive oxygen species, p53, the heat-shock protein, and NF-E2 related factor 2 (NRF2) [49]. Accumulating evidence suggests that ferroptotic cells attract and activate innate immune cells, such as neutrophils, and are efficiently engulfed by phagocytes [50]. Also, ferroptotic cells release HMGB-1, one of the well-known DAMPs, as a “find-me” signal, likely resulting in promoted immune cell recruitment [51]. If MWA truly induces ferroptosis, it might have another option to activate the immune response to ICIs.

3.4. Cryoablation

Cryoablation stimulated antitumor immunity together with immunoadjuvant therapy in a rat HCC model, leading to prolonged survival [52]. In a small series of 13 patients with unresectable liver tumors who underwent cryoablation, a subset of patients showed tumor necrosis, not only in directly ablated tumors but also in distant tumors, suggesting that cryoablation might induce the abscopal effect [53]. Furthermore, pretreatment serum tumor necrosis factor-alpha (TNF-α) and IL-10 levels were associated with the emergence of an abscopal effect. Zeng et al. showed that the proportions of PD-L1+ monocytes and PD-1+ CD8 T cells were positively correlated with the HCC stage [54]. Interestingly, the proportions were reduced after cryoablation, suggesting that cryoablation might modulate the systemic inflammatory status. Distinct immune modulation between cryoablation and hyperthermal techniques has yet to be confirmed in HCC. Inflammation induced by cryoablation may be greater than that induced by RFA in rat liver [55]. On the other hand, compared to RFA, cryoablation augmented both pro- and anti-inflammatory cytokines, including IL-1β, IL-5, IL-6, and IL-10, and did not reduce immunosuppressive regulatory T cells, although RFA reduced them, in a colon cancer mouse model [56].

3.5. Transarterial Embolization

Transarterial embolization therapy is the standard treatment for intermediate-stage HCC. HCC predominantly receives blood supply from the hepatic artery rather than from the portal vein, which is the major blood feeder for non-tumoral liver tissue. Therefore, the delivery of embolic agents, such as lipiodol, with or without anticancer drugs (TACE), drug-eluting bead (DEB)-TACE (DEB-TACE), or yttrium-90 (Y90)-loaded radioactive microspheres (transarterial radioembolization (TARE)), from a catheter placed in tumor-feeding arteries effectively leads to cancer lethality with minimal non-tumor liver injury. The hypoxic tumor state or cell death after transarterial embolization therapy is believed to drastically activate the intratumoral immune response through abundant antigen release. Differences in the mixed antitumor drugs or loaded materials may induce distinct immune responses. For TACE, doxorubicin or cisplatin is typically used to generate an emulsion with lipiodol. Cisplatin downregulates PD-L2 in human dendritic cells, whereas doxorubicin promotes immunogenic cell death and the clonal expansion of immunosuppressive MDSCs [57]. The type of embolic material also affects the magnitude of the immune response [58,59].

3.6. TACE/DEB-TACE

TACE appears to induce a systemic response in HCC by expanding AFP-specific CD4 T cells in the peripheral blood [60]. Tischfield et al. showed that the number of infiltrating CD3, CD4, and CD8 T cells, as well as the expression of PD-L1, was significantly increased in embolized tumors in a rat HCC model [58]. However, these findings remain controversial in human HCC. Consistent with the animal experiment, elevated PD-L1 expression was confirmed in human resected HCC after TACE [61]. However, another study demonstrated that CD4 and CD8 T cells, as well as immunosuppressive regulatory T cells, were reduced in human post-TACE HCCs compared to those without TACE [62]. To characterize the immune microenvironment post-TACE, Tan et al. subjected resected HCC samples with or without preoperative TACE to single-cell RNA sequencing [63]. Inconsistent with the above-mentioned rodent model observations, the authors found that the number of CD8 T cells was reduced after TACE compared to that in paired pre-TACE HCC tissues, whereas the triggering receptor expressed on myeloid cells 2 (TREM2)+ tumor-associated macrophages (TAMs), characterized by low expression of CXCL9, were increased. Mechanistically, TREM2+ TAM preferentially expressed galectin-1 encoded by LGALS1, which can hamper the functionality of CD8 T cells [64]. Moreover, galectin-1 induces the overexpression of PD-L1 in vascular endothelial cells, functioning as an obstacle to the migration of CD8 T cells into the tumor site. In two mouse HCC models, systemic TREM2 knockout enhanced the antitumor effect of the anti-PD-L1 antibody.
It is well known that malignant cells preferentially metabolize glucose through glycolysis to produce energy even under conditions of high oxygen, the so-called Warburg effect. Therefore, lactic acid concentration in malignancy is generally high, creating microenvironmental acidosis, although this may be etiology-dependent in HCC [65,66]. Tumor-derived lactic acid inhibits the migration of T cells and monocytes and facilitates immune evasion by tumors [67,68]. Conventional doxorubicin-mixed TACE normalizes the acidic environment in an animal model of HCC, which may lead to an ICI-susceptible microenvironment [69].

3.7. TARE

In the TARE procedure, Y90-loaded radioactive microspheres are transarterially injected into HCC and emit high-energy β-radiation to destroy tumor cells. As with conventional TACE and DEB-TACE, it has been reported that TARE can induce immunogenic cell death [70]. In addition, TARE has been shown to have a long-lasting antitumor effect (3–6 months) despite its short half-life (~60 h) [71,72]. Chew et al. showed that TARE activates the local and systemic immune response involving T cells, NK cells, natural killer T (NKT) cells, and APCs [73]. Interestingly, PD-1+ and TIM3+ CD8 T cells in peripheral blood from patients who showed sustained antitumor effect after Y90 TARE maintained their capability to express pro-inflammatory cytokines interferon-γ and TNF-α when stimulated ex vivo, suggesting that anti-PD-1 or anti-TIM3 ICIs after TARE, especially in sustained responders, may reactivate the antitumor effect as the sequential therapy. Indeed, an independent group showed that Y90 TARE followed by anti-PD-1 and anti-LAG3 inhibitors after one month induced synergistic immune-mediated HCC control [74]. Impaired lymphocyte function due to high radiation activity should also be considered as a side effect [75].

3.8. Radiotherapy

As the abscopal effect was originally reported as an unexpected regression in lesions outside the irradiated tumor in 1953 [76], radiotherapy can activate systemic and local immune responses by releasing antigens, DAMPs, and cytokines from dying cancer cells, as well as increasing major histocompatibility complex (MHC) class I on tumor cells [77]. Durvalumab has already been approved as a maintenance therapy after chemoradiation for stage III non-small cell lung cancer [78,79]. For HCC, Sung et al. developed a mathematical model to simulate the percentage of tumor volume irradiated to synergize antitumor ability with ICIs, and concluded that irradiating 90% of tumor cells in addition to ICIs yields an incremental benefit between 33% and 71% compared to that without irradiation [80]. Radiotherapy also triggers immune responses via gasdermin-E-mediated pyroptosis [81]. Furthermore, the cyclic guanosine monophosphate–adenosine monophosphate synthase (cGAS)-stimulator of interferon gene (STING) pathway has been reported to be the key to understanding radiotherapy-mediated immune responses. Du et al. reported that radiotherapy upregulated PD-L1 expression in HCC by activating the intrinsic cGAS-STING pathway, leading to immune evasion; therefore, combination therapy with anti-PD-L1 blockade potentiated the antitumor effect of radiotherapy [82]. Meanwhile, the activated cGAS-STING pathway in M1 macrophages promotes T cell recruitment to HCC, which may enhance ICI efficacy [83]. Anti-VEGF sorafenib may enhance the immune response to radiotherapy by downregulating the signal transducer and activator of transcription (STAT) pathways [84]. To induce these synergistic effects, gut dysbiosis may have to be modulated. Li et al. reported that cyclic di-AMP, an emerging second messenger of bacteria, serves as an agonist of STINGs and activates the cGAS-STRING-interferon I pathway, resulting in the suppression of antigen presentation and impairment of effector T cell functions [85]. During radiotherapy, organs outside the target tumor can be irradiated, which may lead to an immunosuppressive tumor microenvironment. Wang et al. reported that ionizing radiation induces immunosuppressive MDSC expansion, contributing to diminished antitumor immunity by ICIs through the YTH N6-methyladenosine RNA binding protein F2 (YTHDF2)-nuclear factor-kappa B (NFκB) axis [86].

3.9. In-Progress Clinical Trials of ICI Plus Locoregional Therapies

With these promising experimental and preclinical studies, dozens of early- to late-phase clinical trials investigating the combination therapies of ICIs and locoregional therapies are ongoing (Table 1).

3.9.1. Ablation Therapy

A phase 2 clinical trial evaluated the combination of RFA and anti-CTLA-4 tremelimumab in 32 patients with Barcelona Clinic Liver Cancer (BCLC) B/C HCC (NCT01853618) [98]. No severe side effects were observed, and 26% of the patients showed a partial response (PR). Interestingly, tumor biopsies after six weeks showed increased CD8 T cells in patients showing a clinical benefit. Median time to tumor progression (TTP) and overall survival (OS) were 7.4 and 12.3 months, respectively. Another proof-of-concept trial (n = 50) also suggested that the addition of ablation therapy to anti-PD-1 pembrolizumab or nivolumab increased the objective response rate (ORR) (24% vs. 10% in combination therapy and anti-PD-1 monotherapy, respectively) and achieved a prolonged median OS (16.9 vs. 5.0 months, respectively) with acceptable toxicity profiles in patients with advanced HCC (NCT03939975) [90]. In the phase 2 IR11330 trial, 48 patients with advanced HCC after at least one line failed systemic therapy received subtotal ablation, defined as the complete treatment of 1–5 lesions by hyperthermal ablation and intentionally leaving residual lesions intact, followed by anti-PD-1 toripalimab initiation on days 3 or 14 after ablation therapy or toripalimab monotherapy (NCT03864211). The ORRs of ablation therapy plus toripalimab on days 3 or 14, and toripalimab monotherapy were 38%, 31%, and 19%, respectively [92].
Given the high frequency of recurrence, even after curative treatment, of early stage HCCs (76% and 70% 5 years after curative RFA and surgical resection, respectively) [111,112], adjuvant ICI therapies for at-risk patients after curative treatment have been intensively tested in prospective clinical trials, with an expected immune-activated state after curative treatment. The phase 3 IMbrave050 trial showed that adjuvant atezolizumab plus bevacizumab significantly improved recurrence-free survival (RFS) compared to active recurrence surveillance in patients with early but high-risk HCC after curative RFA and surgical resection (hazard ratio (HR), 0.72) (NCT04102098) [87]. Phase 3 CheckMate 9DX (NCT03383458), KEYNOTE-937 (NCT03867084) [88], and EMERALD-2 (NCT03847428) [89] trials are currently in progress to evaluate the efficacy of adjuvant nivolumab, pembrolizumab, and durvalumab in improving RFS after curative treatment in patients with early-stage HCC, respectively.

3.9.2. TACE

To enhance the immune response against intermediate- and advanced-stage HCC, combination therapies of TACE and ICI have been investigated in prospective clinical trials. In a phase 1/2 clinical trial, TACE followed by anti-CTLA-4 tremelimumab had a higher PR rate and prolonged OS with acceptable adverse events (NCT01853618) [98]. Anti-PD-1 pembrolizumab following TACE achieved 10.8 months of progression-free survival (PFS) from the first TACE (NCT03397654) in the phase 1/2 PETAL trial [96]. The phase 2 IMMUTACE trial evaluating the effects of TACE in combination with nivolumab for intermediate-stage HCC resulted in a complete response (CR) and a PR of 16% and 55%, respectively. At a median follow-up of 20 months, the median PFS was 7.2 months (NCT03572582) [94,113].

3.9.3. TARE

In a prospective open-label, phase 1 clinical trial of nivolumab plus Y90 TARE in patients with advanced HCC, 9 (81%) and 6 (46%) out of 11 patients exhibited stable disease and reduced serum AFP levels, respectively (NCT02837029) [103]. In a phase 2 trial in which 40 patients underwent Y90 TARE plus nivolumab, one (3%) and ten (28%) patients showed CR and PR, respectively (NCT03033446) [101]. A phase 2 NASIR-HCC trial evaluated the combination of TARE and nivolumab in 42 patients with unresected HCC and demonstrated an objective response rate of 42% and a median TTP and OS of 8.8 and 20.9 months, respectively (NCT03380130) [100]. A prospective multicenter single-arm phase 2 clinical trial investigated pembrolizumab plus Y90 TARE in patients with HCC that was likely refractory to TARE alone, defined as a multifocal disease, branch portal vein thrombosis, and/or diffuse distribution (NCT03099564) [102]. Despite the refractory nature, the median PFS, TTP, and OS were 8.6, 9.9, 22.0 months, respectively.

3.9.4. Stereotactic Body Radiotherapy (SBRT)

In a multicenter phase 1 clinical trial, patients with advanced or unresected HCC received either nivolumab alone or nivolumab plus ipilimumab, followed by SBRT. Clinical outcomes favored the nivolumab plus ipilimumab arm compared with nivolumab alone, with an ORR of 57% (four of seven patients) and 0% (zero of six patients), a median PFS of 11.6 and 2.7 months, and a median OS of 41.6 versus 4.7 months, respectively. In the combined immunotherapy group, the 3-year survival rate was 57%, with an acceptable safety profile (NCT032033040) [108]. The single-arm phase 2 START-FIT trial investigated sequential TACE and SBRT followed by anti-PD-L1 avelumab in patients with locally advanced hepatocellular carcinoma who were unsuitable for curative treatment (NCT03817736) [109]. Among the 33 enrolled patients, 14 (42%) and 4 (12%) achieved radiological CR and curative treatment, respectively. In a single-arm trial including 21 patients with unresectable HCC, SBRT plus camrelizumab demonstrated a median PFS and OS of 5.8 and 14.2 months after a median follow-up of 19.7 months, respectively [114].

4. Conclusions

The emergence of ICIs has dramatically and rapidly changed the therapeutic landscape for malignancies, including HCC. As summarized, ICIs will be used for early- to late-stage HCC, given their broad antitumor mode of action. Therefore, an in-depth understanding of ICIs from bench to bedside is required to effectively realize their potential. To further maximize the synergistic ability of ICIs and locoregional therapy, the treatment sequences and timing must be optimized. Immune-enhancing strategies, such as cancer vaccination and nanomedicine via HCC-targeting peptides, may also improve ICI efficacy [115,116]. In addition, biomarkers for identifying patients who would benefit the most from combination therapies are an unmet need to efficiently use limited medical resources [117]. The accumulating evidence indicates that immune evasion plays a pivotal role, not only in fully developed HCC, but also in de novo HCC development from premalignant lesions, suggesting that combination therapy may be immune preventive [118,119]. Elucidating these unanswered questions will eventually improve the prognosis for HCC patients. We are witnessing the dawn of a revolution in cancer therapy using ICIs.

Author Contributions

Y.T.: Investigation, Writing—Original draft, Visualization; N.F.: Project administration, Writing—Original Review & Editing; T.T.; Visualization; S.M. and H.N.: Supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by JSPS KAKENHI Grant Number 23K16770 (Y.T.), 21H02892 (H.N.), AMED Grant Number JP23fk0210130, JP23fk0210090, JP23fk0210115, the Princess Takamatsu Cancer Research Fund, Daiichi Sankyo Foundation of Life Science, and Takeda Science Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef]
  2. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef] [PubMed]
  3. Singal, A.G.; Llovet, J.M.; Yarchoan, M.; Mehta, N.; Heimbach, J.K.; Dawson, L.A.; Jou, J.H.; Kulik, L.M.; Agopian, V.G.; Marrero, J.A.; et al. AASLD Practice Guidance on prevention, diagnosis, and treatment of hepatocellular carcinoma. Hepatology 2023. [Google Scholar] [CrossRef] [PubMed]
  4. Fujiwara, N.; Friedman, S.L.; Goossens, N.; Hoshida, Y. Risk factors and prevention of hepatocellular carcinoma in the era of precision medicine. J. Hepatol. 2018, 68, 526–549. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, Y.T.; Fujiwara, N.; Yang, J.D.; Hoshida, Y. Risk stratification and early detection biomarkers for precision HCC screening. Hepatology 2023, 78, 319–362. [Google Scholar] [CrossRef] [PubMed]
  6. Park, J.W.; Chen, M.; Colombo, M.; Roberts, L.R.; 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]
  7. Shiina, S.; Tateishi, R.; Arano, T.; Uchino, K.; Enooku, K.; Nakagawa, H.; Asaoka, Y.; Sato, T.; Masuzaki, R.; Kondo, Y.; et al. Radiofrequency ablation for hepatocellular carcinoma: 10-year outcome and prognostic factors. Am. J. Gastroenterol. 2012, 107, 569–577; quiz 578. [Google Scholar] [CrossRef]
  8. 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]
  9. 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]
  10. Qin, S.; Chan, S.L.; Gu, S.; Bai, Y.; Ren, Z.; Lin, X.; Chen, Z.; Jia, W.; Jin, Y.; Guo, Y.; et al. Camrelizumab plus rivoceranib versus sorafenib as first-line therapy for unresectable hepatocellular carcinoma (CARES-310): A randomised, open-label, international phase 3 study. Lancet 2023, 402, 1133–1146. [Google Scholar] [CrossRef]
  11. Abou-Alfa, G.K.; Lau, G.; Kudo, M.; Chan, S.L.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.-K.; Dao, T.V.; Toni, E.N.D.; et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022, 1, EVIDoa2100070. [Google Scholar] [CrossRef]
  12. Cappuyns, S.; Corbett, V.; Yarchoan, M.; Finn, R.S.; Llovet, J.M. Critical Appraisal of Guideline Recommendations on Systemic Therapies for Advanced Hepatocellular Carcinoma: A Review. JAMA Oncol. 2023. [Google Scholar] [CrossRef]
  13. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutierrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  14. Curigliano, G.; Gelderblom, H.; Mach, N.; Doi, T.; Tai, D.; Forde, P.M.; Sarantopoulos, J.; Bedard, P.L.; Lin, C.C.; Hodi, F.S.; et al. Phase I/Ib Clinical Trial of Sabatolimab, an Anti-TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti-PD-1 Antibody, in Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 3620–3629. [Google Scholar] [CrossRef] [PubMed]
  15. Finn, R.S.; Ryoo, B.-Y.; Hsu, C.-H.; Li, D.; Burgoyne, A.; Cotter, C.; Badhrinarayanan, S.; Wang, Y.; Yin, A.; Edubilli, T.R.; et al. Results from the MORPHEUS-liver study: Phase Ib/II randomized evaluation of tiragolumab (tira) in combination with atezolizumab (atezo) and bevacizumab (bev) in patients with unresectable, locally advanced or metastatic hepatocellular carcinoma (uHCC). J. Clin. Oncol. 2023, 41, 4010. [Google Scholar] [CrossRef]
  16. D’Alessio, A.; Fulgenzi, C.A.M.; Nishida, N.; Schonlein, M.; von Felden, J.; Schulze, K.; Wege, H.; Gaillard, V.E.; Saeed, A.; Wietharn, B.; et al. Preliminary evidence of safety and tolerability of atezolizumab plus bevacizumab in patients with hepatocellular carcinoma and Child-Pugh A and B cirrhosis: A real-world study. Hepatology 2022, 76, 1000–1012. [Google Scholar] [CrossRef]
  17. Fulgenzi, C.A.M.; Cheon, J.; D’Alessio, A.; Nishida, N.; Ang, C.; Marron, T.U.; Wu, L.; Saeed, A.; Wietharn, B.; Cammarota, A.; et al. Reproducible safety and efficacy of atezolizumab plus bevacizumab for HCC in clinical practice: Results of the AB-real study. Eur. J. Cancer 2022, 175, 204–213. [Google Scholar] [CrossRef] [PubMed]
  18. Kudo, M. Prioritized Requirements for First-Line Systemic Therapy for Hepatocellular Carcinoma: Broad Benefit with Less Toxicity. Liver Cancer 2023, 12, 1–6. [Google Scholar] [CrossRef]
  19. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571. [Google Scholar] [CrossRef]
  20. Van Allen, E.M.; Miao, D.; Schilling, B.; Shukla, S.A.; Blank, C.; Zimmer, L.; Sucker, A.; Hillen, U.; Foppen, M.H.G.; Goldinger, S.M.; et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015, 350, 207–211. [Google Scholar] [CrossRef]
  21. Ji, R.R.; Chasalow, S.D.; Wang, L.; Hamid, O.; Schmidt, H.; Cogswell, J.; Alaparthy, S.; Berman, D.; Jure-Kunkel, M.; Siemers, N.O.; et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol. Immunother. 2012, 61, 1019–1031. [Google Scholar] [CrossRef] [PubMed]
  22. Bagaev, A.; Kotlov, N.; Nomie, K.; Svekolkin, V.; Gafurov, A.; Isaeva, O.; Osokin, N.; Kozlov, I.; Frenkel, F.; Gancharova, O.; et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021, 39, 845–865.e7. [Google Scholar] [CrossRef] [PubMed]
  23. Fujiwara, N.; Nakagawa, H. Clinico-histological and molecular features of hepatocellular carcinoma from nonalcoholic fatty liver disease. Cancer Sci. 2023, 114, 3825–3833. [Google Scholar] [CrossRef] [PubMed]
  24. Ruiz de Galarreta, M.; Bresnahan, E.; Molina-Sanchez, P.; Lindblad, K.E.; Maier, B.; Sia, D.; Puigvehi, M.; Miguela, V.; Casanova-Acebes, M.; Dhainaut, M.; et al. beta-Catenin Activation Promotes Immune Escape and Resistance to Anti-PD-1 Therapy in Hepatocellular Carcinoma. Cancer Discov. 2019, 9, 1124–1141. [Google Scholar] [CrossRef]
  25. Murai, H.; Kodama, T.; Maesaka, K.; Tange, S.; Motooka, D.; Suzuki, Y.; Shigematsu, Y.; Inamura, K.; Mise, Y.; Saiura, A.; et al. Multiomics identifies the link between intratumor steatosis and the exhausted tumor immune microenvironment in hepatocellular carcinoma. Hepatology 2023, 77, 77–91. [Google Scholar] [CrossRef]
  26. Magen, A.; Hamon, P.; Fiaschi, N.; Soong, B.Y.; Park, M.D.; Mattiuz, R.; Humblin, E.; Troncoso, L.; D’Souza, D.; Dawson, T.; et al. Intratumoral dendritic cell-CD4(+) T helper cell niches enable CD8(+) T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat. Med. 2023, 29, 1389–1399. [Google Scholar] [CrossRef]
  27. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef]
  28. Yamada, T.; Fujiwara, N.; Kubota, N.; Matsushita, Y.; Nakatsuka, T.; Kurosaki, S.; Minami, T.; Tateishi, R.; Ichida, A.; Arita, J.; et al. Lenvatinib recruits cytotoxic GZMK+CD8 T cells in hepatocellular carcinoma. Hepatol. Commun. 2023, 7, e0209. [Google Scholar] [CrossRef]
  29. 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]
  30. Scheffer, S.R.; Nave, H.; Korangy, F.; Schlote, K.; Pabst, R.; Jaffee, E.M.; Manns, M.P.; Greten, T.F. Apoptotic, but not necrotic, tumor cell vaccines induce a potent immune response in vivo. Int. J. Cancer 2003, 103, 205–211. [Google Scholar] [CrossRef]
  31. Nelson, B.E.; Adashek, J.J.; Lin, S.H.; Subbiah, V. The abscopal effect in patients with cancer receiving immunotherapy. Med 2023, 4, 233–244. [Google Scholar] [CrossRef] [PubMed]
  32. Abou-Alfa, G.K.; Fan, J.; Heo, J.; Arai, Y.; Erinjeri, J.P.; Kuhl, C.K.; Lencioni, R.; Ren, Z.; Zeng, A.; Evans, B.; et al. 727TiP A randomised phase III study of tremelimumab (T) plus durvalumab (D) with or without lenvatinib combined with concurrent transarterial chemoembolisation (TACE) versus TACE alone in patients (pts) with locoregional hepatocellular carcinoma (HCC): EMERALD-3. Ann. Oncol. 2022, 33, S874. [Google Scholar] [CrossRef]
  33. 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 2022, 11, 209–218. [Google Scholar] [CrossRef] [PubMed]
  34. Tateishi, R.; Fujiwara, N. Precision Locoregional Therapies for Hepatocellular Carcinoma: Percutaneous Ablation and Radiotherapy. In Hepatocellular Carcinoma: Translational Precision Medicine Approaches; Hoshida, Y., Ed.; Humana Press: New York, NY, USA; Springer Nature: Cham, Switzerland, 2019; pp. 195–224. [Google Scholar]
  35. Chu, K.F.; Dupuy, D.E. Thermal ablation of tumours: Biological mechanisms and advances in therapy. Nat. Rev. Cancer 2014, 14, 199–208. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, M.; Duan, Y.; Yang, M.; Guo, Y.; Li, F.; Wang, J.; Si, T. The analysis of immunogenic cell death induced by ablation at different temperatures in hepatocellular carcinoma cells. Front. Cell Dev. Biol. 2023, 11, 1146195. [Google Scholar] [CrossRef] [PubMed]
  37. Ali, M.Y.; Grimm, C.F.; Ritter, M.; Mohr, L.; Allgaier, H.P.; Weth, R.; Bocher, W.O.; Endrulat, K.; Blum, H.E.; Geissler, M. Activation of dendritic cells by local ablation of hepatocellular carcinoma. J. Hepatol. 2005, 43, 817–822. [Google Scholar] [CrossRef]
  38. den Brok, M.H.; Sutmuller, R.P.; Nierkens, S.; Bennink, E.J.; Frielink, C.; Toonen, L.W.; Boerman, O.C.; Figdor, C.G.; Ruers, T.J.; Adema, G.J. Efficient loading of dendritic cells following cryo and radiofrequency ablation in combination with immune modulation induces anti-tumour immunity. Br. J. Cancer 2006, 95, 896–905. [Google Scholar] [CrossRef]
  39. Mizukoshi, E.; Nakamoto, Y.; Arai, K.; Yamashita, T.; Sakai, A.; Sakai, Y.; Kagaya, T.; Yamashita, T.; Honda, M.; Kaneko, S. Comparative analysis of various tumor-associated antigen-specific t-cell responses in patients with hepatocellular carcinoma. Hepatology 2011, 53, 1206–1216. [Google Scholar] [CrossRef]
  40. Yang, M.; Yang, X.; Wang, S.; Xu, L.; Ke, S.; Ding, X.; Sun, W.; Gao, J. HMGB1-induced endothelial cell pyroptosis is involved in systemic inflammatory response syndrome following radiofrequency ablation of hepatic hemangiomas. Am. J. Transl. Res. 2019, 11, 7555–7567. [Google Scholar]
  41. Grover, A.; Sanseviero, E.; Timosenko, E.; Gabrilovich, D.I. Myeloid-Derived Suppressor Cells: A Propitious Road to Clinic. Cancer Discov. 2021, 11, 2693–2706. [Google Scholar] [CrossRef]
  42. Colligan, S.H.; Amitrano, A.M.; Zollo, R.A.; Peresie, J.; Kramer, E.D.; Morreale, B.; Barbi, J.; Singh, P.K.; Yu, H.; Wang, J.; et al. Inhibiting the biogenesis of myeloid-derived suppressor cells enhances immunotherapy efficacy against mammary tumor progression. J. Clin. Investig. 2022, 132, e158661. [Google Scholar] [CrossRef] [PubMed]
  43. Arihara, F.; Mizukoshi, E.; Kitahara, M.; Takata, Y.; Arai, K.; Yamashita, T.; Nakamoto, Y.; Kaneko, S. Increase in CD14+HLA-DR -/low myeloid-derived suppressor cells in hepatocellular carcinoma patients and its impact on prognosis. Cancer Immunol. Immunother. 2013, 62, 1421–1430. [Google Scholar] [CrossRef] [PubMed]
  44. Rochigneux, P.; Nault, J.C.; Mallet, F.; Chretien, A.S.; Barget, N.; Garcia, A.J.; Del Pozo, L.; Bourcier, V.; Blaise, L.; Grando-Lemaire, V.; et al. Dynamic of systemic immunity and its impact on tumor recurrence after radiofrequency ablation of hepatocellular carcinoma. Oncoimmunology 2019, 8, 1615818. [Google Scholar] [CrossRef]
  45. Zhao, Y.; Li, K.; Sun, J.; He, N.; Zhao, P.; Zang, C.; Yang, X.; Hu, C.; Long, J.; Zhang, H.; et al. Genomic DNA methylation profiling indicates immune response following thermal ablation treatment for HBV-associated hepatocellular carcinoma. Oncol. Lett. 2020, 20, 677–684. [Google Scholar] [CrossRef]
  46. Mukherjee, S.; Fried, A.; Hussaini, R.; White, R.; Baidoo, J.; Yalamanchi, S.; Banerjee, P. Phytosomal curcumin causes natural killer cell-dependent repolarization of glioblastoma (GBM) tumor-associated microglia/macrophages and elimination of GBM and GBM stem cells. J. Exp. Clin. Cancer Res. 2018, 37, 168. [Google Scholar] [CrossRef]
  47. Mohammadpour, H.; Pourfathollah, A.A.; Nikougoftar Zarif, M.; Shahbazfar, A.A. Irradiation enhances susceptibility of tumor cells to the antitumor effects of TNF-alpha activated adipose derived mesenchymal stem cells in breast cancer model. Sci. Rep. 2016, 6, 28433. [Google Scholar] [CrossRef] [PubMed]
  48. Su, T.; Huang, M.; Liao, J.; Lin, S.; Yu, P.; Yang, J.; Cai, Y.; Zhu, S.; Xu, L.; Peng, Z.; et al. Insufficient Radiofrequency Ablation Promotes Hepatocellular Carcinoma Metastasis Through N6-Methyladenosine mRNA Methylation-Dependent Mechanism. Hepatology 2021, 74, 1339–1356. [Google Scholar] [CrossRef]
  49. Yu, L.; Cheng, M.; Liu, J.; Ye, X.; Wei, Z.; Xu, J.; Xie, Q.; Liang, J. Crosstalk between microwave ablation and ferroptosis: The next hot topic? Front. Oncol. 2023, 13, 1099731. [Google Scholar] [CrossRef]
  50. Li, W.; Feng, G.; Gauthier, J.M.; Lokshina, I.; Higashikubo, R.; Evans, S.; Liu, X.; Hassan, A.; Tanaka, S.; Cicka, M.; et al. Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J. Clin. Investig. 2019, 129, 2293–2304. [Google Scholar] [CrossRef]
  51. Wen, Q.; Liu, J.; Kang, R.; Zhou, B.; Tang, D. The release and activity of HMGB1 in ferroptosis. Biochem. Biophys. Res. Commun. 2019, 510, 278–283. [Google Scholar] [CrossRef]
  52. Mandt, T.; Bangar, A.; Sauceda, C.; Das, M.; Moderbacher, C.; Ghani, M.; Webster, N.; Newton, I. Stimulating Antitumoral Immunity by Percutaneous Cryoablation and Combination Immunoadjuvant Therapy in a Murine Model of Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2023, 34, 1516–1527.e6. [Google Scholar] [CrossRef] [PubMed]
  53. Osada, S.; Imai, H.; Tomita, H.; Tokuyama, Y.; Okumura, N.; Matsuhashi, N.; Sakashita, F.; Nonaka, K. Serum cytokine levels in response to hepatic cryoablation. J. Surg. Oncol. 2007, 95, 491–498. [Google Scholar] [CrossRef] [PubMed]
  54. Zeng, Z.; Shi, F.; Zhou, L.; Zhang, M.N.; Chen, Y.; Chang, X.J.; Lu, Y.Y.; Bai, W.L.; Qu, J.H.; Wang, C.P.; et al. Upregulation of circulating PD-L1/PD-1 is associated with poor post-cryoablation prognosis in patients with HBV-related hepatocellular carcinoma. PLoS ONE 2011, 6, e23621. [Google Scholar] [CrossRef]
  55. Jansen, M.C.; van Hillegersberg, R.; Schoots, I.G.; Levi, M.; Beek, J.F.; Crezee, H.; van Gulik, T.M. Cryoablation induces greater inflammatory and coagulative responses than radiofrequency ablation or laser induced thermotherapy in a rat liver model. Surgery 2010, 147, 686–695. [Google Scholar] [CrossRef] [PubMed]
  56. Mauda-Havakuk, M.; Hawken, N.M.; Owen, J.W.; Mikhail, A.S.; Saxena, A.; Karim, B.; Wakim, P.G.; Pritchard, W.F.; Karanian, J.W.; Wood, B.J. Comparative analysis of the immune response to RFA and cryoablation in a colon cancer mouse model. Sci. Rep. 2022, 12, 18229. [Google Scholar] [CrossRef] [PubMed]
  57. Galluzzi, L.; Buque, 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]
  58. Tischfield, D.J.; Gurevich, A.; Johnson, O.; Gatmaytan, I.; Nadolski, G.J.; Soulen, M.C.; Kaplan, D.E.; Furth, E.; Hunt, S.J.; Gade, T.P.F. Transarterial Embolization Modulates the Immune Response within Target and Nontarget Hepatocellular Carcinomas in a Rat Model. Radiology 2022, 303, 215–225. [Google Scholar] [CrossRef] [PubMed]
  59. Berz, A.M.; Santana, J.G.; Iseke, S.; Gross, M.; Pekurovsky, V.; Laage Gaupp, F.; Savic, L.J.; Borde, T.; Gottwald, L.A.; Boustani, A.M.; et al. Impact of Chemoembolic Regimen on Immune Cell Recruitment and Immune Checkpoint Marker Expression following Transcatheter Arterial Chemoembolization in a VX2 Rabbit Liver Tumor Model. J. Vasc. Interv. Radiol. 2022, 33, 764–774.e764. [Google Scholar] [CrossRef]
  60. Ayaru, L.; Pereira, S.P.; Alisa, A.; Pathan, A.A.; Williams, R.; Davidson, B.; Burroughs, A.K.; Meyer, T.; Behboudi, S. Unmasking of alpha-fetoprotein-specific CD4(+) T cell responses in hepatocellular carcinoma patients undergoing embolization. J. Immunol. 2007, 178, 1914–1922. [Google Scholar] [CrossRef]
  61. Montasser, A.; Beaufrere, A.; Cauchy, F.; Bouattour, M.; Soubrane, O.; Albuquerque, M.; Paradis, V. Transarterial chemoembolisation enhances programmed death-1 and programmed death-ligand 1 expression in hepatocellular carcinoma. Histopathology 2021, 79, 36–46. [Google Scholar] [CrossRef]
  62. Pinato, D.J.; Murray, S.M.; Forner, A.; Kaneko, T.; Fessas, P.; Toniutto, P.; Minguez, 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] [PubMed]
  63. Tan, J.; Fan, W.; Liu, T.; Zhu, B.; Liu, Y.; Wang, S.; Wu, J.; Liu, J.; Zou, F.; Wei, J.; et al. TREM2(+) macrophages suppress CD8(+) T-cell infiltration after transarterial chemoembolisation in hepatocellular carcinoma. J. Hepatol. 2023, 79, 126–140. [Google Scholar] [CrossRef] [PubMed]
  64. Nambiar, D.K.; Aguilera, T.; Cao, H.; Kwok, S.; Kong, C.; Bloomstein, J.; Wang, Z.; Rangan, V.S.; Jiang, D.; von Eyben, R.; et al. Galectin-1-driven T cell exclusion in the tumor endothelium promotes immunotherapy resistance. J. Clin. Investig. 2019, 129, 5553–5567. [Google Scholar] [CrossRef] [PubMed]
  65. Fujiwara, N.; Nakagawa, H.; Enooku, K.; Kudo, Y.; Hayata, Y.; Nakatsuka, T.; Tanaka, Y.; Tateishi, R.; Hikiba, Y.; Misumi, K.; et al. CPT2 downregulation adapts HCC to lipid-rich environment and promotes carcinogenesis via acylcarnitine accumulation in obesity. Gut 2018, 67, 1493–1504. [Google Scholar] [CrossRef] [PubMed]
  66. Enooku, K.; Nakagawa, H.; Fujiwara, N.; Kondo, M.; Minami, T.; Hoshida, Y.; Shibahara, J.; Tateishi, R.; Koike, K. Altered serum acylcarnitine profile is associated with the status of nonalcoholic fatty liver disease (NAFLD) and NAFLD-related hepatocellular carcinoma. Sci. Rep. 2019, 9, 10663. [Google Scholar] [CrossRef] [PubMed]
  67. Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
  68. Goetze, K.; Walenta, S.; Ksiazkiewicz, M.; Kunz-Schughart, L.A.; Mueller-Klieser, W. Lactate enhances motility of tumor cells and inhibits monocyte migration and cytokine release. Int. J. Oncol. 2011, 39, 453–463. [Google Scholar] [CrossRef]
  69. Savic, L.J.; Schobert, I.T.; Peters, D.; Walsh, J.J.; Laage-Gaupp, F.M.; Hamm, C.A.; Tritz, N.; Doemel, L.A.; Lin, M.; Sinusas, A.; et al. Molecular Imaging of Extracellular Tumor pH to Reveal Effects of Locoregional Therapy on Liver Cancer Microenvironment. Clin. Cancer Res. 2020, 26, 428–438. [Google Scholar] [CrossRef]
  70. Lugade, A.A.; Sorensen, E.W.; Gerber, S.A.; Moran, J.P.; Frelinger, J.G.; Lord, E.M. Radiation-induced IFN-gamma production within the tumor microenvironment influences antitumor immunity. J. Immunol. 2008, 180, 3132–3139. [Google Scholar] [CrossRef]
  71. Salem, R.; Lewandowski, R.J.; Mulcahy, M.F.; Riaz, A.; Ryu, R.K.; Ibrahim, S.; Atassi, B.; Baker, T.; Gates, V.; Miller, F.H.; et al. Radioembolization for hepatocellular carcinoma using Yttrium-90 microspheres: A comprehensive report of long-term outcomes. Gastroenterology 2010, 138, 52–64. [Google Scholar] [CrossRef]
  72. Sangro, B.; Inarrairaegui, M.; Bilbao, J.I. Radioembolization for hepatocellular carcinoma. J. Hepatol. 2012, 56, 464–473. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [PubMed]
  74. Rivoltini, L.; Bhoori, S.; Camisaschi, C.; Bergamaschi, L.; Lalli, L.; Frati, P.; Citterio, D.; Castelli, C.; Mazzaferro, V. Y(90)-radioembolisation in hepatocellular carcinoma induces immune responses calling for early treatment with multiple checkpoint blockers. Gut 2023, 72, 406–407. [Google Scholar] [CrossRef] [PubMed]
  75. 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] [PubMed]
  76. Mole, R.H. Whole body irradiation; radiobiology or medicine? Br. J. Radiol. 1953, 26, 234–241. [Google Scholar] [CrossRef]
  77. Chen, L.; Zhang, R.; Lin, Z.; Tan, Q.; Huang, Z.; Liang, B. Radiation therapy in the era of immune treatment for hepatocellular carcinoma. Front. Immunol. 2023, 14, 1100079. [Google Scholar] [CrossRef]
  78. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef]
  79. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Overall Survival with Durvalumab after Chemoradiotherapy in Stage III NSCLC. N. Engl. J. Med. 2018, 379, 2342–2350. [Google Scholar] [CrossRef]
  80. Sung, W.; Hong, T.S.; Poznansky, M.C.; Paganetti, H.; Grassberger, C. Mathematical Modeling to Simulate the Effect of Adding Radiation Therapy to Immunotherapy and Application to Hepatocellular Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2022, 112, 1055–1062. [Google Scholar] [CrossRef]
  81. Cao, W.; Chen, G.; Wu, L.; Yu, K.N.; Sun, M.; Yang, M.; Jiang, Y.; Jiang, Y.; Xu, Y.; Peng, S.; et al. Ionizing Radiation Triggers the Antitumor Immunity by Inducing Gasdermin E-Mediated Pyroptosis in Tumor Cells. Int. J. Radiat. Oncol. Biol. Phys. 2023, 115, 440–452. [Google Scholar] [CrossRef]
  82. Du, S.S.; Chen, G.W.; Yang, P.; Chen, Y.X.; Hu, Y.; Zhao, Q.Q.; Zhang, Y.; Liu, R.; Zheng, D.X.; Zhou, J.; et al. Radiation Therapy Promotes Hepatocellular Carcinoma Immune Cloaking via PD-L1 Upregulation Induced by cGAS-STING Activation. Int. J. Radiat. Oncol. Biol. Phys. 2022, 112, 1243–1255. [Google Scholar] [CrossRef] [PubMed]
  83. Ma, H.; Kang, Z.; Foo, T.K.; Shen, Z.; Xia, B. Disrupted BRCA1-PALB2 interaction induces tumor immunosuppression and T-lymphocyte infiltration in HCC through cGAS-STING pathway. Hepatology 2023, 77, 33–47. [Google Scholar] [CrossRef] [PubMed]
  84. Cheng, C.C.; Ho, A.S.; Peng, C.L.; Chang, J.; Sie, Z.L.; Wang, C.L.; Chen, Y.L.; Chen, C.Y. Sorafenib suppresses radioresistance and synergizes radiotherapy-mediated CD8(+) T cell activation to eradicate hepatocellular carcinoma. Int. Immunopharmacol. 2022, 112, 109110. [Google Scholar] [CrossRef] [PubMed]
  85. Li, Z.; Zhang, Y.; Hong, W.; Wang, B.; Chen, Y.; Yang, P.; Zhou, J.; Fan, J.; Zeng, Z.; Du, S. Gut microbiota modulate radiotherapy-associated antitumor immune responses against hepatocellular carcinoma Via STING signaling. Gut Microbes 2022, 14, 2119055. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, L.; Dou, X.; Chen, S.; Yu, X.; Huang, X.; Zhang, L.; Chen, Y.; Wang, J.; Yang, K.; Bugno, J.; et al. YTHDF2 inhibition potentiates radiotherapy antitumor efficacy. Cancer Cell 2023, 41, 1294–1308.e1298. [Google Scholar] [CrossRef]
  87. Hack, S.P.; Spahn, J.; Chen, M.; Cheng, A.L.; Kaseb, A.; Kudo, M.; Lee, H.C.; Yopp, A.; Chow, P.; Qin, S. IMbrave 050: A Phase III trial of atezolizumab plus bevacizumab in high-risk hepatocellular carcinoma after curative resection or ablation. Future Oncol. 2020, 16, 975–989. [Google Scholar] [CrossRef]
  88. Goyal, L.; Vogel, A.; Zhu, A.X.; Cheng, A.-L.; Yau, T.; Zhou, J.; Uppot, R.N.; Kim, E.; Malhotra, U.; Siegel, A.B.; et al. P024 KEYNOTE-937 trial in progress: Adjuvant pembrolizumab for hepatocellular carcinoma and complete radiologic response after surgical resection or local ablation. Gut 2021, 70, A22. [Google Scholar] [CrossRef]
  89. Knox, J.; Cheng, A.; Cleary, S.; Galle, P.; Kokudo, N.; Lencioni, R.; Park, J.; Zhou, J.; Mann, H.; Morgan, S.; et al. A phase 3 study of durvalumab with or without bevacizumab as adjuvant therapy in patients with hepatocellular carcinoma at high risk of recurrence after curative hepatic resection or ablation: EMERALD-2. Ann. Oncol. 2019, 30, iv59–iv60. [Google Scholar] [CrossRef]
  90. 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]
  91. Vogel, A.; Waidmann, O.; Müller, T.; Siegler, G.M.; Goetze, T.O.; Toni, E.N.D.; Gonzalez-Carmona, M.A.; Hausner, G.; Geissler, M.; Weikersthal, L.F.v.; et al. IMMULAB: A phase II trial of immunotherapy with pembrolizumab in combination with local ablation for patients with early-stage hepatocellular carcinoma (HCC). J. Clin. Oncol. 2023, 41, 555. [Google Scholar] [CrossRef]
  92. Shi, L.; Zhou, C.; Long, X.; Li, H.; Chen, C.; Peng, C.; Li, P.; Li, J.; Gu, S.; Liang, B.; et al. 949P Thermal ablation plus toripalimab in patients with advanced hepatocellular carcinoma: Phase I results from a multicenter, open-label, controlled phase I/II trial (IR11330). Ann. Oncol. 2021, 32, S826. [Google Scholar] [CrossRef]
  93. Kudo, M.; Guo, Y.; Hua, Y.; Zhao, M.; Xing, W.; Zhang, Y.; Liu, R.; Ren, Z.; Gu, S.; Lin, Z.; et al. TALENTACE: A phase III, open-label, randomized study of on-demand transarterial chemoembolization combined with atezolizumab + bevacizumab or on-demand transarterial chemoembolization alone in patients with untreated hepatocellular carcinoma. J. Clin. Oncol. 2022, 40, TPS487. [Google Scholar] [CrossRef]
  94. Saborowski, A.; Waldschmidt, D.; Hinrichs, J.; Ettrich, T.J.; Martens, U.M.; Mekolli, A.; Toni, E.N.D.; Berg, T.; Geißler, M.; Hausner, G.; et al. IMMUTACE: A biomarker-orientated phase II, single-arm, open-label AIO study of transarterial chemoembolization (TACE) in combination with nivolumab performed for intermediate-stage hepatocellular carcinoma (HCC; AIO-HEP-0217)—Updated efficacy results. J. Clin. Oncol. 2022, 40, 4116. [Google Scholar] [CrossRef]
  95. Xiaoyun, Z.; Zhu, X.; Feng, X.; Han, W.; Yan, M.L.; Xie, F.; Zhang, S.; Zhang, Y.; Jiang, X.; Peng, W.; et al. 715P The safety and efficacy of lenvatinib combined with TACE and PD-1 inhibitors (Len-TAP) versus TACE alone in the conversion resection for initially unresectable hepatocellular carcinoma: Interim results from a multicenter prospective cohort study. Ann. Oncol. 2022, 33, S870. [Google Scholar] [CrossRef]
  96. Fulgenzi, C.A.M.; Cortellini, A.; D’Alessio, A.; Thomas, R.; Tait, P.; Ross, P.J.; Young, A.-M.; Talbot, T.; Goldin, R.; Ward, C.; et al. A phase Ib study of pembrolizumab following trans-arterial chemoembolization (TACE) in hepatocellular carcinoma (HCC): PETAL. J. Clin. Oncol. 2022, 40, e16195. [Google Scholar] [CrossRef]
  97. Harding, J.J.; Yarmohammadi, H.; Reiss, K.A.; Chou, J.F.; Capanu, M.; Do, R.K.G.; Khalil, D.; Dika, I.H.E.; Giardina, J.D.; Merghoub, T.; et al. Nivolumab (NIVO) and drug eluting bead transarterial chemoembolization (deb-TACE): Preliminary results from a phase I study of patients (pts) with liver limited hepatocellular carcinoma (HCC). J. Clin. Oncol. 2020, 38, 525. [Google Scholar] [CrossRef]
  98. 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]
  99. He, A.R.; Toskich, B. The ROWAN study: Safety and efficacy of transarterial radioembolization with Y-90 glass microspheres and the STRIDE regimen in hepatocellular carcinoma. J. Clin. Oncol. 2023, 41, TPS622. [Google Scholar] [CrossRef]
  100. 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 2022, 10, e005457. [Google Scholar] [CrossRef]
  101. 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]
  102. McRee, A.J.; Helft, P.R.; Harris, W.P.; Sanoff, H.K.; Johnson, M.; Yu, M.; O’Neil, B. A study of pembrolizumab (pembro) in combination with Y90 radioembolization in patients (pts) with poor prognosis hepatocellular carcinoma (HCC) with preserved liver function. J. Clin. Oncol. 2022, 40, 422. [Google Scholar] [CrossRef]
  103. Fenton, S.E.; Kircher, S.M.; Mulcahy, M.F.; Mahalingam, D.; Salem, R.; Lewandowski, R.; Kulik, L.; Benson, A.B.; Kalyan, A. A phase I study of nivolumab (NIVO) in combination with TheraSphere (Yttrium-90) in patients with advanced hepatocellular cancer. J. Clin. Oncol. 2021, 39, e16183. [Google Scholar] [CrossRef]
  104. Chen, Y.; Yang, P.; Du, S.; Zhou, J.; Huang, C.; Zhu, W.; Hu, Y.; Yu, Y.; Liu, T.; Zeng, Z. A phase II study of stereotactic body radiotherapy (SBRT) combined with sintilimab in patients with recurrent or oligometastatic hepatocellular carcinoma (HCC). J. Clin. Oncol. 2022, 40, 4071. [Google Scholar] [CrossRef]
  105. Wang, K.; Yu, H.M.; Xiang, Y.J.; Cheng, Y.Q.; Ni, Q.Z.; Guo, W.X.; Shi, J.; Feng, S.; Zhai, J.; Cheng, S.Q. Efficacy and safety of radiotherapy combined with atezolizumab plus bevacizumab in treating hepatocellular carcinoma with portal vein tumour thrombus: A study protocol. BMJ Open 2022, 12, e064688. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, B.; Yue, J.; Shi, X.; Cui, K.; Li, L.; Zhang, C.; Sun, P.; Zhong, J.; Li, Z.; Zhao, L. Protocol of notable-HCC: A phase Ib study of neoadjuvant tislelizumab with stereotactic body radiotherapy in patients with resectable hepatocellular carcinoma. BMJ Open 2022, 12, e060955. [Google Scholar] [CrossRef]
  107. Brown, T.J.; Minn, A.J.; Carpenter, E.L.; Ben-Josef, E.; Karasic, T.B. A phase I clinical trial of stereotactic body radiotherapy with atezolizumab and bevacizumab in advanced hepatocellular carcinoma. J. Clin. Oncol. 2023, 41, TPS626. [Google Scholar] [CrossRef]
  108. Juloori, A.; Katipally, R.R.; Lemons, J.M.; Singh, A.K.; Iyer, R.; Robbins, J.R.; George, B.; Hall, W.A.; Pitroda, S.P.; Arif, F.; et al. Phase 1 Randomized Trial of Stereotactic Body Radiation Therapy Followed by Nivolumab plus Ipilimumab or Nivolumab Alone in Advanced/Unresectable Hepatocellular Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2023, 115, 202–213. [Google Scholar] [CrossRef]
  109. Chiang, C.L.; Chiu, K.W.H.; Chan, K.S.K.; Lee, F.A.S.; Li, J.C.B.; Wan, C.W.S.; Dai, W.C.; Lam, T.C.; Chen, W.; Wong, N.S.M.; et al. Sequential transarterial chemoembolisation and stereotactic body radiotherapy followed by immunotherapy as conversion therapy for patients with locally advanced, unresectable hepatocellular carcinoma (START-FIT): A single-arm, phase 2 trial. Lancet Gastroenterol. Hepatol. 2023, 8, 169–178. [Google Scholar] [CrossRef]
  110. Chiang, C.L.; Chang, S.L.; Chan, S.-K.; Lee, A.S.; Chiu, K.W.H.; Yeung, V.T.Y.; Wong, N.S.M.; Lee, V.W.Y.; Lau, V.W.H.; Man, N.K.; et al. Preliminary results of sequential transarterial chemoembolization and stereotactic body radiotherapy followed by immunotherapy using single tremelimumab regular interval durvalumab in locally advanced, unresectable hepatocellular carcinoma (START-FIT using STRIDE): A single-arm, phase II study. J. Clin. Oncol. 2023, 41, 4124. [Google Scholar] [CrossRef]
  111. Yu, J.; Yu, X.L.; Han, Z.Y.; Cheng, Z.G.; Liu, F.Y.; Zhai, H.Y.; Mu, M.J.; Liu, Y.M.; Liang, P. Percutaneous cooled-probe microwave versus radiofrequency ablation in early-stage hepatocellular carcinoma: A phase III randomised controlled trial. Gut 2017, 66, 1172–1173. [Google Scholar] [CrossRef]
  112. Sherman, M. Recurrence of hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 2045–2047. [Google Scholar] [CrossRef] [PubMed]
  113. Number, P. IMMUTACE: A Phase 2 Single-Arm, Open-Label Study of Transarterial Chemoembolization in Combination With Nivolumab Performed for Intermediate-Stage Hepatocellular Carcinoma. Gastroenterol. Hepatol. 2021, 17, 16–17. [Google Scholar]
  114. Li, J.X.; Su, T.S.; Gong, W.F.; Zhong, J.H.; Yan, L.Y.; Zhang, J.; Li, L.Q.; He, M.L.; Zhang, R.J.; Du, Y.Q.; et al. Combining stereotactic body radiotherapy with camrelizumab for unresectable hepatocellular carcinoma: A single-arm trial. Hepatol. Int. 2022, 16, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
  115. Wu, C.H.; Lan, C.H.; Wu, K.L.; Wu, Y.M.; Jane, W.N.; Hsiao, M.; Wu, H.C. Hepatocellular carcinoma-targeted nanoparticles for cancer therapy. Int. J. Oncol. 2018, 52, 389–401. [Google Scholar] [CrossRef]
  116. Repáraz, D.; Aparicio, B.; Llopiz, D.; Hervás-Stubbs, S.; Sarobe, P. Therapeutic Vaccines against Hepatocellular Carcinoma in the Immune Checkpoint Inhibitor Era: Time for Neoantigens? Int. J. Mol. Sci. 2022, 23, 2022. [Google Scholar] [CrossRef]
  117. Fujiwara, N.; Kobayashi, M.; Fobar, A.J.; Hoshida, A.; Marquez, C.A.; Koneru, B.; Panda, G.; Taguri, M.; Qian, T.; Raman, I.; et al. A blood-based prognostic liver secretome signature and long-term hepatocellular carcinoma risk in advanced liver fibrosis. Med 2021, 2, 836–850.e10. [Google Scholar] [CrossRef]
  118. Pfister, D.; Nunez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A.; et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 2021, 592, 450–456. [Google Scholar] [CrossRef]
  119. Fujiwara, N.; Kubota, N.; Crouchet, E.; Koneru, B.; Marquez, C.A.; Jajoriya, A.K.; Panda, G.; Qian, T.; Zhu, S.; Goossens, N.; et al. Molecular signatures of long-term hepatocellular carcinoma risk in nonalcoholic fatty liver disease. Sci. Transl. Med. 2022, 14, eabo4474. [Google Scholar] [CrossRef]
Figure 1. Expected response to immune checkpoint inhibitors (ICIs) in “hot”, “excluded”, and “cold” tumors. Brown (3,3′-diaminobenzidine (DAB)) staining represents CD8. These “hot”, “excluded”, and “cold” tumor phenotypes are determined according to the absolute abundance of T cells. The abundance of infiltrated immune cells that exert antitumor activity is expected to be positively correlated with susceptibility to ICI. Created with BioRender.com accessed on 18 October 2023.
Figure 1. Expected response to immune checkpoint inhibitors (ICIs) in “hot”, “excluded”, and “cold” tumors. Brown (3,3′-diaminobenzidine (DAB)) staining represents CD8. These “hot”, “excluded”, and “cold” tumor phenotypes are determined according to the absolute abundance of T cells. The abundance of infiltrated immune cells that exert antitumor activity is expected to be positively correlated with susceptibility to ICI. Created with BioRender.com accessed on 18 October 2023.
Cancers 15 05072 g001
Figure 2. Immune responses against cancers after immune checkpoint inhibitors and locoregional therapy in the cancer–immunity cycle. The cancer–immunity cycle consists of six steps. The red, blue, and green molecules represent plausible molecular modifications by ablation, transarterial embolization, and radiotherapy, respectively. Created with BioRender.com accessed on 12 September 2023. LCKs, lymphocyte-specific protein tyrosine kinases; TERT, telomerase reverse transcriptase; HMGB-1, high mobility group box 1; IL, interleukin; CXCL10, C-X-C motif chemokine ligand 10; NK, natural killer; MDSC, myeloid-derived immunosuppressive cells; PD-1, programmed death-1; TIM3, T cell immunoglobulin mucin-3; APC, antigen-presenting cells; CTLA4, cytotoxic T-lymphocyte antigen 4; VEGF, vascular endothelial growth factor; PD-L1, programmed death ligand-1; TREM2, triggering receptor expressed on myeloid cells 2; TAM, tumor-associated macrophage; MHC, major histocompatibility complex.
Figure 2. Immune responses against cancers after immune checkpoint inhibitors and locoregional therapy in the cancer–immunity cycle. The cancer–immunity cycle consists of six steps. The red, blue, and green molecules represent plausible molecular modifications by ablation, transarterial embolization, and radiotherapy, respectively. Created with BioRender.com accessed on 12 September 2023. LCKs, lymphocyte-specific protein tyrosine kinases; TERT, telomerase reverse transcriptase; HMGB-1, high mobility group box 1; IL, interleukin; CXCL10, C-X-C motif chemokine ligand 10; NK, natural killer; MDSC, myeloid-derived immunosuppressive cells; PD-1, programmed death-1; TIM3, T cell immunoglobulin mucin-3; APC, antigen-presenting cells; CTLA4, cytotoxic T-lymphocyte antigen 4; VEGF, vascular endothelial growth factor; PD-L1, programmed death ligand-1; TREM2, triggering receptor expressed on myeloid cells 2; TAM, tumor-associated macrophage; MHC, major histocompatibility complex.
Cancers 15 05072 g002
Table 1. In-progress clinical trials of combination therapies of locoregional therapies and ICIs.
Table 1. In-progress clinical trials of combination therapies of locoregional therapies and ICIs.
Clinical Trial Identifier (Study Acronym) *Study Phase#Pt.LocationsEligible HCC StageCombined ICI and TKIORRDCRMedian PFS, RFSOSTRAE Clinical Trial Status
(As of 15 September 2023)
Reference
Ablation
(or surgical resection)
NCT04102098 (IMbrave050)3668GlobalAdjuvantAtezolizumab
+ bevacizumab
--22.1 m-41.0%Active, not recruiting[87]
NCT03383458
(CheckMate 9DX)
3545GlobalAdjuvantNivolumab-----Active, not recruiting
NCT03867084
(KEYNOTE-937)
3950GlobalAdjuvantPembrolizumab-----Active, not recruiting[88]
NCT03847428
(EMERALD-2)
3908GlobalAdjuvantDurvalumab
+/− bevacizumab
-----Active, not recruiting[89]
NCT04727307
(AB-LATE02)
2202FranceNeoadjuvantAtezolizumab
+ bevacizumab
-----Recruitingn.a.
NCT03939975250ChinaBCLC B/CPembrolizumab or nivolumab
or toripalimab
24.0%68.0%5.0 m16.9 m14.0%Completed[90]
NCT03753659
(IMMULAB)
230GermanyBCLC APembrolizumab--17.4 m--Active, not recruiting[91]
NCT03864211
(IR11330)
1/2145ChinaBCLC CToripalimab31.2–37.5%---25.0%Active, not recruiting[92]
NCT046524401/230ChinaBCLC A/BTislelizumab-----Recruitingn.a.
TACE
NCT04340193
(CheckMate 74W)
326GlobalBCLC B/CNivolumab + ipilimumab-----Active, not recruitingn.a.
NCT04712643 (TALENTACE)3342China, JapanBCLC B/CAtezolizumab
+ bevacizumab
-----Active, not recruiting[93]
NCT05301842 (EMERALD-3)3725GlobalBCLC B/CDurvalumab + tremelimumab
+/− lenvatinib
-----Recruiting[32]
NCT04268888
(TACE-3)
2/3522United KingdomBCLC BNivolumab-----Recruitingn.a.
NCT03572582 (IMMUTACE)249GermanyBCLC BNivolumab71.4%75.5%7.2 m28.3 m34.7%Completed[94]
NCT04814030
(AIPD1)
280ChinaBCLC CSintilimab-----Recruitingn.a.
NCT05751343255ChinaBCLC B/CAtezolizumab
+ bevacizumab
-----Recruitingn.a.
NCT03638141230United StatesBCLC BDurvalumab + tremelimumab-----Recruitingn.a.
NCT049978501/2142ChinaBCLC B/CCamrelizumab
or sintilimab
+ lenvatinib
78.9%94.4%17.1 m-53.5%Enrolling by invitation[95]
NCT03397654 (PETAL)1/226United KingdomBCLC BPembrolizumab--10.8 m-21.0%Active, not recruiting[96]
NCT03143270120United StatesBCLC BNivolumab22.2%100.0%---Active, not recruiting[97]
Ablation and TACE
NCT018536181/261United StatesBCLC B/CTremelimumab26.3%-7.4 m12.3 m-Completed[98]
NCT04220944145ChinaBCLC B/CSintilimab-----Recruitingn.a.
TARE
NCT05377034 (STRATUM)2176SingaporeBCLC B/CAtezolizumab
+ bevacizumab
-----Recruitingn.a.
NCT05063565 (ROWAN)2100United States, SpainBCLC B/CDurvalumab + tremelimumab-----Recruiting[99]
NCT03380130
(NASIR-HCC)
241SpainBCLC B/CNivolumab41.5%92.7%9.0 m20.9 m21.4%Completed[100]
NCT03033446240SingaporeBCLC B/CNivolumab30.6%---6.0%Active, not recruiting[101]
NCT03099564130United StatesBCLC B/CPembrolizumab27.0%84.7%8.6 m22.0 m-Active, not recruiting[102]
NCT04605731132United StatesBCLC B/CDurvalumab + tremelimumab-----Recruitingn.a.
NCT02837029127United StatesBCLC B/CNivolumab-82.0%---Completed[103]
NCT0381256212United StatesNeoadjuvantNivolumab-----Active, not recruitingn.a.
SBRT
NCT04167293 (ISBRT01)2/3116ChinaBCLC CSintilimab-----Recruitingn.a.
NCT04913480237Hong KongBCLC B/CDurvalumab-----Recruitingn.a.
NCT03857815230ChinaBCLC BSintilimab96.0%---4.0%Recruiting[104]
NCT05396937242ChinaBCLC CAtezolizumab
+ bevacizumab
-----Recruiting[105]
NCT03316872230CanadaBCLC B/CPembrolizumab-----Recruitingn.a.
NCT052863201/227TaiwanBCLC B/CPembrolizumab
+ lenvatinib
Not yet recruitingn.a.
NCT04857684120United StatesNeoadjuvantAtezolizumab
+ bevacizumab
-----Recruitingn.a.
NCT05185531 (Notable-HCC)120ChinaNeoadjuvantTislelizumab-----Recruiting[106]
NCT05096715120United StatesBCLC B/CAtezolizumab
+ bevacizumab
-----Not yet recruitingn.a.
NCT05488522118United StatesBCLC B/CAtezolizumab
+ bevacizumab
-----Recruiting[107]
NCT03203304114United StatesBCLC B/CNivolumab + ipilimumab57.1%-11.6 m41.6 m61.6%Terminated[108]
TACE and SBRT
NCT03817736
(START-FIT)
233Hong KongBCLC B/CAvelumab42.4%---33.3%Active, not recruiting[109]
NCT04988945233Hong KongBCLC B/CDurvalumab + tremelimumab81.3%---31.3%Recruiting[110]
* Data were obtained from ClinicalTrials.gov (https://clinicaltrials.gov/) accessed on 15 September 2023. #Pt., the number of patients. Percentages of grade 3 or 4 treatment-related adverse events are shown. ICI, immune checkpoint inhibitor; HCC, hepatocellular carcinoma; TKI, tyrosine kinase inhibitor; ORR, objective response rate; DCR, disease control rate; PFS, progression-free survival; RFS, recurrence-free survival; OS, overall survival; TRAE, treatment-related adverse event; BCLC, Barcelona Clinic Liver Cancer; n.a., not available.
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

Tamai, Y.; Fujiwara, N.; Tanaka, T.; Mizuno, S.; Nakagawa, H. Combination Therapy of Immune Checkpoint Inhibitors with Locoregional Therapy for Hepatocellular Carcinoma. Cancers 2023, 15, 5072. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15205072

AMA Style

Tamai Y, Fujiwara N, Tanaka T, Mizuno S, Nakagawa H. Combination Therapy of Immune Checkpoint Inhibitors with Locoregional Therapy for Hepatocellular Carcinoma. Cancers. 2023; 15(20):5072. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15205072

Chicago/Turabian Style

Tamai, Yasuyuki, Naoto Fujiwara, Takamitsu Tanaka, Shugo Mizuno, and Hayato Nakagawa. 2023. "Combination Therapy of Immune Checkpoint Inhibitors with Locoregional Therapy for Hepatocellular Carcinoma" Cancers 15, no. 20: 5072. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15205072

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