ABSTRACT
Radiotherapy (RT) is one of the primary treatment modalities in managing cancer patients. Recently, combined RT and immunotherapy (IT) (i.e., radio-IT [RIT]) have been aggressively investigated in managing cancer patients. However, several issues in conducting RIT are challenging, such as incorporating advanced irradiation techniques, predictive/prognostic biomarkers, and other treatment modalities. Several clinical efforts and novel biomarkers have been introduced and developed to solve these challenges. For example, stereotactic radiosurgery/stereotactic radiotherapy, stereotactic body radiotherapy/stereotactic ablative body radiotherapy, and FLASH-RT have been applied for delivering precise irradiation to lung and liver tumors in conjunction with IT. Besides, several novel IT agents and incorporations of other therapies, such as targeted and thermal therapies, have been further investigated. The present study reviewed the emerging challenges of RIT in modern oncology. We also evaluated clinical practice, bench research, and multimodality treatments. In addition to several clinically applicable biomarkers, we emphasize the roles of advanced irradiation techniques and epigenetic modification as predictive/prognostic biomarkers and potential therapeutic targets. For example, 6(m) A-based epigenetic agents demonstrate the potential to enhance the treatment effects of RIT. However, further prospective randomized trials should be conducted to confirm their roles.
KEYWORDS: Epigenetic modification, Immunotherapy, Outcome prediction, Radiotherapy, Toxicity
INTRODUCTION
Radiotherapy in oncology
Radiotherapy (RT) is effective in managing cancer patients [1,2]. In radiobiology, RT attacks cells, mainly damaging the DNA [3]. Cancer cells generally demonstrate an impaired ability to repair their postirradiation DNA damage compared with normal cells [4]. As a result, irradiated cancer cells present more significant apoptosis and postmitotic death than normal cells. Post-irradiation damaged or dead cancer cells frequently released broken double-strand DNA (dsDNA) fragments into the peri-tumor microenvironment or peripheral blood circulation. Biologically, these released dsDNA fragments enrich cancer-specific mutations. If these mutations are significant enough in quality and amount, the anticancer host immune reaction may be apparent via the presentation of antigen-presenting cells (APC). The postirradiation abscopal effect partly supports this hypothesis in several clinical observations [5]. Hence, RT is recognized as a crucial treatment modality in synergizing the treatment effect of modern immunotherapy (IT) [6].
Immunotherapy in oncology
IT has been used to manage malignant melanoma patients for decades [7]. However, interferon-based IT is limited in clinical oncology mainly due to its constraints of treatment toxicities. Recently, after the introduction of PD-1/PD-L1 and CTLA-4 immune checkpoint blockages [8], IT has gained a considerable advance in managing cancer patients [9]. As mentioned above, combined RT and IT demonstrate a synergistic anticancer effect. Initially, this combination was mainly applied to cancer patients with recurrent and metastatic diseases. Recently, IT has extended its clinical application to several new indications. For example, IT is tested in the context of a neoadjuvant setting in patients with advanced [10] or resectable [11] head-and-neck cancers, intending to replace the role of systemic chemotherapy.
Combined radiotherapy and immunotherapy for managing cancer patients
Combined RT and IT (i.e., radio-IT [RIT]) is a hot trend in modern oncology [6]. As mentioned above, one promising reason for applying RIT is to activate the potential abscopal effect of RT to enhance the treatment effects of IT [12]. Clinically, this combination is useful in managing several types of cancer patients, such as head and neck [13], lung [12], liver [14], glioblastoma [15,16], melanoma [17,18], lymphoma [19], breast [20,21], uterine cervix [22,23], ovarian [24], urinary bladder [25,26], prostate [27], renal cell carcinoma [28], esophagus [29], and pancreas [30].
Many metastatic cancer patients are also benefits from RIT, including brain [31] and liver metastases [32]. However, for lung cancer patients, PD-1/PD-L1-based IT with or without RT may increase the risk of treatment-related pneumonitis [33]. Besides, current RIT still has several challenges that are required to be further investigated, such as the introduction of advanced irradiation techniques, investigation of predictive/prognostic biomarkers, and incorporation of other treatment modalities [34].
Epigenetics in oncology
Epigenetic modifications regulate gene function without altering its DNA sequence [35]. Several types of epigenetic modifications have been identified, such as DNA methylation [36], histone modification [37], and noncoding RNA regulations (e.g., micro-RNA [miRNA or miR] [38], long-noncoding RNA (lncRNA), and circular RNA [circRNA]) [39]. Biologically, epigenetic modification is a crucial investigating field in oncology [40], involving progression [41] and metastatic processes [42], carcinogenesis [43,44], angiogenesis [45], migration/invasion [46], and immune suppression and modulation [47,48]. Clinically, application in serving as diagnostic/predictive/prognostic biomarkers is a promising research topic in cancer epigenetics. For example, the lncRNA-miRNA network is recognized as potential cancer biomarkers [49,50]. More notably, epigenetic-based agents have been actively investigated as one potential anti-cancer treatment modality [51], particularly for managing treatment-resistant/recurrent patients [52].
One good point for investigating epigenetic-based biomarkers in oncology is that advanced epigenetic-exploring techniques are actively developing for pan-cancer noninvasive screening [53], liquid biopsy [54], and high-performance detection [55]. Hence, the present study aimed to review clinical challenges and available biomarkers of combined RT and IT in managing cancers. Emerging challenges of epigenetic application in potential biomarkers and therapeutic targets for RIT are also reviewed.
CLINICAL CHALLENGES AND EMERGING ISSUES FOR TREATMENT ADVANCE, OUTCOME PREDICTION, AND TOXICITIES PREVENTION OF RADIO-IMMUNOTHERAPY
Clinical challenges in treatment advance, outcome prediction, and toxicities prevention of radio-immunotherapy
Several factors affect RIT’s treatment response and patient prognosis in managing cancer patients, including patient, cancer, and treatment factors [Table 1]. For example, for rectal cancer patients with a bulky unresectable liver metastasis that showed enriched PD-L1 expression, stereotactic body radiotherapy (SBRT) may be a better irradiation technique than conventional RT in conjunction with IT. However, several issues are still challenging in clinical practice, especially when the incorporation of other treatment modalities is under consideration, such as chemotherapy, targeted therapy, and thermal ablation.
Table 1.
Factors | Description | Reference |
---|---|---|
Patient factor | ||
Performance status (e.g., ECOG) | ECOG performance score (i.e., 2 vs. 0–1) is an independent prognostic factor for OS and PFS | [56] |
Cancer factor | ||
Lung cancer | RIT is effective in managing SCLC and NSCLC patients, even in those with brain or liver metastases | [9,12,32,57,58,59,60,61,62,63,64,65,66,67,68] |
Esophagus cancer | RIT is synergized in managing esophagus cancer patients; further trials are required | [29] |
Breast cancer | RIT is effective in managing triple-negative breast cancer, even for metastatic patients | [20,21] |
Head-and-neck cancers | RIT is helpful and challenging in patients with primary, recurrent, and metastatic HNCs | [69,70,71,72,73] |
GYN cancers | RIT is effective in managing patients with cervical and ovarian cancers | [22,23,24] |
GU cancers | RIT is helpful in patients with prostate, urinary bladder, and renal cell carcinoma | [25,27,28] |
RT techniques | ||
SABR/SBRT | Lung SABR/SBRT shows better outcomes than conventional RT | [74,75,76,77] |
SRS/SRT | Combined SRS/SRT and IT is helpful in lung cancer with liver or brain metastases, which improves neurological outcomes | [31,32,62] |
Charged-particle irradiation | Charged-particle irradiation may enhance the RIT treatment effect with limited toxicities | [78] |
FLASH-RT | Combined FLASH-RT and IT in managing cancer patients are under investigation | [79] |
BNCT | Combined BNCT and IT may be a benefit in tumor control in vivo | [80] |
Combined therapy** | ||
Chemotherapy | Several chemotherapy agents enhance RIT effects | [81,82,83,84] |
Targeted therapy | Several targeted therapy enhances RIT effects, such as MEK inhibitors | [81,82,85] |
Thermal ablation | Combined thermal ablation may further enhance the treatment effects of IT | [86] |
m(6) A epigenetic modifier | Applying m (6) A epigenetic modifier, such as FTO* inhibitors, can reprogram RNA epitranscriptome and enhance the IT effect | [87,88] |
Novel IT agents*** | ||
Nanoparticle-based IT | Nanoparticle-based IT may enhance RIT treatment effects | [89,90,91] |
NKTR-214 IT | Combined NKTR-214 immunotherapy and RT to stimulate systemic CD8(+) T-cell responses may cure multi-focal cancers [92] | [92] |
Dendritic cell-based IT | The role of DC-based IT in conjunction with RT and chemotherapy is still not well defined [93]. Further randomized trials are required | [93] |
*FTO, an m(6) A demethylase, utilizes the FTO-mediated epigenetic regulation to evade immune surveillance, **Multimodality therapy is the keystone in managing cancer patients, particularly those with advanced diseases. Combined treatments may enhance tumor response with the cost of increasing toxicities, ***Several novel IT agents are potential for clinical use in conjunction with RT, such as tumor vaccine, cytokine-based therapy, and adoptive T-cell therapy [6]. ECOG: Eastern Cooperative Oncology Group, OS: Overall survival, PFS: Progression-free survival, RIT: Radio-immunotherapy, IT: Immunotherapy, SCLC: Small cell lung cancer, NSCLC: Non-small cell lung cancer, HNCs: Head-and-neck cancers, SABR: Stereotactic ablative body radiotherapy, SBRT: Stereotactic body radiotherapy, SRS: Stereotactic radiosurgery, SRT: Stereotactic (hypofractionated) radiotherapy, RT: Radiotherapy, BNCT: Boron neutron capture therapy, GU: Genitourinary cancer, GYN: Gynecology cancer
Clinical challenges in treatment advance of radio-immunotherapy, including advanced radiotherapy techniques and novel immunotherapy agent combinations
IT and RT showed a zigzag association in their synergic effects on treatment benefits. That is, RT demonstrates the role of immune sensitizer for IT [94], and IT also functions as a radiation sensitizer for local RT [95,96]. As a result, few studies suggested that IT followed by RT may be feasible in some specific clinical conditions. However, treatment sequencing about RIT is generally recommended as RT followed by IT. The biological reason is that RT enhances cancer-specific dsDNA exposure to APC and then synergizes the treatment effects of IT [97]. Clinical data mainly supported this type of treatment sequence.
Many studies have shown that patients who received RT combined with IT significantly benefit in overall survival when compared with those who received IT or RT alone [98]. Generally, a higher dose per fraction of RT may enhance cancer cell killing, improve clinical outcomes [99], and trigger the immune effect. For example, the combination of hypofractionated RT (>5 Gy per fraction) with IT has been reported to prolong patient survival [100]. Besides, a phase II clinical trial indicated that pembrolizumab given within 7 days after SBRT, which consisted of 3 fractions of 8 Gy, improves progression-free and overall survival in patients with metastatic NSCLC [101]. However, a prior report indicated that irradiation doses exceeding 12–18 Gy may trigger the exonuclease Trex 1, which diminishes radiation-induced immunogenicity by breaking down cytosolic DNA [102].
The sequence of RT and IT has also been demonstrated to influence the effectiveness of treatment [57]. In a phase III clinical trial (PACIFIC trial), stage III non-small cell lung cancer patients who received concurrent chemoradiotherapy were randomized into two groups: one receiving durvalumab and the other receiving placebo. Their results showed that durvalumab treatment led to substantially extended progression-free survival compared to the placebo group [103]. Recently, they reported cumulative 4-and 5-year clinical outcome data, indicating that adding IT after concurrent chemoradiotherapy increases both progression-free and overall survival [103,104,105,106].
In another study involving metastatic non-small cell lung cancer, a Phase II trial (PEMBRO-RT; NCT02492568) investigated the efficacy of immunotherapy (pembrolizumab) following RT. The initial dose of pembrolizumab was administered after the final dose of RT (24 Gy in 3 fractions). They observed positive outcomes in the PD-L1–negative subgroup, showing notable improvements in both progression-free and overall survival [101]. Conversely, another trial (NCT02444741) investigated the concurrent treatment of IT (pembrolizumab) and RT (either 50 Gy in 4 fractions or 45 Gy in 15 fractions). However, no significant differences were found in terms of objective response rates or progression-free survival [107]. While a pooled analysis of these two randomized trials showed adding IT to RT significantly increased responses and outcomes in patients with metastatic non-small cell lung cancer [57], clinical outcomes may be largely influenced by age, tumor stage, cancer type, and the specific IT drug used.
Besides, preclinical studies showed the optimal sequence may vary depending on the specific immunomodulatory agent used. For instance, RT may demonstrate the highest synergy with anti-PD-L1 when administered simultaneously [108]. In contrast, administering a transforming growth factor-beta (TGF-β) inhibitor before RT might enhance the survival rate in vivo [109]. In a preclinical study comparing the sequencing of anti-CTLA-4 and anti-OX40 in relation to RT, the most favorable outcomes were observed when anti-CTLA-4 was administered before RT. However, the highest percentage of survival was noted in the group receiving anti-OX40 1 day after RT [110]. Currently, the sequencing of IT and RT remains a subject of debate, with uncertainty regarding its effects across different cancer types. More clinical data are needed.
When combined with IT, lymphocyte-sparing irradiation should be critically considered [111], particularly in vulnerable elderly cancer patients. Lymphocyte-sparing RT limits irradiation dose to lymphocyte-enriched tissue/organs, such as large blood vessels, noninvolved lymph nodes, spleen, and bone marrow. Clinically, a high accumulated dose of RT to the above tissue/organs induces transient or persistent lymphocytopenia, including depletion of CD8(+) T-cells, which may vastly reduce the treatment effect of IT. As a result, for medical-fit patients, it is wise to limit irradiation dose to lymphoid-enriched organs at risk (OARs) according to the principle of “as low as reasonably achievable [112],” particularly in patients treated with combined pelvic irradiation and IT [113,114]. In this regard, advanced RT techniques, such as intensity-modulated radiotherapy [115] and volumetric-modulated arc therapy [116], are clinically helpful.
In contrast, for patients with bulky tumors, applying advanced RT techniques to deliver high tumor dose with maximum tolerated dose to nonlymphoid-enriched adjacent OARs for achieving the ultimate tumor control may be considered [117,118]. Advanced RT techniques are useful in RIT, such as stereotactic radiosurgery (SRS)/stereotactic radiotherapy [31] and SBRT/stereotactic ablative body radiotherapy [74,75,76,77,119] [Table 1]. The main reason is that advanced irradiation techniques largely improved therapeutic gains – increasing tumor control and decreasing treatment toxicities simultaneously. Furthermore, combined SRS and IT have been observed to enhance tumor control and improve neurological outcomes in lung cancer patients with brain metastases [31].
Some modern RT techniques demonstrated better clinical outcomes than conventional RT in the context of the RIT setting [17], such as charged particle irradiation [78], proton therapy [120], and FLASH-RT [79,121]. However, these RT techniques should be further tested in randomized clinical trials to demarcate their actual effective sizes.
Several novel IT agents are potential for clinical use in conjunction with RT, such as tumor vaccine, cytokine-based therapy, adoptive T-cell therapy, and nanoparticle-based IT [6]. Concurrently investigating potential predictive biomarkers is the trend in managing cancer patients treated with RIT [122]. For example, combined NKTR-214 IT and RT to stimulate systemic CD8(+) T-cell responses are tested for curing multi-focal cancers [92]. In this regard, the amount and function quality of activated CD8(+) T-cells may play crucial roles in predicting treatment response [123].
On the bench side, several signaling pathways have been reported to be bridged in RT and IT, such as the IFN-JAK-STAT axis [124] and well-known cGAS/STING signaling [125]. In this regard, epigenetic modification has been observed to play a role in conjunction with immune checkpoint inhibitors in several cancers, such as colorectal cancer [126] and melanoma [127].
Challenges of outcome prediction of radio-immunotherapy
Exploring clinically useful RIT biomarkers for outcome prediction is crucial and challenging in cancer patients [122,128]. Some outcome-predicting biomarkers have been reported, such as PD-L1 expression level, tumor mutation burden (TMB) [129,130,131], and imaging biomarkers [132,133] [Table 2]. Recently, combined biomarkers have been introduced clinically, such as the Integration of oncogene-based genomic profiling, tumor mutational burden, and PD-L1 expression [152,154]. In addition to the above biomarkers, one potential type of outcome-predicting biomarkers is epigenetic-based regulating factors. For example, the m6A RNA methylation has been observed to associate with the immune microenvironment, TMB, and PD-L1 level, predicting response to anti-PD-L1 IT [134]. Furthermore, the miRNA signature also demonstrates a value in predicting response to IT [122].
Table 2.
Potential biomarkers | Description | Reference |
---|---|---|
| ||
Epigenetic-based potential biomarkers | ||
m6A RNA methylation | The m6A RNA methylation associates with the immune microenvironment, TMB, and PD-L1 level, predicting response to anti-PD-L1 IT Besides, developing m6A RNA epigenetic agents is a potential direction to the synergy treatment effect of IT | [87,88,134] |
miRNA signature | Several miRNAs and their associated gene expression/signature are potential for predicting/prognostic biomarkers in RIT | [49,50,122,135,136,137] |
miR-16-5p | Serum exosomal miR-16-5p regulates PD-L1 expression, serving as a biomarker for PD-L1-based IT | [135] |
miR-195/miR-497 | miR-195/miR-497 regulates PD-L1 (i.e., CD274) expression in triple-negative breast cancer | [136,138] |
IMS | A high immune microenvironment score shows a better response to PD1/PDL1-based IT | [139] |
5mC score | A high 5mC score predicts low sensitivity to IT, neoadjuvant chemotherapy, and RT | [140] |
lncRNA | Several immune-related lncRNAs predict prognosis and treatment response | [49,50,141,142,143] |
circRNA | Has_circ_0006692 promoted NSCLC progression via the mir-205-5p/CDK10 axis and might serve as a prognostic biomarker and therapeutic target | [39] |
| ||
Nonepigenetic-based potential biomarkers | ||
| ||
TCR | Post-RT-released cancer dsDNAs activate the cGas-STING pathway and host T-cells. The function of activated T-cells plays an essential role in RIT | [144] |
CD8(+) T-cells | CD8(+) T-cell signature plays a role in the outcome prediction of IT | [123] |
PNI | PNI and NLR may predict treatment response in IT | [56] |
NLR | Elevated NLR was a poor predictor for OS in advanced and brain-metastatic NSCLC | [9,56,145] |
TMB | High TMB predicts better IT outcomes in several cancers, such as gastric cancer and NSCLC | [129,130,131] |
MSI-H | IT is effective in MSI-H cancer patients | [146] |
MMR-P | IT alone is ineffective in patients with MMR-P mCRC, even in conjunction with RT | [146,147] |
PD-L1 expression | PD-L1 expression is clinically applied for prescribing PD-L1-based IT | [131,148,149] |
TME score | TME score predicts prognosis and treatment response to IT in breast cancer patients | [150] |
Serum tumor markers | Dynamic changes of serum tumor markers, such as CEA, CA-125, and SCC-Ag, may be prognostic in IT | [151] |
KMT2C/TP53 co-mutation | KMT2C/TP53 co-mutation in conjunction with PD-L1 expression and TMB may be effective biomarkers in IT | [152] |
Imaging biomarkers | PET-based imaging biomarkers, such as (18) F-FET, (18) F-FDG, and preclinical [(64) Cu] NOTA-CD8a PET*, help monitor the treatment response of IT and RT | [132,133,153] |
*The evidence from [(64) Cu] NOTA-CD8a PET is based on preclinical in vivo experiments [153], revealing that identifying novel appropriate isotopes of PET for clinical application is challenging in terms of precisely targeting, clinical availableness, and cost-effectiveness. TCR: T-cell receptors, PNI: Prognostic nutrition index, NLR: Neutrophil-to-lymphocyte ratio, TMB: Tumor mutational burden, MSI-H: Microsatellite instability-high, PD-L1: Programmed death ligand-1, IR: Ionizing radiation, IT: Immunotherapy, IMS: Immune microenvironment score, ceRNA: Competitive endogenous RNA, NSCLC: Non-small cell lung cancer, lncRNA: Long noncoding RNA, MMR-P: Mismatch repair-proficient, mCRC: Metastatic colorectal cancer, TME: Tumor microenvironment, PET: Positron emission tomography, miR: MicroRNA, also termed miRNA, OS: Overall survival, RT: Radiotherapy
Remarkably, developing imaging biomarkers is particularly attractive for radiation oncologists [155]. The main reason is that the visible intra-tumor metabolic-hot part is crucial for guiding precise high-dose irradiation. Clinically, some metabolic-function-based Imaging biomarkers are introduced, such as (18) F-FET [132], (18) F-FDG [133], and preclinical [(64) Cu] NOTA-CD8a [153] positron emission tomography (PET). Note that evidence from [(64) Cu] NOTA-CD8a PET is based on preclinical in vivo experiments [153], revealing that identifying novel appropriate isotopes of PET for clinical application is challenging in terms of precisely targeting, clinical availableness, and cost-effectiveness.
Challenge of toxicity prevention for radio-immunotherapy
RT and IT have their treatment toxicities in managing cancer patients, respectively. Clinically, some treatment toxicities may be enhanced by applying RIT. For example, radiation-associated cardiovascular disease (RACVD) is a well-recognized late sequela in irradiated cancer survivors. In this regard, IT may also result in cardiovascular dysfunctions [156]. Hence, when IT is delivered in conjunction with RT, the risk of RACVD may be enhanced [157]. Besides, other RT-associated side effects may be enhanced with IT, such as lung fibrosis [158], particularly in patients treated with lung, breast, and esophagus RT. These toxicity enhancements may impair clinical outcomes and patients’ life quality.
As mentioned previously, some agents may help to prevent the occurrence of RACVD, such as statins (HMG-CoA reductase inhibitors) [159] and candesartan (an angiotensin II receptor antagonist) [160]. However, more clinical data are required to confirm whether these agents still benefit from preventing cardiovascular toxicities in the context of RIT.
Emerging challenge: Further multimodality combinations, including epigenetic agents, should be tested in randomized clinical trial settings
RIT gains clinical success in managing several types of cancer patients. However, post-RIT survived cancer cells may obtain acquired radioresistance and develop other strategies to evade immune surveillance [161]. To avoid such a condition, applying multimodality management, such as chemotherapy, targeted therapy [81,82], local intervention (e.g., thermal ablation [86]), or epigenetic modifier [87], may be considered in medically suitable patients who burden advanced cancer diseases. For example, cancer cells may modulate the TGF-β activation [162] or utilize the FTO-mediated epigenetic regulation to evade immune surveillance [87]. In the latter condition, applying agents to inhibit the activity of FTO, a m (6) A demethylase, can reprogram RNA epitranscriptome and synergize the treatment effect of IT [87].
Combined chemotherapy or targeted therapy’s role in enhancing IT’s treatment effect is also interesting in clinical oncology [81,82]. For example, combined short-course RT and IT are evaluated in conjunction with chemotherapy in the context of total neoadjuvant therapy for patients with local-regionally advanced rectal cancer [163]. If medical fit, one potential treatment direction is combining chemoradiotherapy and IT in patients with locoregionally advanced diseases [83]. However, the above new combinations of multimodality therapies planned to combine with RIT should be further tested in randomized clinical trials to define their effective sizes in tumor control and the potential harms of treatment toxicity.
Emerging challenge: The role of epigenetic modification in radiotherapy and immunotherapy in terms of potential biomarkers and therapeutic targets
As mentioned above, in addition to clinically applicable biomarkers, such as PD-L1 expression and TMB [129,130,131], several studies investigated novel predictive/prognostic biomarkers for RIT. Of these, endogenous noncoding RNAs, e.g., miRNA and lncRNA, that function in modulating gene expression grasp interests in both outcome-predicting biomarkers and potential treatment targets [164].
Emerging challenge of bench studies for outcome prediction in radio-immunotherapy, focusing on epigenetic biomarkers
Detecting miRNA levels in plasma or serum is actively investigated in outcome prediction and disease follow-up. Several miRNAs have been reported to show potential roles in predicting outcomes in cancer patients treated with RIT [49,50], such as miR-16-5p [135] and miR-195/miR-497 [136] [Table 2]. In epigenetics, one biological effect of circular RNAs (circRNAs) is to act as miRNA sponges that attenuate the function of pair-matched miRNAs [165]. As a result, circRNAs have been reported as one candidate of epigenetic biomarkers in RIT, e.g., Has_circ_0006692 [39]. Besides, other epigenetic-based biomarkers are potential for clinical application in RIT, such as 5 mC score [140] and lncRNA [49,50]. Note that the competitive endogenous RNA (ceRNA) network analysis should be considered to investigate the role of miRNA or lncRNA [141]. Besides, as mentioned previously [166], applying epigenetic biomarkers in clinical practice have some limitations and challenges. For example, different test timing may show different results that affect interpretations. Hence, at least two test time points should be implemented and denoted as pretreatment references, i.e., before initiating RT and IT, respectively.
Emerging challenge of bench studies for epigenetically therapeutic targets in radio-immunotherapy
Epigenetic regulation plays an active role in RT. For example, epigenetic dysregulation has been recognized to involve cancer radioresistance [167]. Remarkably, epigenetic regulators may reprogram the epigenome and restore radiosensitivity [168]. Similarly, applying epigenetic agents, such as HDAC inhibitors [169], to IT also gains ongoing interest [170]. Several epigenetically biological targets are investigated in RIT, such as PD-L1 [171], YTH domain containing 2 [172], and m6A RNA modification.
Of these, m6A RNA modification has been reported to be involved in cancer treatment resistance [88], and applying m6A RNA epigenetic agents may reverse the resistance and restore treatment effect [87]. Besides, DNA methylation of some target genes has been reported to correlate with immune infiltration and survival [173], serving as potential candidates for treatment targets. However, DNA-methylation-based epigenetic therapy has a challenge for clinical application. That is, targeted methylation/de-methylation is difficult to be achieved [174]. Unfortunately, this type of epigenetic modification is essential to leading DNA-methylation-based epigenetic therapy being clinically helpful and applicable. Remarkably, targeting noncoding RNAs, such as miRNA and lncRNA, are the preferred epigenetic modulation in cancer epigenetic-based therapy [175]; however, the role in the context of RIT should be further demarcated.
CONCLUSION
Combined RT and IT are one active treatment direction in managing cancer patients. Further efforts from clinical trials are ongoing to define the best combination, such as incorporating advanced RT techniques, novel IT agents, and other treatment modalities. Besides, bench and clinical studies are emergently required to improve the prediction of treatment response and clinical outcomes. Integrating other treatment modalities in conjunction with RIT, such as chemotherapy, targeted therapy, and other local therapies (e.g.,, thermal ablation), is warranted and ongoing. On ClinicalTrials.gov [176], more than 800 clinical trials are registered for investigating challenging issues of RIT in several cancer diseases. These data show a still hot trend of combined RIT in oncology. Results from these prospective trials are expected. Further investigations to explore effective epigenetics-based biomarkers and potential treatment targets are encouraged.
Data availability statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Financial support and sponsorship
The present study was supported by the Ministry of Science and Technology, Taiwan (Grant Number, 106-2923-B-194-001-MY3), and the Buddhist Tzu Chi Medical Foundation (Grant Number, TCMMP 105-09-02 [-04], TCMMP 106-02-02 [-04], TCMF-A 111-10, TCMF-A 112-04, TCMF-A 108-06, and DTCRD 106 [2]-E-18).
Conflicts of interest
There are no conflicts of interest.
REFERENCES
- 1.Clinical Practice Guidelines in Oncology; 2019. [(NCCN Guidelines™) [[Last accessed on 2018 Oct 09]]. Available from: https://www.nccn.org/professionals/physician_gls/default.aspx .
- 2.Halperin EC, Wazer DE, Perez CA, Brady LW. 7th ed. United States: Lippincott Williams & Wilkins; 2018. Perez and Brady's principles and practice of radiation oncology. [Google Scholar]
- 3.Islam MT. Radiation interactions with biological systems. Int J Radiat Biol. 2017;93:487–93. doi: 10.1080/09553002.2017.1286050. [DOI] [PubMed] [Google Scholar]
- 4.Czajkowski D, Szmyd R, Gee HE. Impact of DNA damage response defects in cancer cells on response to immunotherapy and radiotherapy. J Med Imaging Radiat Oncol. 2022;66:546–59. doi: 10.1111/1754-9485.13413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Theelen WS, de Jong MC, Baas P. Synergizing systemic responses by combining immunotherapy with radiotherapy in metastatic non-small cell lung cancer: The potential of the abscopal effect. Lung Cancer. 2020;142:106–13. doi: 10.1016/j.lungcan.2020.02.015. [DOI] [PubMed] [Google Scholar]
- 6.Yu S, Wang Y, He P, Shao B, Liu F, Xiang Z, et al. Effective combinations of immunotherapy and radiotherapy for cancer treatment. Front Oncol. 2022;12:809304. doi: 10.3389/fonc.2022.809304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mitchell MS, Jakowatz J, Harel W, Dean G, Stevenson L, Boswell WD, et al. Increased effectiveness of interferon alfa-2b following active specific immunotherapy for melanoma. J Clin Oncol. 1994;12:402–11. doi: 10.1200/JCO.1994.12.2.402. [DOI] [PubMed] [Google Scholar]
- 8.Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res. 2013;19:1021–34. doi: 10.1158/1078-0432.CCR-12-2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sheng J, Li H, Yu X, Yu S, Chen K, Pan G, et al. Efficacy of PD-1/PD-L1 inhibitors in patients with non-small cell lung cancer and brain metastases: A real-world retrospective study in China. Thorac Cancer. 2021;12:3019–31. doi: 10.1111/1759-7714.14171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Shibata H, Saito S, Uppaluri R. Immunotherapy for head and neck cancer: A paradigm shift from induction chemotherapy to neoadjuvant immunotherapy. Front Oncol. 2021;11:727433. doi: 10.3389/fonc.2021.727433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gutiérrez Calderón V, Cantero González A, Gálvez Carvajal L, Aguilar Lizarralde Y, Rueda Domínguez A. Neoadjuvant immunotherapy in resectable head and neck cancer: Oral cavity carcinoma as a potential research model. Ther Adv Med Oncol. 2021;13 doi: 10.1177/1758835920984061. 1758835920984061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ratnayake G, Shanker M, Roberts K, Mason R, Hughes BG, Lwin Z, et al. Prior or concurrent radiotherapy and nivolumab immunotherapy in non-small cell lung cancer. Asia Pac J Clin Oncol. 2020;16:56–62. doi: 10.1111/ajco.13242. [DOI] [PubMed] [Google Scholar]
- 13.Xing DT, Khor R, Gan H, Wada M, Ermongkonchai T, Ng SP. Recent research on combination of radiotherapy with targeted therapy or immunotherapy in head and neck squamous cell carcinoma: A review for radiation oncologists. Cancers (Basel) 2021;13:5716. doi: 10.3390/cancers13225716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Dong D, Zhu X, Wang H, Li L, Wan M, Li S, et al. Prognostic significance of albumin-bilirubin score in patients with unresectable hepatocellular carcinoma undergoing combined immunotherapy and radiotherapy. J Med Imaging Radiat Oncol. 2022;66:662–70. doi: 10.1111/1754-9485.13398. [DOI] [PubMed] [Google Scholar]
- 15.Werlenius K, Stragliotto G, Strandeus M, Blomstrand M, Carén H, Jakola AS, et al. A randomized phase II trial of efficacy and safety of the immunotherapy ALECSAT as an adjunct to radiotherapy and temozolomide for newly diagnosed glioblastoma. Neurooncol Adv. 2021;3 doi: 10.1093/noajnl/vdab156. vdab156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lara-Velazquez M, Shireman JM, Lehrer EJ, Bowman KM, Ruiz-Garcia H, Paukner MJ, et al. A comparison between chemo-radiotherapy combined with immunotherapy and chemo-radiotherapy alone for the treatment of newly diagnosed glioblastoma: A systematic review and meta-analysis. Front Oncol. 2021;11:662302. doi: 10.3389/fonc.2021.662302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tagliaferri L, Lancellotta V, Fionda B, Mangoni M, Casà C, Di Stefani A, et al. Immunotherapy and radiotherapy in melanoma: A multidisciplinary comprehensive review. Hum Vaccin Immunother. 2022;18:1903827. doi: 10.1080/21645515.2021.1903827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Moyers JT, Chong EG, Peng J, Tsai HH, Sufficool D, Shavlik D, et al. Real world outcomes of combination and timing of immunotherapy with radiotherapy for melanoma with brain metastases. Cancer Med. 2021;10:1201–11. doi: 10.1002/cam4.3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Guerini AE, Filippi AR, Tucci A, Simontacchi G, Re A, Guaineri A, et al. Le roi est mort, vive le roi: New roles of radiotherapy in the treatment of lymphomas in combination with immunotherapy. Clin Lymphoma Myeloma Leuk. 2022;22:e135–48. doi: 10.1016/j.clml.2021.09.005. [DOI] [PubMed] [Google Scholar]
- 20.Zhang R, Yang Y, Dong W, Lin M, He J, Zhang X, et al. D-mannose facilitates immunotherapy and radiotherapy of triple-negative breast cancer via degradation of PD-L1. Proc Natl Acad Sci U S A. 2022;119:e2114851119. doi: 10.1073/pnas.2114851119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.David S, Tan J, Siva S, Karroum L, Savas P, Loi S. Combining radiotherapy and immunotherapy in metastatic breast cancer: Current status and future directions. Biomedicines. 2022;10:821. doi: 10.3390/biomedicines10040821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yang X, Ren H, Fu J. Combinations of radiotherapy with immunotherapy in cervical cancer. J Cancer. 2022;13:1480–9. doi: 10.7150/jca.65074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Feng CH, Mell LK, Sharabi AB, McHale M, Mayadev JS. Immunotherapy with radiotherapy and chemoradiotherapy for cervical cancer. Semin Radiat Oncol. 2020;30:273–80. doi: 10.1016/j.semradonc.2020.05.003. [DOI] [PubMed] [Google Scholar]
- 24.Herrera FG, Irving M, Kandalaft LE, Coukos G. Rational combinations of immunotherapy with radiotherapy in ovarian cancer. Lancet Oncol. 2019;20:e417–33. doi: 10.1016/S1470-2045(19)30401-2. [DOI] [PubMed] [Google Scholar]
- 25.Wilkins A, Ost P, Sundahl N. Is there a benefit of combining immunotherapy and radiotherapy in bladder cancer? Clin Oncol (R Coll Radiol) 2021;33:407–14. doi: 10.1016/j.clon.2021.02.014. [DOI] [PubMed] [Google Scholar]
- 26.Daro-Faye M, Kassouf W, Souhami L, Marcq G, Cury F, Niazi T, et al. Combined radiotherapy and immunotherapy in urothelial bladder cancer: Harnessing the full potential of the anti-tumor immune response. World J Urol. 2021;39:1331–43. doi: 10.1007/s00345-020-03440-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ollivier L, Labbé M, Fradin D, Potiron V, Supiot S. Interaction between modern radiotherapy and immunotherapy for metastatic prostate cancer. Front Oncol. 2021;11:744679. doi: 10.3389/fonc.2021.744679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sun X, Gan L, Na A, Ge L, Chen B, Liu J. Combination with stereotactic body radiotherapy offers a promising strategy to overcome resistance to immunotherapy in advanced renal cell cancer. J Oncol. 2019;2019:1483406. doi: 10.1155/2019/1483406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sardaro A, Ferrari C, Carbonara R, Altini C, Lavelli V, Rubini G. Synergism between immunotherapy and radiotherapy in esophageal cancer: An overview of current knowledge and future perspectives. Cancer Biother Radiopharm. 2021;36:123–32. doi: 10.1089/cbr.2020.3643. [DOI] [PubMed] [Google Scholar]
- 30.Mahmood J, Shukla HD, Soman S, Samanta S, Singh P, Kamlapurkar S, et al. Immunotherapy, radiotherapy, and hyperthermia: A combined therapeutic approach in pancreatic cancer treatment. Cancers (Basel) 2018;10:469. doi: 10.3390/cancers10120469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Enright TL, Witt JS, Burr AR, Yadav P, Leal T, Baschnagel AM. Combined immunotherapy and stereotactic radiotherapy improves neurologic outcomes in patients with non-small-cell lung cancer brain metastases. Clin Lung Cancer. 2021;22:110–9. doi: 10.1016/j.cllc.2020.10.014. [DOI] [PubMed] [Google Scholar]
- 32.Corrao G, Marvaso G, Ferrara R, Lo Russo G, Gugliandolo SG, Piperno G, et al. Stereotatic radiotherapy in metastatic non-small cell lung cancer: Combining immunotherapy and radiotherapy with a focus on liver metastases. Lung Cancer. 2020;142:70–9. doi: 10.1016/j.lungcan.2020.02.017. [DOI] [PubMed] [Google Scholar]
- 33.Aiad M, Fresco K, Prenatt Z, Tahir A, Ramos-Feliciano K, Stoltzfus J, et al. Comparison of pneumonitis rates and severity in patients with lung cancer treated by immunotherapy, radiotherapy, and immunoradiotherapy. Cureus. 2022;14:e25665. doi: 10.7759/cureus.25665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Luna J, Zafra J, Areses Manrique MC, Rodríguez A, Sotoca A, Fírvida JL, et al. New challenges in the combination of radiotherapy and immunotherapy in non-small cell lung cancer. World J Clin Oncol. 2021;12:983–99. doi: 10.5306/wjco.v12.i11.983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lee JE, Kim MY. Cancer epigenetics: Past, present and future. Semin Cancer Biol. 2022;83:4–14. doi: 10.1016/j.semcancer.2021.03.025. [DOI] [PubMed] [Google Scholar]
- 36.Wisnieski F, Geraldis JC, Santos LC, Leal MF, Calcagno DQ, Gigek CO, et al. Differential regulation of LRRC37A2 in gastric cancer by DNA methylation. Epigenetics. 2022;17:110–6. doi: 10.1080/15592294.2021.1878724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Peng J, Ma Y, Zhao X, Yang X, Wang H. Constitutive ?-catenin overexpression represses lncrna MIR100HG transcription via HDAC6-mediated histone modification in colorectal cancer. Mol Cancer Res. 2022;20:949–59. doi: 10.1158/1541-7786.MCR-21-0923. [DOI] [PubMed] [Google Scholar]
- 38.Mortazavi D, Sohrabi B, Mosallaei M, Nariman-Saleh-Fam Z, Bastami M, Mansoori Y, et al. Epi-miRNAs: Regulators of the histone modification machinery in human cancer. J Oncol 2022. 2022 doi: 10.1155/2022/4889807. 4889807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Liao J, Chen Z, Luo X, Su Y, Huang T, Xu H, et al. Hsa_circ_0006692 promotes lung cancer progression via miR-205-5p/CDK19 axis. Genes (Basel) 2022;13:846. doi: 10.3390/genes13050846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kim TW. Cinnamaldehyde induces autophagy-mediated cell death through ER stress and epigenetic modification in gastric cancer cells. Acta Pharmacol Sin. 2022;43:712–23. doi: 10.1038/s41401-021-00672-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lian Y, Meng L, Ding P, Sang M. Epigenetic regulation of MAGE family in human cancer progression-DNA methylation, histone modification, and non-coding RNAs. Clin Epigenetics. 2018;10:115. doi: 10.1186/s13148-018-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yu B, Yu X, Xiong J, Ma M. Methylation modification, alternative splicing, and noncoding RNA play a role in cancer metastasis through epigenetic regulation. Biomed Res Int 2021. 2021 doi: 10.1155/2021/4061525. 4061525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Naqvi SM, Kim Y. Epigenetic modification by galactic cosmic radiation as a risk factor for lung cancer: Real world data issues. Transl Lung Cancer Res. 2019;8:116–8. doi: 10.21037/tlcr.2019.01.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Pang B, Sui S, Wang Q, Wu J, Yin Y, Xu S. Upregulation of DLEU1 expression by epigenetic modification promotes tumorigenesis in human cancer. J Cell Physiol. 2019;234:17420–32. doi: 10.1002/jcp.28364. [DOI] [PubMed] [Google Scholar]
- 45.Aspriţoiu VM, Stoica I, Bleotu C, Diaconu CC. Epigenetic regulation of angiogenesis in development and tumors progression: Potential implications for cancer treatment. Front Cell Dev Biol. 2021;9:689962. doi: 10.3389/fcell.2021.689962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Li Y, Wan X, Wei Y, Liu X, Lai W, Zhang L, et al. LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion. Oncol Rep. 2016;35:3586–92. doi: 10.3892/or.2016.4729. [DOI] [PubMed] [Google Scholar]
- 47.McCaw TR, Randall TD, Arend RC. Overcoming immune suppression with epigenetic modification in ovarian cancer. Transl Res. 2019;204:31–8. doi: 10.1016/j.trsl.2018.06.003. [DOI] [PubMed] [Google Scholar]
- 48.Sulek JE, Robinson SP, Petrossian AA, Zhou S, Goliadze E, Manjili MH, et al. Role of epigenetic modification and immunomodulation in a murine prostate cancer model. Prostate. 2017;77:361–73. doi: 10.1002/pros.23275. [DOI] [PubMed] [Google Scholar]
- 49.Volovat SR, Volovat C, Hordila I, Hordila DA, Mirestean CC, Miron OT, et al. MiRNA and LncRNA as potential biomarkers in triple-negative breast cancer: A review. Front Oncol. 2020;10:526850. doi: 10.3389/fonc.2020.526850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lv Y, Wang Y, Zhang Z, Bao J, Su H. Potentials of long non-coding RNAs as biomarkers of colorectal cancer. Clin Transl Oncol. 2022;24:1715–31. doi: 10.1007/s12094-022-02834-7. [DOI] [PubMed] [Google Scholar]
- 51.Liu Z, Gao Y, Li X. Cancer epigenetics and the potential of epigenetic drugs for treating solid tumors. Expert Rev Anticancer Ther. 2019;19:139–49. doi: 10.1080/14737140.2019.1552139. [DOI] [PubMed] [Google Scholar]
- 52.Li GH, Qu Q, Qi TT, Teng XQ, Zhu HH, Wang JJ, et al. Super-enhancers: A new frontier for epigenetic modifiers in cancer chemoresistance. J Exp Clin Cancer Res. 2021;40:174. doi: 10.1186/s13046-021-01974-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yang Y, Qin Y, Zeng Z, Lu Y, Gong Y, Li Y, et al. The pan-cancer noninvasive screening and early detection by epigenetic techniques. Epigenomics. 2021;13:649–52. doi: 10.2217/epi-2021-0063. [DOI] [PubMed] [Google Scholar]
- 54.Xu Q, Ye L, Huang L, Zhou L, Chen X, Ye M, et al. Serum exosomal miRNA might be a novel liquid biopsy to identify leptomeningeal metastasis in non-small cell lung cancer. Onco Targets Ther. 2021;14:2327–35. doi: 10.2147/OTT.S291611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jamialahmadi K, Azghandi M, Javadmanesh A, Zardadi M, Shams Davodly E, Kerachian MA. A DNA methylation panel for high performance detection of colorectal cancer. Cancer Genet. 2021;252-253:64–72. doi: 10.1016/j.cancergen.2020.12.007. [DOI] [PubMed] [Google Scholar]
- 56.Zhang Y, Jin J, Tang M, Li P, Zhou LN, Du YP, et al. Prognostic nutritional index predicts outcome of PD-L1 negative and MSS advanced cancer treated with PD-1 inhibitors. Biomed Res Int 2022. 2022 doi: 10.1155/2022/6743126. 6743126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Theelen WS, Chen D, Verma V, Hobbs BP, Peulen HM, Aerts JG, et al. Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: A pooled analysis of two randomised trials. Lancet Respir Med. 2021;9:467–75. doi: 10.1016/S2213-2600(20)30391-X. [DOI] [PubMed] [Google Scholar]
- 58.Yang Y, Deng L, Yang Y, Zhang T, Wu Y, Wang L, et al. Efficacy and safety of combined brain radiotherapy and immunotherapy in non-small-cell lung cancer with brain metastases: A systematic review and meta-analysis. Clin Lung Cancer. 2022;23:95–107. doi: 10.1016/j.cllc.2021.06.009. [DOI] [PubMed] [Google Scholar]
- 59.Zhu J, Yuan Y, Wan X, Yin D, Li R, Chen W, et al. Immunotherapy (excluding checkpoint inhibitors) for stage I to III non-small cell lung cancer treated with surgery or radiotherapy with curative intent. Cochrane Database Syst Rev. 2021;12 doi: 10.1002/14651858.CD011300.pub3. CD011300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Suwinski R. Combination of immunotherapy and radiotherapy in the treatment of brain metastases from non-small cell lung cancer. J Thorac Dis. 2021;13:3315–22. doi: 10.21037/jtd-2019-rbmlc-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Shang S, Liu J, Verma V, Wu M, Welsh J, Yu J, et al. Combined treatment of non-small cell lung cancer using radiotherapy and immunotherapy: Challenges and updates. Cancer Commun (Lond) 2021;41:1086–99. doi: 10.1002/cac2.12226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Scoccianti S, Olmetto E, Pinzi V, Osti MF, Di Franco R, Caini S, et al. Immunotherapy in association with stereotactic radiotherapy for non-small cell lung cancer brain metastases: Results from a multicentric retrospective study on behalf of AIRO. Neuro Oncol. 2021;23:1750–64. doi: 10.1093/neuonc/noab129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Onn A, Gottfried T, Stemmer A, Appel S, Lawrence YR, Urban D, et al. Real-world analysis of the impact of radiotherapy on immunotherapy efficacy in non-small cell lung cancer. Cancers (Basel) 2021;13:2800. doi: 10.3390/cancers13112800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Miyasaka Y, Sato H, Okano N, Kubo N, Kawamura H, Ohno T. A promising treatment strategy for lung cancer: A combination of radiotherapy and immunotherapy. Cancers (Basel) 2021;14:203. doi: 10.3390/cancers14010203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Liu G, Lv X, Ding Y, Guo Y. Precise medicine of programmed cell death-1/programmed cell death 1 ligand 1 inhibitor immunotherapy combined radiotherapy for inoperable advanced lung cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021;100:e26367. doi: 10.1097/MD.0000000000026367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Chodavadia PA, Jacobs CD, Wang F, Salama JK, Kelsey CR, Clarke JM, et al. Synergy between early-incorporation immunotherapy and extracranial radiotherapy in metastatic non-small cell lung cancer. Transl Lung Cancer Res. 2021;10:261–73. doi: 10.21037/tlcr-20-537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Stanic K, Vrankar M, But-Hadzic J. Consolidation radiotherapy for patients with extended disease small cell lung cancer in a single tertiary institution: Impact of dose and perspectives in the era of immunotherapy. Radiol Oncol. 2020;54:353–63. doi: 10.2478/raon-2020-0046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wu M, Liu J, Wu S, Liu J, Wu H, Yu J, et al. Systemic immune activation and responses of irradiation to different metastatic sites combined with immunotherapy in advanced non-small cell lung cancer. Front Immunol. 2021;12:803247. doi: 10.3389/fimmu.2021.803247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sari SY, Yilmaz MT, Aktas BY, Aksoy S, Gullu I, Cengiz M, et al. Results of concurrent radiotherapy and immunotherapy in recurrent and metastatic head and neck cancer: A single-center experience. Oral Oncol. 2022;124:105658. doi: 10.1016/j.oraloncology.2021.105658. [DOI] [PubMed] [Google Scholar]
- 70.Dua D, Kelly C, Kovarik J, Iqbal MS. The role of combining immunotherapy with primary (Chemo) radiotherapy in curative treatment settings of the head and neck cancer. Asia Pac J Clin Oncol. 2022;18:e3–10. doi: 10.1111/ajco.13583. [DOI] [PubMed] [Google Scholar]
- 71.Qian JM, Schoenfeld JD. Radiotherapy and immunotherapy for head and neck cancer: Current evidence and challenges. Front Oncol. 2020;10 doi: 10.3389/fonc.2020.608772. 608772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Plavc G, Jesenko T, Oražem M, Strojan P. Challenges in combining immunotherapy with radiotherapy in recurrent/metastatic head and neck cancer. Cancers (Basel) 2020;12:3197. doi: 10.3390/cancers12113197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Merlano MC, Merlotti AM, Licitra L, Denaro N, Fea E, Galizia D, et al. Activation of immune responses in patients with relapsed-metastatic head and neck cancer (CONFRONT phase I-II trial): Multimodality immunotherapy with avelumab, short-course radiotherapy, and cyclophosphamide. Clin Transl Radiat Oncol. 2018;12:47–52. doi: 10.1016/j.ctro.2018.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Akanda ZZ, Neeson PJ, John T, Barnett S, Hanna GG, Miller A, et al. A narrative review of combined stereotactic ablative radiotherapy and immunotherapy in metastatic non-small cell lung cancer. Transl Lung Cancer Res. 2021;10:2766–78. doi: 10.21037/tlcr-20-1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Chen D, Patel RR, Verma V, Ramapriyan R, Barsoumian HB, Cortez MA, et al. Interaction between lymphopenia, radiotherapy technique, dosimetry, and survival outcomes in lung cancer patients receiving combined immunotherapy and radiotherapy. Radiother Oncol. 2020;150:114–20. doi: 10.1016/j.radonc.2020.05.051. [DOI] [PubMed] [Google Scholar]
- 76.Chicas-Sett R, Morales-Orue I, Castilla-Martinez J, Zafra-Martin J, Kannemann A, Blanco J, et al. Stereotactic ablative radiotherapy combined with immune checkpoint inhibitors reboots the immune response assisted by immunotherapy in metastatic lung cancer: A systematic review. Int J Mol Sci. 2019;20:2173. doi: 10.3390/ijms20092173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Mattes MD, Eubank TD, Almubarak M, Wen S, Marano GD, Jacobson GM, et al. A prospective trial evaluating the safety and systemic response from the concurrent use of radiation therapy with checkpoint inhibitor immunotherapy in metastatic non-small cell lung cancer. Clin Lung Cancer. 2021;22:268–73. doi: 10.1016/j.cllc.2021.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Marcus D, Lieverse RI, Klein C, Abdollahi A, Lambin P, Dubois LJ, et al. Charged particle and conventional radiotherapy: Current implications as partner for immunotherapy. Cancers (Basel) 2021;13:1468. doi: 10.3390/cancers13061468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhang Y, Ding Z, Perentesis JP, Khuntia D, Pfister SX, Sharma RA. Can rational combination of ultra-high dose rate flash radiotherapy with immunotherapy provide a novel approach to cancer treatment? Clin Oncol (R Coll Radiol) 2021;33:713–22. doi: 10.1016/j.clon.2021.09.003. [DOI] [PubMed] [Google Scholar]
- 80.Trivillin VA, Langle YV, Palmieri MA, Pozzi EC, Thorp SI, Benitez Frydryk DN, et al. Evaluation of local, regional and abscopal effects of boron neutron capture therapy (BNCT) combined with immunotherapy in an ectopic colon cancer model. Br J Radiol. 2021;94:20210593. doi: 10.1259/bjr.20210593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Seo I, Lee HW, Byun SJ, Park JY, Min H, Lee SH, et al. Neoadjuvant chemoradiation alters biomarkers of anticancer immunotherapy responses in locally advanced rectal cancer. J Immunother Cancer. 2021;9:e001610. doi: 10.1136/jitc-2020-001610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mo DC, Huang JF, Luo PH, Huang SX, Wang HL. The efficacy and safety of combination therapy with immune checkpoint inhibitors in non-small cell lung cancer: A meta-analysis. Int Immunopharmacol. 2021;96:107594. doi: 10.1016/j.intimp.2021.107594. [DOI] [PubMed] [Google Scholar]
- 83.Rallis KS, Lai Yau TH, Sideris M. Chemoradiotherapy in cancer treatment: Rationale and clinical applications. Anticancer Res. 2021;41:1–7. doi: 10.21873/anticanres.14746. [DOI] [PubMed] [Google Scholar]
- 84.Wang Y, Shen L, Wan J, Zhang H, Wu R, Wang J, et al. Short-course radiotherapy combined with CAPOX and Toripalimab for the total neoadjuvant therapy of locally advanced rectal cancer: A randomized, prospective, multicentre, double-arm, phase II trial (TORCH) BMC Cancer. 2022;22:274. doi: 10.1186/s12885-022-09348-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Limagne E, Nuttin L, Thibaudin M, Jacquin E, Aucagne R, Bon M, et al. MEK inhibition overcomes chemoimmunotherapy resistance by inducing CXCL10 in cancer cells. Cancer Cell. 2022;40:136–52.e12. doi: 10.1016/j.ccell.2021.12.009. [DOI] [PubMed] [Google Scholar]
- 86.Rangamuwa K, Leong T, Weeden C, Asselin-Labat ML, Bozinovski S, Christie M, et al. Thermal ablation in non-small cell lung cancer: A review of treatment modalities and the evidence for combination with immune checkpoint inhibitors. Transl Lung Cancer Res. 2021;10:2842–57. doi: 10.21037/tlcr-20-1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu Y, Liang G, Xu H, Dong W, Dong Z, Qiu Z, et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab. 2021;33:1221–33.e11. doi: 10.1016/j.cmet.2021.04.001. [DOI] [PubMed] [Google Scholar]
- 88.Shriwas O, Mohapatra P, Mohanty S, Dash R. The impact of m6A RNA modification in therapy resistance of cancer: Implication in chemotherapy, radiotherapy, and immunotherapy. Front Oncol. 2020;10:612337. doi: 10.3389/fonc.2020.612337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Castro F, Pinto ML, Pereira CL, Serre K, Barbosa MA, Vermaelen K, et al. Chitosan/γ-PGA nanoparticles-based immunotherapy as adjuvant to radiotherapy in breast cancer. Biomaterials. 2020;257:120218. doi: 10.1016/j.biomaterials.2020.120218. [DOI] [PubMed] [Google Scholar]
- 90.Selvaraja VK, Gudipudi DK. Fundamentals to clinical application of nanoparticles in cancer immunotherapy and radiotherapy. Ecancermedicalscience. 2020;14:1095. doi: 10.3332/ecancer.2020.1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.American Joint Committee on Cancer, editor. AJCC cancer staging manual. 5th ed. Philadelphia: Lippincott-Raven; 1997. [Google Scholar]
- 92.Walker JM, Rolig AS, Charych DH, Hoch U, Kasiewicz MJ, Rose DC, et al. NKTR-214 immunotherapy synergizes with radiotherapy to stimulate systemic CD8(+) T cell responses capable of curing multi-focal cancer. J Immunother Cancer. 2020;8:e000464. doi: 10.1136/jitc-2019-000464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Nava S, Lisini D, Frigerio S, Bersano A. Dendritic cells and cancer immunotherapy: The adjuvant effect. Int J Mol Sci. 2021;22:12339. doi: 10.3390/ijms222212339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Keam S, Gill S, Ebert MA, Nowak AK, Cook AM. Enhancing the efficacy of immunotherapy using radiotherapy. Clin Transl Immunology. 2020;9:e1169. doi: 10.1002/cti2.1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Liu Z, Zhao Q, Zheng Z, Liu S, Meng L, Dong L, et al. Vascular normalization in immunotherapy: A promising mechanisms combined with radiotherapy. Biomed Pharmacother. 2021;139:111607. doi: 10.1016/j.biopha.2021.111607. [DOI] [PubMed] [Google Scholar]
- 96.Vanneste BG, Van Limbergen EJ, Dubois L, Samarska IV, Wieten L, Aarts MJ, et al. Immunotherapy as sensitizer for local radiotherapy. Oncoimmunology. 2020;9:1832760. doi: 10.1080/2162402X.2020.1832760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Buchwald ZS, Wynne J, Nasti TH, Zhu S, Mourad WF, Yan W, et al. Radiation, immune checkpoint blockade and the abscopal effect: A critical review on timing, dose and fractionation. Front Oncol. 2018;8:612. doi: 10.3389/fonc.2018.00612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Trommer-Nestler M, Marnitz S, Kocher M, Rueß D, Schlaak M, Theurich S, et al. Robotic stereotactic radiosurgery in melanoma patients with brain metastases under simultaneous anti-PD-1 treatment. Int J Mol Sci. 2018;19:2653. doi: 10.3390/ijms19092653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Aboudaram A, Modesto A, Chaltiel L, Gomez-Roca C, Boulinguez S, Sibaud V, et al. Concurrent radiotherapy for patients with metastatic melanoma and receiving anti-programmed-death 1 therapy: A safe and effective combination. Melanoma Res. 2017;27:485–91. doi: 10.1097/CMR.0000000000000386. [DOI] [PubMed] [Google Scholar]
- 100.Wang SJ, Jhawar SR, Rivera-Nunez Z, Silk AW, Byun J, Miller E, et al. The association of radiation dose-fractionation and immunotherapy use with overall survival in metastatic melanoma patients. Cureus. 2020;12 doi: 10.7759/cureus.8767. e8767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Theelen WS, Peulen HM, Lalezari F, van der Noort V, de Vries JF, Aerts JG, et al. Effect of pembrolizumab after stereotactic body radiotherapy versus 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. 2019;5:1276–82. doi: 10.1001/jamaoncol.2019.1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Vanpouille-Box C, Formenti SC, Demaria S. TREX1 dictates the immune fate of irradiated cancer cells. Oncoimmunology. 2017;6:e1339857. doi: 10.1080/2162402X.2017.1339857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2017;377:1919–29. doi: 10.1056/NEJMoa1709937. [DOI] [PubMed] [Google Scholar]
- 104.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018;379:2342–50. doi: 10.1056/NEJMoa1809697. [DOI] [PubMed] [Google Scholar]
- 105.Faivre-Finn C, Vicente D, Kurata T, Planchard D, Paz-Ares L, Vansteenkiste JF, et al. Four-year survival with durvalumab after chemoradiotherapy in stage III NSCLC-an update from the pacific trial. J Thorac Oncol. 2021;16:860–7. doi: 10.1016/j.jtho.2020.12.015. [DOI] [PubMed] [Google Scholar]
- 106.Spigel DR, Faivre-Finn C, Gray JE, Vicente D, Planchard D, Paz-Ares L, et al. Five-year survival outcomes from the pacific trial: Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. J Clin Oncol. 2022;40:1301–11. doi: 10.1200/JCO.21.01308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Welsh J, Menon H, Chen D, Verma V, Tang C, Altan M, et al. Pembrolizumab with or without radiation therapy for metastatic non-small cell lung cancer: A randomized phase I/II trial. J Immunother Cancer. 2020;8:e001001. doi: 10.1136/jitc-2020-001001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74:5458–68. doi: 10.1158/0008-5472.CAN-14-1258. [DOI] [PubMed] [Google Scholar]
- 109.Young KH, Newell P, Cottam B, Friedman D, Savage T, Baird JR, et al. TGFβ inhibition prior to hypofractionated radiation enhances efficacy in preclinical models. Cancer Immunol Res. 2014;2:1011–22. doi: 10.1158/2326-6066.CIR-13-0207. [DOI] [PubMed] [Google Scholar]
- 110.Young KH, Baird JR, Savage T, Cottam B, Friedman D, Bambina S, et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One. 2016;11:e0157164. doi: 10.1371/journal.pone.0157164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lambin P, Lieverse RI, Eckert F, Marcus D, Oberije C, van der Wiel AM, et al. Lymphocyte-sparing radiotherapy: The rationale for protecting lymphocyte-rich organs when combining radiotherapy with immunotherapy. Semin Radiat Oncol. 2020;30:187–93. doi: 10.1016/j.semradonc.2019.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hiniker SM, Donaldson SS. ALARA: In radiation oncology and diagnostic imaging alike. Oncology (Williston Park) 2014;28:247–8. [PubMed] [Google Scholar]
- 113.Zhou P, Zhang Y, Luo S, Zhang S. Pelvic bone marrow sparing radiotherapy for cervical cancer: A systematic review and meta-analysis. Radiother Oncol. 2021;165:103–18. doi: 10.1016/j.radonc.2021.10.015. [DOI] [PubMed] [Google Scholar]
- 114.Huang W, Dang J, Li Y, Cui HX, Lu WL, Jiang QF. Effect of pelvic bone marrow sparing intensity modulated radiation therapy on acute hematologic toxicity in rectal cancer patients undergoing chemo-radiotherapy. Front Oncol. 2021;11:646211. doi: 10.3389/fonc.2021.646211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Huang J, Gu F, Ji T, Zhao J, Li G. Pelvic bone marrow sparing intensity modulated radiotherapy reduces the incidence of the hematologic toxicity of patients with cervical cancer receiving concurrent chemoradiotherapy: A single-center prospective randomized controlled trial. Radiat Oncol. 2020;15:180. doi: 10.1186/s13014-020-01606-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Chen JL, Wang MC, Huang YS, Huang CY, Pan CK, Hsu CY, et al. Extended-field bone marrow sparing radiotherapy for primary chemoradiotherapy in cervical cancer patients with para-aortic lymphadenopathy: Volumetric-modulated arc therapy versus helical tomotherapy. J Xray Sci Technol. 2020;28:111–24. doi: 10.3233/XST-190593. [DOI] [PubMed] [Google Scholar]
- 117.Ozyigit G, Selek U. Switzerland: Springer Nature Switzerland AG; 2019. Radiation oncology: A case-based review. [Google Scholar]
- 118.Ohri N. Radiotherapy dosing for locally advanced non-small cell lung carcinoma: MTD or “ALARA”? Front Oncol. 2017;7:205. doi: 10.3389/fonc.2017.00205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chen LC, Lin HY, Lee MS, Chiou WY, Huang LW, Chew CH, et al. Effectiveness of individual audio-visual coaching, respiratory modulated stereotactic body radiotherapy for localized hepatocellular carcinoma: Analysis of 29 cases from a single academic radiotherapy center. Tzu Chi Med J. 2021;33:380–7. doi: 10.4103/tcmj.tcmj_247_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lee HJ, Jr, Zeng J, Rengan R. Proton beam therapy and immunotherapy: An emerging partnership for immune activation in non-small cell lung cancer. Transl Lung Cancer Res. 2018;7:180–8. doi: 10.21037/tlcr.2018.03.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Borghini A, Vecoli C, Labate L, Panetta D, Andreassi MG, Gizzi LA. FLASH ultra-high dose rates in radiotherapy: Preclinical and radiobiological evidence. Int J Radiat Biol. 2022;98:127–35. doi: 10.1080/09553002.2022.2009143. [DOI] [PubMed] [Google Scholar]
- 122.Meng L, Xu J, Ye Y, Wang Y, Luo S, Gong X. The combination of radiotherapy with immunotherapy and potential predictive biomarkers for treatment of non-small cell lung cancer patients. Front Immunol. 2021;12:723609. doi: 10.3389/fimmu.2021.723609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sun R, Sundahl N, Hecht M, Putz F, Lancia A, Rouyar A, et al. Radiomics to predict outcomes and abscopal response of patients with cancer treated with immunotherapy combined with radiotherapy using a validated signature of CD8 cells. J Immunother Cancer. 2020;8:e001429. doi: 10.1136/jitc-2020-001429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Shi LZ, Bonner JA. Bridging radiotherapy to immunotherapy: The IFN-JAK-STAT axis. Int J Mol Sci. 2021;22:12295. doi: 10.3390/ijms222212295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sato H, Demaria S, Ohno T. The role of radiotherapy in the age of immunotherapy. Jpn J Clin Oncol. 2021;51:513–22. doi: 10.1093/jjco/hyaa268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tong H, Wei H, Smith AO, Huang J. The role of m6A epigenetic modification in the treatment of colorectal cancer immune checkpoint inhibitors. Front Immunol. 2021;12:802049. doi: 10.3389/fimmu.2021.802049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Nanamori H, Sawada Y. Epigenetic modification of PD-1/PD-L1-mediated cancer immunotherapy against melanoma. Int J Mol Sci. 2022;23:1119. doi: 10.3390/ijms23031119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.van Hooren L, Handgraaf SM, Kloosterman DJ, Karimi E, van Mil LW, Gassama AA, et al. CD103(+) regulatory T cells underlie resistance to radio-immunotherapy and impair CD8(+) T cell activation in glioblastoma. Nat Cancer. 2023;4:665–81. doi: 10.1038/s43018-023-00547-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chang X, Ge X, Zhang Y, Xue X. The current management and biomarkers of immunotherapy in advanced gastric cancer. Medicine (Baltimore) 2022;101:e29304. doi: 10.1097/MD.0000000000029304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Nan Z, Guoqing W, Xiaoxu Y, Yin M, Xin H, Xue L, et al. The predictive efficacy of tumor mutation burden (TMB) on nonsmall cell lung cancer treated by immune checkpoint inhibitors: A systematic review and meta-analysis. Biomed Res Int 2021. 2021 doi: 10.1155/2021/1780860. 1780860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Deng H, Zhao Y, Cai X, Chen H, Cheng B, Zhong R, et al. PD-L1 expression and tumor mutation burden as pathological response biomarkers of neoadjuvant immunotherapy for early-stage non-small cell lung cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 2022;170:103582. doi: 10.1016/j.critrevonc.2022.103582. [DOI] [PubMed] [Google Scholar]
- 132.Galldiks N, Abdulla DS, Scheffler M, Wolpert F, Werner JM, Hüllner M, et al. Treatment monitoring of immunotherapy and targeted therapy using (18) F-FET PET in patients with melanoma and lung cancer brain metastases: Initial experiences. J Nucl Med. 2021;62:464–70. doi: 10.2967/jnumed.120.248278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Vekens K, Everaert H, Neyns B, Ilsen B, Decoster L. The value of (18) F-FDG PET/CT in predicting the response to PD-1 blocking immunotherapy in advanced NSCLC patients with high-level PD-L1 expression. Clin Lung Cancer. 2021;22:432–40. doi: 10.1016/j.cllc.2021.03.001. [DOI] [PubMed] [Google Scholar]
- 134.Liu X, Ma C, Liu H, Sun Z, Luo J. M6A regulator expression patterns predict the immune microenvironment and prognosis of non-small cell lung cancer. J Cancer Res Clin Oncol. 2022;148:2803–14. doi: 10.1007/s00432-022-04032-y. [DOI] [PubMed] [Google Scholar]
- 135.Chen HL, Luo YP, Lin MW, Peng XX, Liu ML, Wang YC, et al. Serum exosomal miR-16-5p functions as a tumor inhibitor and a new biomarker for PD-L1 inhibitor-dependent immunotherapy in lung adenocarcinoma by regulating PD-L1 expression. Cancer Med. 2022;11:2627–43. doi: 10.1002/cam4.4638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yang L, Cai Y, Zhang D, Sun J, Xu C, Zhao W, et al. miR-195/miR-497 regulate CD274 expression of immune regulatory ligands in triple-negative breast cancer. J Breast Cancer. 2018;21:371–81. doi: 10.4048/jbc.2018.21.e60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Wu Q, Yang Z, Shi Y, Fan D. MiRNAs in human cancers: The diagnostic and therapeutic implications. Curr Pharm Des. 2014;20:5336–47. doi: 10.2174/1381612820666140128204914. [DOI] [PubMed] [Google Scholar]
- 138.Tao Z, Xu S, Ruan H, Wang T, Song W, Qian L, et al. MiR-195/-16 family enhances radiotherapy via T cell activation in the tumor microenvironment by blocking the PD-L1 immune checkpoint. Cell Physiol Biochem. 2018;48:801–14. doi: 10.1159/000491909. [DOI] [PubMed] [Google Scholar]
- 139.Lin Y, Pan X, Zhao L, Yang C, Zhang Z, Wang B, et al. Immune cell infiltration signatures identified molecular subtypes and underlying mechanisms in gastric cancer. NPJ Genom Med. 2021;6:83. doi: 10.1038/s41525-021-00249-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hu J, Othmane B, Yu A, Li H, Cai Z, Chen X, et al. 5mC regulator-mediated molecular subtypes depict the hallmarks of the tumor microenvironment and guide precision medicine in bladder cancer. BMC Med. 2021;19:289. doi: 10.1186/s12916-021-02163-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yang CZ, Hu LH, Huang ZY, Deng L, Guo W, Liu S, et al. Comprehensive analysis of an immune infiltrate-related competitive endogenous RNA network reveals potential prognostic biomarkers for non-small cell lung cancer. PLoS One. 2021;16:e0260720. doi: 10.1371/journal.pone.0260720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Liu X, Wang Z, Wang L, Wang Y, Wang Y, Yang S, et al. Signature involved in immune-related lncRNA pairs for predicting the immune landscape of cervical cancer. Int J Biol Markers. 2022;37:191–9. doi: 10.1177/03936155221091832. [DOI] [PubMed] [Google Scholar]
- 143.Luo Y, Yang J, Yu J, Liu X, Yu C, Hu J, et al. Long non-coding RNAs: Emerging roles in the immunosuppressive tumor microenvironment. Front Oncol. 2020;10:48. doi: 10.3389/fonc.2020.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Baxevanis CN, Gritzapis AD, Voutsas IF, Batsaki P, Goulielmaki M, Adamaki M, et al. T-cell repertoire in tumor radiation: The emerging frontier as a radiotherapy biomarker. Cancers (Basel) 2022;14:2674. doi: 10.3390/cancers14112674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Öjlert ÅK, Nebdal D, Lund-Iversen M, Åstrøm Ellefsen R, Brustugun OT, Gran JM, et al. Immune checkpoint blockade in the treatment of advanced non-small cell lung cancer –Predictors of response and impact of previous radiotherapy. Acta Oncol. 2021;60:149–56. doi: 10.1080/0284186X.2020.1854851. [DOI] [PubMed] [Google Scholar]
- 146.Liu N, Shan F, Ma M. Strategic enhancement of immune checkpoint inhibition in refractory colorectal cancer: Trends and future prospective. Int Immunopharmacol. 2021;99:108017. doi: 10.1016/j.intimp.2021.108017. [DOI] [PubMed] [Google Scholar]
- 147.Segal NH, Cercek A, Ku G, Wu AJ, Rimner A, Khalil DN, et al. Phase II single-arm study of durvalumab and tremelimumab with concurrent radiotherapy in patients with mismatch repair-proficient metastatic colorectal cancer. Clin Cancer Res. 2021;27:2200–8. doi: 10.1158/1078-0432.CCR-20-2474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bassanelli M, Ramella S, Zeuli M, Ceribelli A. Radiotherapy and immunotherapy: The power of the teamwork for the treatment of NSCLC. Anticancer Res. 2022;42:2241–7. doi: 10.21873/anticanres.15704. [DOI] [PubMed] [Google Scholar]
- 149.Xiong L, Cai Y, Zhou X, Dai P, Wei Y, Zhao J, et al. Optimum immunotherapy method according to PD-L1 expression in advanced lung cancer: A network meta-analysis. Future Oncol. 2022;18:883–96. doi: 10.2217/fon-2021-1217. [DOI] [PubMed] [Google Scholar]
- 150.Tian Y, Wang J, Wen Q, Gao A, Huang A, Li R, et al. The significance of tumor microenvironment score for breast cancer patients. Biomed Res Int 2022. 2022 doi: 10.1155/2022/5673810. 5673810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Zhang Z, Yuan F, Chen R, Li Y, Ma J, Yan X, et al. Dynamics of serum tumor markers can serve as a prognostic biomarker for Chinese advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors. Front Immunol. 2020;11:1173. doi: 10.3389/fimmu.2020.01173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Shi Y, Lei Y, Liu L, Zhang S, Wang W, Zhao J, et al. Integration of comprehensive genomic profiling, tumor mutational burden, and PD-L1 expression to identify novel biomarkers of immunotherapy in non-small cell lung cancer. Cancer Med. 2021;10:2216–31. doi: 10.1002/cam4.3649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kristensen LK, Christensen C, Alfsen MZ, Cold S, Nielsen CH, Kjaer A. Monitoring CD8a(+) T cell responses to radiotherapy and CTLA-4 blockade using [(64) Cu] NOTA-CD8a PET imaging. Mol Imaging Biol. 2020;22:1021–30. doi: 10.1007/s11307-020-01481-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Negrao MV, Skoulidis F, Montesion M, Schulze K, Bara I, Shen V, et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J Immunother Cancer. 2021;9:e002891. doi: 10.1136/jitc-2021-002891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Seiwert TY, Foster CC, Blair EA, Karrison TG, Agrawal N, Melotek JM, et al. OPTIMA: A phase II dose and volume de-escalation trial for human papillomavirus-positive oropharyngeal cancer. Ann Oncol. 2019;30:297–302. doi: 10.1093/annonc/mdy522. [DOI] [PubMed] [Google Scholar]
- 156.Totzeck M, Schuler M, Stuschke M, Heusch G, Rassaf T. Cardio-oncology – Strategies for management of cancer-therapy related cardiovascular disease. Int J Cardiol. 2019;280:163–75. doi: 10.1016/j.ijcard.2019.01.038. [DOI] [PubMed] [Google Scholar]
- 157.Ryan TD, Nagarajan R, Godown J. Pediatric cardio-oncology: Development of cancer treatment-related cardiotoxicity and the therapeutic approach to affected patients. Curr Treat Options Oncol. 2019;20:56. doi: 10.1007/s11864-019-0658-x. [DOI] [PubMed] [Google Scholar]
- 158.Omer H, Sulieman A, Alzimami K. Risks of lung fibrosis and pneumonitis after postmastectomy electron radiotherapy. Radiat Prot Dosimetry. 2015;165:499–502. doi: 10.1093/rpd/ncv111. [DOI] [PubMed] [Google Scholar]
- 159.Boulet J, Peña J, Hulten EA, Neilan TG, Dragomir A, Freeman C, et al. Statin use and risk of vascular events among cancer patients after radiotherapy to the thorax, head, and neck. J Am Heart Assoc. 2019;8:e005996. doi: 10.1161/JAHA.117.005996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Gulati G, Heck SL, Ree AH, Hoffmann P, Schulz-Menger J, Fagerland MW, et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): A 2×2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur Heart J. 2016;37:1671–80. doi: 10.1093/eurheartj/ehw022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Raimondi C, Carpino G, Nicolazzo C, Gradilone A, Gianni W, Gelibter A, et al. PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system? Oncoimmunology. 2017;6:e1315488. doi: 10.1080/2162402X.2017.1315488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, et al. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight. 2018;3:122591. doi: 10.1172/jci.insight.122591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Wang YQ, Shen LJ, Wan JF, Zhang H, Wang Y, Wu X, et al. Short-course radiotherapy combined with CAPOX and PD-1 inhibitor for the total neoadjuvant therapy of locally advanced rectal cancer: The preliminary single-center findings of a prospective, multicentre, randomized phase II trial (TORCH) Zhonghua Wei Chang Wai Ke Za Zhi. 2023;26:448–58. doi: 10.3760/cma.j.cn441530-20230107-00010. [DOI] [PubMed] [Google Scholar]
- 164.Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 2016;1:15004. doi: 10.1038/sigtrans.2015.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wang L, Meng X, Li G, Zhou Q, Xiao J. Circular RNAs in cardiovascular diseases. Adv Exp Med Biol. 2018;1087:191–204. doi: 10.1007/978-981-13-1426-1_15. [DOI] [PubMed] [Google Scholar]
- 166.Lee MS, Liu DW, Hung SK, Yu CC, Chi CL, Chiou WY, et al. Emerging challenges of radiation-associated cardiovascular dysfunction (RACVD) in modern radiation oncology: Clinical practice, bench investigation, and multidisciplinary care. Front Cardiovasc Med. 2020;7:16. doi: 10.3389/fcvm.2020.00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Lin HY, Huang TH, Chan MW. Aberrant epigenetic modifications in radiation-resistant head and neck cancers. Methods Mol Biol. 2015;1238:321–32. doi: 10.1007/978-1-4939-1804-1_17. [DOI] [PubMed] [Google Scholar]
- 168.Oronsky B, Scicinski J, Kim MM, Cabrales P, Salacz ME, Carter CA, et al. Turning on the radio: Epigenetic inhibitors as potential radiopriming agents. Biomolecules. 2016;6:32. doi: 10.3390/biom6030032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Conte M, De Palma R, Altucci L. HDAC inhibitors as epigenetic regulators for cancer immunotherapy. Int J Biochem Cell Biol. 2018;98:65–74. doi: 10.1016/j.biocel.2018.03.004. [DOI] [PubMed] [Google Scholar]
- 170.Flower KJ, Ghaem-Maghami S, Brown R. Is there a role for epigenetic enhancement of immunomodulatory approaches to cancer treatment? Curr Cancer Drug Targets. 2018;18:5–15. doi: 10.2174/1568009617666170206105131. [DOI] [PubMed] [Google Scholar]
- 171.Palicelli A, Croci S, Bisagni A, Zanetti E, De Biase D, Melli B, et al. What do we have to know about PD-L1 expression in prostate cancer? A systematic literature review. Part 5: Epigenetic regulation of PD-L1. Int J Mol Sci. 2021;22:12314. doi: 10.3390/ijms222212314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Zhang C, Guo C, Li Y, Ouyang L, Zhao Q, Liu K. The role of YTH domain containing 2 in epigenetic modification and immune infiltration of pan-cancer. J Cell Mol Med. 2021;25:8615–27. doi: 10.1111/jcmm.16818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Li D, Zhao W, Zhang X, Lv H, Li C, Sun L. NEFM DNA methylation correlates with immune infiltration and survival in breast cancer. Clin Epigenetics. 2021;13:112. doi: 10.1186/s13148-021-01096-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Zeng XQ, Wang J, Chen SY. Methylation modification in gastric cancer and approaches to targeted epigenetic therapy (review) Int J Oncol. 2017;50:1921–33. doi: 10.3892/ijo.2017.3981. [DOI] [PubMed] [Google Scholar]
- 175.Gómez F, Gonzalez-Castaño DM, Fernández NG, Pardo-Montero J, Schüller A, Gasparini A, et al. Development of an ultra-thin parallel plate ionization chamber for dosimetry in flash radiotherapy. Med Phys. 2022;49:4705–14. doi: 10.1002/mp.15668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Clinicaltrials. Government. 2022. [[Last accessed n 2018 Oct 09]]. Available from: https://www.clinicaltrials.gov/ct2/home .
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.