Volume 44, Issue 7 pp. 739-760
REVIEW
Open Access

Beyond success: unveiling the hidden potential of radiotherapy and immunotherapy in solid tumors

Yuze Wu

Yuze Wu

Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China

Search for more papers by this author
Ming Yi

Ming Yi

Department of Breast Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China

Search for more papers by this author
Mengke Niu

Mengke Niu

Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China

Search for more papers by this author
Binghan Zhou

Binghan Zhou

Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China

Search for more papers by this author
Qi Mei

Corresponding Author

Qi Mei

Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China

Correspondence

Kongming Wu, Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, P. R. China.

Email: [email protected]

Qi Mei, Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, P. R. China.

Email: [email protected]

Search for more papers by this author
Kongming Wu

Corresponding Author

Kongming Wu

Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, P. R. China

Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China

Correspondence

Kongming Wu, Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, P. R. China.

Email: [email protected]

Qi Mei, Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, P. R. China.

Email: [email protected]

Search for more papers by this author
First published: 05 June 2024
Citations: 8

Abstract

Immunotherapy, particularly with immune checkpoint inhibitors, has significantly transformed cancer treatment. Despite its success, many patients struggle to respond adequately or sustain long-lasting clinical improvement. A growing consensus has emerged that radiotherapy (RT) enhances the response rate and overall efficacy of immunotherapy. Although combining RT and immunotherapy has been extensively investigated in preclinical models and has shown promising results, establishing itself as a dynamic and thriving area of research, clinical evidence for this combination strategy over the past five years has shown both positive and disappointing results, suggesting the need for a more nuanced understanding. This review provides a balanced and updated analysis of the combination of immunotherapy and RT. We summarized the preclinical mechanisms through which RT boosts antitumor immune responses and mainly focused on the outcomes of recently updated clinical trials, including those that may not have met expectations. We investigated the optimization of the therapeutic potential of this combined strategy, including key challenges, such as fractionation and scheduling, lymph node irradiation, and toxicity. Finally, we offered insights into the prospects and challenges associated with the clinical translation of this combination therapy, providing a realistic perspective on the current state of research and potential future directions.

List of abbreviations

  • ICI
  • immune checkpoint inhibitor
  • RT
  • radiotherapy
  • IMRT
  • intensity-modulated radiotherapy
  • SBRT
  • stereotactic body radiotherapy
  • ISABR
  • immunotherapy and stereotactic ablative radiotherapy
  • IGRT
  • image-guided radiotherapy
  • CTLA-4
  • cytotoxic T lymphocyte-associated protein 4
  • HNSCC
  • head and neck squamous cell carcinoma
  • NSCLC
  • non-small cell lung cancer
  • RIT
  • radioimmunotherapy
  • OS
  • overall survival
  • TME
  • tumor microenvironment
  • APC
  • antigen-presenting cell
  • TAA
  • tumor-associated antigen
  • DAMP
  • damage-associated molecular pattern
  • HMGB-1
  • high-mobility group box 1
  • ATP
  • adenosine triphosphate
  • DLN
  • draining lymph node
  • MHC
  • major histocompatibility complex
  • dsDNA
  • double-stranded DNA
  • cGAS
  • cyclic GMP-AMP synthase
  • cGAMP
  • cyclic GMP-AMP
  • TBK1
  • TANK-binding kinase 1
  • IRF3
  • interferon regulatory factor 3
  • IFN
  • interferon
  • TNF
  • tumor necrosis factor
  • TREX1
  • three prime repair exonuclease 1
  • MDSC
  • myeloid-derived suppressor cell
  • TAM
  • tumor associated macrophage
  • LPS
  • lipopolysaccharide
  • iNOS
  • inducible nitric oxide synthase
  • CSF1
  • macrophage colony-stimulating factor
  • SABR
  • stereotactic ablative radiotherapy
  • CR
  • complete response
  • BED
  • biologically effective dose
  • MPR
  • Major pathological response
  • HFRT
  • hypofractionated radiotherapy
  • ORR
  • overall response rate
  • PDAC
  • pancreatic ductal adenocarcinoma
  • NSCLC
  • non-small cell lung cancer
  • PD-1
  • programmed cell death protein 1
  • PD-L1
  • programmed death-ligand 1
  • DC
  • dendritic cell
  • MHC
  • major histocompatibility complex
  • NK
  • natural killer
  • Treg
  • regulatory T cell
  • LA
  • locally advanced
  • CRT
  • chemo- radiotherapy
  • cCRT
  • concurrent chemo- radiotherapy
  • sCRT
  • sequential chemo- radiotherapy
  • CFRT
  • conventionally fractionated radiotherapy
  • STING
  • stimulator of interferon genes
  • CXCL
  • C-X-C motif chemokine ligand
  • TIL
  • tumor infiltrating lymphocytes
  • Foxp3
  • forkhead box protein P3
  • TGF-β
  • transforming growth factor-β
  • CCL
  • C-C motif chemokine
  • CCR
  • C-C chemokine receptor type
  • YTHDF2
  • YT521B homology domain family 2
  • AE
  • adverse effects
  • TPS
  • tumor proportion score
  • SCLC
  • small cell lung cancer
  • LDHRT
  • low-dose hypofractionated radiotherapy
  • MSS
  • microsatellite stable
  • 1 BACKGROUND

    Immunotherapy, specifically immune checkpoint inhibitors (ICIs), offers a new paradigm for treating several solid tumors, including melanoma [1, 2], non-small cell lung cancer (NSCLC) [3, 4], and head and neck squamous cell carcinoma (HNSCC) [5]. However, most patients with cancer do not respond optimally to immunotherapy alone [6, 7]. Accordingly, combining immunotherapy with other established cancer treatments, including radiotherapy (RT), has garnered considerable attention [8-12].

    RT, a widely used and efficacious cancer treatment modality, can enhance both localized and systemic antitumor immune responses [13, 14]. RT has evolved considerably over time, driven by advancements in diagnostic imaging and delivery techniques. A pivotal leap occurred in the development of electron linacs in the 1960s and the 1970s. The evolution of RT techniques, including intensity-modulated RT, stereotactic body RT (SBRT), image-guided RT, and proton therapy, has ushered in a new era of precision radiation for solid tumors, with lower toxicities and higher conformality of the radiation fields targeting the tumor [14-18].

    The concept of radioimmunotherapy was first proposed in 2005, triggering many preclinical studies that explored the potential synergy between RT and immunotherapy [19]. However, evidence from relevant clinical trials is limited. In 2016, Bernstein et al. [20] introduced the definitive concept of immunotherapy and stereotactic ablative RT (ISABR). In 2018, they further advocated comprehensive irradiation of multiple lesions in the ISABR field [21] (Figure 1).

    Details are in the caption following the image
    Timeline depicting important events in the development of the combination of RT and immunotherapy. Abbreviations: ICI, immune checkpoint inhibitor; IMRT, intensity-modulated radiotherapy; ISABR, immunotherapy and stereotactic ablative RT; NSCLC, non-small cell lung cancer; RT, radiotherapy.

    First, a secondary analysis of the KEYNOTE-001 trial (NCT01295827) provided intriguing insights into this combination strategy at the clinical trial level. The primary objective of the phase I KEYNOTE-001 trial was to assess the safety and antitumor activity of pembrolizumab (an anti-programmed cell death protein 1 [anti-PD-1] antibody) in patients with advanced NSCLC [22]. Shaverdian et al. [23] assessed patients with advanced NSCLC who had received RT before pembrolizumab treatment. They found that the overall survival (OS) and progression-free survival (PFS) were significantly longer in patients who had previously received RT than in those who had not, with an acceptable safety profile. This benefit was observed despite the significant interval of 9.5 months between RT and pembrolizumab treatment. Another major milestone occurred when Antonia et al. [24] found that in the PACIFIC trial, patients who started durvalumab (anti-programmed death-ligand 1 [anti-PD-L1] antibody) within 2 weeks after completing chemo-RT (CRT) survived longer than those who started durvalumab at 4 weeks. The PACIFIC trial, a randomized phase III trial (NCT02125461), enrolled patients with stage III unresectable NSCLC who received at least 2 cycles of platinum-based CRT. These patients were then assigned to receive durvalumab or placebo. This trial demonstrated improved OS and PFS in patients with NSCLC receiving durvalumab post-CRT [24, 25]. The latest analyses demonstrated robust and sustained OS and durable PFS benefits [26]. PACIFIC-R (NCT03798535) is a large, real-world, retrospective study of patients who received the PACIFIC regimen. Better real-world PFS outcomes were also observed among patients who received durvalumab closer to the end of RT, which is consistent with the findings from the PACIFIC trial [27]. These findings provide compelling evidence supporting the potential of RT to elicit a systemic antitumor immune response, inducing an abscopal effect. However, certain factors, including PD-1/PD-L1, impede RT-induced abscopal effects, highlighting the role of ICIs in enhancing the efficacy of RT.

    The purpose of this review is to provide a comprehensive and balanced examination of the combination of RT and immunotherapy in cancer treatment. We aim to present an overview of the numerous clinical trials, such as the SPRINT, DOLPHIN, PEMBRO-RT, and MDACC trials, which have released their findings in the past five years. Our goal is to shed light on the successes and setbacks of these trials, highlighting the need for a nuanced understanding of this combination therapy. In addition to summarizing the preclinical mechanisms that enhance antitumor immune responses through RT, our objective extends to exploring the potential optimization of this combined strategy, including challenges such as fractionation and scheduling, lymph node irradiation, and toxicity management. Ultimately, this review seeks to provide insights into the potential and hurdles of translating this combination therapy into clinical practice, offering a realistic view of the current state of research and possible future directions.

    2 MECHANISMS AND PRECLINICAL EVIDENCE

    Several reviews have discussed the preclinical mechanisms of synergy between RT and immunotherapy [28-30]. In this section, we present an updated summary of recent preclinical studies on the impact of RT on the immune system via in situ vaccination and immune reprogramming.

    2.1 In situ vaccination

    The immune mechanisms triggered by RT encompass three essential processes: immunogenic cell death and arousal of antigen-presenting cells, T cell priming in lymph nodes, and effector T cells homing to tumors [31, 32] (Figure 2). RT-damaged tumor cells release various tumor-associated antigens and damage-associated molecular patterns (DAMPs), including high-mobility group box 1 [33, 34] and adenosine triphosphate [35, 36]. Increased DAMPs activate dendritic cells (DCs) and trigger the MyD88 pathway, inducing a cascade of cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), IL-6, and IL-8 [37, 38]. Furthermore, RT enhances the expression of calreticulin on the cell surface, acting as an “eat me” signal [39-41], and upregulates the expression of major histocompatibility complex (MHC) class I on tumor cells [42, 43]. RT enhances antigen cross-presentation within draining lymph nodes (DLNs) [43, 44]. During this process, activated DCs migrate to DLNs, where they present antigens to T cells [45]. After education, T cells, mainly CD8+ T cells, leave the DLNs and circulate throughout the body, patrol for tumor antigens, and target both irradiated and non-irradiated tumor deposits, thereby promoting the regression of distant tumors, an intriguing phenomenon known as the abscopal response [46-49].

    Details are in the caption following the image
    In-situ vaccination induced by RT. In-situ vaccination induced by RT encompasses three key procedures: exposure of tumor antigen and activation of APCs, T cell priming in lymph nodes, and effector T cells home to tumors. Abbreviations: APCs, antigen-presenting cells; DAMP, damage-associated molecular pattern molecules; DCs, dendritic cells; HMGB-1, high-mobility group box 1; IFN-β, interferon-β; MHC, major histocompatibility complex; RT, radiotherapy; STING, stimulator of interferon genes; TNF-α, tumor necrosis factor-α;.

    Cellular responses after RT are intricate and multifaceted, involving various signaling pathways that affect immune system function. There is a recurring consensus that non-tumor cell stimulator of interferon genes (STING) is a crucial factor [50, 51] (Figure 3). Radiation-generated cytoplasmic double-stranded DNA fragments trigger cyclic GMP-AMP (cGAMP) synthase activation, leading to the synthesis of the secondary messenger cGAMP [52]. This, in turn, recruits TANK-binding kinase 1 and IκB kinase [53, 54], initiating the transcription of inflammatory cytokines, especially interferon-β (IFN-β) [50, 55]. Notably, Vanpouille-Box et al. [56] showed that RT ranging from 12 Gy to 18 Gy activates three prime repair exonuclease 1 (TREX1) within tumor cells, thereby orchestrating the degradation of radiation-induced cytoplasmic double-stranded DNA.

    Details are in the caption following the image
    The mechanism of radiation-induced activation of the cGAS-STING pathway. Radiation-generated cytoplasmic dsDNA fragments trigger cGAS activation. Then, cGAS dimerizes and synthesizes the dinucleotide secondary messenger cGAMP, which binds to the STING at the ER. STING oligomerize and translocate to the Golgi, which involves in COP complexes. In the ER-Golgi intermediate compartment, STING recruits TBK1 and IKK. Together with STING, TBK1 is allowed to co-activate the IRF3 by phosphorylation. The phosphorylated IRF3 polymerizes and translocate to the nucleus to induce the expression of IFN-β, which are crucial for therapeutic responses to RT in immune cells. In addition, IKK activates NF-κB pathway and subsequently promotes transcription of inflammatory cytokines IL-6, IFN-β, TNF-α, and IL-1β. Abbreviations: IFN-β, interferon-β; RT, radiotherapy; STING, stimulator of interferon genes; TNF-α, tumor necrosis factor-α; TREX1, three prime repair exonuclease 1.

    2.2 Immune reprogramming

    RT also induces immune reprogramming by dynamically altering the immune milieu in response to treatment [57]. This causes the release of chemokines, including C-X-C motif chemokine ligand 9 (CXCL9) [58], CXCL10 [58, 59], and CXCL16 [60, 61], leading to the infiltration and accumulation of immune cells and reprogramming of the tumor microenvironment (TME) [62-64]. DCs [46], CD8+ T cells [65], natural killer (NK) cells [66], as well as regulatory T cells (Tregs) [67, 68] and myeloid-derived suppressor cells (MDSCs) [69], are involved in this process (Figure 4).

    Details are in the caption following the image
    RT reprogrammed the tumor microenvironment. RT induces the upregulation of MHC class I and NK2GD, thereby promoting a potent cytotoxic response from CD8+ T cells and NK cells. RT on DCs increases the expression of CCL19 and CCL21 and mediate migration of DCs. RT induced macrophage differentiation into M2 phenotype. RT increased Tregs through release of adenosine by tumor cells as well IL-10. Moreover, RT recruited MDSCs via the CCR2 pathway. Abbreviations: CCL, C-C motif chemokine; CCR, C-C chemokine receptor type; DC, dendritic cell; IFN-β, interferon-β; MDSC, myeloid-derived suppressor cell; MHC, major histocompatibility complex; NK cell, natural killer cell; RT, radiotherapy; TAM, tumor-associated macrophage; TGF-β, transforming growth factor-β; Treg, regulatory T cell.

    Immune cells exhibit differential sensitivities to radiation. For instance, immature DCs can tolerate radiation doses of 10 Gy to 30 Gy, while retaining their viability and functionality. However, mature DCs, which are crucial for immune activation, may be more radiation-sensitive [70]. Tregs are more resistant to RT than other lymphocytes [71, 72]. Recently, irradiated DCs were found to decrease the secretion of IL-12 and IL-23 cytokines, a reduction, in turn, mitigated by irradiated fibroblasts [73].

    Significant CD4+ T helper 1 and CD8+ T cytotoxic 1 polarization was observed in tumor DLNs after RT [74]. To delve deeper into the underlying potentiation of RT and ICIs, Rudqvist et al. [75] identified the separate contributions of each therapy (RT and anti-cytotoxic T-lymphocyte-associated antigen 4 [anti-CTLA-4] antibody) to the T cell population. They found that the anti-CTLA-4 antibody expanded CD4+ T helper 1 cells and RT expanded exhausted CD8+ T cells. However, in the combination group, Tregs were reduced while CD8 effector memory, early activation, and precursor exhausted T cells were expanded compared to those in the control and monotherapy groups.

    NK cells are cytotoxic innate lymphoid cells essential for the innate immunosurveillance of tumors [76, 77]. NK cells were activated by irradiated tumor cells and could regulate the response to RT and CTLA-4 blockade [66, 78]. Combining RT with the adoptive transfer of NK cells has been shown to prolong survival compared with that of RT alone [79].

    Tumor-associated macrophages (TAMs) are the most abundant tumor-infiltrating lymphocytes in the TME [80-83]. TAMs are phenotypically and functionally diverse and can be broadly divided into two types: pro-inflammatory M1 and anti-inflammatory M2 macrophages [84-87]. RT can shift macrophage differentiation to the M1 phenotype, indirectly increasing tumor infiltrating lymphocytes (TIL) frequency [88, 89]. Moreover, depletion of TAMs can reverse immunosuppression and promote RT efficacy [90].

    Tregs, characterized by high forkhead box protein P3 (Foxp3) and CD25 expression, inhibit antitumor immunity, thereby promoting tumor development [91-93]. Different doses of RT can increase the expression of CTLA‑4 on Tregs as well as the level of transforming growth factor-β (TGF-β) secreted by Tregs [94]. Moreover, in a murine model of HNSCC, RT upregulated C-C motif chemokine 2 (CCL2) chemokine production in tumor cells, resulting in the C-C chemokine receptor type 2 (CCR2)-dependent accumulation of CCR2+ Tregs. This reduces the efficacy of RT [95]. Treg depletion combined with RT can significantly enhance immune-promoting effects, as well as reduce tumor burden and improve OS [44, 96, 97].

    MDSCs are a cluster of cells with immunosuppressive effects that are classified into 2 distinct subsets: polymorphonuclear and monocytic MDSCs [98-100]. RT induces MDSC expansion and recruitment in murine models and humans [69]. Interestingly, a transient significant increase in the percentage of MDSCs was observed 3 days after RT, which then decreased at day 14 after RT [101]. The upregulation of CCL2, CCL7, and CCL12 after high-dose radiation (20 Gy) led to the accumulation of CCR2+ MDSC in the TME [102]. The CCR2 blockade abrogates RT-induced MDSCs [102, 103]. A recent study found that RT-induced YT521B homology domain family 2 (YTHDF2) expression and YTHDF2 deficiency reversed the accumulation of MDSC following local RT, improving the effects of combined RT and/or anti-PD-L1 treatment [104].

    In summary, while RT-recruited immunosuppressive cells can potentially hinder immune-stimulatory responses, it has been observed that RT can ultimately increase the effector-to-suppressor cell ratio in the TME [33]. This, combined with the upregulation of PD-L1 expression induced by RT, suggests that combining RT with ICIs could be a promising strategy for treating solid tumors [105-108] (Table 1).

    TABLE 1. Preclinical studies of RT in combination with immunotherapy.
    Preclinical study Tumor model RT Immunotherapy Results
    Chen et al., 2021 [182] Lewis lung carcinoma 4 fractions of 10 Gy anti-PD-L1 Suppressed the tumor growth, improved survival, and prolonged immune memory
    Hong et al., 2020 [183] MFC gastric cancer 3 fractions of 5 Gy anti-PD-1 Improved antitumor activities in RT-insensitive tumor models
    Philippou et al., 2020 [184] TRAMP-C1 and MyC-CaP prostate cancer 3 fractions of 5 Gy anti-PD-L1 Tumor growth delay and immune cell changes
    Grapin et al., 2019 [185] Subcutaneous CT26 colorectal carcinoma 3 fractions of 8 Gy, 2 fractions of 18 Gy and 1 fraction of 16.4 Gy anti-PD-L1, anti-TIGIT 3 fractions of 8 Gy were the most effective protocol
    Oweida et al., 2018 [186] MOC2 and LY2 squamous cell carcinoma 1 fraction of 20 Gy, 3 fractions of 8 Gy anti-PD-L1, anti-TIM-3 Targeting Tregs enhances therapeutic response to RT and dual immune checkpoint blockade
    Rodriguez-Ruiz et al., 2017 [187]

    Subcutaneous

    MC38 colon adenocarcinoma

    3 fractions of 8 Gy anti-PD1, anti-CD137 or both Brachytherapy with immunotherapy can potentiate the abscopal effect
    Young et al., 2016 [188] Subcutaneous CT26 colorectal carcinoma 1 fraction of 20 Gy anti-CTLA-4 and anti-TNFRSF4 Ideal timing of RT is dependent on the mechanism of action of the respective immunotherapy utilized
    Hao et al., 2016 [189] Subcutaneous LLC 1 fraction of 6 Gy anti-CD40 RT combined with anti-CD40 boosts the abscopal effect
    Habets et al., 2016 [190] Subcutaneous 67NR mammary carcinoma 3 fractions of 8 Gy Flt3L HFRT with Flt3L induces abscopal effect
    Twyman-Saint et al., 2015 [191] Subcutaneous TSA and B16-F10 3 fractions of 8 Gy or 1 fraction of 20 Gy anti-CTLA-4, anti-PD-1, and anti-PD-L1 Anti-CTLA-4 decreased Tregs, anti-PD-L1 reinvigorates exhausted T cells, and RT increases the TCR repertoire
    Deng et al., 2014 [105] Subcutaneous TUBO mammary carcinoma and MC38 1 fraction of 12 Gy anti-PD-L1

    Increased T-cell infiltration boosts

    abscopal effect

    Lee et al., 2009 [45] Subcutaneous 4T1 and B16-CCR7 2 fractions of 12 Gy TNFSF14 Increased abscopal effect was CD8+ T cell-mediated
    Dewan et al., 2009 [192] Subcutaneous 4T1 and MC38 5 fractions of 6 Gy, 3 fractions of 8 Gy, or 1 fraction of 20 Gy anti-CTLA-4 Fractionated but no single fraction RT with immunotherapy induced abscopal effect
    Demaria et al., 2005 [193] Subcutaneous 4T1 mammary carcinoma 2 fractions of 12 Gy anti-CTLA-4 A promising new strategy against poorly immunogenic metastatic cancers
    Demaria et al., 2004 [194] Subcutaneous 67NR 1 fraction of 2-6 Gy Flt3L Synergism through T cell-mediated abscopal effect
    Chakravarty et al., 1999 [195] Subcutaneous LLC 60 Gy Flt3L Improved survival and reduced pulmonary metastases
    • Abbreviations: CTLA-4, cytotoxic T-lymphocyte associated protein 4; Flt3L, Flt3 ligand; HFRT, hypofractionated radiotherapy; LLC, Lewis lung carcinoma; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; RT, radiation therapy; TCR, T cell receptor; TIGIT, T cell immunoreceptor with immunoglobulin and ITIM domain; TIM-3, T cell immunoglobulin and mucin domain-3; TNFRSF4, TNF receptor superfamily member 4; Treg, regulatory T cell.

    3 EVIDENCE FROM CLINICAL TRIALS

    Previous case reports and limited clinical trials have demonstrated promising results when combining RT with immunotherapy [109-112]. Despite the recent disclosure of the results of several clinical trials, the future of this field remains uncertain. SBRT, also known as SABR, is the most commonly used treatment in clinical trials [113]. In this section, we discussed the results of clinical trials and findings revealed over the past five years, focusing primarily on NSCLC, where the most promising results have been observed. We also summarized the relevant clinical trials combining RT with ICIs, with a predominant emphasis on NSCLC (Table 2) and other solid tumors (Table 3).

    TABLE 2. Clinical trials combining RT with immunotherapy in NSCLC.
    Condition NCT number Status Phase Immunotherapy RT (dose and fractionation) Timing and sequencing
    Early-stage NSCLC NCT03383302 Unknown Phase I/II Nivolumab SBRT (N/A) RT prior to immunotherapy
    Early-stage NSCLC NCT04271384 Recruiting Phase II Nivolumab SBRT (N/A) Concurrent
    Stage I NSCLC NCT03446547 Unknown Phase II Durvalumab SBRT (N/A) RT prior to immunotherapy
    Stage I NSCLC NCT04944173 Not yet recruiting Phase II Durvalumab SBRT (N/A) Concurrent
    Stage I-II NSCLC NCT03383302 Unknown Phase I/II Nivolumab SBRT (N/A) RT prior to immunotherapy
    Stage I-IIA NSCLC NCT03110978 Active, not recruiting Phase II Nivolumab SBRT (N/A) Concurrent
    Stage III NSCLC NCT05157542 Recruiting Phase I Durvalumab Low dose RT (N/A) Concurrent
    Stage II-III NSCLC NCT04310020 Recruiting Phase II Atezolizumab HFRT (N/A) RT prior to immunotherapy
    Metastatic NSCLC NCT03168464 Completed Phase I/II Ipilimumab/nivolumab SBRT (30 Gy in 6 fractions) Concurrent
    Stage IV NSCLC NCT01860430 Completed Phase I Pembrolizumab Palliative EBRT (N/A) Concurrent
    Stage IV NSCLC NCT02303990 Completed Phase I Pembrolizumab SBRT (N/A) Concurrent
    Locally advanced or metastatic NSCLC NCT05111197 Recruiting Phase III anti-PD-1 or anti-PD-L1 SBRT (24 Gy in 3 fractions) Concurrent
    Advanced NSCLC NCT02839265 Completed Phase II FLT3 ligand (CDX-301) SBRT (N/A) Concurrent
    Advanced NSCLC NCT04491084 Recruiting Phase I/II FLT3 ligand (CDX-301) and anti-CD40 antibody (CDX-1140) SBRT (N/A) Concurrent
    Metastatic NSCLC NCT03825510 Completed Phase II Nivolumab/Pembrolizumab SBRT (N/A) RT prior to immunotherapy
    Stage III NSCLC NCT04577638 Active, not recruiting Phase II Nivolumab IMRT (N/A) Concurrent
    Stage IV NSCLC NCT03867175 Active, not recruiting Phase III Pembrolizumab SBRT (N/A) Concurrent
    Stage IV NSCLC NCT02608385 Active, not recruiting Phase I Pembrolizumab SBRT (N/A) Concurrent
    Stage IV NSCLC NCT02221739 Completed Phase I/II Ipilimumab IMRT (30 Gy in 5 fractions) Concurrent
    Metastatic NSCLC NCT03224871 Completed Phase I IL-2 HFRT (N/A) Concurrent
    NSCLC NCT03217071 Completed Phase II Pembrolizumab SBRT (a single 12 Gy dose) Immunotherapy prior to RT
    Metastatic NSCLC NCT02463994 Completed Early phase I MPDL3280A (Anti-PD-L1) IMRT (N/A) RT prior to immunotherapy
    Stage IV NSCLC NCT03812549 Completed Phase I Sintilimab (Anti-PD-1) SBRT (N/A) Immunotherapy prior to RT
    Stage IIIb or Stage IV NSCLC NCT00879866 Completed Phase I EMD 521873 (immunocytokine) RT (20 Gy in 5 fractions) RT prior to immunotherapy
    NSCLC NCT02221739 Completed Phase I/II Ipilimumab IMRT (N/A) Concurrent
    Metastatic NSCLC NCT03158883 Completed Early phase I Avelumab SBRT (N/A) N/A
    Stage IIIa/b NSCLC NCT02434081 Completed Phase II Nivolumab RT (66 Gy in 33 fractions) Concurrent

    Advanced NSCLC

    (PEMBRO-RT)

    NCT02492568 Completed Phase II Pembrolizumab SBRT (24 Gy in 3 fractions) RT prior to Immunotherapy
    Stage IV NSCLC NCT04929041 Recruiting Phase II/III ICIs SBRT (N/A) Concurrent
    Stage IV NSCLC NCT03223155 Active, not recruiting Phase I Ipilimumab/nivolumab SBRT (N/A) Concurrent and sequential
    Metastatic colorectal cancer or NSCLC NCT02888743 Active, not recruiting Phase II Tremelimumab and durvalumab High/low dose RT (N/A) Immunotherapy prior to RT
    Metastatic or locally advanced NSCLC NCT05000710 Recruiting Phase II Tremelimumab and durvalumab Low dose RT (N/A) Concurrent
    NSCLC with brain metastases NCT04889066 Not yet recruiting Phase II Durvalumab SBRT (N/A) Concurrent
    NSCLC NCT02444741 Active, not recruiting Phase I/II Pembrolizumab SBRT (N/A) or conventional RT (N/A) Concurrent
    NSCLC (KEYNOTE-799) NCT03631784 Active, not recruiting Phase II Pembrolizumab RT (60 Gy in 30 daily fractions) Concurrent
    Stage II/III NSCLC NCT04013542 Recruiting Phase I Ipilimumab and nivolumab RT (N/A) Concurrent
    Stage IV NSCLC NCT03705403 Recruiting Phase II L19-IL2 SBRT (N/A) Concurrent
    Metastatic NSCLC NCT05034055 Not yet recruiting Phase II Atezolizumab/tiragolumab SBRT (N/A) RT prior to immunotherapy
    • Abbreviations: SBRT, stereotactic body radiation therapy; RT, radiation therapy; EBRT, external beam radiation therapy; NSCLC, non-small cell lung cancer; IMRT, intensity modulated radiation therapy; L19-IL2, human recombinant scFv fragment directed against fibronectin containing extra domain, designated L19, combined with the pro-inflammatory interleukin-2; CIRT, carbon ion radiotherapy; HFRT, hypofractionated radiotherapy; HDCRT, high-dose conformal radiotherapy; ICI, immune checkpoint inhibitor; N/A, not applicable.
    TABLE 3. Clinical trials about the combination of RT and immunotherapy in other solid tumors.
    Condition NCT number Status Phase Immunotherapy RT (dose and fractionation) Timing and sequencing
    Metastatic castration-resistant prostate cancer NCT01807065 Completed Phase II Sipuleucel-T SBRT (N/A) RT prior to immunotherapy
    Metastatic castration-resistant prostate cancer NCT01818986 Completed Phase II Sipuleucel-T SBRT (N/A) Concurrent
    Glioblastoma NCT02313272 Completed Phase I/II Pembrolizumab EBRT (N/A) Concurrent
    Glioblastoma NCT02968940 Completed Phase II Avelumab HFRT (30 Gy in 5 fractions) N/A
    Advanced melanoma NCT01497808 Completed Phase I Ipilimumab Palliative SBRT (N/A) RT prior to immunotherapy
    Advanced melanoma NCT01449279 Completed Phase I Ipilimumab Palliative EBRT (N/A) Immunotherapy prior to RT
    Advanced melanoma NCT01689974 Terminated Phase II Ipilimumab SBRT (30Gy in 5 fractions) Immunotherapy starts day 4 of RT
    Advanced melanoma NCT01557114 Terminated Phase I Ipilimumab EBRT (dose escalation, 9, 15, 18, 24 Gy in 3 fractions) RT starts week 4 of immunotherapy
    Advanced melanoma NCT02406183 Completed Phase I Ipilimumab SBRT (N/A) Immunotherapy prior to RT

    Melanoma with

    brain metastases

    NCT01703507 Completed Phase I Ipilimumab SBRT (N/A) or whole brain EBRT (N/A) Concurrent

    Melanoma with

    brain metastases

    NCT02115139 Completed Phase II Ipilimumab Whole brain EBRT (30 Gy in 10 fractions) Concurrent
    Melanoma with brain metastases NCT02097732 Terminated Phase II Ipilimumab SBRT (N/A) Immunotherapy prior to RT
    Stage III-IVb HNSCC NCT01935921 Completed Phase I Ipilimumab IMRT (N/A) Concurrent
    HNSCC NCT04220775 Completed Phase I/II Bintrafusp alfa SBRT (N/A) Immunotherapy prior to RT
    HNSCC NCT02684253 Completed Phase II Nivolumab SBRT (27 Gy in 3 fractions) Concurrent
    Metastatic clear cell renal cell carcinoma NCT01896271 Completed Phase II IL-2 (proleukin) SBRT (N/A) Concurrent
    Renal cell carcinoma NCT03065179 Completed Phase II Nivolumab/ipilimumab SBRT (N/A) Concurrent
    Metastatic urothelial cancer NCT02826564 Completed Phase I Pembrolizumab SBRT (N/A) Concurrent and sequential
    Soft tissue sarcoma NCT01347034 Completed Phase II Autologous DC intra-tumoral vaccination EBRT (N/A) Concurrent

    Stage IV soft tissue sarcoma

    NCT02180698 Completed Phase I TLR4 Palliative EBRT (N/A) Concurrent
    Metastatic breast cancer NCT01862900 Completed Phase I MEDI6469 (anti-OX40) SBRT (3 dose escalation cohorts of 15 Gy, 20 Gy or 25 Gy to liver or lung metastases) Concurrent
    Metastatic breast cancer NCT01421017 Completed Phase I/II TLR7 imiquimod SBRT (6 Gy in 5 fractions) Concurrent
    Advanced solid tumors with liver and lung metastases NCT02239900 Completed Phase I Ipilimumab

    SBRT (50 Gy in 4 fractions to 1-4 lesion[s])

    Immunotherapy prior to RT
    Recurrent or metastatic solid tumors NCT02318771 Unknown Phase I Pembrolizumab SBRT (N/A) Concurrent
    Metastatic colorectal cancer NCT02437071 Active, not recruiting Phase II Pembrolizumab IMRT (N/A) Concurrent

    Colorectal cancer with

    liver Metastases

    NCT03101475 Completed Phase II Durvalumab SBRT (10 Gy in 3 fractions) RT starts day 8 of immunotherapy
    Locally advanced rectal cancer NCT04663763 Not yet recruiting Phase II Sintilimab(anti-PD-1)/capecitabine/Oxaliplatin Shor-course RT (25 Gy in 5 fractions) RT prior to immunotherapy
    Penile cancer NCT03686332 Active, not recruiting Phase II Atezolizumab RT (33 fractions of 1.5 Gy or 1.8 Gy) Concurrent
    Metastatic anaplastic thyroid cancer NCT03122496 Completed Phase I Durvalumab/tremelimumab SBRT (27 Gy in 3 fractions) Immunotherapy prior to RT
    Solid tumors NCT02086721 Completed Phase I L19-IL2 (immunocytokine) SBRT (N/A) Concurrent
    Solid tumors NCT05097781 Recruiting Phase II Anti-PD-1 antibody RT (N/A) N/A
    Solid tumors NCT05229614 Recruiting Phase II Pembrolizumab CIRT (N/A) Immunotherapy prior to RT
    Solid tumors NCT02239900 Completed Phase I Ipilimumab SBRT (N/A) Concurrent and sequential
    Solid tumors NCT03220854 Completed Phase II Anti-PD-1/PD-L1 antibody SBRT (N/A) N/A
    Solid tumors NCT02987166 Completed Phase I Pembrolizumab HDCRT (N/A) Concurrent and sequential
    Solid tumors NCT02474186 Completed Phase I/II GM-CSF RT (35 Gy in 10 fractions) Concurrent
    Solid tumors NCT03313804 Recruiting Phase II ICIs SBRT (N/A) Immunotherapy prior to RT
    • Abbreviations: CIRT, carbon ion radiotherapy; DC, dendritic cell; EBRT, external beam radiation therapy; GM-CSF, granulocyte-macrophage colony-stimulating factor; HDCRT, high-dose conformal radiotherapy; HFRT, hypofractionated radiotherapy; HNSCC, head and neck squamous cell carcinoma; ICI, immune checkpoint inhibitor; IMRT, intensity modulated radiation therapy; L19-IL2, human recombinant scFv fragment directed against fibronectin containing extra domain, designated L19, combined with the pro-inflammatory interleukin-2; mCRPC, metastatic castration-resistant prostate cancer; N/A, not applicable; RT, radiation therapy; SBRT, stereotactic body radiation therapy.

    3.1 Early-stage NSCLC

    The RTOG 0236 trial showed that SBRT achieved a high rate of tumor control in patients with medically inoperable early-stage NSCLC [114]. These patients are typically treated with definitive SBRT as the standard of care [115]. However, a longer follow-up period revealed additional cancer recurrences [116, 117]. Preclinical evidence suggests that ICIs combined with SBRT may facilitate preoperative immunotherapy without compromising antitumor efficacy, making it a safer option for neoadjuvant therapy [118].

    An open-label phase II trial (NCT02904954) compared durvalumab alone with durvalumab plus SBRT (3 × 8 Gy) for early-stage NSCLC [119]. Major pathological response (MPR) rates were significantly higher in the durvalumab plus SBRT group (16/30 patients) than in the durvalumab alone group (2/30 patients). Notably, half of the patients in the dual therapy group with MPR showed complete pathological response [119].

    A recent open-label phase II trial (NCT03110978) evaluated the efficacy and safety of SBRT alone and in combination with nivolumab for early-stage NSCLC. Compared with that of the SBRT group, the combination therapy group showed significantly improved four-year event-free survival with tolerable toxicity [120]. ISABR (NCT03148327), a multicenter prospective clinical trial, recently reported the safety and efficacy of the combination of SBRT and durvalumab in 18 patients [121]. These results suggest that pulmonary toxicity risk is the greatest concern for combination therapy [121].

    3.2 Oligometastases

    The concept of an oligometastatic state was first postulated in 1995 [122]. This hypothesis posits that metastases may be limited to specific organs in limited numbers [123], implying potential curability with localized interventions, including RT or surgery [124]. Earlier evidence supports the safety and efficacy of SBRT for oligometastases [125]. In limited metastatic NSCLC, SBRT before maintenance chemotherapy significantly improved PFS compared with maintenance chemotherapy alone [126]. The SABR-COMET phase II trial showed that SBRT improved OS [127].

    Harnessing the innate and adaptive immunity is vital for restricting metastatic development [128-130]. Pitroda et al. [131] identified that the upregulation of immune-related genes in colorectal liver metastases was associated with better clinical outcomes. Accordingly, the integration of immunotherapy with SBRT has been proposed for oligometastases [132] (Table 4). Luke et al. [133] conducted a phase I trial and found that pembrolizumab and SBRT combination therapy had an acceptable safety profile. A subsequent clinical study (NCT02316002) showed improved PFS with reduced quality of life after SBRT for oligometastatic NSCLC [134].

    TABLE 4. Clinical trials about the combination of RT and immunotherapy in oligometastatic.
    Condition NCT number Status Phase Immunotherapy RT (dose and fractionation) Timing and sequencing
    Melanoma NCT01565837 Unknown Phase II Ipilimumab SBRT (N/A) Immunotherapy prior to RT
    Melanoma NCT01416831 Active, not recruiting Phase II IL-2 SBRT (N/A) Concurrent
    Melanoma NCT02107755 Unknown Phase II Ipilimumab SBRT (N/A) SBRT starts 5 weeks after first dose of ipilimumab
    Solid tumors NCT05259319 Not recruiting Phase I Atezolizumab and tiragolumab SBRT (24 Gy in 3 fractions) Sequential
    NSCLC NCT03275597 Terminated Phase I Durvalumab and tremelimumab SBRT (N/A) RT prior to immunotherapy
    NSCLC NCT03965468 Active, not recruiting Phase II Durvalumab SBRT (N/A) Concurrent
    NSCLC NCT04549428 Recruiting Phase II Atezolizumab RT (a single fraction of 8 Gy) Concurrent
    NSCLC NCT04238169 Recruiting Phase II Toripalimab SBRT (N/A) Concurrent
    • Abbreviations: IL-2, interleukin-2; N/A, not applicable; NSCLC, non-small cell lung cancer; RT, radiation therapy; SBRT, stereotactic body radiation therapy.

    The results from a randomized phase II trial have established the use of SBRT to all lesions, becoming the standard of care for patients with oligometastatic NSCLC [135]. A multicenter prospective observational study aimed to determine whether concomitant anti-PD-1 and SABR could enhance tumor response in metastatic NSCLC and melanoma. In this study, all patients received concurrent pembrolizumab or nivolumab and SABR to 1 to 5 lesions, with the anti-PD-1 treatment continuing until further progression, unacceptable toxicity, or a medical/patient decision to discontinue. The objective response rate (ORR) was 42%, and the median PFS was 14.2 months [136]. This approach achieved high response rates and extended the clinical benefits of immunotherapy by delaying further progression and developing a new systemic therapy.

    3.3 Locally advanced (LA)-NSCLC

    Approximately one-third of patients with NSCLC are initially diagnosed with LA disease [137]. Based on the finding of the PACIFIC trial [24, 25], concurrent CRT (cCRT) followed by consolidation durvalumab (the PACIFIC regime) became the standard of care for patients with LA-NSCLC. With updated suboptimal 5-year OS and PFS rates [26], novel treatment strategies are under investigation to improve clinical outcomes. One such approach was the GEMSTONE-301 phase III trial (NCT03728556) [138] and the PACIFIC-6 phase II trial (NCT03693300) [139]. Both trials demonstrated that ICI after sequential CRT (sCRT) is an effective consolidation therapy for LA-NSCLC, suggesting that sCRT followed by ICI could be an alternative for patients unsuitable for the PACIFIC regimen [140].

    Trials exploring the use of ICIs concurrently with cCRT or sCRT have also been conducted. KEYNOTE-799 is a nonrandomized phase II trial of pembrolizumab concurrent with cCRT as the initial therapy for the treatment of LA-NSCLC, with an ORR of 70.5% [141]. The PACIFIC-2 trial (NCT03519971) randomized patients to receive durvalumab or a placebo concurrently with CRT. On November 14, 2023, the news that durvalumab administered concurrently with CRT failed to achieve statistical significance for PFS compared with CRT alone was announced [142]. The failure of the PACIFIC-2 trial could be attributed to several factors, including the toxicity caused by immature treatment regime. Thus, the optimal regime of RT still requires further exploration and research.

    Furthermore, the induction of ICIs before CRT in patients with LA-NSCLC is currently being explored in clinical trials. The prospective AFT-16 study (NCT03102242) evaluated the safety and efficacy of atezolizumab before CRT. The primary endpoint of the disease control rate at 12 weeks was 77.4% [143]. Recently, the analysis of secondary endpoints was updated. The median PFS was 23.7 months. The median OS is not yet estimable [144]. Owing to the encouraging PFS and OS rates without unexpected safety signals, further studies are warranted. In a retrospective study, patients with LA-NSCLC received standard of care or induction ICIs, followed by standard of care. Although the OS and PFS rates between the 2 groups were similar, the induction ICIs group had a significantly lower distant metastasis rate [145].

    For patients unable to complete the PACIFIC regimen due to chemotherapy-induced adverse effects (AEs), combining RT with immunotherapy can reduce toxicity while maintaining survival [146]. The SPRINT study (NCT03523702) is a prospective phase II trial that tested sequential pembrolizumab and RT. Patients with LA-NSCLC (n = 25) having PD-L1 tumor proportion score (TPS) ≥ 50% were enrolled. The primary endpoint was PFS. The actuarial 1-year PFS rate was 74% and the actuarial 1-year OS rate was 95% [147]. The promising initial results suggest that sequential pembrolizumab and RT may be an alternative treatment approach for patients in this setting. Further clinical trials should be designed to optimize the SPRINT regimen and compare it with the standard of care. The DOLPHIN study was a nonrandomized, single-arm, phase II trial. Patients with PD-L1 positive, LA-NSCLC received RT (60 Gy) concurrently with durvalumab, followed by maintenance durvalumab therapy. The 12-month PFS rate was 72.1%, far exceeding the 28% set under the original hypothesis. The median PFS was 25.6 months and the ORR was 90.9% [148]. This study is expected to support further development of phase III clinical trials. The START-NEW-ERA study (NCT05291780) was a single-arm phase II trial that explored the efficacy of SBRT combined with immunotherapy in patients with LA-NSCLC. The median OS was 55 months [148, 149]. Early outcomes suggest that SBRT followed by ICIs may be a suitable treatment regimen for these patients. The TRADE-hypo study (NCT04351256), a prospective randomized phase II trial, addressed the safety and efficacy of durvalumab combined with either conventional (30 × 2.00 Gy) or hypofractionated (20 × 2.75 Gy) RT. The primary end point was ORR [150]. Preliminary results were showcased at the 2024 ESMO meeting. Interim futility analysis was conducted in the conventional RT arm and was positive with 11/18 patients achieving tumor response (5 stable disease, 2 progressive disease). This suggests that for patients who are not suitable for chemotherapy, a novel combination of duvalizumab and conventional chest RT could potentially be beneficial. Additional safety, efficacy, and biomarker data are expected to be provided in May 2025.

    3.4 Metastatic NSCLC

    The PEMBRO-RT (NCT02492568) and MDACC (NCT02444741) trials provided solid evidence for combining RT and ICIs for metastatic NSCLC. Both trials enrolled patients with metastatic NSCLC who were randomly assigned to receive either pembrolizumab alone or SBRT. In the phase II PEMBRO-RT trial, pembrolizumab was administered within 7 days of the completion of SBRT (3 × 8 Gy). The primary endpoint of 12-weeks ORR was improved from 20% in the pembrolizumab alone arm to 50% in the pembrolizumab after RT arm [151]. Intriguingly, subgroup analyses showed the largest benefit from the addition of RT in patients with PD-L1-negative tumors, which significantly improved PFS and OS. In the phase I/II MDACC trial, SBRT was 50 Gy/4 fractions or 45 Gy/15 fractions. No significant differences in ORR or PFS were observed; however, exploratory analyses suggested that, for patients with low PD-L1 expression, a longer median PFS was observed in the ICIs plus RT group [152]. None of the trials met the preset criteria for meaningful clinical benefit owing to the small sample size. A pooled analysis showed significantly longer median PFS (9.0 vs. 4.4 months) and median OS (19.2 vs. 8.7 months) in the pembrolizumab plus RT group, with no new safety concerns [153]. Unfortunately, this pooled study could not draw conclusions regarding the optimal dose and timing of RT to induce a distant response, which should be further confirmed in specialized, large, randomized trials.

    While RT combined with ICIs shows promise for NSCLC, the enhanced antitumor effect appears to be limited to other tumor types [154]. Indeed, evidence to date has shown that combination therapy has limited benefits for most patients with HNSCC. A phase II trial (NCT02684253) enrolled patients with metastatic HNSCC who were randomly assigned to receive nivolumab alone or nivolumab in combination with SBRT. Regrettably, no improvement in clinical outcomes or evidence of the abscopal effect was found [155]. In a phase II study involving 18 patients with relapsed small cell lung cancer (SCLC) (NCT02701400), durvalumab and tremelimumab were administered with or without SBRT (3 × 9 Gy). SBRT was administered as an immune sensitizer prior to ICIs treatment. However, neither OS nor PFS showed a significant difference in the two arms [156]. In a recent trial (NCT03104439) involving patients with hepatocellular carcinoma and portal vein tumor thrombus, camrelizumab and apatinib were administered with or without SBRT (36-40 Gy/6-8 Gy). Longer median OS (12.7 vs. 8.6 months) and median PFS (4.6 vs. 2.5 months) were observed in the ICIs plus SBRT group [157]. This combination regimen showed clinical benefits with an acceptable safety profile, and may be a promising first-line therapy for patients with hepatocellular carcinoma and portal vein tumor thrombus. Regarding immune desert tumors, including microsatellite-stable colorectal cancer and pancreatic ductal adenocarcinoma, despite their limited response to immunotherapy alone, a phase II trial (NCT03104439) showed that RT can enhance the immunotherapy response even in these cases [158].

    Collectively, RT combined with ICIs has demonstrated encouraging outcomes in patients with NSCLC and certain solid tumors. However, many clinical studies investigating the efficacy of combining RT with ICIs have not included ICI or RT monotherapy. This omission obstructed the ability to discern a synergistic therapeutic benefit from combination therapy compared with the effects of either monotherapy independently.

    4 CHALLENGES AND CONCERNS

    4.1 Fractionation and scheduling

    For decades, conventionally fractionated radiotherapy (CFRT) has typically been delivered at doses of 1.8 to 2.0 Gy per fraction, 5 days per week, for 5-8 weeks. In contrast, hypofractionated radiotherapy (HFRT) delivers large doses in one-fifth fractions and is increasingly used in clinical practice. The most prominent example of an HFRT is SBRT. Biological differences between SBRT and CFRT exist [159]. Low-dose irradiation with a single dose (0.5-1.0 Gy) has been suggested to modulate the TME and activate immune responses [88, 160, 161]. Low-dose HFRT refers to a higher dose per fraction but a lower total dose of radiation for cancer treatment, both used in preclinical models [88] and clinical trials [162, 163].

    Preclinical studies have emphasized the fractionation and scheduling of RT with ICIs to establish a long-lasting antitumor immune response [164, 165]. The optimal regimen of RT combined with ICIs to maximize the antitumor immune response remains controversial [166]. Recent clinical trials have evaluated the safety and effectiveness of HFRT and low-dose hypofractionated radiotherapy (LDHRT) in combination with ICIs. However, in patients with microsatellite stable (MSS) colorectal cancer, the abscopal effect was not observed in either radiation regime, and the median PFS and OS were limited [167]. A randomized study (NCT02888743) in 2022 evaluated ICIs combined with LDFRT or HFRT in patients with metastatic NSCLC [168]. The study did not identify any significant benefit in ORR for either the LDFRT or HFRT regimens, and no significant differences in PFS or OS were observed between the treatment arms.

    A latest phase I/II clinical trial (NCT02239900) compared the administration of concurrent or sequential SBRT with ipilimumab in patients with metastatic cancer [169, 170]. All patients received ipilimumab and were randomly assigned to 5 treatment groups based on tumor size and location. The group that received sequential ipilimumab with SBRT to the lungs showed the highest rate of clinical benefit. A recent systematic literature review sought to reach a consensus among experts on the combination of SBRT and ICIs [171]. The consensus reached was that anti-PD-L1 or anti-PD-1 treatment should continue during SBRT delivery without omission of treatment cycles and that nivolumab plus ipilimumab should not be administered on the same day as SBRT. However, in an observational cohort study of patients with early-stage NSCLC, all patients were treated with SBRT (27 Gy/1 fraction or 50 Gy/5 fractions) targeting different lung lesions [172]. This large retrospective analysis found no statistically significant differences in the 5-year OS or 5-year PFS rates, raising the question of whether the fraction and location matter. Indeed, irradiation of liver metastases in patients with NSCLC has been shown to result in stronger activation of antitumor immunity than irradiation of pulmonary metastases [170]. Several ongoing clinical trials have persistently scrutinized diverse regimens of RT combined with ICIs [173, 174]. Emerging evidence will furnish precise directives for an optimal regime.

    4.2 DLNs irradiation and dose

    Notably, the total dose of the DLNs irradiation is a topic worthy of discussion. DLNs serve as the site where DCs prime antigen-specific CD8+ T cells, and irradiation of lymphoid organs can result in lymphopenia [45, 175]. A study published in 2018 highlighted that irradiation of the DLN impeded adaptive immune responses and attenuated the efficacy of SBRT and ICIs [67]. A concept termed lymphocyte-sparing RT has been proposed that advocates the sparing of lymphocytes whenever possible [176]. Therefore, precautionary irradiation of the DLNs is recommended [177]. A nonrandomized phase II trial (NCT01463423) enrolled patients into 3 groups based on the tumor stage. The results suggest that individual doses and fractionation of SBRT, including doses lower than those routinely administered, are associated with local tumor control [178]. Therefore, individualized dosing should be considered in future studies.

    4.3 Toxicity

    Pneumonitis and radiation pneumonitis are the most common AEs associated with the administration of CRT [141]. The overall incidence of pneumonitis in the PACIFIC trial was 33.9% [25, 27]. In real-world studies, the incidences of all-grade and grade ≥3 pneumonitis were 35% and 6%, respectively [179]. The incidence of pneumonitis varies with race and age [146, 179]. The MDACC trial found that most AEs were self-limiting, and no patient in the ICI combined with SBRT group experienced grade 4 or 5 toxic effects [152]. However, another multicenter analysis demonstrated that SBRT with concurrent ICI increased the risk of grade 3 pneumonitis compared with that of SBRT alone [180]. Closer monitoring should be considered in patients who are administered ICIs and RT. Differentiating between RT-induced pneumonitis and immune-related pneumonitis is challenging [27]. Radiomics holds great promise in aiding correct diagnosis [181]. To date, evidence indicates that the observed risk of severe toxicity for SBRT plus anti-PD1/PD-L1 monotherapy is low [171], and it remains to be seen whether the combination simply increases or amplifies toxicity. In the absence of objective data showing that simultaneous administration leads to a significant increase in toxicity, RT combined with ICI is a reasonable strategy in clinical practice.

    5 CONCLUSIONS

    The immune system's capacity to reject a tumor depends on the presence of neoantigens within cancer cells. RT increases the number of neoantigens via in situ vaccination. Additionally, RT reprograms the TME to foster a durable and systemic immune response. Ongoing studies have explored precision RT based on gene expression profiles to complement the precision of cancer medicine using immunotherapy.

    In conclusion, the integration of RT with immunotherapy represents a paradigm shift in cancer treatment. This field awaits the results of the ongoing clinical trials. Over the past five years, promising clinical trial outcomes have been predominantly observed in NSCLC. Conversely, trials investigating other solid malignancies, including HNSCC and colorectal cancer, are limited in number, and the outcomes have been less encouraging. The inherent heterogeneity of tumors may dictate disparate responses to the combination of RT and immunotherapy. A severe limitation of ongoing clinical trials is that they are not biomarker-driven. Biomarker implementation and the identification of distinct patient subsets are priorities. However, challenges, including determining the optimal dosing, fraction, and schedule with the lowest toxicity of the combination therapy, remain. Further research is imperative to refine combination therapy and identify predictive biomarkers for the individualized treatment of solid tumors.

    AUTHOR CONTRIBUTIONS

    Yuze Wu drafted the manuscript and prepared the figures. Ming Yi, Mengke Niu, and Binghan Zhou helped in collecting the related literatures and participated in discussion. Kongming Wu and Qi Mei designed the review and revised the manuscript. All authors read and approved the final manuscript.

    ACKNOWLEDGEMENTS

    This work was supported by the National Natural Science Foundation of China (82073370 and 82272794).

      CONFLICT OF INTEREST STATEMENT

      The authors declare that they have no competing interests.

      ETHICS APPROVAL AND CONSENT TO PARTICIPATE

      Not applicable.

      DATA AVAILABILITY STATEMENT

      Not applicable.

        The full text of this article hosted at iucr.org is unavailable due to technical difficulties.