Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

Skip to main content

Carbon ion irradiation mobilizes antitumor immunity: from concept to the clinic

Abstract

Carbon ion radiotherapy (CIRT), a type of particle therapy, is at the forefront of clinical oncology treatments due to its superior physical properties and biological performance. Although CIRT has demonstrated outstanding therapeutic outcomes in clinical settings, the biological mechanisms underpinning its effects, particularly its immunogenic potential and the superiority of its induced antitumor immune response compared to photon radiotherapy, remain areas of active investigation. This review summarizes the latest research progress on the mechanisms of antitumor immune responses triggered by CIRT and discusses preclinical and clinical studies related to combined CIRT and immunotherapy (CCIT). Against the backdrop of extensive research and significant clinical efficacy achieved by combining radiotherapy with immunotherapy, this review provides a theoretical foundation for a better understanding of the superior tumor cell-killing effects of CIRT and the underlying immunological mechanisms. Further insights into the factors affecting the efficacy, toxic effects, and developmental limitations of this combination therapy mode will be instrumental in guiding the conduction of CCIT studies.

Introduction

Photon radiotherapy (PhRT) remains the most commonly used form of radiotherapy today. Limited by its physical and biological characteristics, despite the assistance of computer technology and imaging methods to optimize radiotherapy plans, traditional radiotherapy still faces unresolved issues such as poor therapeutic effects and severe side effects [1, 2]. The American physicist Robert Rathbun Wilson first proposed the use of heavy charged particles for cancer treatment in 1946 [3]. Subsequently, researchers at the Lawrence Berkeley Laboratory (LBL) in the United States initiated studies from 1975 to 1992 on treating cancer patients with heavy ions, including carbon ions [3,4,5]. Since 1994, Japan has established the world’s first medical facility dedicated to heavy ion radiotherapy at the National Institute of Radiological Sciences (NIRS) in Chiba, starting the use of carbon ions for treatment and scientific research and gradually becoming the largest carbon ion radiotherapy (CIRT) center in the world [6]. Currently, institutions with CIRT capabilities are mainly located in Asia and Europe, with the Mayo Clinic announcing the establishment of the first carbon ion radiation facility in the United States at Florida in 2020 [7]. CIRT plays an irreplaceable role in malignant tumors traditionally resistant to radiotherapy, such as bone and soft-tissue sarcomas, locally recurrent rectal cancer and pancreatic cancer [6, 8, 9].

Tumor immune escape is a severe issue that plays a significant role in tumor development and resistance to immunotherapy. The immune system is involved in every stage of tumor occurrence and progression, which can be divided into the elimination, equilibrium, and escape phases, known as the three “E” stages [10, 11]. The immune escape mechanism evolved by tumors enables tumors to survive despite the host immune system’s attempts to combat them [11]. Although immunotherapy has been successful in treating many advanced cancers, a portion of patients still do not respond or develop resistance after treatment, leading to therapeutic failure [12,13,14]. Consequently, a primary focus of current research has been to identify partners that enhance the efficacy of immunotherapy while maintaining acceptable side effects.

Notably, the combination of immunotherapy and radiotherapy has garnered considerable attention in recent years. Immunotherapy tends to be less effective against solid tumors with high tumor burdens in advanced stages, while radiotherapy, in addition to its debulking effect, can also stimulate the release of tumor antigens, induce immunogenic cell death (ICD), and reshape the tumor immune microenvironment (TIME), thereby reactivating the anti-tumor immune response [15,16,17]. Conversely, immunotherapy can amplify the immune response, significantly increase the occurrence of abscopal effects, and prolong patient survival. The immunotherapy and radiotherapy is synergistic, commonly referred to as radioimmunotherapy [15, 18]. However, the results of clinical trials for combined PhRT and immunotherapy (CPIT) have not always supported its superior effectiveness to either therapy alone [19,20,21]. This limitation may be due to the suboptimal dose delivery of PhRT, its substantial toxicity, and the insufficient specificity or intensity of the immune response it generates, making PhRT less compatible with immunotherapy. Preliminary evidence suggests that CIRT may enhance antitumor immunity more effectively than PhRT, potentially improving radioimmunotherapy outcomes [22,23,24]. Based on the limited data available, without strictly limiting the target patient population and clinical characteristics, this paper aims to provide a comprehensive review of the antitumor immune responses triggered by CIRT, revisit the related research on combined CIRT and immunotherapy (CCIT), and discuss the factors affecting the efficacy, toxic reactions, and developmental limitations of CCIT to offer a broader reference for the conduction of CCIT clinical research and its clinical application.

CIRT enhances the antitumor immune response

Radiation-mediated antitumor immune responses include adaptive and innate immune mechanisms. Initially, within an immunopermissive tumor microenvironment, ionizing radiation facilitates the generation or exposure of tumor antigens and the release of damage-associated molecular patterns (DAMPs). These tumor antigens are captured, processed, and presented to T cells by antigen-presenting cells (APCs), subsequently activating T cells to proliferate, infiltrate the tumor, and kill tumor cells, accompanied by further release of tumor antigens from the tumor [15, 25, 26]. Additionally, innate immune cells such as natural killer (NK) cells and related immune molecules in the microenvironment also play a critical role in this process [25, 26]. Although in CPIT, PhRT acts as an ignition, shifting the tumor from “cold” to “hot” and enhancing the likelihood of an abscopal effect, achieving a complete cure remains challenging [27]. Although not systematically summarized, abscopal effects have occurred in clinical cases where CIRT was used to treat advanced tumors with metastasis or recurrent tumors with multiple lymph node metastases [26, 28,29,30,31]. In preclinical experimental models, CIRT also seems to induce abscopal effects [32]. Notably, despite similarities in immune responses following exposure to low and high linear energy transfer (LET) radiations, CIRT, which has superior physical and biological characteristics compared to PhRT, may elicit even stronger antitumor immunity [22, 33]. Here, we summarize the CIRT-mobilized antitumor immune response and its related molecular mechanisms from three levels: immunogenicity, tumor immune microenvironment and systemic tumor immune environment.

Increased immunogenicity of tumor cells caused by CIRT

Complex DNA damage and repair

While radiotherapy promotes the generation and exposure of tumor antigens, recent clinical studies on CPIT have revealed that the immune response elicited by photon-induced tumor antigens often falls short of eradicating the primary tumor (let alone effective abscopal effects), with many patients showing either no response or short-lived responses [34, 35]. Compared to photon radiation, carbon ions induce a greater frequency and complexity of DNA double-strand breaks (DSBs), with clustered DSBs or multiple DSBs representing further increases in overall damage complexity and mutagenicity, as clustered DSBs carry a greater risk of misrepair. Unrepaired DNA damage molecules during mitosis lead to various chromosomal-level distortions and genomic instability [36,37,38,39]. Studies have explored the impact of isoeffective doses of 295 keV/μm carbon ions versus 1.3 MeV or 0.2 keV/μm photons on chromosomes, demonstrating that carbon ions increase the yield of chromosome shattering and chromothripsis [40]. Moreover, lost DNA or chromosome fragments (manifested as micronuclei, etc.) migrate from the nucleus to the cytoplasm during interphase after carbon ion irradiation [36]. This process is more pronounced with carbon ions than with photon irradiation, as these ions more effectively induce cytoplasmic double-stranded DNA (dsDNA) or micronuclei [41,42,43]. Recently, we observed a more pronounced decrease in LNC EBLN3P expression in tumor cells following carbon ion irradiation compared to X-ray irradiation [44]. Further investigation indicated that the downregulation of LNC EBLN3P in response to carbon ions enhances radiation-induced mitochondrial damage via the Keap1/Nrf2/HO-1 pathway [45], which leads to changes in membrane permeability and facilitates mitochondrial DNA (mtDNA) release into the cytoplasm [46, 47]. Aberrantly exposed dsDNA or mtDNA in the cytoplasm can be recognized by the DNA receptor cyclic GMP-AMP synthase (cGAS), triggering the cGAS/STING pathway and inducing a type I interferon (IFN-1) response [48]. Through transcriptome analysis of cancer cells and in vitro and in vivo experiments, it has been demonstrated that compared to low-LET radiation, carbon ions with high LET significantly induced stronger cGAS-STING-IFN signals, which also indirectly demonstrated the enhancement of the above process [43, 49,50,51].

To ensure genomic stability, cells (including tumor cells) undergo complex DNA damage repair (DDR) processes after DNA damage, and the DDR after carbon ion irradiation involves proteins different from those after photon irradiation [52, 53]. Compared to the classical nonhomologous end-joining pathway used after low-LET irradiation, these error-prone alternative DNA repair pathways become more prevalent after CIRT, increasing the likelihood of gene mutation or repair failure [54]. Therefore, it can be speculated that carbon ion radiation has greater potential to induce the generation of antigenic neoepitopes than photon radiation [36, 55].

Non-lethal effects

During the process of DNA repair following irradiation, cells undergo cell cycle checkpoints, such as G1/S, intra-S, and G2/M checkpoints. Due to the unique nature of DNA damage under high-LET carbon ion action, studies have shown that carbon ion radiation is more effective at reducing the cell proliferation index and increasing the expression of cell cycle inhibitors than X-rays [56]. Furthermore, carbon ion irradiation induces more prolonged G2/M arrest than does photon irradiation at the same physical dose (isodose) [57,58,59]. Cell cycle arrest induced by ionizing radiation, including carbon ions, seems to be related to tumor immunity. For example, ionizing radiation activates the ATM/p53/p21 pathway, inhibiting CDK4/6 and causing G1/S arrest [60, 61]; however, effective inhibition of CDK4/6 can promote tumor immunogenicity and enhance T-cell activation through various mechanisms [62, 63]. Particularly, the effectiveness of carbon ions is less dependent on the cell cycle compared to photons [64].

The DNA damage inflicted by carbon ion radiation is complex and challenging to repair, resulting in lower repair efficiency and longer-lasting damage compared to the DNA damage induced by X-rays, which is predominantly repaired within 24 h with most residual damage associated with telomeric DNA [65]. A study with human uveal melanoma 92–1 cells demonstrated that heavy ions more effectively induced senescence than X-rays, as shown by measurements of senescence-associated β-galactosidase and cell proliferation [66]. Cell senescence, a stress-induced process, can participate in antitumor immune responses or radiation resistance processes by activating the senescence-associated secretory phenotype (SASP) and triggering the secretion of related cytokines [67].

Mitotic catastrophe is considered a cytostatic mechanism preceding cytotoxic processes such as apoptosis and necrosis or as a consequence of aberrant mitotic progression [68]. Daijiro and colleagues reported that carbon ions induce mitotic catastrophe more effectively than the isodose of X-rays by assessing clonogenic cell death with 4’,6-diamidino-2-phenylindole dihydrochloride staining [69]. This may be related to the inhibition of NBS1/BRCA1-mediated γ-tubulin protein monoubiquitination by high-LET carbon ions, thereby enhancing excessive replication of centrioles in tumor cells [70]. Mitotic catastrophe leads to the formation of giant nuclei, multinucleation, and micronuclei and triggers the cGAS/STING pathway, contributing to genomic instability of tumor cells and triggering antitumor immunity after recognition by the immune system [57, 69, 70].

Immunogenic cell death

Ionizing radiation causes DSBs through direct ionization of target molecules or indirect action by generating free radicals. Nucleases, proteases, and other enzymes are activated, leading to chromatin degradation, histone efflux, nuclear contraction and fragmentation, and ultimately cell death [71]. Additionally, factors such as membrane structural damage and disruption of cellular energy metabolism are also important causes of cell death induced by ionizing radiation [72, 73]. Radiation damage caused by low-LET radiation, such as photons, is mainly mediated by the formation of radical oxygen species [74]. After carbon ion irradiation, due to the severity of DNA damage and the inefficiency of DNA repair, tumor cells (even cancer stem cells) that are too late or unable to repair the damage will undergo multiple types of cell death (p53-independent apoptosis, necrosis, necroptosis, ferroptosis, etc.). The above processes after carbon ion irradiation are more significant than those after the same dose of photon radiation [75,76,77,78]. These ICD modes allow tumor cells to produce or release antigens, becoming antigen sources; thus, these mechanisms are also known as in situ autovaccination [79,80,81,82,83,84,85].

The occurrence of ICD by tumor cells releases meaningful tumor antigens and also effectively coordinates the spatial–temporal release of DAMPs, thereby triggering another characteristic of ICD, including exposure to calreticulin (CRT), extracellular release of high mobility group box 1 (HMGB1) and adenosine triphosphate (ATP). These events promote APC recruitment, phagocytic activity, and maturation, enabling them to engulf antigenic material, migrate to lymph nodes, and initiate cytotoxic T lymphocyte (CTL)-dependent immune responses, namely, adjuvanticity [25, 86]. The mechanisms of DAMPs in immune cell activation are not further elaborated here but can be found in the relevant literature [35, 87]. Compared to isodose photon irradiation, carbon ion irradiation leads to a more significant increase in HMGB1 in a dose-independent manner [88]. However, under isoeffective dose irradiation, both groups exhibited comparable increases in HMGB1 [89]. Additionally, under isoeffective dose conditions, exposure to HMGB1 increases with increasing LET of carbon ions [90]. As one of the most critical markers of ICD, the translocation of calreticulin to the cell surface (ecto-CRT) promotes APC phagocytosis and activates antitumor immunity. In lung adenocarcinoma, tongue squamous carcinoma, and nasopharyngeal carcinoma cells, the level of ecto-CRT induced by 2 Gy and 4 Gy carbon ion irradiation is greater than that induced by isodose protons and photons [91]. Furthermore, this advantage exists in normoxic tumor cells, whereas under hypoxic conditions, the expression level of CRT is significantly upregulated at baseline (0 Gy), and radiation cannot further increase CRT expression. This situation may be due to a competitive relationship between hypoxia-induced endoplasmic reticulum stress (ER stress) and radiation-induced ER stress, leading to the upregulation of CRT [92, 93]. Although the above studies were conducted in vitro, obvious induction of ICD characteristics by carbon ions was also observed through in vivo detection using sera from tumor-bearing mice or tumor samples [50, 94, 95].

Immunorecognition-related molecules

Alternatively, the formation of immunogenic niches is crucial for immune responses. After photon irradiation, the expression of intercellular adhesion molecule-1 (ICAM-1) and major histocompatibility complex class I antigen (MHC-I) on the cell membrane surface increases, enhancing the ability of T cells to recognize and kill tumor cells and leading to the aggregation of effector T cells at the tumor site [96, 97]. It has been reported that C3H/HeSlc mice bearing NR-S1 squamous cell carcinoma cells were irradiated with carbon ions. Immunohistochemistry showed that ICAM1 expression increased in a time-dependent manner after irradiation [98]. After carbon ion irradiation in MC38 colorectal tumor cells, the expression of MHC class I genes was upregulated and was significantly greater than that in response to the same dose of photons [99], which is conducive to the formation of effective immune synapses between T cells, APCs and tumor cells.

CIRT reshapes the tumor immune microenvironment

Even in the presence of sufficient antigenicity and adjuvanticity, the immune microenvironment of tumor cells is the primary determinant to initiate adaptive immune responses, referred to as the TIME [81, 100]. Tumor cells induce the infiltration of immunosuppressive cells into the TIME by secreting inhibitory cytokines, and immunosuppressive cells can also recruit more of the same type of cells by secreting these factors, leading to an immunosuppressive vicious cycle [101,102,103]. Once the tumor progresses to the immune escape stage (according to the immunoediting theory), an TIME that promotes malignant tumor progression, drug resistance, and metastasis is eventually established under the action of various immunosuppressive cells (such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and tumor-associated neutrophils (TANs)) and immunosuppressive molecules (such as transforming growth factor-β (TGF-β), IL-10, Fas ligand (FasL), and immune checkpoint molecules) through multiple complex mechanisms that inhibit antitumor immunity, maintain or promote self-immunosuppressive phenotypes, and facilitate malignant tumor progression [104, 105]. Here, we investigated the immunological effects of CIRT on the composition and function of TIME.

The distribution and function of immune cells such as T cells, Tregs, and mononuclear phagocytes and the levels of immunostimulatory or inhibitory molecules in the TIME are important indicators for evaluating the effect of radiation on the TIME [106, 107]. PhRT has been reported to reduce the infiltration of Tregs and MDSCs into tumors and increase the infiltration and activation of CD4+ T and CD8+ T cells [15]. In contrast, compared with PhRT, CIRT elicits a limited induction of immunosuppressive cells such as MDSCs, and inhibits M2-like polarization [108, 109]. Additionally, carbon ions can increase the infiltration of CD8+ T cells, CD4+ T cells, macrophages, and natural killer cells into the TIME and increase the abundance of immunostimulatory molecules such as interferon-gamma (IFN-γ), IL-2, and IL-1β [43, 50, 51, 108,109,110]. Compared with X-rays, carbon ions relatively reduced the exposure levels of immunosuppressive factors IL-10 and TGF-β [88].

In terms of function, immunosuppressive cell subgroups play a crucial role in inhibiting the TIME [107]. MDSCs reduce the formation and function of NK cells by producing inhibitory cytokines, thus downregulating NKG2D expression and IFN-γ secretion [111]. Simultaneously, Tregs can downregulate NKG2D expression on NK cells via membrane-bound TGF-β or directly suppress NKG2D activity upon contact with NK cells [112], which are crucial cytotoxic cells in tumor innate immunity that directly kill tumor cells in a non-MHC-restricted manner using perforin and granzymes, making their functional status necessary for antitumor immune responses [113]. Carbon ion irradiation significantly upregulates Klrk1 gene expression to activate the NKG2D/NKG2D-Ls pathway and enhance the cytotoxic pathway mediated by NK cells [114]. This finding suggested that inhibiting Tregs or MDSCs combined with CIRT can strengthen the immune response by increasing NK cell cytotoxicity. TAMs, which can function as specialized APCs to induce antitumor immunity by presenting tumor antigens, are also involved in the formation of a suppressive TIME [115, 116]. Conrad et al. compared the effects of 250 kV X-rays and 9.8 MeV/u carbon ions on RAW 264.7 macrophages at isodose and reported that carbon ions induce only a slight decrease in macrophage viability but exhibit greater enhancement of phagocytic activity than X-rays [117]. Carbon ion irradiation enhances CTL sensitivity to tumor cell recognition and killing, similar to the modulatory effect of photons at isoeffective doses [118]. A study simulating the effects of space radiation (including carbon ions) on Jurkat cells, a T-cell model widely used in space radiation studies, revealed that 1 Gy of carbon ion irradiation causes a smaller decrease in IL-2 secretion by T cells than photons at isodose, suggesting that CIRT may have less immunosuppressive effects on T cells [119]. Studies have reported that carbon ion irradiation does not significantly increase the number of dendritic cells (DCs) or promote functions such as phagocytosis, migration, and IL-12 secretion [120]. However, carbon ions trigger IFN-γ and IL-12 in DCs treated with lipopolysaccharide (LPS), suggesting the potential long-lasting induction of adaptive immune responses after carbon ion irradiation [121].

Tumors can induce a decrease in oxygen pressure (even below 2%, while the physiological range of normal tissues is 3–7.4%) in the TIME through a series of mechanisms, leading to the generation of hypoxic microenvironments [122]. Conversely, hypoxia, as a cellular stress factor, extensively affects multiple cellular metabolic pathways, thereby correlating with tumor invasive phenotypes and treatment resistance and serving as a critical factor in tumor microenvironment (TME) immune suppression [123, 124]. Hypoxia induces multiple effects by upregulating hypoxia-inducible factor-1α (HIF-1α), such as promoting angiogenesis, tumor invasion, and metastasis, as well as increasing Treg numbers and upregulating programmed cell death-ligand 1 (PD-L1) on tumor cells [125]. Interestingly, although PhRT can increase tumor immunogenicity to some extent, this effect is greatly influenced by oxygen pressure in the TIME (oxygen enhancement ratio, OER is 2–3). In contrast, carbon ion beams at the Bragg peak with high LET (up to > 200 keV/μm) primarily exert direct radiation effects, and their immunostimulatory effects are almost unaffected by oxygen pressure [93]. Since the DNA damage caused by carbon ion irradiation is mainly due to direct radiation damage, subsequent micronucleus formation and cGAS-STING activation, carbon ions are more effective than X-rays in inducing DDR defects, regardless of normoxia or hypoxia. Additionally, HIF is involved in the postirradiation DDR process in tumor cells, promoting the generation of hypoxic radiation resistance [126, 127]. Compared to photons, carbon ions significantly reduce HIF expression, thereby inhibiting the DDR to maintain a damaged DNA status, which also optimizes the tumor microenvironment via the cGAS-STING pathway to promote immunogenicity [51, 128, 129]. Furthermore, studies have shown that under hypoxic conditions, high-LET carbon ion beams at the SOBP further increase the radiation sensitivity of DNA repair-deficient Chinese hamster ovary cells [130].

Ionizing radiation also has a certain degree of immunosuppressive effect [131]. Local PhRT can recruit immunosuppressive bone marrow cells, directly promoting tumor growth, and can also upregulate tumor cell PD-L1 expression through the ATM/ATR/Chk1 pathway, activating the programmed cell death protein 1 (PD-1) receptor on inhibitory T cells [132, 133]. Studies have shown that the upregulation of PD-L1 on tumor cells and PD-1 expression on T cells after carbon ion irradiation is greater than that caused by PhRT [51, 99, 134]. However, the development of immune checkpoint inhibitors (ICIs) targeting PD-L1 or PD-1 is relatively mature, which further suggests the necessity of CIRT combined with ICI drugs to further improve the efficacy of treatments for tumors with high immune checkpoint expression.

CIRT improves the systemic tumor immune environment to maintain the foundation of antitumor immunity

Measuring the antitumor immune response should extend beyond the local immune activity within the TIME to encompass the broader systemic tumor immune environment (STIE). The antitumor immune response is primarily mediated by immune effector cells and molecules from both the innate and adaptive immune systems. These immune-active substances circulate between the TIME, immune organs, and other bodily organs, playing a crucial role in the development of the STIE [106, 135]. The STIE serves as the foundation for establishing antitumor immune responses and affects patient prognosis after radiotherapy [136,137,138].

CIRT improves STIE by modulating immune cells and cytokines in the circulatory system (blood, spleen, etc.). Studies have confirmed that compared to PhRT, carbon-ion irradiation results in a greater increase in CD3+, CD4+, and CD8+ T cells; macrophages; and NK cells in the peripheral blood of tumor-bearing mice at an isoeffective dose, while MDSCs decrease more in the bone marrow, peripheral blood, spleen, and tumor tissue [109]. Similarly, research has shown that CIRT expands the populations of CD8+ T cells and effector memory T cells while reducing T-cell exhaustion in the spleens of tumor-bearing mice [43]. Recent studies have shown that low-dose radiation has significant immunostimulatory effects on the TIME [139, 140]. Low-dose irradiation can alleviate tumor-induced immune suppression, resulting in increases in the proportions of CD3+ T cells, CD3+ CD4+ T cells, and CD3+ CD8+ T cells, as well as in the levels of IFN-γ and IL-2 in the peripheral blood after irradiation [141]. Peripheral blood samples were collected from 32 prostate cancer patients before and after receiving CIRT. Analyses of these samples revealed changes in the frequency, proliferation, and cytokine expression of immune cells. Notably, CIRT preserved lymphocyte viability, augmented lymphocyte proliferation, improved T-cell functionality, curtailed the induction of immunosuppressive cells, and decreased the expression of immunosuppressive cytokines [142]. Although the evidence available is limited, this retrospective clinical study, to some extent, demonstrated that carbon-ion immune modulation is beneficial for improving STIE in patients.

The state of STIE affects the occurrence and development of tumors, and the progression of tumors also leads to immune damage in the body [106, 143, 144]. Ionizing radiation, including photons and charged particles, directly enhances antitumor immunity by rapidly and extensively inactivating tumor cells through DNA damage [145]. As noted, carbon ions achieve greater tumor cell inactivation than photons at equivalent doses, exhibiting a higher relative biological effectiveness (RBE), even in hypoxic tumor stem cells. Consequently, they exert a more potent suppressive effect on tumor-induced immune damage [108, 146,147,148,149,150]. Overall, as most cancers are chronic debilitating diseases, and many cancer patients suffer from leukopenia and diminished leukocyte function due to treatment-related toxicities associated with radiotherapy and chemotherapy, the functional state of the immune system declines. This reduction is detrimental to establishing an effective antitumor immune response [151]. Compared to PhRT, CIRT can inhibit the above processes with a superior tumoricidal effect.

The improvement in STIE is partly due to the more precise and limited dose delivery of CIRT compared to PhRT [152]. Lymphocytes are extremely sensitive to ionizing radiation, and in the process of three-dimensional PhRT, circulating lymphocytes are inevitably irradiated, leading to lymphopenia, a common side effect of radiotherapy [35]. CIRT has the characteristic of differential dose distribution, with Bragg peak effect and less lateral scattering, which preserves more normal tissues from being irradiated and delivers a lower dose to normal tissues in the entrance channel, further reducing the dose deposited in circulating lymphocytes and reducing the proportion of chromosomal aberrations in lymphocytes, thus reducing the common complications of radiation-induced lymphopenia [110, 153,154,155,156].

In vivo study of combined CIRT and immunotherapy

Relying solely on tumor ablation rarely results in effective antitumor immunity. In the therapeutic setting, the TIME of most tumors can greatly limit immune-driven ICD [15, 34]. Therefore, unless additional immune-stimulating factors are provided, merely irradiating tumor lesions typically fail to generate a sufficiently strong immune response [157], which is typically evaluated using indicators such as inhibition of tumor metastasis, tumor suppression or rejection, abscopal effects, and survival in tumor-bearing mice [158]. Compared to other low-LET radiation, CIRT has advantages in terms of immunomodulation [23, 35]. Therefore, we propose that the combination of immunotherapy with CIRT, termed CCIT, may elicit a comparable or more effective antitumor immunity compared to its combination with PhRT, known as CPIT. Here, we compiled preclinical studies on the efficacy of CCIT, with or without comparisons to CPIT, detailed in Sects. “CIRT combined with ICIs” to “CIRT combined with DCs” below (Table 1).

Table 1 Preclinical studies demonstrating the antitumor immune response to carbon ion radiotherapy combined with immunotherapy

CIRT combined with ICIs

In the adaptive immune system, with the assistance of CD4+ T cells and DCs, CD8+ T cells are activated to become CTLs, which ultimately exert tumor-killing effects [136]. However, the presence of inhibitory costimulatory molecules (immune checkpoints), such as PD-1/PD-L1 and CTLA-4, renders CTL cells dysfunctional [163]. ICIs are important targeted drugs for various tumor immunotherapies [164]. Notably, both photons and carbon ions increase the expression of tumor PD-L1, with carbon ions showing a more significant increasing trend [51, 134]. Therefore, the combination of CIRT with ICI drugs may lead to more effective synergistic therapeutic strategies. Studies have shown that combining CIRT with dual immune checkpoint blockade therapy (anti-PD-L1 and anti-CTLA-4 antibodies) results in better control of irradiated tumors than CIRT alone (p < 0.01) [95]. Importantly, the complete remission rate of mice with nonirradiated tumors in the combination group was significantly greater (64%) than that in the ICI alone group (20%) (p = 0.0392); the overall survival rate in the combination group was greater than that in the CIRT or ICIs alone groups (vs CIRT, p = 0.0014; vs ICIs, p = 0.0009) [95]. Similarly, Huang et al. reported that combination therapy with ICIs and CIRT significantly inhibited the growth of locally irradiated and distant melanoma tumors compared to CIRT or ICIs alone, improving the survival rate of mice [76]. There are numerous studies on the combination of PhRT with ICIs, with Zhang et al.’s review detailing positive outcomes [157]. In recent years, studies have preliminarily confirmed to varying degrees that CCIT demonstrates similar or superior antitumor immunity compared to CPIT, where tumors were irradiated with isodose or isoeffective doses of carbon ions or photons combined with ICIs, with the detailed experimental designs shown in Table 1.[50, 51, 160,161,162].

The above studies demonstrated that effective immune responses, including abscopal effects, are induced by the combination of CIRT with ICIs and are closely related to the immunomodulatory processes triggered by irradiation. Bioinformatics analysis of infiltrating immune cells, molecular components, and tumor cell transcriptomes revealed that changes in tumor cells and the TIME in the combination group were consistent with that in CIRT alone, as discussed in Sect. “CIRT reshapes the tumor immune microenvironment”. Furthermore, ICI drugs further enhance this advantage. For example, more complex and persistent DNA damage, along with higher levels of cGAS-STING and more significant activation of cell death pathways, was observed in tumor cells. In the TIME at the irradiated site, there was increased infiltration and activation of immune-active cells such as CD45+ and CD8+ cells, NK cells, and macrophages, and the levels of innate immune signaling markers such as IL-2, INF-γ, inflammatory factors, and chemokines were significantly increased. Increased CD8+/GzmB+ and CD4+ tumor-infiltrating lymphocytes (TILs) were also observed in nonirradiated tumor tissues, with a greater frequency of activated naïve T cells in the combination group [50, 51, 76, 95, 160,161,162]. In a recent clinical case, a 24-year-old woman with alveolar soft part sarcoma underwent pelvic tumor irradiation with carbon ions at 67.2 GyE in 16 fractions, followed by pembrolizumab (ICI) administration 20 days after radiotherapy. The results showed significant shrinkage of both irradiated pelvic tumors and nonirradiated leg tumors, with MRI 10 months post-CIRT indicating an 80% reduction in both tumors compared to baseline [165].

CIRT combined with Treg inhibitors

As mentioned earlier, Treg cells can downregulate NK cell surface NKG2D expression via membrane-bound TGF-β or inhibit NK cell activity through cell contact [112]. Recent studies have shown that Treg inhibitors can effectively inhibit Treg activity by specifically blocking the interleukin-2 receptor alpha subunit (IL-2Rα, CD25) on Tregs without affecting the IL-2 signaling pathway on effector T cells [166]. Additionally, CIRT significantly induces Klrk1 gene expression and activates the NKG2D/NKG2D-ls pathway to enhance NK cell-mediated cytotoxic pathways. Therefore, the combination of CIRT with Treg inhibitors (anti-CD25 and niclosamide) significantly increases NK cell infiltration and function in the lung cancer tumor microenvironment, prolonging the survival of Lewis lung cancer-bearing C57BL/6 mice, with superior efficacy compared to either single-agent Treg inhibitor immunotherapy or CIRT (p < 0.001) [114].

CIRT combined with NeoAg RNA-LPX vaccines

In terms of combining active immunotherapy, studies have demonstrated that the combination of RNA-LPX vaccines with conventional PhRT can achieve potent tumor suppression [167]. Salomon et al. further investigated whether the inhibitory effect of the neoAg RNA-LPX vaccine combined with CIRT on murine colorectal tumors was superior to that of the combination with photon radiation. Compared with photon radiation, CIRT at isodose significantly upregulated the expression of MHC class I and extracellular calcium-binding protein in MC38 cells and increased HMGB1 secretion and PD-L1 expression [99], consistent with the trends we summarized earlier. Moreover, immune cell and molecular component analyses revealed greater infiltration of total CD8+ T cells, CD4+ T cells (including neoantigen-specific CD4+ T cells), and NK cells in the combination of CIRT with neoAg RNA-LPX-treated mouse tumors than in all other treatment groups. These T cells showed reduced expression of the inhibitory markers PD-1 and Tim-3, along with high secretion of the cytokines IFN-γ and TNF-α. In terms of tumor control and survival, the neoAg RNA-LPX vaccine showed similar overall therapeutic efficacy in mediating tumor growth inhibition and survival when combined with CIRT or PhRT, even at a lower physical radiation dose for carbon ions than for X-rays [99].

CIRT combined with DCs

DCs, as potent professional APCs in vivo, play a crucial role in presenting and activating naïve T cells and helper T cells after processing tumor antigens released under radiation [168]. Pretreated DCs administered intravenously or intratumorally in combination with CIRT have shown promising results [94, 98, 159]. Significant upregulation of tumor cell DAMPs (such as ecto-CRT), costimulatory molecules (ICAM-1), and antigens, which promote DC maturation, activation, and antigen presentation, thereby enhancing systemic antitumor immunity, was observed after carbon ion irradiation. In a mouse squamous cell carcinoma model, the administration of DCs after carbon ion treatment significantly improved the rate of tumor rejection after secondary tumor inoculation compared to that in the CIRT alone group (70.4% vs 88.5%; p < 0.01) and improved the rate of SCCVII-induced tumor-specific CTLs compared to that in the CIRT alone group [159]. In another study, the early lung metastasis marker S100A8 and the number of lung metastases in the CIRT combined with DC treatment group were significantly lower than those in the CIRT alone group [98]. Ando et al. compared the effects of 290 MeV/u, 70–80 keV/µm carbon ions and 137 Cs γ-rays combined with DCs on tumor immune responses. They first prepared tumor-bearing mice and irradiated them with CIRT at 2 Gy or PhRT at 4 Gy on the seventh day, followed by DC administration 1.5 days after irradiation, and observed the number of lung metastases 2 weeks postirradiation. The results showed that intravenous injection of DCs combined with CIRT significantly suppressed lung metastasis compared to CIRT or DCs combined with PhRT. Compared with those in response to photon irradiation, the levels of ecto-CRT in tumor cells significantly increased after CIRT, and subsequent coculture of these irradiated tumor cells with DCs significantly upregulated the expression levels of CD40 and IL-12 in DCs, indicating improved functional levels. Intravenously injected cocultured DCs into nonirradiated NR-S1 mice exerted antimetastatic effects [94].

Clinical study of CIRT combined with immunotherapy

In summary, as we have come to understand, CCIT provides a potent synergistic approach for tumor treatment, offering new avenues in this continuously evolving field. Currently, clinical trials investigating CCIT are actively underway, mostly targeting recurrent, metastatic, and advanced malignant tumors that are resistant to standard treatments. We identified 4 relevant studies related to CCIT on the ClinicalTrials.gov website. These trials cover various pathologies and immunotherapy characteristics, including CIRT combined with ICI (camrelizumab) for locally recurrent nasopharyngeal carcinoma (NCT04143984), CIRT combined with ICI (pembrolizumab) for stable disease in non-small cell lung cancer, head and neck squamous cell carcinoma, melanoma, and urothelial carcinoma (NCT05229614), SABR CIRT combined with the immunocytokine L19-IL2 for stage IV metastatic non-small cell lung cancer (NCT03705403) [169], and CIRT combined with granulocyte–macrophage colony-stimulating factor (GM-CSF) for hepatocellular carcinoma (NCT02946138) (withdrawn due to enrollment issues). Additionally, we found two studies on the jrct.niph.go.jp website: a phase Ib study registered in 2021 evaluating the safety and efficacy of durvalumab (an anti-PD-L1 antibody) combined with CIRT and weekly cisplatin for locally advanced cervical cancer (jRCT2031210083), which has completed patient recruitment. In the same year, another phase Ib study explored the safety and efficacy of durvalumab (MEDI4736) or tremelimumab combined with CIRT for treating advanced hepatocellular carcinoma patients with macrovascular invasion (jRCT2031210046), but this study is currently being suspended.

Summary and prospects

In summary, the superior immunomodulatory effects of CIRT can be attributed to several factors: first, the enhanced tumor immunogenicity induced by CIRT efficiently activates tumor immune responses; second, CIRT leads to the formation of an immunopermissive TME and suppresses the negative immune regulatory process, which not only promotes the successful construction of an adaptive immune response but also effectively recruits and activates nonspecific innate immune cells (NK, macrophages, NKT, etc.)-mediated antitumor effects; third, tumor shrinkage mediated by the high RBE characteristics of CIRT and precise dose distribution improve the STIE to maintain the basis of antitumor immunity. These theoretical foundations hope to promote the development of research related to CCIT.

In this era of immunotherapy, radiation therapy has ushered in new opportunities. By adjusting the protocols of radiotherapy and immunotherapy to make combinations more compatible with antitumor immune activation and to reduce side effects, the efficacy of radioimmunotherapy can be maximized, which is known as adaptive radioimmunotherapy (ART) [15, 170]. Currently, large fractionation is often recommended for CIRT [171]. Using single high-dose irradiation (such as 8 or 10 Gy), carbon ion irradiation dose-dependently upregulated the expression of the DNA exonuclease three-prime repair exonuclease 1 (TREX1), which degrades cytoplasmic dsDNA, in tumors, thereby inhibiting IFN-1 responses [161, 172]. However, by silencing TREX1 (siRNA), the activation of IFN-1 responses is enhanced under carbon ion radiation, IFN-β levels are significantly increased, and tumor cell apoptosis is enhanced [161]. The method of DC administration may affect the synergistic effect of CIRT combined with DCs. Compared with intratumoral injection of DCs, intravenous injection of DCs combined with CIRT has a greater antimetastatic effect, which may be related to the influence of the TME on intratumorally injected DCs [94]. However, it must be acknowledged that there is currently no standard protocol for determining the optimal dose, administration method, or sequence of CCIT, and further investigations are needed within the context of clinical trials. In terms of safety, it is worth noting that while ICIs combined with CIRT exhibit synergistic efficacy, the occurrence of adverse events does not seem to have a similar synergistic effect. A retrospective analysis revealed that among advanced melanoma patients treated with sequential CIRT combined with ICI, 21% experienced G3 + adverse events, similar to ICI monotherapy, while the frequency of local adverse events was similar to that of CIRT monotherapy [173].

The development and clinical use of CCIT still have a long way to go. Firstly, the rarity of CIRT facilities limits the progress of research and further clinicalization. Currently, the number of institutions globally with corresponding equipment and relatively mature carbon ion treatment planning systems is far less than that for PhRT, greatly limiting the acquisition of experimental samples and clinical data [174]. Clinical data on the combination of carbon ions and immunotherapy are lacking, especially for large-scale RCT trials comparing the efficacy and safety of CCIT with CPIT or combined therapy with protons. Currently, numerous plans for carbon ion facilities are underway or under construction [7]. The immune response induced by carbon ion radiation and its combination with immunotherapy provide evidence of excellent efficacy and safety for further approval and construction. Secondly, while comprehensive statistical data are lacking, as mentioned above, most carbon ion radiobiology research primarily targets tumor cells, with limited studies on immune and stromal components, which are equally crucial for the efficacy of radioimmunotherapy [106]. There is also a pressing need for translational research and development of clinically relevant biomarkers. Additionally, clearer understanding of CIRT’s effects and mechanisms may suggest potential benefits when combined with targeted therapies or chemotherapy. Finally, the overall quantity and quality of CCIT research are lower than those of CPIT-related studies, however, the latter can provide some methodological experience. Moreover, the cost of CIRT is relatively high, and combination therapy with immunotherapy will inevitably incur substantial expenses. However, the radiobiology of carbon ions provides many targets for sensitizing PhRT, such as differential metabolic factors, epigenetic, and posttranslational modification targets. Based on the differential molecular signaling pathways underlying the radiobiological effects, targeted drugs for sensitizing PhRT are expected to be developed, providing a new approach for radioimmunotherapy. Can we replace CCIT with a combination of PhRT, targeted drugs, and immunotherapy in parallel? Only further research can provide answers to these questions.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

APC:

Antigen-presenting cell

ART:

Adaptive radioimmunotherapy

CAF:

Cancer-associated fibroblast

CCIT:

Combined carbon ion radiotherapy and immunotherapy

CPIT:

Combined photon radiotherapy and immunotherapy

CIRT:

Carbon ion radiotherapy

CTL:

Cytotoxic T lymphocyte

cGAS:

Cyclic GMP-AMP synthase

DC:

Dendritic cell

DDR:

DNA damage repair

DAMPs:

Damage-associated molecular patterns

DSBs:

DNA double-strand breaks

ER stress:

Endoplasmic reticulum stress

FasL:

Fas ligand

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

HMGB1:

High mobility group box 1

HIF-1α:

Hypoxia-inducible factor-1α

ICAM-1:

Intercellular adhesion molecule-1

ICI:

Immune checkpoint inhibitor

ICD:

Immunogenic cell death

IFN-1:

Type I interferon

IFN-γ:

Interferon-γ

LET:

Linear energy transfer

LPS:

Lipopolysaccharide

MDSCs:

Myeloid-derived suppressor cells

mtDNA:

Mitochondrial DNA

NIRS:

National Institute of Radiological Sciences

NK cells:

Natural killer cells

OER:

Oxygen enhancement ratio

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed cell death-ligand 1

PhRT:

Photon radiotherapy

RBE:

Relative biological effectiveness

SASP:

Senescence-associated secretory phenotype

STIE:

Systemic tumor immune environment

TAMs:

Tumor-associated macrophages

TANs:

Tumor-associated neutrophils

TGF-β:

Transforming growth factor-β

TIME:

Tumor immune microenvironment

TILs:

Tumor-infiltrating lymphocytes

TME:

Tumor microenvironment

Tregs:

Regulatory T cells

TREX1:

Three-prime repair exonuclease 1

References

  1. Durante M, Loeffler JS. Charged particles in radiation oncology. Nat Rev Clin Oncol. 2010;7:37–43.

    Article  PubMed  Google Scholar 

  2. Olivares-Urbano MA, Griñán-Lisón C, Marchal JA, Núñez MI. CSC radioresistance: a therapeutic challenge to improve radiotherapy effectiveness in cancer. Cells. 2020;9(7):1651. https://doi.org/10.3390/cells9071651.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Halperin EC. Particle therapy and treatment of cancer. Lancet Oncol. 2006;7:676–85.

    Article  CAS  PubMed  Google Scholar 

  4. Tobias CA. Failla memorial lecture. the future of heavy-ion science in biology and medicine. Radiat Res. 1985;103:1–33.

    Article  CAS  PubMed  Google Scholar 

  5. Castro JR. Results of heavy ion radiotherapy. Radiat Environ Biophys. 1995;34:45–8.

    Article  CAS  PubMed  Google Scholar 

  6. Kamada T, et al. Carbon ion radiotherapy in Japan: an assessment of 20 years of clinical experience. Lancet Oncol. 2015;16:e93-100.

    Article  PubMed  Google Scholar 

  7. Beltran C, Amos RA, Rong Y. We are ready for clinical implementation of carbon ion radiotherapy in the USA. J Appl Clin Med Phys. 2020;21:6–9.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Helm A, Fournier C. High-LET charged particles: radiobiology and application for new approaches in radiotherapy. Strahlenther Onkol. 2023;199(12):1225–41. https://doi.org/10.1007/s00066-023-02158-7.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Helm A, Fournier C, Durante M. Particle radiotherapy and molecular therapies: mechanisms and strategies towards clinical applications. Expert Rev Mol Med. 2022;24: e8.

    Article  CAS  PubMed  Google Scholar 

  10. Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. Jnci-J Natl Cancer Inst. 2013;105:256–65.

    Article  CAS  Google Scholar 

  11. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.

    Article  CAS  PubMed  Google Scholar 

  12. Jiang X, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18:10.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Fong L, et al. Adenosine 2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov. 2020;10:40–53.

    Article  CAS  PubMed  Google Scholar 

  14. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Galluzzi L, Aryankalayil MJ, Coleman CN, Formenti SC. Emerging evidence for adapting radiotherapy to immunotherapy. Nat Rev Clin Oncol. 2023;20:543–57.

    Article  PubMed  Google Scholar 

  16. Rodriguez-Ruiz ME, Vitale I, Harrington KJ, Melero I, Galluzzi L. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol. 2020;21:120–34.

    Article  CAS  PubMed  Google Scholar 

  17. Golden EB, Marciscano AE, Formenti SC. Radiation therapy and the in situ vaccination approach. Int J Radiat Oncol Biol Phys. 2020;108:891–8.

    Article  PubMed  Google Scholar 

  18. Vanneste BGL, et al. Immunotherapy as sensitizer for local radiotherapy. Oncoimmunology. 2020;9:1832760.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Lee NY, et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol. 2021;22:450–62.

    Article  CAS  PubMed  Google Scholar 

  20. Omuro A, et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: an international randomized phase III trial. Neuro Oncol. 2023;25:123–34.

    Article  CAS  PubMed  Google Scholar 

  21. Lim M, et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. 2022;24:1935–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Keisari Y, Kelson I. The potentiation of anti-tumor immunity by tumor abolition with alpha particles, protons, or carbon ion radiation and its enforcement by combination with immunoadjuvants or inhibitors of immune suppressor cells and checkpoint molecules. Cells. 2021;10(2):228. https://doi.org/10.3390/cells10020228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhou Z, Guan B, Xia H, Zheng R, Xu B. Particle radiotherapy in the era of radioimmunotherapy. Cancer Lett. 2023;567: 216268.

    Article  CAS  PubMed  Google Scholar 

  24. Marcus D, Lieverse RIY, Klein C, Abdollahi A, Lambin P, Dubois LJ, Yaromina A. Charged particle and conventional radiotherapy: current implications as partner for immunotherapy. Cancers. 2021;13(6):1468. https://doi.org/10.3390/cancers13061468.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Galluzzi L, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. 2020;8:e000337.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Durante M, Reppingen N, Held KD. Immunologically augmented cancer treatment using modern radiotherapy. Trends Mol Med. 2013;19:565–82.

    Article  CAS  PubMed  Google Scholar 

  27. Ngwa W, et al. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer. 2018;18:313–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Durante M, Brenner DJ, Formenti SC. Does heavy ion therapy work through the immune system? Int J Radiat Oncol Biol Phys. 2016;96:934–6.

    Article  PubMed  Google Scholar 

  29. Ebner DK, Kamada T, Yamada S. Abscopal effect in recurrent colorectal cancer treated with carbon-ion radiation therapy: 2 case reports. Adv Radiat Oncol. 2017;2:333–8.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Zhang YS, et al. Bystander effect and abscopal effect in recurrent thymic carcinoma treated with carbon-ion radiation therapy: a case report. World J Clinic Cases. 2021;9:6538–43.

    Article  CAS  Google Scholar 

  31. Nakao A, Takiuchi H. A case of lung and lymphnode metastasis treated with carbon ion radiotherapy after radical nephrectomy for renal cell carcinoma. Hinyokika kiyo Acta urologica Japonica. 2008;54:345–7.

    PubMed  Google Scholar 

  32. Huang Q, et al. Biological guided carbon-ion microporous radiation to tumor hypoxia area triggers robust abscopal effects as open field radiation. Front Oncol. 2020;10: 597702.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Sudo, M., Tsutsui, H. and Fujimoto, J. Carbon Ion Irradiation Activates Anti-Cancer Immunity. International journal of molecular sciences 25 (2024).

  34. Pointer KB, Pitroda SP, Weichselbaum RR. Radiotherapy and immunotherapy: open questions and future strategies. Trends in cancer. 2022;8:9–20.

    Article  CAS  PubMed  Google Scholar 

  35. Helm A, Totis C, Durante M, Fournier C. Are charged particles a good match for combination with immunotherapy? Current knowledge and perspectives. Int Rev Cell Mol Biol. 2023;376:1–36.

    Article  CAS  PubMed  Google Scholar 

  36. Nickoloff JA, Sharma N, Taylor L. Clustered DNA double-strand breaks: biological effects and relevance to cancer radiotherapy. Genes. 2020;11(1):99. https://doi.org/10.3390/genes11010099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li P, et al. Mitotic DNA damages induced by carbon-ion radiation incur additional chromosomal breaks in polyploidy. Toxicol Lett. 2014;230:36–47.

    Article  CAS  PubMed  Google Scholar 

  38. Niimi A, et al. Identification of DNA double strand breaks at chromosome boundaries along the track of particle irradiation. Genes Chromosom Cancer. 2016;55:650–60.

    Article  CAS  PubMed  Google Scholar 

  39. Asaithamby A, Hu B, Chen DJ. Unrepaired clustered DNA lesions induce chromosome breakage in human cells. Proc Natl Acad Sci USA. 2011;108:8293–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pantelias A, Zafiropoulos D, Cherubini R, Sarchiapone L, De Nadal V, Pantelias GE, Georgakilas AG, Terzoudi GI. Interphase cytogenetic analysis of G0 lymphocytes exposed to α-particles, C-ions, and protons reveals their enhanced effectiveness for localized chromosome shattering—a critical risk for chromothripsis. Cancers. 2020;12(9):2336. https://doi.org/10.3390/cancers12092336.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hirayama R, Uzawa A, Obara M, Takase N, Koda K, Ozaki M, Noguchi M, Matsumoto Y, Li H, Yamashita K, Koike S, Ando K, Shirai T, Matsufuji N, Furusawa Y. Determination of the relative biological effectiveness and oxygen enhancement ratio for micronuclei formation using high-LET radiation in solid tumor cells: an in vitro and in vivo study. Mut Res/Gene Toxicol Environ Mutagene. 2015;793:41–7. https://doi.org/10.1016/j.mrgentox.2015.08.003.

    Article  CAS  Google Scholar 

  42. Masunaga S, et al. Responses of total and quiescent cell populations in solid tumors to carbon ion beam irradiation (290 MeV/u) in vivo. Radiat Med. 2008;26:270–7.

    Article  PubMed  Google Scholar 

  43. Hu W, et al. Carbon ion irradiation exerts antitumor activity by inducing cGAS-STING activation and immune response in prostate cancer-bearing mice. Cancer Med. 2024;13: e6950.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tang H, Huang H, Guo Z, Huang H, Niu Z, Ji Y, Zhang Y, Bian H, Wentao H. Heavy ion-responsive lncRNA EBLN3P functions in the radiosensitization of non-small cell lung cancer cells mediated by TNPO1. Cancers. 2023;15(2):511. https://doi.org/10.3390/cancers15020511.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tang H, Liu S, Yan X, Jin Y, He X, Hao Huang L, Liu WH, Anqing W. Inhibition of LNC EBLN3P enhances radiation-induced mitochondrial damage in lung cancer cells by targeting the keap1/Nrf2/HO-1 Axis. Biology. 2023;12(9):1208. https://doi.org/10.3390/biology12091208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Meyer JN, Leuthner TC, Luz AL. Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology. 2017;391:42–53.

    Article  CAS  PubMed  Google Scholar 

  47. Walsh DWM, et al. Live cell imaging of mitochondria following targeted irradiation in situ reveals rapid and highly localized loss of membrane potential. Sci Rep. 2017;7:46684.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Yang C, Liang Y, Liu N, Sun M. Role of the cGAS-STING pathway in radiotherapy for non-small cell lung cancer. Radiation Oncol. 2023. https://doi.org/10.1186/s13014-023-02335-z.

    Article  Google Scholar 

  49. Du J, et al. Comparative analysis of the immune responses in cancer cells irradiated with x-ray, proton and carbon-ion beams. Biochem Biophys Res Commun. 2021;585:55–60.

    Article  CAS  PubMed  Google Scholar 

  50. Zhou H, et al. Carbon ion radiotherapy triggers immunogenic cell death and sensitizes melanoma to anti-PD-1 therapy in mice. Oncoimmunology. 2022;11:2057892.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Guo YA, et al. Carbon ion irradiation induces DNA damage in melanoma and optimizes the tumor microenvironment based on the cGAS-STING pathway. J Cancer Res Clin. 2023. https://doi.org/10.1007/s00432-023-04577-6.

    Article  Google Scholar 

  52. Mohamad O, Sishc B, Saha J, Pompos A, Rahimi A, Story M. Carbon ion radiotherapy: a review of clinical experiences and preclinical research, with an emphasis on DNA damage/repair. Cancers. 2017;9(6):66. https://doi.org/10.3390/cancers9060066.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Rødland GE, Temelie M, Mariampillai AE, Hauge S, Gilbert A, Chevalier F, Savu DI, Syljuåsen RG. Potential benefits of combining proton or carbon ion therapy with DNA damage repair inhibitors. Cells. 2024;13(12):1058. https://doi.org/10.3390/cells13121058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gerelchuluun A, et al. The major DNA repair pathway after both proton and carbon-ion radiation is NHEJ, but the HR pathway is more relevant in carbon ions. Radiat Res. 2015;183:345–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mladenova, V., Mladenov, E., Stuschke, M. & Iliakis, G. DNA Damage Clustering after Ionizing Radiation and Consequences in the Processing of Chromatin Breaks. Molecules (Basel, Switzerland) 27 (2022).

  56. Fournier C, et al. Interrelation amongst differentiation, senescence and genetic instability in long-term cultures of fibroblasts exposed to different radiation qualities. Radiother Oncol. 2007;83:277–82.

    Article  CAS  PubMed  Google Scholar 

  57. Maalouf M, et al. Different mechanisms of cell death in radiosensitive and radioresistant p53 mutated head and neck squamous cell carcinoma cell lines exposed to carbon ions and x-rays. Int J Radiat Oncol Biol Phys. 2009;74:200–9.

    Article  CAS  PubMed  Google Scholar 

  58. Tsuboi K, et al. Cell cycle checkpoint and apoptosis induction in glioblastoma cells and fibroblasts irradiated with carbon beam. J Radiat Res. 2007;48:317–25.

    Article  CAS  PubMed  Google Scholar 

  59. Li S, et al. Carbon ion induces cell death and G2/M arrest through pRb/E2F1Chk2/Cdc2 signaling pathway in x-ray resistant B16F10 melanoma cells. Dose-response : A Public Int Hormesis Soc. 2022;20:15593258221092364.

    Article  CAS  Google Scholar 

  60. Paull TT. Mechanisms of ATM activation. Annu Rev Biochem. 2015;84:711–38.

    Article  CAS  PubMed  Google Scholar 

  61. Engeland K. Cell cycle regulation: p53–p21-RB signaling. Cell Death Differ. 2022;29:946–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Deng J, et al. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 2018;8:216–33.

    Article  CAS  PubMed  Google Scholar 

  63. Goel S, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548:471–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Perez RL, Nicolay NH, Wolf J-C, Frister M, Schmezer P, Weber K-J, Huber PE. DNA damage response of clinical carbon ion versus photon radiation in human glioblastoma cells. Radiotherapy Oncol. 2019;133:77–86. https://doi.org/10.1016/j.radonc.2018.12.028.

    Article  CAS  Google Scholar 

  65. Rossiello F, Herbig U, Longhese MP, Fumagalli M, d’Adda di Fagagna F. Irreparable telomeric DNA damage and persistent DDR signalling as a shared causative mechanism of cellular senescence and ageing. Curr Opinion Gene Develop. 2014;26:89–95.

    Article  CAS  Google Scholar 

  66. Zhang X, et al. Both complexity and location of DNA damage contribute to cellular senescence induced by ionizing radiation. PLoS ONE. 2016;11: e0155725.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Chibaya L, Snyder J, Ruscetti M. Senescence and the tumor-immune landscape: Implications for cancer immunotherapy. Semin Cancer Biol. 2022;86:827–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Eriksson D, Stigbrand T. Radiation-induced cell death mechanisms. Tumour Biol: J Int Soc Oncodevelop Biol Med. 2010;31:363–72.

    Article  Google Scholar 

  69. Kobayashi D, et al. Mitotic catastrophe is a putative mechanism underlying the weak correlation between sensitivity to carbon ions and cisplatin. Sci Rep. 2017;7:40588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Shimada M, Hirayama R, Komatsu K. High LET radiation amplifies centrosome overduplication through a pathway of γ-tubulin monoubiquitination. Int J Radiat Oncol Biol Phys. 2013;86:358–65.

    Article  CAS  PubMed  Google Scholar 

  71. Jiao Y, Cao F, Liu H. Radiation-induced cell death and its mechanisms. Health Phys. 2022;123:376–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Olive PL. The role of DNA single- and double-strand breaks in cell killing by ionizing radiation. Radiat Res. 1998;150:S42-51.

    Article  CAS  PubMed  Google Scholar 

  73. Philchenkov A. Radiation-induced cell death: signaling and pharmacological modulation. Crit Rev Oncog. 2018;23:13–37.

    Article  PubMed  Google Scholar 

  74. Riley PA. Free radicals in biology: oxidative stress and the effects of ionizing radiation. Int J Radiat Biol. 1994;65:27–33.

    Article  CAS  PubMed  Google Scholar 

  75. Zheng X, et al. PERK regulates the sensitivity of hepatocellular carcinoma cells to high-LET carbon ions via either apoptosis or ferroptosis. J Cancer. 2022;13:669–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Huang Q, et al. Carbon ion radiotherapy combined with immunotherapy: synergistic anti-tumor efficacy and preliminary investigation of ferroptosis. Cancer Immunol Immunotherapy : CII. 2023;72:4077–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Jin X, et al. Carbon ions induce autophagy effectively through stimulating the unfolded protein response and subsequent inhibiting Akt phosphorylation in tumor cells. Sci Rep. 2015;5:13815.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Sudo M, et al. Autophagy inhibition increased sensitivity of pancreatic cancer cells to carbon ion radiotherapy. Cell Phys Biochem: Int J Experiment Cell Phys, Biochem Pharm. 2023;57:212–25.

    Article  CAS  Google Scholar 

  79. Demaria S, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58:862–70.

    Article  PubMed  Google Scholar 

  80. Formenti SC, Demaria S. Radiation therapy to convert the tumor into an in situ vaccine. Int J Radiat Oncol Biol Phys. 2012;84:879–80.

    Article  PubMed  Google Scholar 

  81. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111.

    Article  CAS  PubMed  Google Scholar 

  82. Gamrekelashvili J, Greten TF, Korangy F. Immunogenicity of necrotic cell death. Cell Mol Life Sci. 2015;72:273–83.

    Article  CAS  PubMed  Google Scholar 

  83. Galluzzi L, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25:486–541.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Vandenabeele P, Vandecasteele K, Bachert C, Krysko O, Krysko DV. Immunogenic apoptotic cell death and anticancer immunity. Adv Exp Med Biol. 2016;930:133–49.

    Article  CAS  PubMed  Google Scholar 

  85. Meier P, Legrand AJ, Adam D, Silke J. Immunogenic cell death in cancer: targeting necroptosis to induce antitumour immunity. Nat Rev Cancer. 2024. https://doi.org/10.1038/s41568-024-00674-x.

    Article  PubMed  Google Scholar 

  86. De Martino M, Daviaud C, Vanpouille-Box C. Radiotherapy: an immune response modifier for immuno-oncology. Semin Immunol. 2021;52: 101474.

    Article  PubMed  Google Scholar 

  87. Garg AD, et al. Molecular and translational classifications of DAMPs in immunogenic cell death. Front Immunol. 2015;6:588.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Ran J, et al. Irradiation-induced changes in the immunogenicity of lung cancer cell lines: based on comparison of x-rays and carbon ions. Front Public Health. 2021;9: 666282.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Yoshimoto Y, et al. Carbon-ion beams induce production of an immune mediator protein, high mobility group box 1, at levels comparable with x-ray irradiation. J Radiat Res. 2015;56:509–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Onishi M, et al. High linear energy transfer carbon-ion irradiation increases the release of the immune mediator high mobility group box 1 from human cancer cells. J Radiat Res. 2018;59:541–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Huang Y, et al. Comparison of the effects of photon, proton and carbon-ion radiation on the ecto-calreticulin exposure in various tumor cell lines. Ann Transl Med. 2019;7:542.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Cubillos-Ruiz JR, Bettigole SE, Glimcher LH. Tumorigenic and immunosuppressive effects of endoplasmic reticulum stress in cancer. Cell. 2017;168:692–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Huang Y, et al. The impacts of different types of radiation on the CRT and PDL1 expression in tumor cells under normoxia and hypoxia. Front Oncol. 2020;10:1610.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Ando K, et al. Intravenous dendritic cell administration enhances suppression of lung metastasis induced by carbon-ion irradiation. J Radiat Res. 2017;58:446–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Takahashi Y, et al. Carbon ion irradiation enhances the antitumor efficacy of dual immune checkpoint blockade therapy both for local and distant sites in murine osteosarcoma. Oncotarget. 2019;10:633–46.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Chakraborty M, et al. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Can Res. 2004;64:4328–37.

    Article  CAS  Google Scholar 

  97. Reits EA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ohkubo Y, et al. Combining carbon ion radiotherapy and local injection of α-galactosylceramide-pulsed dendritic cells inhibits lung metastases in an in vivo murine model. Int J Radiat Oncol Biol Phys. 2010;78:1524–31.

    Article  CAS  PubMed  Google Scholar 

  99. Salomon N, et al. Carbon ion and photon radiation therapy show enhanced antitumoral therapeutic efficacy with neoantigen RNA-LPX vaccines in preclinical colon carcinoma models. Int J Radiat Oncol Biol Phys. 2023. https://doi.org/10.1016/j.ijrobp.2023.12.042.

    Article  PubMed  Google Scholar 

  100. Fridman WH, Zitvogel L, Sautès-Fridman C, Kroemer G. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017;14:717–34.

    Article  CAS  PubMed  Google Scholar 

  101. Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. FEBS J. 2019;286:4160–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Novitskiy SV, et al. TGF-β receptor II loss promotes mammary carcinoma progression by Th17 dependent mechanisms. Cancer Discov. 2011;1:430–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Wei X, et al. Reciprocal expression of IL-35 and IL-10 defines two distinct effector treg subsets that are required for maintenance of immune tolerance. Cell Rep. 2017;21:1853–69.

    Article  CAS  PubMed  Google Scholar 

  104. Zhu S, Wang Y, Tang J, Cao M. Radiotherapy induced immunogenic cell death by remodeling tumor immune microenvironment. Front Immunol. 2022;13:1074477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Shevtsov M, Sato H, Multhoff G, Shibata A. Novel approaches to improve the efficacy of immuno-radiotherapy. Front Oncol. 2019;9:156.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Xu L, et al. Reshaping the systemic tumor immune environment (STIE) and tumor immune microenvironment (TIME) to enhance immunotherapy efficacy in solid tumors. J Hematol Oncol. 2022;15:87.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15:409–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Chiblak S, Tang Z, Lemke D, Knoll M, Dokic I, Warta R, Moustafa M, Mier W, Brons S, Rapp C, Muschal S, Seidel P, Bendzsus M, Adeberg S, Wiestler OD, Haberkorn U, Debus J, Herold-Mende C, Wick W, Abdollahi A. Carbon irradiation overcomes glioma radioresistance by eradicating stem cells and forming an antiangiogenic and immunopermissive niche. JCI Insight. 2019. https://doi.org/10.1172/jci.insight.123837.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Zhou H, et al. Carbon ion radiotherapy boosts anti-tumour immune responses by inhibiting myeloid-derived suppressor cells in melanoma-bearing mice. Cell Death Discov. 2021;7:332.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Spina CS, et al. Differential immune modulation with carbon-ion versus photon therapy. Int J Radiat Oncol Biol Phys. 2021;109:813–8.

    Article  PubMed  Google Scholar 

  111. Sadegh L, Chen PW, Brown JR, Han Z, Niederkorn JY. NKT cells act through third party bone marrow-derived cells to suppress NK cell activity in the liver and exacerbate hepatic melanoma metastases. Int J Cancer. 2015;137:1085–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol. 2023;20:1002–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Ghiringhelli F, Ménard C, Martin F, Zitvogel L. The role of regulatory T cells in the control of natural killer cells: relevance during tumor progression. Immunol Rev. 2006;214:229–38.

    Article  CAS  PubMed  Google Scholar 

  114. Wang J, et al. Carbon ion ((12)C(6+)) irradiation induces the expression of Klrk1 in lung cancer and optimizes the tumor microenvironment based on the NKG2D/NKG2D-Ls pathway. Cancer Lett. 2021;521:178–95.

    Article  CAS  PubMed  Google Scholar 

  115. Allavena P, Sica A, Garlanda C, Mantovani A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol Rev. 2008;222:155–61.

    Article  CAS  PubMed  Google Scholar 

  116. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 2017;14:399–416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Conrad S, Ritter S, Fournier C, Nixdorff K. Differential effects of irradiation with carbon ions and x-rays on macrophage function. J Radiat Res. 2009;50:223–31.

    Article  CAS  PubMed  Google Scholar 

  118. Hartmann L, et al. Photon versus carbon ion irradiation: immunomodulatory effects exerted on murine tumor cell lines. Sci Rep. 2020;10:21517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Miranda S, Vermeesen R, Radstake WE, Parisi A, Ivanova A, Baatout S, Tabury K, Baselet B. Lost in space? Unmasking the T cell reaction to simulated space stressors. Int J Mol Sci. 2023;24(23):16943. https://doi.org/10.3390/ijms242316943.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. König, L. et al. Influence of photon, proton and carbon ion irradiation on differentiation, maturation and functionality of dendritic cells. Frontiers in bioscience (Scholar edition) 14, 2 (2022).

  121. Zhang P, et al. Effects of 12C6+ heavy ion radiation on dendritic cells function. Med Sci Monit. 2018;24:1457–63.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Wu Q, et al. Hypoxia-inducible factors: master regulators of hypoxic tumor immune escape. J Hematol Oncol. 2022;15:77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Gray LH, Conger AD, Ebert M, Hornsey S, Scott OC. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol. 1953;26:638–48.

    Article  CAS  PubMed  Google Scholar 

  124. McKeown SR. Defining normoxia, physoxia and hypoxia in tumours-implications for treatment response. Br J Radiol. 2014;87:20130676.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Ma SR, et al. Blockade of adenosine A2A receptor enhances CD8(+) T cells response and decreases regulatory T cells in head and neck squamous cell carcinoma. Mol Cancer. 2017;16:99.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Srivastava N, Usmani SS, Subbarayan R, Saini R, Pandey PK. Hypoxia: syndicating triple negative breast cancer against various therapeutic regimens. Front Oncol. 2023;13:1199105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Calvo-Asensio I, Dillon ET, Lowndes NF, Ceredig R. The Transcription factor Hif-1 enhances the radio-resistance of mouse MSCs. Front Physiol. 2018;9:439.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Subtil FS, et al. Carbon ion radiotherapy of human lung cancer attenuates HIF-1 signaling and acts with considerably enhanced therapeutic efficiency. Faseb J. 2014;28:1412–21.

    Article  CAS  PubMed  Google Scholar 

  129. Wozny A-S, Gauthier A, Alphonse G, Malésys C, Varoclier V, Beuve M, Brichart-Vernos D, Magné N, Vial N, Ardail D, Nakajima T, Rodriguez-Lafrasse C. Involvement of HIF-1α in the detection, signaling, and repair of DNA double-strand breaks after photon and carbon-ion irradiation. Cancers. 2021;13(15):3833. https://doi.org/10.3390/cancers13153833.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Cartwright IM, Cathy S, Haskins JS, Salinas VA, Sunada S, Hao Y, Uesaka M, Hirakawa H, Chen DJ, Fujimori A, Kato TA. DNA repair deficient chinese hamster ovary cells exhibiting differential sensitivity to charged particle radiation under aerobic and hypoxic conditions. Int J Mol Sci. 2018;19(8):2228. https://doi.org/10.3390/ijms19082228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wennerberg E, Lhuillier C, Vanpouille-Box C, Pilones KA, García-Martínez E, Rudqvist N-P, Formenti SC, Demaria S. Barriers to radiation-induced in situ tumor vaccination. Front Immunol. 2017. https://doi.org/10.3389/fimmu.2017.00229.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Kalbasi A, et al. Tumor-derived CCL2 mediates resistance to radiotherapy in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2017;23:137–48.

    Article  CAS  PubMed  Google Scholar 

  133. Deng LF, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Investig. 2014;124:687–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Permata TBM, et al. High linear energy transfer carbon-ion irradiation upregulates PD-L1 expression more significantly than x-rays in human osteosarcoma U2OS cells. J Radiat Res. 2021;62:773–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Hiam-Galvez KJ, Allen BM, Spitzer MH. Systemic immunity in cancer. Nat Rev Cancer. 2021;21:345–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 2011;29:235–71.

    Article  CAS  PubMed  Google Scholar 

  137. Yang ZR, et al. Peripheral lymphocyte subset variation predicts prostate cancer carbon ion radiotherapy outcomes. Oncotarget. 2016;7:26422–35.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Davuluri R, et al. Lymphocyte nadir and esophageal cancer survival outcomes after chemoradiation therapy. Int J Radiat Oncol Biol Phys. 2017;99:128–35.

    Article  PubMed  Google Scholar 

  139. Zhou L, et al. Low-dose radiation therapy mobilizes antitumor immunity: new findings and future perspectives. Int J Cancer. 2024;154:1143–57.

    Article  CAS  PubMed  Google Scholar 

  140. Herrera FG, Romero P, Coukos G. Lighting up the tumor fire with low-dose irradiation. Trends Immunol. 2022;43:173–9.

    Article  CAS  PubMed  Google Scholar 

  141. Chen Y, Wang C, He M, Zhang H, Chen X. Effect of low dose heavy ion irradiation on subset percentage and cytokines expression of peripheral blood lymphocytes in patients with pancreatic cancer. Zhonghua zhong liu za zhi [Chinese J Oncol]. 2014;36:435–9.

    PubMed  Google Scholar 

  142. Hu W, et al. Immunomodulatory effects of carbon ion radiotherapy in patients with localized prostate cancer. J Cancer Res Clin Oncol. 2023;149:4533–45.

    Article  CAS  PubMed  Google Scholar 

  143. Casbon AJ, et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci USA. 2015;112:E566-575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16:717–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Demple B, Harrison L. Repair of oxidative damage to DNA: enzymology and biology. Annu Rev Biochem. 1994;63:915–48.

    Article  CAS  PubMed  Google Scholar 

  146. Chiblak S, et al. Radiosensitivity of patient-derived glioma stem cell 3-dimensional cultures to photon, proton, and carbon irradiation. Int J Radiat Oncol Biol Phys. 2016;95:112–9.

    Article  PubMed  Google Scholar 

  147. Oonishi K, et al. Different effects of carbon ion beams and X-rays on clonogenic survival and DNA repair in human pancreatic cancer stem-like cells. Radiother Oncol. 2012;105:258–65.

    Article  CAS  PubMed  Google Scholar 

  148. Liang S, Zhou G, Wentao H. Research progress of heavy ion radiotherapy for non-small-cell lung cancer. Int J Mol Sci. 2022;23(4):2316. https://doi.org/10.3390/ijms23042316.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Brownstein JM, et al. Characterizing the potency and impact of carbon ion therapy in a primary mouse model of soft tissue sarcoma. Mol Cancer Ther. 2018;17:858–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Takahashi A, et al. Effects of accelerated carbon-ions on growth inhibition of transplantable human esophageal cancer in nude mice. Cancer Lett. 1998;122:181–6.

    Article  CAS  PubMed  Google Scholar 

  151. Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous and multifaceted. Nat Rev Immunol. 2022;22:173–87.

    Article  CAS  PubMed  Google Scholar 

  152. Kraft, G. The radiobiological and physical basis for radiotherapy with protons and heavier ions. Strahlentherapie und Onkologie: Organ der Deutschen Rontgengesellschaft... [et al] 166, 10–13 (1990).

  153. Friedrich T, Henthorn N, Durante M. Modeling radioimmune response-current status and perspectives. Front Oncol. 2021;11: 647272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Durante M, et al. X-rays versus carbon-ion tumor therapy: cytogenetic damage in lymphocytes. Int J Radiation Oncol Biol Phys. 2000;47:793–8.

    Article  CAS  Google Scholar 

  155. Pignalosa D, et al. Chromosome inversions in lymphocytes of prostate cancer patients treated with x-rays and carbon ions. Radiother Oncol. 2013;109:256–61.

    Article  CAS  PubMed  Google Scholar 

  156. Hartel C, et al. Chromosomal aberrations in peripheral blood lymphocytes of prostate cancer patients treated with IMRT and carbon ions. Radiother Oncol. 2010;95:73–8.

    Article  CAS  PubMed  Google Scholar 

  157. Zhang Z, Liu X, Chen D, Yu J. Radiotherapy combined with immunotherapy: the dawn of cancer treatment. Signal Transduct Target Ther. 2022;7:258.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Shimokawa T, Ma L, Ando K, Sato K, Imai T. The future of combining carbon-ion radiotherapy with immunotherapy: evidence and progress in mouse models. Int J Part Ther. 2016;3:61–70.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Matsunaga A, et al. Carbon-ion beam treatment induces systemic antitumor immunity against murine squamous cell carcinoma. Cancer-Am Cancer Soc. 2010;116:3740–8.

    Google Scholar 

  160. Helm A, et al. Reduction of lung metastases in a mouse osteosarcoma model treated with carbon ions and immune checkpoint inhibitors. Int J Radiat Oncol Biol Phys. 2021;109:594–602.

    Article  PubMed  Google Scholar 

  161. Liu R, et al. Carbon ion irradiation combined with PD-1 inhibitor trigger abscopal effect in lewis lung cancer via a threshold dose. J Cancer. 2024;15:2245–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Hartmann L, et al. Carbon ion irradiation plus CTLA4 blockade elicits therapeutic immune responses in a murine tumor model. Cancer Lett. 2022;550: 215928.

    Article  CAS  PubMed  Google Scholar 

  163. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Okamoto M, Sato H, Gao X, Ohno T. Pembrolizumab after carbon ion radiation therapy for alveolar soft part sarcoma shows a remarkable abscopal effect: a case report. Adv Radiat Oncol. 2022;7: 100893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Solomon I, et al. CD25-T(reg)-depleting antibodies preserving IL-2 signaling on effector T cells enhance effector activation and antitumor immunity. Nature Cancer. 2020;1:1153–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Salomon N, et al. Local radiotherapy and E7 RNA-LPX vaccination show enhanced therapeutic efficacy in preclinical models of HPV16(+) cancer. Cancer Immun, Immun: CII. 2022;71:1975–88.

    Article  CAS  Google Scholar 

  168. Fu C, Jiang A. Dendritic cells and CD8 T cell immunity in tumor microenvironment. Front Immunol. 2018;9:3059.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Lieverse RIY, et al. Stereotactic ablative body radiotherapy (SABR) combined with immunotherapy (L19-IL2) versus standard of care in stage IV NSCLC patients, ImmunoSABR: a multicentre, randomised controlled open-label phase II trial. BMC Cancer. 2020;20:557.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Yan D, Vicini F, Wong J, Martinez A. Adaptive radiation therapy. Phys Med Biol. 1997;42:123–32.

    Article  CAS  PubMed  Google Scholar 

  171. Durante M. New challenges in high-energy particle radiobiology. Br J Radiol. 2014;87:20130626.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Vanpouille-Box C, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Cavalieri S, et al. Toxicity of carbon ion radiotherapy and immune checkpoint inhibitors in advanced melanoma. Radiother Oncol: J Euro Soc Therapeutic Radiol Oncol. 2021;164:1–5.

    Article  CAS  Google Scholar 

  174. Liang X, Mohammadi H, Moreno KC, Beltran CJ, Holtzman AL. Heavy ion particle therapy in modern day radiation oncology. Hematol Oncol Clin North Am. 2024. https://doi.org/10.1016/j.hoc.2024.11.007.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and a project funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

Funding

This work was funded by the National Natural Science Foundation of China (No. 12475350, No. 32071243 and No. 12205215); the Innovation and Entrepreneurship Training Program for College Students of Jiangsu Province (No. 202210285183Y) and College Student Innovation Training Program Project of Soochow University (No. 202410285101Z).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, Shanghai Liu, Siqi Liang and Wentao Hu; Methodology, Xiangyang He, Lu Liu; Software, Xiangyang He; Investigation, Shanghai Liu; Writing—original draft preparation, Shanghai Liu; Writing—review and editing, Wentao Hu, Anqing Wu; Visualization, Lu Liu; Supervision, Wentao Hu, Anqing Wu; Project administration and funding acquisition, Shanghai Liu, Anqing Wu and Wentao Hu.

Corresponding authors

Correspondence to Anqing Wu, Lu Liu or Wentao Hu.

Ethics declarations

Ethical approval and consent to participate

Not applicable.

Consent for publication

All authors have read and agreed to the published version of the manuscript.

Competing interests

The authors declare no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, S., He, X., Liang, S. et al. Carbon ion irradiation mobilizes antitumor immunity: from concept to the clinic. Radiat Oncol 20, 85 (2025). https://doi.org/10.1186/s13014-025-02647-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13014-025-02647-2

Keywords