
Combination of radiotherapy and ICIs in advanced hepatocellular carcinoma: A systematic review of current evidence and future prospects (Review)
- Authors:
- Published online on: May 14, 2025 https://doi.org/10.3892/ol.2025.15088
- Article Number: 342
-
Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Hepatocellular carcinoma (HCC) is the sixth most common malignant tumor in the world and the third greatest cause of cancer-associated mortality (1). The global incidence rate of HCC is expected to exceed 1 million cases by 2025, mainly driven by chronic viral hepatitis and metabolic related liver disease, of which metabolic related liver disease cases account for 15–20% in western countries (2). HCC often remains asymptomatic in the early stages, therefore, diagnosis typically occurs in the late stages, and the 5-year survival rate of patients with advanced HCC (aHCC) is <15%, underscoring the limitations of current aHCC treatment options (3).
Since its confirmation in the SHARP trial, sorafenib, a multi-kinase inhibitor, has been routinely used as the first-line treatment for aHCC (4). However, the effectiveness of sorafenib is limited. Compared with the placebo, it increased the median overall survival (mOS) by 2.8 months. However, it typically has side effects (hand-foot skin reaction in 30–60% of patients, diarrhea in 40–55%) that adversely affect the quality of life (4). Due to these constraints, new treatments for patients with aHCC have been researched to increase survival rates and quality of life (5).
Immune checkpoint inhibitors (ICIs) have potential in treating aHCC because of their immune system modulatory activity (5,6). The CheckMate 040 and KEYNOTE-224 trials (7,8) revealed that nivolumab and pembrolizumab are both safe and effective in treating aHCC.
Despite advances in immunotherapy and targeted therapy for aHCC, a considerable proportion of patients are unable to obtain effective treatment because of variables such as the complicated tumor microenvironment (TME) of HCC (9). As a result, novel therapeutic strategies integrate multiple existing therapeutic approaches to use the complementary advantages of each strategy while addressing their limitations. Radiotherapy (RT) + ICI combinations have shown promise in non-small cell lung cancer and melanoma, improving survival by modulating the TME (10). Therefore, RT + ICI combinations in HCC may enhance tumor response to immunotherapy. Clinicians have hypothesized combining RT and ICIs as a potential synergistic method to improve the treatment of aHCC (11–13).
The justification for combining RT and ICIs in treating aHCC stems from their complementary mechanisms and antitumor efficacy. This combination therapy may be a more effective and comprehensive method of combating aHCC (14). RT has traditionally been considered a local treatment, but has been discovered to promote systemic immune responses via the ‘abscopal effect’, in which the tumor in the irradiated area degenerates due to immune activation (15,16). Radiation-induced cell death causes the production of tumor-associated antigens and damage-associated molecular patterns (DAMPs), which initiate an antitumor immune response (17,18). Furthermore, radiation can promote tumor immunogenicity by increasing expression of major histocompatibility complex class I and T cell infiltration and altering the TME (11,19). These radiation-induced changes convert immunologically ‘cold’ tumors (lacking immune cell infiltration and antigen presentation) into ‘hot’ tumors (with immune recognition and attack capacity), thereby increasing their susceptibility to ICIs (20,21). Moreover, ICIs boost the systemic immune response, overcoming the limitations of radiation, which may not totally eradicate malignancy.
RT can modulate the TME, leading to increased infiltration of CD8+ T cells and production of effector cytokines such as IFN-γ and decreased levels of regulatory T cells (Tregs) (22,23). These changes enhance the efficacy of ICIs by improving immune system recognition of tumor cells (24). Furthermore, preclinical investigations have shown synergistic effects of RT and ICIs in aHCC models, with increased antitumor effects observed when these treatments are combined (22,25).
However, there are challenges associated with optimizing the combination of RT and ICIs for aHCC (26,27). Determining the appropriate sequencing, dosage and timing of RT in comparison with those of ICIs is key (28,29). Additionally, discovering predictive biomarkers for response to combination therapy is essential for tailored treatment approaches (30).
The present review aimed to summarize processes through which RT and ICIs may interact to improve clinical outcomes and methods to enhance this combination technique, as well as the potential of low-dose radiation treatment (LDRT) as a complementary or alternative strategy to high-dose regimens, particularly when paired with ICIs (31–33). Furthermore, the present review aimed to highlight the role of advanced imaging techniques and specialized equipment in accurately targeting tumor tissue and minimizing damage to surrounding healthy tissue, which is key for the successful implementation of LDRT (34,35).
Additionally, the present review aimed to summarize clinical trials assessing the safety and efficacy of combining RT and ICIs in aHCC, including those that have assessed the impact of different sequencing and timing strategies on treatment outcomes (36–39). Identification and validation of reliable biomarkers for predicting response to combined RT and ICIs may improve patient selection and therapy optimization (40–47).
RT in HCC
Historically, the use of external beam radiation therapy (EBRT) for HCC has been limited because of the radiosensitivity of the liver and the risk of radiation-induced liver disease (RILD) (48). Attempts to cure HCC with conventional radiation have shown limited efficacy and severe toxicity because it is challenging to provide an adequate radiation dose to tumors without destroying the surrounding healthy liver tissue (49,50). Developments in imaging and radiation transmission technology improved the accuracy of tumor targeting (51–53). Steroetactic body radiation therapy (SBRT) and three-dimensional conformal radiation have promise in the treatment of HCC (54,55). According to reports, local control rates have improved, and hazardous reactions are now acceptable (56,57). Blomgren et al (58), for example, reported that high-dose fractionated radiation achieves local tumor suppression in different types of liver cancer, including HCC, with manageable side effects.
Despite these advancements, irradiation for HCC has limitations, including radiation-induced hepatotoxicity, variability in tumor radiation sensitivity and technical complexity (59,60). Technical complexity refers to the challenges involved in accurately targeting tumors in the liver while sparing surrounding healthy tissue. This is difficult due to the anatomical variations of the liver, its proximity to key structures and the need to optimize radiation planning to minimize damage to healthy liver tissue while ensuring sufficient tumor dose delivery. These challenges require advanced imaging technology and highly precise radiation delivery systems, which can vary depending on tumor size, location and liver function (61).
Liver toxicity has switched from ‘classical’ to ‘non-classical’ RILD, with the former primarily characterized by widespread damage due to whole-liver irradiation, while the latter manifests as localized damage and elevated liver enzymes. This is often associated with an increased production of pro-inflammatory cytokines, such as TNF-α and interleukins, which contribute to hepatocyte injury and the activation of fibrogenic pathways. By contrast, non-classical RILD involves more localized liver damage and is associated with elevated levels of transaminases and jaundice (62). This form of toxicity is hypothesized to result from endothelial cell damage, increased vascular permeability and a dysregulated immune response in the irradiated liver region (62). Baseline liver function is the primary predictor of liver toxicity after SBRT, and patients with Child-Pugh grade ≥B8 have a greater risk (63,64). Cárdenes et al (63) revealed that a Child-Pugh score of ≥8 is associated with severe liver toxicity or mortality in ≤6 months. The American Society for Radiation Oncology guidelines establish dosage limits on the basis of various Child-Pugh scores: 15–18, 13–15 and 8–10 Gy for non-cirrhotic, Child-Pugh A and B7 grades, respectively (65,66). Technical advancements have markedly decreased these constraints. Modern EBRT employs three-dimensional computed tomography (CT) and intensity-modulated RT, as well as improved imaging technology, to portray tumors while preserving normal liver tissue (67,68). These approaches accommodate differing dosage distributions, allowing the dose of the tumor-specific treatment to be gradually increased while minimizing exposure to healthy liver parenchyma (69) (Fig. 1).
Chemotherapy and targeted molecular therapies are less popular in the treatment of aHCC due to key limitations. Chemotherapy exhibits poor efficacy, with high rates of chemo-resistance and considerable toxicity, particularly in patients with underlying liver dysfunction, resulting in minimal survival benefits (70). Targeted therapies, such as sorafenib and other tyrosine kinase inhibitors, offer modest improvements in OS while being associated with substantial side effects and a narrow therapeutic window (71). Additionally, resistance to targeted therapies often develops, driven by tumor heterogeneity and the immunosuppressive TME. These factors, combined with their lack of impact on modulating the TME, render chemotherapy and targeted molecular therapies less favorable compared with emerging strategies such as the combination of RT and ICIs. By contrast, RT combined with ICIs is preferred for aHCC (27). RT techniques such as SBRT and proton therapy provide precise dose delivery, ideal for targeting small- and medium-sized tumors in complex locations, making it more effective for tumors that are large or irregularly shaped (72). Second, RT enhances immune activation by altering the TME, promoting immune cell infiltration and improving tumor recognition, which synergizes with ICIs to boost immune responses, particularly in advanced stages where immune activity is often suppressed (73). Additionally, combining RT with ICIs helps overcome resistance to immunotherapy by enhancing T cell activation and preventing immune exhaustion (27). Moreover, RT + ICIs offers comprehensive treatment by targeting both primary tumors and metastases and providing broader control over both systemic and local disease, unlike therapies such as radioembolization, which are more suited for localized disease or portal vein invasion. Additionally, compared with tyrosine kinase inhibitors (such as sorafenib), which primarily target angiogenesis, RT + ICI combinations address both local tumor control and systemic immune activation, offering a dual therapeutic advantage (74). Finally, ongoing clinical trials show promising results, indicating that RT + ICIs provides enhanced antitumor activity and safety for patients with aHCC (36,75). Furthermore, careful patient selection, precise treatment planning and interdisciplinary collaboration are needed to improve outcomes and minimize adverse effects.
The combination of radiotherapy and systemic treatments, particularly ICIs, represents a promising frontier in the treatment of aHCC. Radiation can affect the TME, improve immune system identification of tumor cells and interact with ICIs (24). Clinical trials are being performed to determine the efficacy and safety of RT in combination with ICIs and other systemic treatments (36,75). This holistic strategy aims to improve the local and systemic control of aHCC while also addressing primary tumors and potential metastatic illness (Fig. 2).
ICIs in HCC
The introduction of ICIs has transformed the therapeutic landscape for HCC, particularly for patients with advanced-stage illness. Traditional systemic medicines provide some survival advantage for these patients, emphasizing the need for innovative therapeutic options. ICIs have emerged as promising medications that induce an efficient antitumor response via the human immune system (4,25).
The ICI revolution in HCC is based on immune checkpoint regulation, specifically PD-1/PD-L1 and CTLA-4 (76,77). Tumors frequently exploit these molecular pathways, which are necessary for maintaining immunological homeostasis, to avoid immune surveillance. Checkpoint inhibitors revive depleted T cells by eliminating inhibitory signals, boosting antitumor immunity (78). (Fig. 3).
PD-1 drugs, including nivolumab and pembrolizumab, have demonstrated clinical success in treating aHCC. The CheckMate 040 and KEYNOTE-224 studies (8,79) reported objective response rates of 14–20 and 17%, respectively, indicating the possibility of disrupting the PD-1 axis. However, these response rates underscore the limited efficacy in treating HCC relative to other types of cancer (80). This variability in patient reactions highlights the complicated immunological landscape of aHCC and presence of immune subgroups within the patient population. The relatively low curative effect may be attributed to several factors, including the inherent immunosuppressive environment, the high heterogeneity of HCC tumors, the possibility of chronic liver diseases that impair immune function and the presence of immunosuppressive cells in the TME (81). Furthermore, HCC tumors may lack sufficient novel antigens or tumor mutation burdens to elicit robust immune responses (82). This requires further research into the mechanisms of response and resistance, as well as the creation of more tailored ICIs, to overcome these limits and enhance the treatment outcomes for patients with HCC.
Given the poor efficacy of ICIs as monotherapies, researchers have investigated combination methods with other treatment modalities, such as RT and anti-antigenic drugs, to improve therapeutic outcomes (83–86). A notable example is the IMbrave 150 study (87), which assessed the combination of atezolizumab (an anti-PD-L1 antibody) and bevacizumab [an anti-vascular endothelial growth factor (VEGF) drug]. After a median follow-up of 61.9 months, the mOS was 21.0 months [95% confidence interval (CI): 10.4–31.6 months]. The OS rates for 3, 4 and 5 years were 36.4, 25.7 and 25.7%, respectively (88). This combination is superior to sorafenib in the first-line treatment of aHCC, and exhibits synergistic potential. This technique improves the killing effect of T cells on tumor cells and modulates the tumor vascular system, potentially enhancing drug delivery and T cell infiltration, allowing the simultaneous treatment of multiple aspects of tumor biology (89).
The most recent advancements in ICIs emphasize the prospect of PD-1/PD-L1 inhibition, and the concurrent investigation of CTLA-4 blockade has potential, particularly in combination therapy (23,90,91). The HIMALAYA study (92), which used durvalumab (an anti-PD-L1 antibody) and tremelimumab (an anti-CTLA-4 antibody), highlights the concept of dual checkpoint blocking in aHCC. This method targets independent but complementary immune regulatory pathways, potentially overcoming resistance mechanisms and increasing the breadth and depth of antitumor immune responses.
The synergy of these pathways can be attributed to their distinct effects on T cell priming and effector activity. CTLA-4 suppression impacts the priming phase in lymphoid organs, whereas PD-1/PD-L1 blocking improves effector T cell activity (93,94). This complementary process provides a theoretical foundation for development of future combined therapy options.
Rationale for combining RT + ICIs
For patients with aHCC, who frequently have few therapeutic options and a poor prognosis, combining RT + ICIs may provide clinical advantages. In addition to enhancing local tumor management, the synergistic effect can trigger systemic antitumor responses, leading to the resolution of primary tumors and metastatic lesions (95,96) (Fig. 4). This strategy is based on radiation-induced immunogenic cell death (ICD), a unique form of cell death that can effectively promote the immune system to target tumor cells.
Following RT, DAMPs are produced as a result of radiation-induced cell death. DAMPs, such as calreticulin, high mobility group box 1 protein (HMGB1) and ATP, are released from tumor cells undergoing necrosis or apoptosis. These molecules serve a key role in modulating immune responses in HCC by acting as endogenous ligands for pattern-recognition receptors (PRRs), including toll-like receptors, on immune cells (97–99). Following receptor activation, DAMPs initiate innate immune responses, which are key for the subsequent activation of adaptive immunity (97). This innate-to-adaptive immune transition is key in the context of ICI therapy, as it helps bridge the gap between the two immune systems.
DAMPs are key for the maturation of dendritic cells (DCs) and the enhancement of antigen presentation (98,100). They promote the activation of DCs and other antigen-presenting cells, improving T cell priming and activation (101,102). Garg et al (97) have revealed that DAMPs, such as surface-exposed calreticulin, secreted ATP and passively released HMGB1, interact with phagocytosis receptors, purinergic receptors and PRRs, respectively. These interactions are key for inducing ICD, leading to the activation of potent anticancer immunity (97,103–106). This helps overcome the immune suppression in the advanced stages of HCC. Immune activation is an important strategy to overcome immune suppression in aHCC (107). Chen et al (108) demonstrated that heat shock protein 70 (HSP70) levels (a DAMP) are markedly higher in the serum of patients with HCC compared with healthy individuals, while antibody levels remain unchanged, suggesting that HSP70 may counter immune suppression via non-antibody mechanisms. Ren et al (109) revealed that during HCC development, damaged or dying cells release DAMPs, which serve as ligands for cyclic GMP-AMP synthase (cGAS). This activates the cGAS-stimulator of interferon genes signaling pathway, potentially enhancing the anti-tumor immune response of the body (109). Furthermore, when synergized with ICIs such as PD-1/PD-L1 inhibitors, DAMPs enhance immune responses by restoring T cell function and counteracting immune exhaustion (97).
RT further illustrates the importance of DAMPs in immune activation. Radiation-induced ICD results in the release of DAMPs, such as calreticulin, HMGB1 and ATP, which enhance the ability of the immune system to target tumor cells. These DAMPs activate both innate and adaptive immune responses, effectively transforming irradiated tumors into in situ vaccines (99,110). Calreticulin, which translocates to the cell surface following radiation, serves as an ‘eat-me’ signal for DCs, prompting them to recognize and engulf dying tumor cells. HMGB1 and ATP stimulate immune activation by binding receptors on DCs, leading to the initiation of robust anti-tumor immunity (98).
The complementary effects of radiation-induced DAMPs (such as calreticulin, HMGB1, ATP) on the TME further elucidate the synergistic interaction between RT and ICIs. Singh et al (111) reported that intercellular adhesion molecule 1 (ICAM-1) serves a key role in breast carcinoma metastasis, promoting adhesion between breast cancer and endothelial cells, enhancing circulating tumor cell cluster formation and tumor cell survival. RT can modify the TME by altering cytokine secretion patterns, increasing the expression of adhesion molecules such as ICAM-1 and vascular CAM-1, and altering chemokine gradients (111). These modifications increase the recruitment and activation of effector immune cells, such as cytotoxic T lymphocytes and natural killer cells. Moreover, ICIs avoid T cell failure by inhibiting inhibitory signals (PD-1/PD-L1 and CTLA-4), increasing long-term antitumor activity and enhancing these effects. Exosomes regulate tumor growth, invasion, metastasis, angiogenesis and immune therapy resistance, serving a key role in intercellular communication in the TME (112,113). They carry proteins, DNA, microRNA and long non-coding RNA, altering recipient cell functions and phenotypes. RT factors enhance or inhibit these effects, influencing tumor progression and increasing their vulnerability to ICIs (114).
Preclinical investigations have established the existence of synergistic effects of RT + ICIs in models of aHCC (22,23,115). Pedros et al (115) revealed that in mouse models of melanoma and prostate cancer, the CTLA4 signaling pathway within Treg cells is key for suppressing tumor immunity. Tregs lacking relevant key proteins weaken their inhibitory function, leading to inhibited tumor growth, increased levels of effector T cells and enhanced function (115). Studies have indicated increased infiltration of CD8+ T cells and production of effector cytokines, including IFN-γ and decreased Tregs within the TME (22,23). Furthermore, radiation stimulates the diversification of the T cell receptor (TCR) pool, identifying a broader range of tumor antigens (116,117). Binder et al (117) revealed that RT stimulates the diversification of TCR pool and expands the recognition range of tumor antigens. When combined with ICIs (such as anti-CTLA-4), RT can increase the diversity of TCR clones within tumors and enhance the anti-tumor effect of the immune system (116,117). This diversification decreases the chance of immune escape and increases the persistent response.
Clinically, the abscopal effect, a phenomenon in which local RT causes regression of distant unirradiated tumors, demonstrates the efficacy of this combination method in systemic treatment (118). Although uncommon, the abscopal effect demonstrates that combining RT + ICIs induces broad antitumor immunity. ICIs have the ability to elicit broad antitumor immune responses. For patients with aHCC with numerous foci and metastases, this combined therapy may provide a solution for both local and distant tumor burden (119).
The continuous updating of technology and developments in treatment procedures provide a foundation for further improving the overall effectiveness of RT and ICIs. Advanced radiation procedures, such as SBRT, accurately administer high radiation doses while causing minimum injury to adjacent tissue (120). When combined with novel ICIs, these strategies provide therapeutic advantages, including tailored cancer vaccines, off-the-shelf T cell therapies, and oncolytic viruses, which operate through complementary but non-overlapping mechanisms (113). Furthermore, treatment planning and prediction can be improved by computational technologies such as radiomics and artificial intelligence.
For patients with aHCC, RT in conjunction with ICIs may improve survival rates and quality of life by balancing local tumor focus resolution with systemic metastasis management. Future research should refine treatment regimens, identify resistance mechanisms and develop biomarkers that can predict treatment efficacy.
RT + ICIs in aHCC
The combined strategy of RT + ICIs in treating aHCC is increasingly supported by preclinical and clinical evidence due to the synergistic effect of radiation-induced immunomodulation and IC suppression (84,121). Preclinical studies have clarified how RT increases tumor immunogenicity (10,22,122). Radiation, for example, causes ICD, as evidenced by the release of DAMPs, including calreticulin, HMGB1 and ATP. These DAMPs stimulate DC maturation and antigen presentation, effectively bridging innate and acquired immunity (100,123). Furthermore, RT increases tumor-associated antigen expression and upregulates key MHC-like molecules, making cytotoxic T cells more visible in the tumor (124).
In aHCC-specific models, the combination of RT + ICIs has demonstrated substantial antitumor activity (22,125). Combining radiation with anti-PD-1 or anti-CTLA-4 antibodies has increased CD8+ T cell infiltration and decreased Tregs within the TME (116,126,127). Twyman-Saint et al (116) reported the tumor regression effect of anti-CTLA4 antibody and radiation therapy on patients with metastatic melanoma, and reproduced this effect in a mouse model. Drug resistance is associated with the upregulation of PD-L1 and T cell depletion in melanoma cells (115). The optimal therapeutic response requires the combination of RT, anti-CTLA4 and anti-PD-L1/PD-1 (116,126,127). Also, RT changes the immune-suppressing TME by decreasing immune-suppressing cytokines and myeloid-derived suppressor cells (10,128).
Early-phase trials have investigated the safety of RT + ICIs and preliminary efficacy in patients with aHCC, translating these preclinical findings into clinical settings (36,129). Juloori et al (36) conducted a phase I trial in patients with aHCC, comparing SBRT followed by nivolumab alone or nivolumab with ipilimumab (36). This study revealed adequate safety. Within 6 months of starting SBRT, 15.4% of patients experienced dose-limiting effects. The combination of SBRT with nivolumab and ipilimumab had ORR of 57%, a median progression-free survival time of 11.6 months and a mOS time of 41.6 months. Tai et al (129) conducted a phase I trial in patients with aHCC, evaluating the combination of SBRT and nivolumab (129). This combination was well tolerated, and the toxicity was moderate, consisting primarily of fatigue and skin events. Preliminary efficacy results suggested an encouraging ORR.
Other phase II trials have evaluated the efficacy of RT + ICI combinations in larger populations (38,130,131). Li et al (38) evaluated a combined therapy of SBRT, camrelizumab (an immune checkpoint inhibitor) and apatinib (a targeted anti-angiogenic drug) in HCC patients with portal vein tumor thrombus. The study reported a median overall survival of 12.7 months (95% CI: 10.2-not reached) and a median progression-free survival of 4.6 months (95% CI: 3.3–7.0). While the treatment was generally tolerable, over 22% of patients experienced grade 3 or higher adverse effects, such as hypertension or liver enzyme elevations.
Owing to the lack of extensive phase III data, retrospective analyses provide key insight into the true efficacy of RT-ICIs (37,132,133). Ning et al (134) conducted a retrospective analysis on the outcomes of 36 patients with aHCC treated with RT and anti-PD-1 drugs and reported markedly higher ORR and OS rates when compared with ICIs alone.
Optimizing the combination of RT and ICIs for aHCC remains an issue. Determining the appropriate sequencing, dosage and timing of RT in comparison with those of ICIs is key (28,29). Furthermore, discovering predictive biomarkers for response to combination medication is key for tailored treatment (80).
Clinical trials will likely offer high-level data to demonstrate the efficacy and safety of combined RT + ICIs in aHCC (Table I). These studies are key for future clinical practice and to redefine the landscape of aHCC treatment.
Optimizing RT + ICIs in aHCC treatment
The synergistic combination of RT and ICIs is a feasible technique for improving treatment for patients with aHCC. Numerous variables must be addressed to obtain the best therapeutic benefit while minimizing toxicity, including dosage, timing, staging and patient-specific characteristics.
The dosage and manner of RT must be optimized to maximize synergy with ICIs in patients with aHCC. Historically, higher doses/fraction, such as those utilized in hypofractionated SBRT, have been employed to produce strong ICD and increase antitumor immune responses (135,136). Robbins et al (137) used a 40 Gy regimen delivered in five parts, demonstrating a move toward providing greater doses over shorter treatment duration to enhance immune activation and improve patient compliance (137).
There is interest in the immunostimulant properties of LDRT (31). LDRT at a dose of <2 Gy increases T cell infiltration, improves antigen presentation, regulates the tumor vasculature and induces phenotypical changes in tumor cells, increasing their susceptibility to immune-mediated death (32,33). LDRT may enhance the immune response while causing less damage to healthy tissue compared to conventional high-dose radiation therapy. The appropriate dosage range for balancing immune activation and tumor management is an important area for further study, as LDRT may provide a complementary or alternative strategy to high-dose regimens, particularly when paired with ICIs.
The choice of RT mode is important in maximizing treatment outcomes. Advanced technologies, such as proton treatment and SBRT, provide precision targeting, maximizing the tumor dose while minimizing damage to adjacent healthy tissue. Several factors influence the choice between these techniques, including the size and location of the tumor, the liver function of the patient and the specific immunological goals (34,35). Integrating LDRT as a supplement to these RT techniques may affect immune activation. Given the ability of LDRT to regulate the TME to promote immune-mediated tumor elimination (138), its combination may improve the overall therapeutic efficacy of these targeted RT technologies. Although LDRT holds promise, its precise delivery and monitoring presents challenges that limit its wider clinical application (139). The technical complexity of LDRT requires highly accurate targeting of tumor tissue while minimizing damage to surrounding healthy tissue, demanding advanced imaging techniques and specialized equipment, which may not be available in all clinical settings. Additionally, the variability in tumor shape, locations and movement, such as respiratory motion, complicates consistent treatment delivery, necessitating real-time adjustments (140). Achieving the required dosimetric precision is a challenge, as high doses of radiation delivered to small, localized areas can cause toxicity to nearby structures (141). Furthermore, real-time monitoring of treatment effects on both tumor and normal tissue remains difficult, as current imaging technology may not provide sufficient resolution or immediate feedback, leading to potential inaccuracies.
The sequence and timing of RT and ICIs are key factors in enhancing the synergy of combination therapy in patients with aHCC. ICI after radiotherapy is more effective than taking ICI before radiotherapy in other tumors (142). This technique exploits immunostimulatory effects, increasing immunotherapeutic drug efficacy (126,143–145). Juloori et al (36) initiated ICIs 14 days after SBRT completion to capitalize on the peak period of radiation-induced antigen release and immune cell infiltration, which normally occurs 1–2 weeks after RT. By delivering ICIs during this window, treatment takes advantage of the increased immunogenicity caused by RT.
By contrast, RT following ICI therapy has resulted in less notable effects. ICIs alone may not fully activate the immune system to exploit subsequent RT-induced ICD and antigen presentation (116). As a result, ICIs after RT remains the most commonly used approach in clinical practice. Theelen et al (146), which examined the efficacy of pembrolizumab with or without RT in patients with metastatic non-small cell lung cancer, lends support to this viewpoint; 564 individuals receiving pembrolizumab were evaluated, with 89 (15.8%) receiving RT either before or during therapy. There was no notable difference in the OS rate between patients who underwent RT (mOS;10.6 months) and patients who did not receive RT (mOS;10.9 months), with an adjusted hazard ratio of 1.13 (95% CI: 0.82–1.54; P=0.46). Similarly, the two groups had no notable difference in progression-free survival or ORR (147). These findings indicate that RT after or during ICI therapy may not provide considerable benefits in terms of survival or tumor response.
Determining the best sequence and timing of RT and ICIs is important in aHCC management (143). The timing of these procedures influences immediate treatment outcomes and may have an impact on the long-term prognoses of patients. To achieve the best clinical results, the treatment plan must be tailored to each patient and the immune dynamics of the TME, while also identifying and validating reliable biomarkers for predicting response to RT + ICIs (30).
Identifying biomarkers that predict response to RT + ICIs is important for patient selection and therapy optimization. Circulating tumor DNA is a potential biomarker that allows non-invasive monitoring of tumor load and molecular alterations throughout treatment (40–42). Analysis of cytokine profiles and circulating immune cell subsets offer information regarding systemic immunological activation and overall immune status, potentially indicating ICI timing and dose. In a study of 11 individuals receiving radioembolization or systemic therapy, assessments at 16 time points revealed that changes in tumor methylation score (TMS) more accurately predict tumor progression than α-fetoprotein (AFP), with Area Under the Receiver Operating Characteristic curve (AUROC) values of 0.800 and 0.783, respectively. Moreover, combining AFP and TMS into a composite score improved the accuracy of distinguishing tumor evolution, with an AUROC of 0.892 (43).
Tumor tissue biomarkers, such as immune cell infiltration patterns, molecular expression of ICs and tumor mutation and novel antigen loads, directly indicate TME features and susceptibility to combined RT + ICI methods (44,45). Imaging modalities such as CT, magnetic resonance imaging (MRI) and positron emission tomography (PET) serve a key role in improving RT planning by providing detailed, high-resolution images of the tumor and surrounding structures. CT imaging is essential for precise localization and accurate dose delivery as it allows 3D visualization of the size and position of the tumor. MRI is useful in delineating liver tumors due to its high soft tissue contrast, helping to distinguish tumors from adjacent liver tissue, which is key in liver-directed therapy. PET imaging, combined with CT, offers insight into tumor metabolism and activity, allowing more accurate identification of active tumor regions that may require higher radiation doses. These imaging techniques, when integrated into RT planning, enable more accurate tumor targeting, improved dose distribution and decreased toxicity to surrounding healthy tissue (46,47).
Biomarker discovery and application may improve the understanding of the synergistic mechanism of RT and ICIs in aHCC, optimize treatment timing and sequence and improve patient selection accuracy and clinical efficacy. This holistic strategy demonstrates potential for enhancing customized medicine and providing more effective treatment options for patients with aHCC.
Toxicity and safety
With the widespread use of immune checkpoint inhibitors (ICIs), the incidence of immune-related adverse events (irAEs) has risen, affecting 60–80% of patients, with severe cases in 10–15%. Active management strategies, such as corticosteroids, are effective in addressing these events (148,149). Fatigue, dermatological responses, colitis, hepatitis, pneumonitis and endocrinopathy are among the most common AEs (150,151). Although the incidence of AEs may increase, these can be effectively managed with close monitoring and appropriate interventions, hence optimizing safety (152,153) (Fig. 5).
Clinical trials have assessed the safety of combining RT and ICIs in the treatment of aHCC and support the viability of this strategy (36–39). There was no notable difference between the rates of irAEs in patients who received a combination treatment plan and those who received only ICIs. These findings suggest that RT does not worsen immune-associated toxicity. Hepatotoxicity has garnered attention among irAEs since both can impair liver function (154,155). However, combination therapy does not always result in a disproportionate increase in hepatotoxicity (132,134,156). Trials of the treatment of aHCC with RT and ICI therapy have revealed that the incidence of severe hepatotoxicity is often tolerable (132,134,156). The aforementioned studies have also stressed the importance of regular hepatic function monitoring and aggressive risk-reduction methods. Additionally, Li et al (156) conducted a multicenter trial to evaluate the safety of a combination of RT + ICIs for patients with portal vein tumor thrombus: The findings revealed that the combined treatment was practical and had manageable safety. Compared with the historical control group that received either method alone, the number of AEs did not increase appreciably. The aforementioned study revealed that attentive monitoring and prompt treatment are key for improving patient outcomes.
The aforementioned clinical investigations indicate that while RT + ICIs may pose additional risks, these side effects are typically controllable and preclude the practicality of a combined strategy. Numerous studies have shown that the safety of this combination therapy can be improved through proactive techniques such as dose modification, vigilant monitoring and patient education (157,158). This enables practitioners to increase treatment outcomes while still ensuring patient safety. The combination of RT + ICIs is a promising therapeutic option for patients with aHCC.
Future directions and limitations
Combination strategies of RT + ICIs have improved the efficacy of aHCC treatment and novel combinations and strategies may maximize the benefits of currently available treatment options.
Combination of trans arterial chemoembolisation (TACE), RT and ICIs for the treatment of aHCC uses synergistic effects to improve therapeutic efficacy. TACE decreases the tumor load and releases tumor antigens, whereas SBRT increases the anticancer immune response by inducing ICD and influencing the TME. Subsequent ICI therapy increases T cell-mediated antitumor immunity, potentially extending the therapeutic window. The START-FIT trial investigated sequential TACE, SBRT and avelumab; 55% of patients exhibited tumors that decreased to a level that would allow surgical intervention and 42% achieved complete remission (75). This novel method improves local tumor control while also combating off-site disease development via systemic immune activation. This synergy occurs through increased tumor antigen presentation, PD-L1 expression levels following radiation and recruitment of tumor-infiltrating lymphocytes. Larger randomized controlled studies are needed to confirm the efficacy and safety of triple therapy as a strategy to downstage initially unresectable aHCC, making it eligible for curative treatments such as surgery or ablation.
Researchers have explored other triple therapies, including RT, ICIs and other drugs, such as anti-VEGF therapy or new immunomodulators designed to target aspects of tumor biology and the immune microenvironment at the same time, potentially leading to more potent and durable antitumor responses (37,84,159).
As understanding of the biology of aHCC and patient-specific variables increases, there is a greater emphasis on establishing tailored treatments (160,161). Tailoring therapy seeks to maximize the efficacy of combining RT + ICIs while limiting toxicity. The development and validation of biomarkers predictive of response to combination therapy is a key focus (162,163). Several possible biomarkers are being investigated, including the tumor mutational load, PD-L1 expression and genetic changes (164,165). The goal is to create a comprehensive biomarker panel that will guide treatment selection and sequencing, ensuring patients receive the most suitable and effective treatment.
There is emphasis on assessing therapies beyond clinical efficacy, including patient-centered outcomes such as quality of life, accessibility and cost-effectiveness (166,167). Understanding the real-world applicability of these medicines is key to turning clinical trial results into meaningful benefits for the patient population. Involving patients in research and clinical decision-making ensures their opinions and priorities are considered when developing and implementing treatment. Shared decision-making models encourage collaborative discussion between clinicians and patients to consider individual preferences, quality of life implications and life circumstances, resulting in increased patient satisfaction, treatment adherence and overall outcomes (168,169).
Constraints have hampered the widespread use and optimization of combined RT + ICIs in aHCC treatment. Determining adequate markers for the prognosis of combination therapy is difficult because of the large number of parameters to examine, including tumor features, liver function, general performance status, biomarker profiles and prior treatment history. HCC tumor heterogeneity may result in varying responses to RT and ICIs, making precise treatment outcome prediction difficult.
Another challenge in implementing RT + ICIs in clinical settings is the lack of established treatment procedures. Key topics requiring additional investigation include radiation dose and segmentation schemes, treatment timing and sequencing, ICIs and target volume specification. It is key to have standardized protocols for clinical trials and consistent methods across institutions.
Methodological limitations impede research on the combination of RT + ICIs, affecting the universality and robustness of findings. Several studies (37,133) have limited sample sizes, which decreases their statistical power and ability to establish confident findings. The paucity of large-scale randomized controlled trials comparing combination therapy with standard medication raises questions regarding its true efficacy and safety. The heterogeneity of patient populations and differences in measurement results and toxicity reports hinder the interpretation and comparison. Short-term follow-up may not demonstrate long-term outcomes, late toxicity or delayed responses to ICIs.
Addressing these problems is key for promoting the combination of RT and ICIs in the treatment of aHCC. Future research should prioritize large-scale, multicenter randomized controlled trials and use defined patient groups and standardized programs. Developing and validating predictive biomarkers will improve patient selection, allow individualized treatment options, maximize therapeutic effects and reduce unnecessary toxicity. Studying the optimal radiation dose, segmentation method and treatment sequence may aid in maximizing the synergistic effect of RT and ICIs.
Conclusion
RT + ICIs represents a substantial advancement in treating aHCC. RT increases tumor antigenicity and eliminates immunosuppressive barriers, which improves the efficacy of ICIs (170). Clinical evidence from early-phase trials reveals that combination treatment improves antitumor responses while maintaining tolerable safety (126,142–144). However, issues such as optimum patient selection, dosage technique, therapy sequencing and toxicity control persist. Large-scale randomized controlled studies should be prioritized to confirm therapeutic advantages and enhance treatment strategies. The incorporation of biomarker-driven approaches is key for personalizing medicines to patient and tumor features, potentially increasing response rates while minimizing side effects. Furthermore, prioritizing patient-centered outcomes, such as quality of life, and real-world applicability (e.g., effectiveness in diverse patient populations, treatment accessibility, and long-term safety), is critical for evaluating the practical impact of these therapies. Continuous research efforts to improve this combination, identify resistance mechanisms and develop predictive biomarkers are needed. By addressing the present constraints and supporting tailored treatment options, longer-lasting responses, enhanced OS rate and improved quality of life may be obtained.
Acknowledgements
Not applicable.
Funding
The present study was supported by the 2024 research project from the Technology Department of Sichuan Provincial Science and Central Guidance Local Exploration Fund Project (grant. no. 24ZYZYTS0262).
Availability of data and materials
Not applicable.
Authors' contributions
RC, OJ and LS conceived the study. RC and XY performed the literature review. XZ and RC wrote the manuscript. RC and CM edited the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare they have no competing interests.
Glossary
Abbreviations
Abbreviations:
aHCC |
advanced hepatocellular carcinoma |
SBRT |
stereotactic body radiation therapy |
ICI |
immune checkpoint inhibitor |
PD-1 |
targeted programmed death-1 |
PD-L1 |
programmed death-ligand 1 |
CTLA-4 |
T lymphocyte-associated protein 4 |
OS |
overall survival |
DAMP |
damage-associated molecular pattern |
MHC |
major histocompatibility complex |
EBRT |
external beam radiation therapy |
RILD |
radiation-induced liver disease |
ICD |
immunogenic cell death |
HMGB1 |
high mobility group protein 1 |
Treg |
regulatory T cell |
TCR |
T cell receptor |
CI |
confidence interval |
LDRT |
low-dose radiation treatment |
CT |
computed tomography |
MRI |
magnetic resonance imaging |
irAE |
immune-related adverse event |
References
Forner A, Llovet JM and Bruix J: Hepatocellular carcinoma. Lancet. 379:1245–1255. 2012. View Article : Google Scholar : PubMed/NCBI | |
Renne SL, Sarcognato S, Sacchi D, Guido M, Roncalli M, Terracciano L and Di Tommaso L: Hepatocellular carcinoma: A clinical and pathological overview. Pathologica. 113:203–217. 2021. View Article : Google Scholar : PubMed/NCBI | |
Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J and Finn RS: Hepatocellular carcinoma. Nat Rev Dis Primers. 7:62021. View Article : Google Scholar : PubMed/NCBI | |
Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, et al: Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 359:378–390. 2008. View Article : Google Scholar : PubMed/NCBI | |
Llovet JM, Montal R, Sia D and Finn RS: Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 15:599–616. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sangro B, Sarobe P, Hervas-Stubbs S and Melero I: Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 18:525–543. 2021. View Article : Google Scholar : PubMed/NCBI | |
El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, Kim TY, Choo SP, Trojan J, Welling TH Rd, et al: Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet. 389:2492–2502. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, Verslype C, Zagonel V, Fartoux L, Vogel A, et al: Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. Lancet Oncol. 19:940–952. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kudo M: Combination cancer immunotherapy in hepatocellular carcinoma. Liver Cancer. 7:20–27. 2018. View Article : Google Scholar : PubMed/NCBI | |
Formenti SC, Rudqvist NP, Golden E, Cooper B, Wennerberg E, Lhuillier C, Vanpouille-Box C, Friedman K, Ferrari de Andrade L, Wucherpfennig KW, et al: Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med. 24:1845–1851. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sharabi AB, Lim M, DeWeese TL and Drake CG: Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 16:e498–e509. 2015. View Article : Google Scholar : PubMed/NCBI | |
Dendy MS, Ludwig JM, Stein SM and Kim HS: Locoregional therapy, immunotherapy and the combination in hepatocellular carcinoma: Future directions. Liver Cancer. 8:326–340. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hsu S, Chao Y, Hu Y, Zhang Y, Hong W, Chen Y, Chen R, Zeng Z and Du S: Radiotherapy enhances efficacy of PD-1 inhibitors in advanced hepatocellular carcinoma: A propensity-matched real-world study. Chin Med J (Engl). 137:1332–1342. 2024. View Article : Google Scholar : PubMed/NCBI | |
Sharabi AB, Nirschl CJ, Kochel CM, Nirschl TR, Francica BJ, Velarde E, Deweese TL and Drake CG: Stereotactic radiation therapy augments antigen-specific PD-1-mediated antitumor immune responses via cross-presentation of tumor antigen. Cancer Immunol Res. 3:345–355. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S and Formenti SC: Using immunotherapy to boost the abscopal effect. Nat Rev Cancer. 18:313–322. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Zhang H, Xin Zhang and Liu Y: Abscopal effect: From a rare phenomenon to a new frontier in cancer therapy. Biomark Res. 12:982024. View Article : Google Scholar : PubMed/NCBI | |
Formenti SC and Demaria S: Combining radiotherapy and cancer immunotherapy: A paradigm shift. J Natl Cancer Inst. 105:256–265. 2013. View Article : Google Scholar : PubMed/NCBI | |
Correction. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. 8:e000337corr12020. View Article : Google Scholar : PubMed/NCBI | |
Kim JY, Kim DH, Kim JH, Lee D, Jeon HB, Kwon SJ, Kim SM, Yoo YJ, Lee EH, Choi SJ, et al: Soluble intracellular adhesion molecule-1 secreted by human umbilical cord blood-derived mesenchymal stem cell reduces amyloid-β plaques. Cell Death Differ. 19:680–691. 2012. View Article : Google Scholar : PubMed/NCBI | |
Demaria S, Coleman CN and Formenti SC: Radiotherapy: Changing the game in immunotherapy. Trends Cancer. 2:286–294. 2016. View Article : Google Scholar : PubMed/NCBI | |
Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, Caux C and Depil S: Cold Tumors: A therapeutic challenge for immunotherapy. Front Immunol. 10:1682019. View Article : Google Scholar : PubMed/NCBI | |
Friedman D, Baird JR, Young KH, Cottam B, Crittenden MR, Friedman S, Gough MJ and Newell P: Programmed cell death-1 blockade enhances response to stereotactic radiation in an orthotopic murine model of hepatocellular carcinoma. Hepatol Res. 47:702–714. 2017. View Article : Google Scholar : PubMed/NCBI | |
Rimassa L, Finn RS and Sangro B: Combination immunotherapy for hepatocellular carcinoma. J Hepatol. 79:506–515. 2023. View Article : Google Scholar : PubMed/NCBI | |
Tojjari A, Yu J and Saeed A: Immunotherapy and radiation therapy combinatorial approaches in hepatocellular carcinoma. Cancers (Basel). 16:10582024. View Article : Google Scholar : PubMed/NCBI | |
Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, et al: Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 382:1894–1905. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dawood ZS, Brown ZJ, Alaimo L, Lima HA, Shaikh C, Katayama ES, Munir MM, Moazzam Z, Endo Y, Woldesenbet S and Pawlik TM: Comparison of tumor response and outcomes of patients with hepatocellular carcinoma after multimodal treatment including immune checkpoint inhibitors-a systematic review and meta-analysis. HPB (Oxford). 26:618–629. 2024. View Article : Google Scholar : PubMed/NCBI | |
Lee YH, Tai D, Yip C, Choo SP and Chew V: Combinational immunotherapy for hepatocellular carcinoma: Radiotherapy, immune checkpoint blockade and beyond. Front Immunol. 11:5687592020. View Article : Google Scholar : PubMed/NCBI | |
Bicak M, Cimen Bozkus C and Bhardwaj N: Checkpoint therapy in cancer treatment: Progress, challenges, and future directions. J Clin Invest. 134:e1848462024. View Article : Google Scholar : PubMed/NCBI | |
Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR and Fu YX: Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 124:687–695. 2014. View Article : Google Scholar : PubMed/NCBI | |
Harding JJ, Nandakumar S, Armenia J, Khalil DN, Albano M, Ly M, Shia J, Hechtman JF, 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. 25:2116–2126. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Zhou J, Wu M, Hu C, Yang J, Li D, Wu P, Chen Y, Chen P, Lin S, et al: Low-dose total body irradiation can enhance systemic immune related response induced by hypo-fractionated radiation. Front Immunol. 10:3172019. View Article : Google Scholar : PubMed/NCBI | |
Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L, et al: Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 24:589–602. 2013. View Article : Google Scholar : PubMed/NCBI | |
Li S, Li K, Wang K, Yu H, Wang X, Shi M, Liang Z, Yang Z, Hu Y, Li Y, et al: Low-dose radiotherapy combined with dual PD-L1 and VEGFA blockade elicits antitumor response in hepatocellular carcinoma mediated by activated intratumoral CD8(+) exhausted-like T cells. Nat Commun. 14:77092023. View Article : Google Scholar : PubMed/NCBI | |
Mizumoto M, Okumura T, Hashimoto T, Fukuda K, Oshiro Y, Fukumitsu N, Abei M, Kawaguchi A, Hayashi Y, Ookawa A, et al: Proton beam therapy for hepatocellular carcinoma: A comparison of three treatment protocols. Int J Radiat Oncol Biol Phys. 81:1039–1045. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, Schipper MJ and Feng M: Outcomes after stereotactic body radiotherapy or radiofrequency ablation for hepatocellular carcinoma. J Clin Oncol. 34:452–459. 2016. View Article : Google Scholar : PubMed/NCBI | |
Juloori A, Katipally RR, Lemons JM, Singh AK, Iyer R, Robbins JR, George B, Hall WA, Pitroda SP, 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. 115:202–213. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Ji X, Sun J, Zhang A, Jia J, Zhang T, Li W and Duan X: Stereotactic body radiotherapy combined with lenvatinib with or without PD-1 inhibitors as initial treatment for unresectable hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 120:1363–1376. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li JX, Su TS, Gong WF, Zhong JH, Yan LY, Zhang J, Li LQ, He ML, Zhang RJ, Du YQ, et al: Combining stereotactic body radiotherapy with camrelizumab for unresectable hepatocellular carcinoma: A single-arm trial. Hepatol Int. 16:1179–1187. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chiang CL, Chan ACY, Chiu KWH and Kong FS: Combined stereotactic body radiotherapy and checkpoint inhibition in unresectable hepatocellular carcinoma: A potential synergistic treatment strategy. Front Oncol. 9:11572019. View Article : Google Scholar : PubMed/NCBI | |
Zhu Q, Xie J, Mei W and Zeng C: Methylated circulating tumor DNA in hepatocellular carcinoma: A comprehensive analysis of biomarker potential and clinical implications. Cancer Treat Rev. 128:1027632024. View Article : Google Scholar : PubMed/NCBI | |
Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, Yi S, Shi W, Quan Q, Li K, et al: Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater. 16:1155–1161. 2017. View Article : Google Scholar : PubMed/NCBI | |
Howell J, Atkinson SR, Pinato DJ, Knapp S, Ward C, Minisini R, Burlone ME, Leutner M, Pirisi M, Buttner R, et al: Identification of mutations in circulating cell-free tumour DNA as a biomarker in hepatocellular carcinoma. Eur J Cancer. 116:56–66. 2019. View Article : Google Scholar : PubMed/NCBI | |
Angeli-Pahim I, Chambers A, Duarte S, Soma D, Beduschi T, Sahin I, Hughes S and Zarrinpar A: Methylated ctDNA quantification: Noninvasive approach to monitoring hepatocellular carcinoma burden. J Am Coll Surg. 238:770–778. 2024. View Article : Google Scholar : PubMed/NCBI | |
Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al: PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 515:568–571. 2014. View Article : Google Scholar : PubMed/NCBI | |
Fridman WH, Zitvogel L, Sautes-Fridman C and Kroemer G: The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 14:717–734. 2017. View Article : Google Scholar : PubMed/NCBI | |
Sun R, Limkin EJ, Vakalopoulou M, Dercle L, Champiat S, Han SR, Verlingue L, Brandao D, Lancia A, Ammari S, et al: A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: An imaging biomarker, retrospective multicohort study. Lancet Oncol. 19:1180–1191. 2018. View Article : Google Scholar : PubMed/NCBI | |
Han X, Guo Y, Ye H, Chen Z, Hu Q, Wei X, Liu Z and Liang C: Development of a machine learning-based radiomics signature for estimating breast cancer TME phenotypes and predicting anti-PD-1/PD-L1 immunotherapy response. Breast Cancer Res. 26:182024. View Article : Google Scholar : PubMed/NCBI | |
Kim J and Jung Y: Radiation-induced liver disease: Current understanding and future perspectives. Exp Mol Med. 49:e3592017. View Article : Google Scholar : PubMed/NCBI | |
Lawrence TS, Robertson JM, Anscher MS, Jirtle RL, Ensminger WD and Fajardo LF: Hepatic toxicity resulting from cancer treatment. Int J Radiat Oncol Biol Phys. 31:1237–1248. 1995. View Article : Google Scholar : PubMed/NCBI | |
Emami B, Lyman J, Brown A, Coia L, Goitein M, Munzenrider JE, Shank B, Solin LJ and Wesson M: Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys. 21:109–122. 1991. View Article : Google Scholar : PubMed/NCBI | |
Rammohan N, Randall JW and Yadav P: History of technological advancements towards MR-Linac: The future of image-guided radiotherapy. J Clin Med. 11:47302022. View Article : Google Scholar : PubMed/NCBI | |
Khaledi N, Khan R and Gräfe JL: Historical progress of stereotactic radiation surgery. J Med Phys. 48:312–327. 2023. View Article : Google Scholar : PubMed/NCBI | |
Khoo VS, Dearnaley DP, Finnigan DJ, Padhani A, Tanner SF and Leach MO: Magnetic resonance imaging (MRI): considerations and applications in radiotherapy treatment planning. Radiother Oncol. 42:1–15. 1997. View Article : Google Scholar : PubMed/NCBI | |
Lawrence TS, Ten Haken RK, Kessler ML, Robertson JM, Lyman JT, Lavigne ML, Brown MB, DuRoss DJ, Andrews JC, Ensminger WD, et al: The use of 3-D dose volume analysis to predict radiation hepatitis. Int J Radiat Oncol Biol Phys. 23:781–788. 1992. View Article : Google Scholar : PubMed/NCBI | |
Timmerman RD, Kavanagh BD, Cho LC, Papiez L and Xing L: Stereotactic body radiation therapy in multiple organ sites. J Clin Oncol. 25:947–952. 2007. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Ke Q, Huang Q, Shao L, Chen J and Wu J: Stereotactic body radiotherapy versus radiofrequency ablation for hepatocellular carcinoma: A systematic review and meta-analysis. Int J Hyperthermia. 37:1313–1321. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wu DH, Liu L and Chen LH: Therapeutic effects and prognostic factors in three-dimensional conformal radiotherapy combined with transcatheter arterial chemoembolization for hepatocellular carcinoma. World J Gastroenterol. 10:2184–2189. 2004. View Article : Google Scholar : PubMed/NCBI | |
Blomgren H, Lax I, Naslund I and Svanstrom R: Stereotactic high dose fraction radiation therapy of extracranial tumors using an accelerator. Clinical experience of the first thirty-one patients. Acta Oncol. 34:861–870. 1995. View Article : Google Scholar : PubMed/NCBI | |
Hall JT, Moon AM, Young M, Tan X, Darawsheh R, Danquah F, Tepper JE and Yanagihara TK: Biochemical safety of SBRT to multiple intrahepatic lesions for hepatocellular carcinoma. J Hepatocell Carcinoma. 11:443–454. 2024. View Article : Google Scholar : PubMed/NCBI | |
Hardy-Abeloos C, Lazarev S, Ru M, Kim E, Fischman A, Moshier E, Rosenzweig K and Buckstein M: Safety and efficacy of liver stereotactic body radiation therapy for hepatocellular carcinoma after segmental transarterial Radioembolization. Int J Radiat Oncol Biol Phys. 105:968–976. 2019. View Article : Google Scholar : PubMed/NCBI | |
Park SH, Kim JC and Kang MK: Technical advances in external radiotherapy for hepatocellular carcinoma. World J Gastroenterol. 22:7311–7321. 2016. View Article : Google Scholar : PubMed/NCBI | |
Koay EJ, Owen D and Das P: Radiation-induced liver disease and modern radiotherapy. Semin Radiat Oncol. 28:321–331. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cárdenes HR, Price TR, Perkins SM, Maluccio M, Kwo P, Breen TE, Henderson MA, Schefter TE, Tudor K, Deluca J and Johnstone PA: Phase I feasibility trial of stereotactic body radiation therapy for primary hepatocellular carcinoma. Clin Transl Oncol. 12:218–225. 2010. View Article : Google Scholar : PubMed/NCBI | |
Culleton S, Jiang H, Haddad CR, Kim J, Brierley J, Brade A, Ringash J and Dawson LA: Outcomes following definitive stereotactic body radiotherapy for patients with Child-Pugh B or C hepatocellular carcinoma. Radiother Oncol. 111:412–417. 2014. View Article : Google Scholar : PubMed/NCBI | |
Apisarnthanarax S, Barry A, Cao M, Czito B, DeMatteo R, Drinane M, Hallemeier CL, Koay EJ, Lasley F, Meyer J, et al: External beam radiation therapy for primary liver cancers: An ASTRO clinical practice guideline. Pract Radiat Oncol. 12:28–51. 2022. View Article : Google Scholar : PubMed/NCBI | |
Mayo CS, Moran JM, Bosch W, Xiao Y, McNutt T, Popple R, Michalski J, Feng M, Marks LB, Fuller CD, et al: American association of physicists in medicine task Group 263: Standardizing nomenclatures in radiation oncology. Int J Radiat Oncol Biol Phys. 100:1057–1066. 2018. View Article : Google Scholar : PubMed/NCBI | |
Schaub SK, Hartvigson PE, Lock MI, Hoyer M, Brunner TB, Cardenes HR, Dawson LA, Kim EY, Mayr NA, Lo SS and Apisarnthanarax S: Stereotactic body radiation therapy for hepatocellular carcinoma: Current trends and controversies. Technol Cancer Res Treat. 17:15330338187902172018. View Article : Google Scholar : PubMed/NCBI | |
Rim CH and Seong J: Application of radiotherapy for hepatocellular carcinoma in current clinical practice guidelines. Radiat Oncol J. 34:160–167. 2016. View Article : Google Scholar : PubMed/NCBI | |
Crane CH and Koay EJ: Solutions that enable ablative radiotherapy for large liver tumors: Fractionated dose painting, simultaneous integrated protection, motion management, and computed tomography image guidance. Cancer. 122:1974–1986. 2016. View Article : Google Scholar : PubMed/NCBI | |
Deng GL, Zeng S and Shen H: Chemotherapy and target therapy for hepatocellular carcinoma: New advances and challenges. World J Hepatol. 7:787–798. 2015. View Article : Google Scholar : PubMed/NCBI | |
Huang A, Yang XR, Chung WY, Dennison AR and Zhou J: Targeted therapy for hepatocellular carcinoma. Signal Transduct Target Ther. 5:1462020. View Article : Google Scholar : PubMed/NCBI | |
Lee SU and Kim TH: Current evidence and the potential role of proton beam therapy for hepatocellular carcinoma. Clin Mol Hepatol. 29:958–968. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wennerberg E, Lhuillier C, Vanpouille-Box C, Pilones KA, García-Martínez E, Rudqvist NP, Formenti SC and Demaria S: Barriers to radiation-induced in situ tumor vaccination. Front Immunol. 8:2292017. View Article : Google Scholar : PubMed/NCBI | |
Lu Z, Polan DF, Wei L, Aryal MP, Fitzpatrick K, Wang C, Cuneo KC, Evans JR, Roseland ME, Gemmete JJ, et al: PET/CT-based absorbed dose maps in (90)Y selective internal radiation therapy correlate with spatial changes in liver function derived from dynamic MRI. J Nucl Med. 65:1224–1230. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chiang CL, Chiu KWH, Chan KSK, Lee FAS, Li JCB, Wan CWS, Dai WC, Lam TC, Chen W, Wong NSM, 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. 8:169–178. 2023. View Article : Google Scholar : PubMed/NCBI | |
Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, Breder V, Edeline J, Chao Y, Ogasawara S, et al: Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: A Randomized, double-blind, phase III trial. J Clin Oncol. 38:193–202. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kudo M, Matilla A, Santoro A, Melero I, Gracian AC, Acosta-Rivera M, Choo SP, El-Khoueiry AB, Kuromatsu R, El-Rayes B, et al: CheckMate 040 cohort 5: A phase I/II study of nivolumab in patients with advanced hepatocellular carcinoma and Child-Pugh B cirrhosis. J Hepatol. 75:600–609. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wherry EJ and Kurachi M: Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 15:486–499. 2015. View Article : Google Scholar : PubMed/NCBI | |
Yau T, Kang YK, Kim TY, El-Khoueiry AB, Santoro A, Sangro B, Melero I, Kudo M, Hou MM, Matilla A, et al: Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: The CheckMate 040 randomized clinical trial. JAMA Oncol. 6:e2045642020. View Article : Google Scholar : PubMed/NCBI | |
Sangro B, Melero I, Wadhawan S, Finn RS, Abou-Alfa GK, Cheng AL, Yau T, Furuse J, Park JW, Boyd Z, et al: Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J Hepatol. 73:1460–1469. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ringelhan M, Pfister D, O'Connor T, Pikarsky E and Heikenwalder M: The immunology of hepatocellular carcinoma. Nat Immunol. 19:222–232. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, Putra J, Camprecios G, Bassaganyas L, Akers N, et al: Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology. 153:812–826. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cheng AL, Hsu C, Chan SL, Choo SP and Kudo M: Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol. 72:307–319. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ning C, Zhang X, Wang Y, Yang X, Yang X, Chao J, Xun Z, Xue J, Wang Y, Sun H, et al: Radiation therapy with combination therapy of immune checkpoint inhibitors and antiangiogenic therapy for hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 118:1461–1471. 2024. View Article : Google Scholar : PubMed/NCBI | |
Song Y, Fu Y, Xie Q, Zhu B, Wang J and Zhang B: Anti-angiogenic agents in combination with immune checkpoint inhibitors: A promising strategy for cancer treatment. Front Immunol. 11:19562020. View Article : Google Scholar : PubMed/NCBI | |
Vafaei S, Zekiy AO, Khanamir RA, Zaman BA, Ghayourvahdat A, Azimizonuzi H and Zamani M: Combination therapy with immune checkpoint inhibitors (ICIs); a new frontier. Cancer Cell Int. 22:22022. View Article : Google Scholar : PubMed/NCBI | |
Cheng AL, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Lim HY, 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. 76:862–873. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kudo M, Finn RS, Galle PR, Zhu AX, Ducreux M, Cheng AL, Ikeda M, Tsuchiya K, Aoki KI, Jia J and Lencioni R: IMbrave150: Efficacy and safety of atezolizumab plus bevacizumab versus sorafenib in patients with barcelona clinic liver cancer stage B unresectable hepatocellular carcinoma: An exploratory analysis of the phase III study. Liver Cancer. 12:238–250. 2022. View Article : Google Scholar : PubMed/NCBI | |
Fukumura D, Kloepper J, Amoozgar Z, Duda DG and Jain RK: Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat Rev Clin Oncol. 15:325–340. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ali S, Arshad M, Summer M, Zulfiqar M, Noor S, Nazakat L and Javed MA: Recent developments on checkpoint inhibitors, CAR T cells, and beyond for T cell-based immunotherapeutic strategies against cancer. J Oncol Pharm Pract. Mar 28–2025.(Epub ahead of print). View Article : Google Scholar | |
Osaki M and Sakaguchi S: Soluble CTLA-4 regulates immune homeostasis and promotes resolution of inflammation by suppressing type 1 but allowing type 2 immunity. Immunity. 58:889–908.e13. 2025. View Article : Google Scholar : PubMed/NCBI | |
Sangro B, Chan SL, Kelley RK, Lau G, Kudo M, Sukeepaisarnjaroen W, Yarchoan M, De Toni EN, Furuse J, Kang YK, et al: Four-year overall survival update from the phase III HIMALAYA study of tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. Ann Oncol. 35:448–457. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wei SC, Duffy CR and Allison JP: Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8:1069–1086. 2018. View Article : Google Scholar : PubMed/NCBI | |
Saygin I, Cakir E, Kazaz SN, Guvercin AR, Eyupoglu I and Ustaoglu MM: Investigation of the status of immune checkpoint molecules (PD-L1 and PD-1) in meningiomas by immunohistochemistry. Turk J Med Sci. 54:735–743. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Liu X, Chen D and Yu J: Radiotherapy combined with immunotherapy: The dawn of cancer treatment. Signal Transduct Target Ther. 7:2582022. View Article : Google Scholar : PubMed/NCBI | |
Chang Y, Chang M, Bao X and Dong C: Advancements in adoptive CAR immune cell immunotherapy synergistically combined with multimodal approaches for tumor treatment. Bioact Mater. 42:379–403. 2024.PubMed/NCBI | |
Garg AD, Nowis D, Golab J, Vandenabeele P, Krysko DV and Agostinis P: Immunogenic cell death, DAMPs and anticancer therapeutics: An emerging amalgamation. Biochim Biophys Acta. 1805:53–71. 2010.PubMed/NCBI | |
Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P and Vandenabeele P: Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 12:860–875. 2012. View Article : Google Scholar : PubMed/NCBI | |
Garg AD, Galluzzi L, Apetoh L, Baert T, Birge RB, Bravo-San Pedro JM, Breckpot K, Brough D, Chaurio R, Cirone M, et al: Molecular and translational classifications of DAMPs in immunogenic cell death. Front Immunol. 6:5882015. View Article : Google Scholar : PubMed/NCBI | |
Galluzzi L, Buque A, Kepp O, Zitvogel L and Kroemer G: Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 17:97–111. 2017. View Article : Google Scholar : PubMed/NCBI | |
Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, et al: Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 13:1050–1059. 2007. View Article : Google Scholar : PubMed/NCBI | |
Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E, et al: Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 15:1170–1178. 2009. View Article : Google Scholar : PubMed/NCBI | |
Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C, Lambrecht BN and Vandenabeele P: Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol. 32:157–164. 2011. View Article : Google Scholar : PubMed/NCBI | |
Palm NW and Medzhitov R: Pattern recognition receptors and control of adaptive immunity. Immunol Rev. 227:221–233. 2009. View Article : Google Scholar : PubMed/NCBI | |
Rock KL and Kono H: The inflammatory response to cell death. Annu Rev Pathol. 3:99–126. 2008. View Article : Google Scholar : PubMed/NCBI | |
Zitvogel L, Kepp O and Kroemer G: Decoding cell death signals in inflammation and immunity. Cell. 140:798–804. 2010. View Article : Google Scholar : PubMed/NCBI | |
Oura K, Morishita A, Tani J and Masaki T: Tumor immune microenvironment and immunosuppressive therapy in hepatocellular carcinoma: A review. Int J Mol Sci. 22:58012021. View Article : Google Scholar : PubMed/NCBI | |
Chen R, Du J, Zhu H and Ling Q: The role of cGAS-STING signalling in liver diseases. JHEP Rep. 3:1003242021. View Article : Google Scholar : PubMed/NCBI | |
Ren B, Luo S, Xu F, Zou G, Xu G, He J, Huang Y, Zhu H and Li Y: The expression of DAMP proteins HSP70 and cancer-testis antigen SPAG9 in peripheral blood of patients with HCC and lung cancer. Cell Stress Chaperones. 22:237–244. 2017. View Article : Google Scholar : PubMed/NCBI | |
Amiri M, Molavi O, Sabetkam S, Jafari S and Montazersaheb S: Stimulators of immunogenic cell death for cancer therapy: Focusing on natural compounds. Cancer Cell Int. 23:2002023. View Article : Google Scholar : PubMed/NCBI | |
Singh V, Kaur R, Kumari P, Pasricha C and Singh R: ICAM-1 and VCAM-1: Gatekeepers in various inflammatory and cardiovascular disorders. Clin Chim Acta. 548:1174872023. View Article : Google Scholar : PubMed/NCBI | |
Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK and Iyer AK: PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: Mechanism, combinations, and clinical outcome. Front Pharmacol. 8:5612017. View Article : Google Scholar : PubMed/NCBI | |
Pandey P, Khan F, Qari HA, Upadhyay TK, Alkhateeb AF and Oves M: Revolutionization in cancer therapeutics via targeting major immune checkpoints PD-1, PD-L1 and CTLA-4. Pharmaceuticals (Basel). 15:3352022. View Article : Google Scholar : PubMed/NCBI | |
Yu S, Jiang S, Zhou Y, Zhu Z and Yang X: Impact of radiation on exosomes in regulating tumor immune microenvironment. Adv Radiat Oncol. 9:1015492024. View Article : Google Scholar : PubMed/NCBI | |
Pedros C, Canonigo-Balancio AJ, Kong KF and Altman A: Requirement of Treg-intrinsic CTLA4/PKCη signaling pathway for suppressing tumor immunity. JCI Insight. 2:e956922017. View Article : Google Scholar : PubMed/NCBI | |
Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM, et al: Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 520:373–377. 2015. View Article : Google Scholar : PubMed/NCBI | |
Binder DC, Fu YX and Weichselbaum RR: Radiotherapy and immune checkpoint blockade: Potential interactions and future directions. Trends Mol Med. 21:463–465. 2015. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Dong Y, Kong L, Shi F, Zhu H and Yu J: Abscopal effect of radiotherapy combined with immune checkpoint inhibitors. J Hematol Oncol. 11:1042018. View Article : Google Scholar : PubMed/NCBI | |
Bae SH, Chun SJ, Chung JH, Kim E, Kang JK, Jang WI, Moon JE, Roquette I, Mirabel X, Kimura T, et al: Stereotactic body radiation therapy for hepatocellular carcinoma: meta-analysis and international stereotactic radiosurgery society practice guidelines. Int J Radiat Oncol Biol Phys. 118:337–351. 2024. View Article : Google Scholar : PubMed/NCBI | |
Schaue D and McBride WH: Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol. 12:527–540. 2015. View Article : Google Scholar : PubMed/NCBI | |
Xiang YJ, Wang K, Zheng YT, Feng S, Yu HM, Li XW, Cheng X, Cheng YQ, Feng JK, Zhou LP, et al: Effects of stereotactic body radiation therapy plus PD-1 inhibitors for patients with transarterial chemoembolization refractory. Front Oncol. 12:8396052022. View Article : Google Scholar : PubMed/NCBI | |
Zhai J, Gu X, Liu Y, Hu Y, Jiang Y and Zhang Z: Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: An update review. Front Pharmacol. 14:11529342023. View Article : Google Scholar : PubMed/NCBI | |
Kroemer G, Galassi C, Zitvogel L and Galluzzi L: Immunogenic cell stress and death. Nat Immunol. 23:487–500. 2022. View Article : Google Scholar : PubMed/NCBI | |
Manda K, Glasow A, Paape D and Hildebrandt G: Effects of ionizing radiation on the immune system with special emphasis on the interaction of dendritic and T cells. Front Oncol. 2:1022012. View Article : Google Scholar : PubMed/NCBI | |
Kim KJ, Lee HW and Seong J: Combination therapy with anti-T-cell immunoglobulin and mucin-domain containing molecule 3 and radiation improves antitumor efficacy in murine hepatocellular carcinoma. J Gastroenterol Hepatol. 36:1357–1365. 2021. View Article : Google Scholar : PubMed/NCBI | |
Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, Stratford IJ, Poon E, Morrow M, Stewart R, et al: Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 74:5458–5468. 2014. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Zhao L, Fu K, Lin Q, Wen X, Jacobson O, Sun L, Wu H, Zhang X, Guo Z, et al: Integrin α(v)β(3)-targeted radionuclide therapy combined with immune checkpoint blockade immunotherapy synergistically enhances anti-tumor efficacy. Theranostics. 9:7948–7960. 2019. View Article : Google Scholar : PubMed/NCBI | |
Shao Y, Han S, Hou Z, Yang C and Zhao Y: Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon. 10:e368392024. View Article : Google Scholar : PubMed/NCBI | |
Tai WM, Yong WP, Lim C, Low LS, Tham CK, Koh TS, Ng QS, Wang WW, Wang LZ, Hartano S, et al: A phase Ib study of selumetinib (AZD6244, ARRY-142886) in combination with sorafenib in advanced hepatocellular carcinoma (HCC). Ann Oncol. 27:2210–2215. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kim BH, Park HC, Kim TH, Koh YH, Hong JY, Cho Y, Sinn DH, Park B and Park JW: Concurrent nivolumab and external beam radiation therapy for hepatocellular carcinoma with macrovascular invasion: A phase II study. JHEP Rep. 6:1009912023. View Article : Google Scholar : PubMed/NCBI | |
Buckstein M, Kim E, Ozbek U, Tabrizian P, Gunasekaran G, Facciuto M, Rosenzweig K, Llovet JM and Schwartz M: Combination transarterial chemoembolization and stereotactic body radiation therapy for unresectable single large hepatocellular carcinoma: Results from a prospective phase 2 trial. Int J Radiat Oncol Biol Phys. 114:221–230. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chiang CL, Chiu KW, Lee FA, Kong FS and Chan AC: Combined stereotactic body radiotherapy and immunotherapy versus transarterial chemoembolization in locally advanced hepatocellular carcinoma: A propensity score matching analysis. Front Oncol. 11:7988322021. View Article : Google Scholar : PubMed/NCBI | |
Chiang CL, Lee FAS, Chan KSK, Lee VWY, Chiu KWH, Ho RLM, Fong JKS, Wong NSM, Yip WWL, Yeung CSY, et al: Survival outcome analysis of stereotactic body radiotherapy and immunotherapy (SBRT-IO) versus SBRT-alone in unresectable hepatocellular carcinoma. Liver Cancer. 13:265–276. 2023.PubMed/NCBI | |
Ning C, Jia J, Zhang X, Sun J, Wang Y, Xue J, Zhang L, Hou X, Yang X, Sang X, et al: Efficacy and safety of subsequent radiotherapy in patients with advanced-stage hepatocellular carcinoma treated with immune checkpoint inhibitors. Hepatobiliary Surg Nutr. 12:882–897. 2023. View Article : Google Scholar : PubMed/NCBI | |
Filatenkov A, Baker J, Mueller AM, Kenkel J, Ahn GO, Dutt S, Zhang N, Kohrt H, Jensen K, Dejbakhsh-Jones S, et al: Ablative tumor radiation can change the tumor immune cell microenvironment to induce durable complete remissions. Clin Cancer Res. 21:3727–3739. 2015. View Article : Google Scholar : PubMed/NCBI | |
Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, Inghirami G, Coleman CN, Formenti SC and Demaria S: DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 8:156182017. View Article : Google Scholar : PubMed/NCBI | |
Robbins JR, Schmid RK, Hammad AY, Gamblin TC and Erickson BA: Stereotactic body radiation therapy for hepatocellular carcinoma: Practice patterns, dose selection and factors impacting survival. Cancer Med. 8:928–938. 2019. View Article : Google Scholar : PubMed/NCBI | |
Herrera FG, Ronet C, Ochoa de Olza M, Barras D, Crespo I, Andreatta M, Corria-Osorio J, Spill A, Benedetti F, Genolet R, et al: Low-Dose radiotherapy reverses tumor immune desertification and resistance to immunotherapy. Cancer Discov. 12:108–133. 2022. View Article : Google Scholar : PubMed/NCBI | |
Petragallo R, Bardach N, Ramirez E and Lamb JM: Barriers and facilitators to clinical implementation of radiotherapy treatment planning automation: A survey study of medical dosimetrists. J Appl Clin Med Phys. 23:e135682022. View Article : Google Scholar : PubMed/NCBI | |
Han-Oh S, Hill C, Kang-Hsin Wang K, Ding K, Wright JL, Alcorn S, Meyer J, Herman J and Narang A: Geometric reproducibility of fiducial markers and efficacy of a patient-specific margin design using deep inspiration breath hold for stereotactic body radiation therapy for pancreatic cancer. Adv Radiat Oncol. 6:1006552021. View Article : Google Scholar : PubMed/NCBI | |
Marks LB, Bentzen SM, Deasy JO, Kong FM, Bradley JD, Vogelius IS, El Naqa I, Hubbs JL, Lebesque JV, Timmerman RD, et al: Radiation dose-volume effects in the lung. Int J Radiat Oncol Biol Phys. 76 (3 Suppl):S70–S76. 2010. View Article : Google Scholar : PubMed/NCBI | |
Higgins K, Hu C, Ross H, Jabbour S, Kozono D, Owonikoko T, Dib E, Brownstein J, Kuzma C and Kotecha R: Concurrent chemoradiation±atezolizumab (atezo) in limited-stage small cell lung cancer (LS-SCLC): results of NRG Oncology/Alliance LU005. Int J Radiat Oncol Biol Phys. 120:S22024. View Article : Google Scholar | |
Young KH, Baird JR, Savage T, Cottam B, Friedman D, Bambina S, Messenheimer DJ, Fox B, Newell P, Bahjat KS, et al: Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One. 11:e01571642016. View Article : Google Scholar : PubMed/NCBI | |
Gunderson AJ and Young KH: Exploring optimal sequencing of radiation and immunotherapy combinations. Adv Radiat Oncol. 3:494–505. 2018. View Article : Google Scholar : PubMed/NCBI | |
Aliru ML, Schoenhals JE, Venkatesulu BP, Anderson CC, Barsoumian HB, Younes AI, K Mahadevan LS, Soeung M, Aziz KE, Welsh JW and Krishnan S: Radiation therapy and immunotherapy: What is the optimal timing or sequencing? Immunotherapy. 10:299–316. 2018. View Article : Google Scholar : PubMed/NCBI | |
Theelen WSME, Peulen HMU, Lalezari F, van der Noort V, de Vries JF, Aerts JGJV, Dumoulin DW, Bahce I, Niemeijer AN, de Langen AJ, et al: Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: Results of the PEMBRO-RT phase 2 Randomized clinical trial. JAMA Oncol. 5:1276–1282. 2019. View Article : Google Scholar : PubMed/NCBI | |
Theelen WSME, Chen D, Verma V, Hobbs BP, Peulen HMU, Aerts JGJV, Bahce I, Niemeijer ALN, Chang JY, de Groot PM, et al: Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: a pooled analysis of two randomised trials. Erratum in: Lancet Respir Med. 9:e292021. | |
Postow MA, Sidlow R and Hellmann MD: Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 378:158–168. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, Zhao S, Das S, Beckermann KE, Ha L, et al: Fatal toxic effects associated with immune checkpoint inhibitors: A systematic review and meta-analysis. JAMA Oncol. 4:1721–1728. 2018. View Article : Google Scholar : PubMed/NCBI | |
Haanen J, Obeid M, Spain L, Carbonnel F, Wang Y, Robert C, Lyon AR, Wick W, Kostine M, Peters S, et al: Management of toxicities from immunotherapy: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 33:1217–1238. 2022. View Article : Google Scholar : PubMed/NCBI | |
Thompson JA, Schneider BJ, Brahmer J, Achufusi A, Armand P, Berkenstock MK, Bhatia S, Budde LE, Chokshi S, Davies M, et al: Management of immunotherapy-related toxicities, version 1.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 20:387–405. 2022. View Article : Google Scholar : PubMed/NCBI | |
Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, et al: Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 381:1535–1546. 2019. View Article : Google Scholar : PubMed/NCBI | |
Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gumus M, Mazieres J, Hermes B, Cay Senler F, Csoszi T, Fulop A, et al: Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N Engl J Med. 379:2040–2051. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sangro B, Chan SL, Meyer T, Reig M, El-Khoueiry A and Galle PR: Diagnosis and management of toxicities of immune checkpoint inhibitors in hepatocellular carcinoma. J Hepatol. 72:320–341. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jácome AA, Castro ACG, Vasconcelos JPS, Silva MHCR, Lessa MAO, Moraes ED, Andrade AC, Lima FMT, Farias JPF, Gil RA, et al: Efficacy and safety associated with immune checkpoint inhibitors in unresectable hepatocellular carcinoma: A meta-analysis. JAMA Netw Open. 4:e21361282021. View Article : Google Scholar : PubMed/NCBI | |
Li G, Zhao Y, Li K, Yang S, Xiang C, Song J, Yang Y, Li G and Dong J: Effectiveness and safety of the PD-1 inhibitor lenvatinib plus radiotherapy in patients with HCC with main PVTT: Real-world data from a tertiary centre. J Hepatocell Carcinoma. 10:2037–2048. 2023. View Article : Google Scholar : PubMed/NCBI | |
Huang Y, Zhang Y, Zhang M, Zhao K, Feng L, Guan J, Dong R, Liu J, Tian D, Liu M, et al: Combined immunotherapy for hepatocellular carcinoma: How to maximize immune checkpoint blockade synergic anti-tumor effect. Crit Rev Oncol Hematol. 189:1040702023. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Xiong L, Li M, Jiang P, Wang J and Li C: Advances in radiotherapy and immunity in hepatocellular carcinoma. J Transl Med. 21:5262023. View Article : Google Scholar : PubMed/NCBI | |
Chen S, Xu B, Wu Z, Wang P, Yu W, Liu Z, Huang X, Wu Y, Li T and Guo W: Pembrolizumab plus lenvatinib with or without hepatic arterial infusion chemotherapy in selected populations of patients with treatment-naive unresectable hepatocellular carcinoma exhibiting PD-L1 staining: A multicenter retrospective study. BMC Cancer. 21:11262021. View Article : Google Scholar : PubMed/NCBI | |
Garin E, Tselikas L, Guiu B, Chalaye J, Edeline J, de Baere T, Assenat E, Tacher V, Robert C, Terroir-Cassou-Mounat M, et al: Personalised versus standard dosimetry approach of selective internal radiation therapy in patients with locally advanced hepatocellular carcinoma (DOSISPHERE-01): A randomised, multicentre, open-label phase 2 trial. Lancet Gastroenterol Hepatol. 6:17–29. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lewandowski RJ and Salem R: Radioembolisation with personalised dosimetry: Improving outcomes for patients with advanced hepatocellular carcinoma. Lancet Gastroenterol Hepatol. 6:2–3. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhu R, Wang X, Sun F, Zhu L and Guo W: Exploring the role of DNA methylation located in cuproptosis-related genes: Implications for prognosis and immune landscape in hepatocellular carcinoma. Front Biosci (Landmark Ed). 29:1232024. View Article : Google Scholar : PubMed/NCBI | |
Zhu J, Tang B, Lv X, Meng M, Weng Q, Zhang N, Li J, Fan K, Zheng L, Fang S, et al: Identifying apoptosis-related transcriptomic aberrations and revealing clinical relevance as diagnostic and prognostic biomarker in hepatocellular carcinoma. Front Oncol. 10:5191802021. View Article : Google Scholar : PubMed/NCBI | |
Nebhan CA and Johnson DB: Predictive biomarkers of response to immune checkpoint inhibitors in melanoma. Expert Rev Anticancer Ther. 20:137–145. 2020. View Article : Google Scholar : PubMed/NCBI | |
Xu H, Liang XL, Liu XG and Chen NP: The landscape of PD-L1 expression and somatic mutations in hepatocellular carcinoma. J Gastrointest Oncol. 12:1132–1140. 2021. View Article : Google Scholar : PubMed/NCBI | |
Parikh ND, Singal AG, Hutton DW and Tapper EB: Cost-effectiveness of hepatocellular carcinoma surveillance: An assessment of benefits and harms. Am J Gastroenterol. 115:1642–1649. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lani L, Stefanini B and Trevisani F: Surveillance for hepatocellular carcinoma in patients with successfully treated viral disease of the liver: A systematic review. Liver Cancer. 13:376–388. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zafar SY, Alexander SC, Weinfurt KP, Schulman KA and Abernethy AP: Decision making and quality of life in the treatment of cancer: A review. Support Care Cancer. 17:117–127. 2009. View Article : Google Scholar : PubMed/NCBI | |
Hamann J, Kissling W and Mendel R: Does it matter whether physicians' recommendations are given early or late in the decision-making process? An experimental study among patients with schizophrenia. BMJ Open. 6:e0112822016. View Article : Google Scholar : PubMed/NCBI | |
Nahm WJ, Bhatt A and Wu J: Role of radiotherapy and its contribution to immunotherapy in hepatocellular carcinoma. Chin Clin Oncol. 12:412023. View Article : Google Scholar : PubMed/NCBI |