Back to Journals » Journal of Hepatocellular Carcinoma » Volume 12
Impact of Timing the Combination of Radiotherapy and PD-1 Inhibitors on Outcomes in Patients with Hepatocellular Carcinoma
Authors Zhong L , Peng W, Sun J, Luo Y, Sheng H, Wu Y, Zhou T, Zhou C, Cao C
Received 26 July 2024
Accepted for publication 15 January 2025
Published 22 January 2025 Volume 2025:12 Pages 123—134
DOI https://doi.org/10.2147/JHC.S480691
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 3
Editor who approved publication: Dr Ahmed Kaseb
Liting Zhong,1,2,* Weiwei Peng,1,2,* Jingyuan Sun,3 Yongyi Luo,3 Hailong Sheng,4 Yi Wu,5 Tonggang Zhou,5 Chaoming Zhou,1,2 Chuanhui Cao1,3
1Department of Oncology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, People’s Republic of China; 2Department of Oncology, Ganzhou People’s Hospital, Ganzhou, Jiangxi, People’s Republic of China; 3Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China; 4Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China; 5Department of Interventional Radiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Chuanhui Cao, Department of Oncology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, No. 16 Meiguan Avenue, Ganzhou, Jiangxi, People’s Republic of China, Email [email protected]
Purpose: The optimal timing for combining radiotherapy with immunotherapy in patients with hepatocellular carcinoma (HCC) remains uncertain and affects treatment efficacy and patient outcomes. This study aimed to evaluate and compare the efficacy and treatment-related adverse events (TRAEs) of synchronously administered radiotherapy and programmed cell death protein (PD)-1 inhibitors and sequential administration in patients with HCC.
Patients and Methods: We retrospectively enrolled 67 patients with HCC who were undergoing liver radiotherapy and PD-1 inhibitor therapy at two medical centers between July 2017 and April 2023. Additionally, we created an experimental tumor model using 6-week-old female mice to validate our findings.
Results: In the concurrent group, the median overall survival was indefinite; however, it was 13 months in the sequential group (95% confidence interval [CI] 6.7– 19.3 months, P=0.010). The median progression-free survival was significantly longer in the concurrent group (12 months, 95% CI 9.5– 14.5 months) than in the sequential group (7 months, 95% CI 1.3– 12.7 months; P=0.043). Grade 3/4 TRAEs occurred in 30.4% (concurrent) and 28.6% (sequential) of patients without any treatment-related deaths. In the mouse model, synchronous treatment significantly inhibited tumor growth compared to sequential treatment (293.4± 45.18 mm3 versus 602.7± 41.68 mm3; P=0.001). Flow cytometry revealed an increased Tregs/CD3+ T cell ratio and a decreased CD8+/Treg cell ratio post-radiotherapy, suggesting an immunosuppressive tumor microenvironment.
Conclusion: Synchronous treatment demonstrated superior efficacy in treating HCC compared to sequential treatment, with manageable adverse events. The rapid increase in Tregs after radiotherapy may contribute to the reduced efficacy of sequential radiotherapy plus PD-1 inhibitors.
Keywords: immunotherapy, treatment efficacy, adverse events, tumor microenvironment
Introduction
Hepatocellular carcinoma (HCC) is a prevalent and deadly malignant tumor that is frequently diagnosed at advanced stages, preventing radical treatment.1,2 Advanced HCC is mainly treated with local therapy combined with systemic treatment. Local therapies include radiotherapy, ablation, hepatic arterial infusion chemotherapy, and transarterial chemoembolization (TACE). Systemic treatments include immune checkpoint inhibitors (ICIs) and targeted drugs. Nevertheless, the combination of local therapy and systemic treatment that best improves the prognosis of patients with HCC remains unclear.
Radiation-induced damage to cancer cells exposes tumor-specific antigens to immune surveillance and facilitates the activation of cytotoxic T cells. Additionally, radiotherapy can activate the tumor immune microenvironment by stimulating the cyclic GMP-AMP synthase/stimulator of interferon genes signaling pathway.3,4 Hence, radiotherapy is considered an optimal complement to immunotherapy. Combining stereotactic body radiation therapy (SBRT) with programmed cell death-1 (PD-1) inhibitors has yielded favorable clinical outcomes in TACE-refractory patients with intermediate-stage HCC.5 Five patients with large unresectable HCC treated with SBRT followed by nivolumab showed a 100% objective response rate (ORR) and tolerable toxicity.6 Therefore, radiotherapy combined with PD-1 inhibitors may serve as a synergistic therapeutic strategy.
One of the key considerations when combining radiotherapy with immunotherapy is the timing of the combination. Although prior studies have examined this aspect, the optimal time window for the combination remains unclear. The PACIFIC trial suggested that patients with unresectable stage III non-small cell lung cancer (NSCLC) receiving devarumab after standard chemoradiotherapy had a significantly improved prognosis.7 Exploratory analyses revealed that patients randomized ≥14 days post-radiotherapy had significantly extended progression-free survival (PFS) and overall survival (OS) compared to those who were randomized ≤14 days post-radiotherapy. Safety profiles were similar across the subgroups.8 However, in patients with stage IV NSCLC, no difference in efficacy was observed between the concurrent and sequential dual ICI therapies and SBRT. Concurrent dual ICIs and SBRT were not more toxic than sequential therapy.9 Multiple studies have shown that radiotherapy combined with ICIs is safe and feasible for melanoma brain metastases (MBM), resulting in a better prognosis than non-synchronous therapy.10–12 However, in contrast to previous research, the ELEKTRA study found that sequencing radiotherapy followed by ICI treatment led to better immunological responses and clinical outcomes in patients with MBM.13
In summary, the results of studies on the timing of radiotherapy combined with ICIs are inconsistent, with some studies showing contradictory results. The optimal timing for combining radiotherapy with PD-1 inhibitors in HCC remains unclear. Therefore, it is crucial to explore the optimal timing of combination therapy in patients with HCC.
This study aimed to compare the efficacy and adverse effects of concurrent and sequential administration of radiotherapy and PD-1 inhibitors in patients with HCC.
Materials and Methods
Patients
Retrospective chart reviews were conducted on patients with HCC who underwent radiotherapy and PD-1 inhibitor treatment at two medical centers (Ganzhou People’s Hospital and Nanfang Hospital, Southern Medical University) between July 2017 and April 2023. The following were the criteria for inclusion: (1) definitive diagnosis of HCC, (2) radiotherapy administered for primary liver cancer or portal vein (hepatic vein) cancer thrombus, (3) receipt of at least two cycles of PD-1 inhibitors, and (4) based on the response evaluation criteria in solid tumors (RECIST) version 1.1, at least one measurable lesion must have been present. There were several exclusion criteria, including: (1) pathological findings of cholangiocarcinoma or mixed-type HCC from biopsy or surgery, (2) previous history of liver transplantation, (3) intracranial metastases, (4) non-completion of radiotherapy, (5) presence of other malignancies, and (6) incomplete patient information.
Patients were categorized into the following two groups based on the timing of radiotherapy plus PD-1 inhibitors: radiotherapy concurrent with PD-1 inhibitors (concurrent group) and sequential radiotherapy plus PD-1 inhibitors (sequential group). The concurrent group comprised patients who received anti-PD-1 therapy within 7 days of the first administration of radiotherapy or during radiotherapy.13–16 In the sequential group, PD-1 inhibitors were administered 1–12 weeks after radiotherapy. The Ethics Committee of Ganzhou People’s Hospital (approval number: 001, 2020) and Nanfang Hospital, Southern Medical University (approval number: NFEC-2020-226) approved this study and waived the need for informed consent due to its retrospective nature. All study protocols complied with the guidelines of the Declaration of Helsinki. All patient data was kept confidential.
Data Collection
Age, sex, alpha-fetoprotein level, Eastern Cooperative Oncology Group score, radiotherapy, PD-1 inhibitor, prior therapy, targeted agents, and hepatitis B virus infection status were extracted from the patients’ medical records. Baseline patient characteristics were documented within 14 days of the first immunotherapy. PD-1 inhibitors include camrelizumab, sintilimab, tislelizumab, pembrolizumab, nivolumab, and toripalimab. Complete response (CR) or partial response (PR), as well as stable disease (SD) or progressive disease (PD), were assessed using contrast-enhanced magnetic resonance imaging or computed tomography according to RECIST v.1.1 or mRECIST. ORR was classified as CR or PR. The disease control rate (DCR) was calculated by adding CR, PR, and SD. OS was measured from the first administration of PD-1 inhibitors to the date of the last follow-up or death. PFS was assessed from the first treatment to the time of PD, last contact, or death. Treatment-related adverse events (TRAEs) were reviewed based on CTCAE v4.03.
Cell Culture
Dulbecco’s Modified Eagle’s Medium (Corning, USA) with 10% fetal bovine serum was used to culture Hepa 1–6 cells obtained from the Shanghai Institute of Biochemistry and Cell Biology. The cell culture was maintained in a constant-temperature incubator set at 37°C with a 5% CO2 atmosphere.
Construction of Subcutaneous Tumor Model
Six-week-old female C57BL/6 mice were procured from the Southern Medical University Animal Resource Center (Guangzhou, China) and randomly allocated to groups. Hepa 1–6 cells were implanted subcutaneously into the C57BL/6 mice’s right hind flank. The formula for calculating tumor volume was length × width2/2. Tumor volumes were measured and recorded every 5 days. Animal ethics approval was granted by the Experimental Animal Ethics Committee of Southern Medical University (SMUL2020118). All animal experiments followed the guidelines of the Institutional Animal Care and Use Committee.
Mice Treatment and Group
Treatment was administered when the tumor size reached approximately 400–500 mm3. In the first, third, and fifth days after the start of the treatment, three fractions of 6 Gy were administered. The small animal radiation research platform (SARRP, Xstrahl) was used for imaging and radiation. The mice in the concurrent group were intraperitoneally administered anti-mouse PD-1 (200 µg, clone RMP1-14, Bio X Cell) as soon as the treatment began on days 1, 4, and 7. Conversely, in the sequential group, 200 µg anti-mouse PD-1 was administered intraperitoneally on days 14, 17, and 20 post-treatment initiation. For the sequential group receiving anti-CD25, mice received intraperitoneal injections of anti-mouse CD25 (500 µg, clone PC-61.5.3, Bio X Cell) simultaneously with anti-mouse PD-1 on days 14, 17, and 20. The mice were monitored until they reached the standard endpoint or died.
Flow Cytometry
After obtaining single cells of the tumor, single-cell suspensions were stained and analyzed following the methods outlined in our previously published articles.17 All antibodies used were as follows: Brilliant Violet 421TM-FOXP3 (MF-14, 126419, Biolegend, California, USA), PerCP/Cyanine5.5 CD45 (30-F11, 103132, Biolegend, California, USA), Brilliant Violet510TM-CD8a (53–6.7, 100752, Biolegend, California, USA), FITC CD3 (17A2, 100204, Biolegend, California, USA), PerCP/Cyanine5.5 Brilliant Violet 421TM NK-1.1 (PK136, 108741, Biolegend, California, USA), APC F4/80 (BM8, 123116, Biolegend, California, USA), PE/Cy7 CD206 (C068C2, 141719, Biolegend, California, USA), PE CD86 (GL-1, 105008, Biolegend, California, USA), FITC CD11b (M1/70, 101206, Biolegend, California, USA), and PPE–CD25 (PC61, 102008, Biolegend, California, USA).
Statistical Analysis
Data were analyzed using the IBM Statistical Package for Social Sciences (SPSS) version 26.0 (IBM Corp., Armonk, New York, USA). The Mann–Whitney U-test was used to compare continuous covariates. Categorical variables were compared using the chi-square test or Fisher’s exact test. The Cochran–Mantel–Haenszel test was used to compare ORR and DCR between treatment groups using a Clopper–Pearson method with 95% confidence intervals (CIs). An analysis of survival was conducted using the Log rank test and Kaplan–Meier method to estimate OS and PFS. Using a mixed-effects model and Tukey’s multiple comparison test, we analyzed mouse subcutaneous tumor volume. We used analysis of variance combined with Tukey’s multiple comparison test to analyze the comparisons between three or more groups. A two-tailed P-value of < 0.05 was considered statistically significant. Plots were made with GraphPad Prism V.8.0.2 (San Diego, CA, USA).
Results
Patient Characteristics
Sixty-seven patients with HCC undergoing radiotherapy and taking PD-1 inhibitors at two medical centers were enrolled. Figure 1 depicts the study’s flow diagram. Patients were categorized into two groups based on the timing of combination therapy: 46 and 21 patients in the concurrent and sequential groups, respectively. Table 1 illustrates the similarities in the patient characteristics between the two groups.
![]() |
Table 1 Patient Characteristics and Treatments |
![]() |
Figure 1 Flow diagram for the present study. |
Clinical Outcome of Concurrent and Sequential Groups
The median follow-up duration was 15 months (range: 4–40 months) and 18 months (range: 1–36 months) in the concurrent and sequential radiotherapy groups, respectively, by the cutoff date of April 2023. Using Kaplan–Meier curves, the OS and PFS were calculated for each group (Figure 2). The median OS was not achieved in the concurrent group. However, it was 13 months (95% CI 6.7–19.3 months) in the sequential group (P=0.010). The survival rates at 6 and 12 months were 94.6% and 84.8%, respectively, in the concurrent group and 78.5% and 49.0%, respectively, in the sequential group. Additionally, the median PFS (12 months, 95% CI 9.5–14.5 months) in the concurrent group was significantly better than that in the sequential group (7 months, 95% CI 1.3–12.7 months; P=0.043). The PFS at 6 months was 87.8% and 55.8% in the concurrent and sequential groups, respectively.
![]() |
Figure 2 Kaplan–Meier curves of (A) overall and (B) progression-free survival between the concurrent radiotherapy group and sequential radiotherapy group. |
Treatment efficacy was evaluated in 66 of the 67 patients. The treatment response was not assessed during weeks 6–8 following the first administration of immunotherapy in one patient. As shown in Table S1, the ORRs were 58.7% (95% CI 43.2–73.0%) and 30.0% (95% CI 11.9–54.3%) in the concurrent group and sequential groups, respectively, based on independent assessment using RECIST 1.1 (P=0.036). The DCR was 97.8% (95% CI 88.5–99.9%) in the concurrent group compared with that in the sequential group (75.0%, 95% CI 50.9–91.3%, P=0.017). The best overall response waterfall plots (RECIST 1.1) are shown in Figure S1. According to the HCC-specific mRECIST, 35 (76.1%) patients in the concurrent group, as compared to 9 (45.0%) in the sequential group, had a CR or PR (Table S2). The best overall response waterfall plots (mRECIST) are shown in Figure S2.
Safety of Concurrent Group and Sequential Group
Overall, 67 patients had at least one TRAE (Table 2). The most common TRAEs of any grade were decreased platelet count (23.9%), increased aspartate aminotransferase (AST) levels (23.9%), increased alanine transaminase (ALT) levels (22.4%), decreased appetite (17.9%), decreased white blood cell (WBC) count (14.9%), and rash (14.9%). Grade 3/4 TRAEs occurred in 14 (30.4%) of the 46 patients who received synchronous radiotherapy plus PD-1 inhibitors, primarily including liver dysfunction (elevated AST, ALT, and bilirubin levels), decreased platelet count, and decreased WBC count. Grade 3/4 TRAEs occurred in six (28.6%) of the 21 patients in the other group of sequential radiotherapy plus PD-1 inhibitors, including increased AST, ALT, decreased WBC, and gastrointestinal hemorrhage. Notably, no treatment-related deaths were reported. Compared with the sequential group, synchronous radiotherapy plus PD-1 inhibitors did not significantly increase the incidence of TRAE.
![]() |
Table 2 Treatment Related Adverse Events in Concurrent Group and Sequential Group |
Mice Model of Concurrent Group and Sequential Group
To further validate the efficacy of the concurrent and sequential treatments, we constructed a subcutaneous tumor model. Figure 3 shows the subcutaneous tumor growth curves for both groups. Radiotherapy combined with concurrent PD-1 inhibitors significantly inhibited tumor growth compared to the sequential approach (293.4±45.18 mm3 vs 602.7±41.68 mm3; P=0.001).
Changes of Tumor-Infiltrating Immune Cells After Radiotherapy
To understand why concurrent radiotherapy with PD-1 inhibitors was more efficacious than sequential radiotherapy plus PD-1 inhibitors, tumor tissues were harvested on days 7, 14, 21, and 28 after the initiation of radiotherapy. Flow cytometry analysis of tumor tissues was conducted to assess changes in tumor-infiltrating immune cells after radiotherapy. The tumor-infiltrating lymphocyte (TIL) Tregs/CD3+ T cell ratio increased on day 7 (6.252%±1.490%) and peaked on day 14 (12.04%±7.028%, P=0.0183), as shown in Figure 4. Conversely, the Tregs/CD3+ T cell ratio gradually decreased on days 21 and 28. On day 7, the number of TIL CD8+ T cells was 99.90±84.23, indicating a decreasing trend with time, reaching the lowest on day 28 (45.20±13.07, P=0.189). Although the difference in TIL CD8+ T cells did not reach statistical significance, the TIL CD8+/Tregs ratio on day 14 significantly declined compared to that on day 7 (9.005±6.422 vs 23.10±7.625; P=0.0134). From day 21 to 28, the TIL CD8+/Tregs ratio decreased further (P=0.5629). Therefore, the TIL Tregs/CD3+ T cell ratio significantly increased, whereas the TIL CD8+ T cell and TIL CD8+/Tregs ratio decreased on day 14. This may explain why tumor control was worse in the sequential group (PD-1 inhibitor administered on day 14 after radiotherapy initiation) than in the concurrent therapy. Additionally, we analyzed the changes in natural killer cells, M1-type macrophages, M2-type macrophages, and myeloid-derived suppressor cells after radiotherapy (Figure S3).
Anti-CD25 reversed tumor growth in the group receiving radiotherapy sequentially plus PD-1 inhibitors (666.9±22.01 mm3 vs 701.3±51.92 mm3; P=0.0064) and achieved tumor control effects similar to those of radiotherapy administered synchronously with PD-1 inhibitors (666.9±22.01 mm3 vs 531.6±46.91 mm3; P=0.0681) (Figure S4).
Discussion
This is the first study to investigate the optimal timing of the combination of radiotherapy and ICIs in HCC. Our findings revealed that radiotherapy administered concurrently with PD-1 inhibitors had better efficacy than sequential radiotherapy plus PD-1 inhibitors alone. Additionally, the synchronous group exhibited tolerable toxic side effects.
Kaplan–Meier analysis and Log rank test indicated better OS and PFS in the concurrent group. These results are consistent with the findings from a study in 2021, which observed that concurrent radiotherapy of brain metastasis combined with ICIs significantly improved OS and PFS compared with the non-concurrent group and that simultaneous combination was an independent prognostic factor.18 Furthermore, a meta-analysis published in Radiotherapy and Oncology demonstrated prolonged OS with concurrent radiotherapy and immunotherapy in patients with brain metastasis.19 According to the RECIST 1.1 criteria, the ORR of the concurrent group (58.7%) surpassed that of the sequential group (30.0%, P=0.036). A Phase II clinical study reported an ORR of 52.4% with SBRT combined with carlizumab for HCC, which is consistent with our findings.20 In this study, no additional toxic effects or safety events were identified. Notably, in both the concurrent and sequential treatment groups, most patients received targeted therapy. Hence, TRAEs caused by targeted drugs were not excluded. Radiotherapy combined with atezolizumab and bevacizumab for HCC has shown that the most common TRAEs include neutropenia, fatigue, and hypertension.21 Additionally, we demonstrated that radiotherapy combined with a PD-1 inhibitor and anti-vascular targeted therapy is safe and feasible for HCC.16
In a mouse model of subcutaneous graft tumors, we demonstrated that synchronous radiotherapy plus PD-1 inhibitors significantly inhibited tumor growth compared to the sequential approach. This finding is consistent with our clinical results. Concurrent PD-L1 inhibitors and radiotherapy are more effective in controlling tumor growth than sequential treatments, as shown in an animal model of colorectal cancer.22 Additionally, another animal experiment in pancreatic cancer found that anti-PD-L1 synchronously combined with radiotherapy significantly improved the antitumor response; however, radiosensitization completely disappeared when anti-PD-L1 was delayed to 7 days after radiotherapy.23 Synchronous radiotherapy combined with anti-PD1/PD-L1 therapy may induce a synergistic antitumor host immune response and improve treatment response.
Previous research by our group showed that simultaneous radiotherapy combined with anti-PD-L1 treatment has a strong immunostimulatory effect, including increasing the number of tumor CD8+ T cells and enhancing the activity of CD8+ T cells, as well as reducing the infiltration of Tregs.17 This study conducted a comprehensive analysis of the alterations in tumor-infiltrating immune cells at various time points following radiation therapy to determine the optimal timing for intervention with ICIs. The ratio of Tregs to CD3+ T cells exhibited a rapid increase from days 7 to 14 post-radiation therapy, whereas the TIL CD8+ T cell and TIL CD8+/Tregs ratio decreased on day 14. Consequently, targeting Tregs may be a critical factor influencing the suboptimal efficacy of sequential PD-1 inhibitor therapy after radiation treatment. Furthermore, we discovered that administration of CD25 antibodies could reverse the diminished effectiveness of sequential anti-PD-1 therapy following radiation. In the immune evasion mechanism of HCC, Tregs suppress the ability of immune cells to kill HCC cells through various pathways, thereby promoting the immune evasion of HCC cells. Tregs are closely related to poor prognosis in patients with HCC. They not only inhibit excessive immune responses to prevent normal function from being impaired but also promote the formation of immune tolerance to protect HCC cells from systemic attacks. Additionally, studies have shown that excessive Tregs in patients with HCC are associated with poor treatment outcomes, as Tregs limit the effect of immunotherapy and weaken the efficacy of ICIs.24 Tregs play a pivotal role in the regrowth of head and neck tumors following radiotherapy combined with ICIs. The depletion of Tregs can enhance antitumor immune responses and induce antitumor memory immunity, leading to tumor shrinkage.25 Clinical studies have associated Tregs with tumor progression, recurrence, and treatment resistance in various malignancies, including colorectal cancer, soft tissue sarcomas, endometrial cancer, and head and neck squamous carcinoma.26–29 Tumors with high oxidative stress promote the enrichment of Tregs, fostering an immunosuppressive microenvironment and resistance to ICIs.30 Radiotherapy not only has positive regulatory effects on the tumor immune microenvironment but also increases the infiltration of Tregs into the tumor microenvironment to produce immunosuppressive effects.31–33 In 2011, a study that evaluated the effects of radiation on Treg cells was published in International Journal of Radiation Oncology, Biology, Physics. The results suggested that Treg cells have a stronger resistance to radiation than other lymphocytes, leading to a prior increase in their numbers and the induction of an immunosuppressive microenvironment.34 This is similar to our findings. Fukushima reported that Treg-directed therapy synergistically improved the efficacy of PD-1 inhibitors in syngeneic murine tumor models.35 Therefore, targeting Tregs may improve the therapeutic potential of radiotherapy in conjunction with immunotherapy.
In our study, we had some limitations. First, the clinical data were constrained by the small sample size and retrospective design. Second, the substantial heterogeneity of the patient population and treatment regimen may affect the interpretation of our findings. Moreover, our in vivo efficacy experiments solely utilized an HCC subcutaneous xenograft model, limiting the generalizability of our findings. Various animal models, including orthotopic models, can provide more robust therapeutic insights. Therefore, further investigation of the underlying molecular mechanisms is warranted to develop a thorough understanding of the therapeutic effects of this treatment regimen.
Conclusion
Synchronous radiotherapy plus PD-1 inhibitors may achieve better tumor control than sequential radiotherapy plus PD-1 inhibitors with manageable TRAEs. Additionally, Treg cell enrichment may contribute to the varying efficacies of the two approaches. Large randomized controlled clinical studies are required to validate the safety and efficacy of synchronous radiotherapy and PD-1 inhibitors.
Abbreviations
PD-1, Programmed Cell Death Protein 1; HCC, Hepatocellular Carcinoma; TRAEs, Treatment-Related Adverse Events; CI, Confidence Interval; RECIST, Response Evaluation Criteria in Solid Tumors; TACE, Trans-Arterial Chemo-Embolization; ICIs, Immune Checkpoint Inhibitors; SBRT, Stereotactic Body Radiation Therapy; ORR, Objective Response Rate; DCR, Disease Control Rate; OS, Overall Survival; PFS, Progression-Free Survival; CTCAE, Common Terminology Criteria for Adverse Events; C57BL/6, A common strain of laboratory mouse; SARRP, Small Animal Radiation Research Platform; TIL, Tumor-Infiltrating Lymphocyte; NSCLC, Non-Small Cell Lung Cancer; mRECIST, Modified Response Evaluation Criteria In Solid Tumors; PC-61.5.3, A monoclonal antibody; NCI, National Cancer Institute; ALT, Alanine Transaminase; WBC, White Blood Cell; Gy, Gray (unit of radiation dose); SPSS, Statistical Package for the Social Sciences; CR, Complete Response; PR, Partial Response; SD, Stable Disease; PD, Progressive Disease; OR, Odds Ratio; C57BL/6, A common strain of laboratory mouse; MBM, Melanoma Brain Metastases; AST, Aspartate Aminotransferase.
Acknowledgments
We thank Dehua Wu for his excellent assistance with revising the manuscript. We thank the associate editor and the reviewers for their useful feedback that improved this paper.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising, or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This study was funded by the National Natural Science Foundation of China (Grant Nos. 82360580, 82073343), Jiangxi Provincial Natural Science Foundation (Grant Nos.20242BAB20418), Ganzhou Municipal Science and Technology Project (Grant Nos. 2022-YB1437, 20222ZDX8175) and Doctoral research start-up fund project of Ganzhou People’sHospital (Grant Nos. BSQD2024009).
Disclosure
The authors declare no competing interests that are relevant to the study.
References
1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi:10.3322/caac.21660
2. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet. 2018;391(10127):1301–1314. doi:10.1016/S0140-6736(18)30010-2
3. Du SS, Chen GW, Yang P, 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(5):1243–1255. doi:10.1016/j.ijrobp.2021.12.162
4. Zhu M, Yang M, Zhang J, et al. Immunogenic cell death induction by ionizing radiation. Front Immunol. 2021;12:705361. doi:10.3389/fimmu.2021.705361
5. Xiang YJ, Wang K, Zheng YT, et al. Effects of stereotactic body radiation therapy plus PD-1 inhibitors for patients with transarterial chemoembolization refractory. Front Oncol. 2022;12:839605. doi:10.3389/fonc.2022.839605
6. Chiang CL, Chan ACY, Chiu KWH, Kong FS. Combined stereotactic body radiotherapy and checkpoint inhibition in unresectable hepatocellular carcinoma: a potential synergistic treatment strategy. Front Oncol. 2019;9:1157. doi:10.3389/fonc.2019.01157
7. Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in Stage III NSCLC. N Engl J Med. 2018;379(24):2342–2350. doi:10.1056/NEJMoa1809697
8. Faivre-Finn C, Spigel DR, Senan S, et al. Impact of prior chemoradiotherapy-related variables on outcomes with durvalumab in unresectable Stage III NSCLC (PACIFIC). Lung Cancer. 2021;151:30–38. doi:10.1016/j.lungcan.2020.11.024
9. Bestvina CM, Pointer KB, Karrison T, et al. A Phase 1 trial of concurrent or sequential ipilimumab, nivolumab, and stereotactic body radiotherapy in patients with Stage IV NSCLC study. J Thorac Oncol. 2022;17(1):130–140. doi:10.1016/j.jtho.2021.08.019
10. Pomeranz Krummel DA, Nasti TH, Izar B, et al. Impact of sequencing radiation therapy and immune checkpoint inhibitors in the treatment of melanoma brain metastases. Int J Radiat Oncol Biol Phys. 2020;108(1):157–163. doi:10.1016/j.ijrobp.2020.01.043
11. Chen L, Douglass J, Kleinberg L, et al. Concurrent immune checkpoint inhibitors and stereotactic radiosurgery for brain metastases in non-small cell lung cancer, melanoma, and renal cell carcinoma. Int J Radiat Oncol Biol Phys. 2018;100(4):916–925. doi:10.1016/j.ijrobp.2017.11.041
12. Anderson ES, Postow MA, Wolchok JD, et al. Melanoma brain metastases treated with stereotactic radiosurgery and concurrent pembrolizumab display marked regression; efficacy and safety of combined treatment. J Immunother Cancer. 2017;5(1):76. doi:10.1186/s40425-017-0282-x
13. Hassel JC, Schank TE, Smetak H, et al. Evaluation of radio-immunotherapy sequence on immunological responses and clinical outcomes in patients with melanoma brain metastases (ELEKTRA). Oncoimmunology. 2022;11(1):2066609. doi:10.1080/2162402X.2022.2066609
14. Gerber DE, Urbanic JJ, Langer C, et al. Treatment design and rationale for a randomized trial of cisplatin and etoposide plus thoracic radiotherapy followed by nivolumab or placebo for locally advanced non-small-cell lung cancer (RTOG 3505). Clin Lung Cancer. 2017;18(3):333–339. doi:10.1016/j.cllc.2016.10.009
15. Luke JJ, Lemons JM, Karrison TG, et al. Safety and clinical activity of pembrolizumab and multisite stereotactic body radiotherapy in patients with advanced solid tumors. J Clin Oncol. 2018;36(16):1611–1618. doi:10.1200/JCO.2017.76.2229
16. Zhong L, Wu D, Peng W, et al. Safety of PD-1/PD-L1 inhibitors combined with palliative radiotherapy and anti-angiogenic therapy in advanced hepatocellular carcinoma. Front Oncol. 2021;11:686621. doi:10.3389/fonc.2021.686621
17. Sheng H, Huang Y, Xiao Y, et al. ATR inhibitor AZD6738 enhances the antitumor activity of radiotherapy and immune checkpoint inhibitors by potentiating the tumor immune microenvironment in hepatocellular carcinoma. J Immunother Cancer. 2020;8(1):e000340. doi:10.1136/jitc-2019-000340
18. Trommer M, Adams A, Celik E, et al. Oncologic outcome and immune responses of radiotherapy with anti-PD-1 treatment for brain metastases regarding timing and benefiting subgroups. Cancers. 2022;14(5):1240. doi:10.3390/cancers14051240
19. Lehrer EJ, Peterson J, Brown PD, et al. Treatment of brain metastases with stereotactic radiosurgery and immune checkpoint inhibitors: an international meta-analysis of individual patient data. Radiother Oncol. 2019;130:104–112. doi:10.1016/j.radonc.2018.08.025
20. Li JX, Su TS, Gong WF, et al. Combining stereotactic body radiotherapy with camrelizumab for unresectable hepatocellular carcinoma: a single-arm trial. Hepatol Int. 2022;16(5):1179–1187. doi:10.1007/s12072-022-10396-7
21. Wang K, Xiang YJ, Yu HM, et al. Intensity-modulated radiotherapy combined with systemic atezolizumab and bevacizumab in treatment of hepatocellular carcinoma with extrahepatic portal vein tumor thrombus: a preliminary multicenter single-arm prospective study. Front Immunol. 2023;14:1107542. doi:10.3389/fimmu.2023.1107542
22. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74(19):5458–5468. doi:10.1158/0008-5472.CAN-14-1258
23. Azad A, Yin Lim S, D’Costa Z, et al. PD-L1 blockade enhances response of pancreatic ductal adenocarcinoma to radiotherapy. EMBO Mol Med. 2017;9(2):167–180. doi:10.15252/emmm.201606674
24. Du G, Dou C, Sun P, Wang S, Liu J, Ma L. Regulatory T cells and immune escape in HCC: understanding the tumor microenvironment and advancing CAR-T cell therapy. Front Immunol. 2024;15:1431211. doi:10.3389/fimmu.2024.1431211
25. Oweida A, Hararah MK, Phan A, et al. Resistance to radiotherapy and PD-L1 blockade is mediated by TIM-3 upregulation and regulatory T-cell infiltration. Clin Cancer Res. 2018;24(21):5368–5380. doi:10.1158/1078-0432.CCR-18-1038
26. Bergsland CH, Jeanmougin M, Moosavi SH, et al. Spatial analysis and CD25-expression identify regulatory T cells as predictors of a poor prognosis in colorectal cancer. Mod Pathol. 2022;35(9):1236–1246. doi:10.1038/s41379-022-01086-8
27. Smolle MA, Herbsthofer L, Granegger B, et al. T-regulatory cells predict clinical outcome in soft tissue sarcoma patients: a clinico-pathological study. Br J Cancer. 2021;125(5):717–724. doi:10.1038/s41416-021-01456-0
28. Kolben T, Mannewitz M, Perleberg C, et al. Presence of regulatory T-cells in endometrial cancer predicts poorer overall survival and promotes progression of tumor cells. Cell Oncol Dordr. 2022;45(6):1171–1185. doi:10.1007/s13402-022-00708-2
29. López-Janeiro Á, Villalba-Esparza M, Brizzi ME, et al. The association between the tumor immune microenvironments and clinical outcome in low-grade, early-stage endometrial cancer patients. J Pathol. 2022;258(4):426–436. doi:10.1002/path.6012
30. Maj T, Wang W, Crespo J, et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol. 2017;18(12):1332–1341. doi:10.1038/ni.3868
31. Ma J, Xin Y, Wang Q, Ding L. Roles of cGAS-STING pathway in radiotherapy combined with immunotherapy for hepatocellular carcinoma. mol Cancer Ther. 2024;23(4):447–453. doi:10.1158/1535-7163.MCT-23-0373
32. Zhao D, Mo Y, Neganova ME, et al. Dual effects of radiotherapy on tumor microenvironment and its contribution towards the development of resistance to immunotherapy in gastrointestinal and thoracic cancers. Front Cell Dev Biol. 2023;11:1266537. doi:10.3389/fcell.2023.1266537
33. Gregucci F, Spada S, Barcellos-Hoff MH, et al. Updates on radiotherapy-immunotherapy combinations: proceedings of 6th annual ImmunoRad conference. Oncoimmunology. 2023;12(1):2222560. doi:10.1080/2162402X.2023.2222560
34. Kachikwu EL, Iwamoto KS, Liao YP, et al. Radiation enhances regulatory T cell representation. Int J Radiat Oncol Biol Phys. 2011;81(4):1128–1135. doi:10.1016/j.ijrobp.2010.09.034
35. Fukushima H, Furusawa A, Takao S, et al. Near-infrared duocarmycin photorelease from a Treg-targeted antibody drug conjugate improves efficacy of PD-1 blockade in syngeneic murine tumor models. OncoImmunology. 2024;13(1):2370544. doi:10.1080/2162402X.2024.2370544
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.