- Review
- Open access
- Published:
Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects
Journal of Experimental & Clinical Cancer Research volume 44, Article number: 31 (2025)
Abstract
Recent advances in oncology research have highlighted the promising synergy between low-dose radiation therapy (LDRT) and immunotherapies, with growing evidence highlighting the unique benefits of the combination. LDRT has emerged as a potent tool for stimulating the immune system, triggering systemic antitumor effects by remodeling the tumor microenvironment. Notably, LDRT demonstrates remarkable efficacy even in challenging metastatic sites such as the liver (uveal) and brain (cutaneous), particularly in advanced melanoma stages. The increasing interest in utilizing LDRT for secondary metastatic sites of uveal, mucosal, or cutaneous melanomas underscores its potential efficacy in combination with various immunotherapies. This comprehensive review traverses the journey from laboratory research to clinical applications, elucidating LDRT’s immunomodulatory role on the tumor immune microenvironment (TIME) and systemic immune responses. We meticulously examine the preclinical evidence and ongoing clinical trials, throwing light on the promising prospects of LDRT as a complementary therapy in melanoma treatment. Furthermore, we explore the challenges associated with LDRT’s integration into combination therapies, addressing crucial factors such as optimal dosage, fractionation, treatment frequency, and synergy with other pharmacological agents. Considering its low toxicity profile, LDRT presents a compelling case for application across multiple lesions, augmenting the antitumor immune response in poly-metastatic disease scenarios. The convergence of LDRT with other disciplines holds immense potential for developing novel radiotherapy-combined modalities, paving the way for more effective and personalized treatment strategies in melanoma and beyond. Moreover, the dose-related toxicities of immunotherapies may be reduced by synergistic amplification of antitumor efficacy with LDRT.
Introduction
In the landscape of advanced melanoma research, both radiation therapy and immunotherapy have ushered in a transformative era in treatment strategies. Immunotherapy has revolutionized the approach to melanoma treatment by using the body’s immune system to target and eradicate metastatic lesions, offering newfound optimism to both patients and healthcare providers. Melanoma, originating from pigment-producing cells known as melanocytes, manifests in various forms, including cutaneous, mucosal, and ocular melanomas. In its advanced stages (stages 3 and 4), the disease disseminates to distant sites such as lymph nodes, lungs, liver, bones, and brain, culminating in metastatic melanoma [1]. Approximately 90% of uveal melanoma metastasizes to the liver, while cutaneous melanoma has the potential to disseminate to various locations, including lymph nodes, lungs, brain, and soft tissue. According to the American Cancer Society (ACS), an estimated 100,640 adults in the United States will receive a melanoma diagnosis in 2024. The American Society of Clinical Oncology (ASCO) reports that around 4% of individuals are diagnosed with melanomas that have progressed to distant sites, representing the most advanced stage of metastatic melanoma. Data from the National Cancer Institute [2] further corroborates these findings. Approximately 8.5% of individuals diagnosed with melanoma experience spread to nearby lymph nodes, and these cases typically carry a somewhat improved prognosis. Uveal melanoma, characterized by its rarity and insidious nature, presents an incidence rate of 4.6 per million. Unfortunately, metastatic uveal melanoma (mUM) is associated with a bleak prognosis, with a 2-year overall survival rate of only 8% [3,4,5,6,7].
Most patients with metastatic uveal melanoma do not achieve favorable outcomes with immunotherapy, unlike those with cutaneous melanoma, which has a five-year overall survival rate of 50%. This disparity is primarily due to several factors, including poor antigen presentation, a low mutational burden, limited neo-antigenicity, deficient T-cell priming and infiltration, T-cell exhaustion, and the suppressive nature of the tumor stroma [4, 8,9,10]. These factors collectively impede the function and entry of immune effector cells, contributing to the challenges encountered in achieving successful treatment responses [4, 11,12,13]. Hence, it is imperative to devise strategies aimed at sensitizing tumors, effectively transforming “cold” or inert tumors into “hot” tumors, to bolster responses to immunotherapy, particularly in cases that have shown non-responsiveness or recurrence. Low-dose radiation therapy (LDRT) has demonstrated the capacity to transiently induce inflammation within tumors, rendering them more conducive and receptive to immunotherapy across a spectrum of malignancies [3, 14,15,16]. Several types of immunotherapies, including novel approaches like tumor-treating fields and oncolytic viruses, have been tested with LDRT (Fig. 1A).
LDRT, typically administered at doses between 0.5 Gy and 2 Gy, enhances the efficacy of immunotherapies, such as immune checkpoint inhibitors (ICI) or cell therapy, by altering immunosuppressive factors. Recent compelling studies have highlighted LDRT’s significant role in reshaping and modulating the tumor immune microenvironment, as illustrated in Fig. 1B. This modification empowers immune effector cells to launch robust responses against cancer cells [3, 14, 17, 18].
High-dose radiation therapy (HDRT) initiates an in-situ tumor vaccination effect (ISV), prompting systemic antitumor immunity and the noteworthy abscopal effect, wherein distant non-irradiated organs experience immune-mediated destruction of metastatic lesions. However, using HDRT poses risks to both normal surrounding tissues and the viability of tumor-infiltrating lymphocytes (TILs). While radiation therapy can modulate the antitumor immune response, the effective radiation dose to immune cells (EDIC) in circulating blood is pivotal for tumor control. Reducing radiation exposure to the immune system holds promise for enhancing survival outcomes. Thus, there is a growing interest in using LDRT owing to its ability to strengthen anticancer immune responses when combined with immunotherapies [19, 20]. Several prospective studies underscore the increasing significance of LDRT in the context of metastatic melanoma. Notably, LDRT applied to liver metastases has demonstrated a significant enhancement in the immunotherapeutic effectiveness of a dual regimen comprising PD-L1 (atezolizumab) and VEGFA blockade (bevacizumab). This augmentation is attributed to the mobilization of CD8+ T cells into the tumor, facilitated by the CXCL10/CXCR3 axis, mainly observed in hepatocellular carcinoma (HCC) [14]. In advanced uveal melanoma, liver metastases predominate and often exhibit resistance to immunotherapy. However, an intriguing clinical observation arose from a stage IV melanoma patient who achieved a complete response following low-dose radiation therapy (LDRT) targeting liver metastases. This patient received a regimen of 5.6 Gy administered over four fractions (1.4 Gy per fraction), encompassing nearly the entire liver [21]. This case highlights that hepatic metastases, typically resistant due to their immunologically cold nature, can be rendered susceptible to treatment through LDRT.
Low-dose radiation therapy (LDRT) combinations with immunotherapies in metastatic melanomas. A Overview of various immunotherapies, including the most recent and clinically relevant strategies, combined with LDRT for treating metastatic melanoma. These combinations explore synergies between LDRT and immunotherapeutic approaches to enhance therapeutic efficacy. B The combination of LDRT with immunotherapies amplifies the antitumor immune response by reprogramming the tumor immune microenvironment (TIME), shifting it from immunosuppressive to immunostimulatory, thereby targeting metastatic lesions effectively
In preclinical models of immunologically cold melanoma, low-dose whole-brain radiation therapy (LD-WBRT) at 4 Gy in a single fraction significantly reduced intracranial tumors and improved survival rates. Conversely, a high-dose radiation-based in-situ vaccination (ISV) regimen, incorporating 12 Gy in a single fraction, immune-cytokines, and anti-CTLA-4, effectively eradicated primary flank tumors but demonstrated limited efficacy against intracranial tumors [22]. ISV strategies harness tumor tissue as a source of antigens to modulate immune responses and induce systemic antitumor immunity, making them particularly promising in overcoming the immunosuppressive microenvironments characteristic of uveal melanoma. In these settings, tumor cells evade antigen presentation and suppress T-cell activation; however, ISV reprograms the tumor microenvironment (TME) into an immunostimulatory state enriched with activated cytotoxic T-cells capable of attacking tumors.
These findings suggest that even challenging metastatic sites, such as brain metastases from advanced cutaneous melanoma, can be made vulnerable to treatment through LDRT. By reshaping the TME and augmenting immunotherapy responsiveness, LDRT holds potential across diverse forms of metastatic melanoma. This review synthesizes evidence supporting LDRT’s capacity to transform resistant tumor sites and advocates for its future integration with immunotherapeutic strategies in cancer treatment.
Mechanisms induced by low-dose radiation therapy (LDRT) to foster an immune-inflamed tumor microenvironment augmenting the effectiveness of antitumoral immunotherapy
The immunosuppressive landscape in solid tumors is characterized by a combination of physical and cellular barriers that inhibit effective anti-tumor immune responses. These include the presence of an inhibitory stroma, immunosuppressive cell populations such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (M2 phenotype). Additionally, the tumor microenvironment (TME) is enriched with inhibitory cytokines like TGF-β, which collectively create a hostile environment that dampens cytotoxic T-cell activity and impairs the overall immune response against the tumor [3, 23, 24]. Like immuno-oncology agents, LDRT can profoundly remodel the TME, fostering conditions that enhance immune effector cell infiltration and promote tumor eradication by modulating the stromal components. LDRT induces systemic antitumor effects through a range of mechanisms, including the activation and recruitment of immune cells, increased cytokine production, redirection of immune responses toward an antitumor phenotype, modulation of gene expression profiles, and disruption of key immunosuppressive pathways within the TME. These multifaceted effects underscore LDRT’s potential to synergize with immunotherapies for improved cancer treatment outcomes [17, 25]. Due to its ability to enhance systemic antitumor effects, there has been increasing interest in using LDRT as a complementary approach to high-dose radiation therapy (HDRT), chemotherapy, immunotherapy, or cell therapy [26]. HDRT and chemotherapy primarily work by directly killing tumor cells, which stimulate the expression of MHC class I molecules within the tumor microenvironment (TME), a critical factor for immune recognition. This is particularly relevant in cases resistant to anti-PD1 therapy, which often exhibit reduced MHC class I expression. Additionally, HDRT facilitates the release of tumor neoantigens, essential for T-cell priming and diversification of the T-cell receptor (TCR) repertoire. When combined with LDRT, these effects can be further amplified, fostering a more robust and targeted immune response against tumors [27]. The immune response generated at oligometastatic sites far from the localized application of HDRT is termed the abscopal effect (Fig. 2). This phenomenon results in the regression of metastatic lesions distant from the irradiated site, showcasing the systemic potential of radiation therapy. However, HDRT can also induce adverse effects, such as T-cell exhaustion and the upregulation of immunosuppressive factors like TGF-β and regulatory T cells (Tregs) [28].
A visual representation of the immune cell cycle illustrating the effects of high-dose and low-dose radiation. (1) High-dose radiation effectively destroys primary tumor cells, (2) facilitating the release of antigens, and (3) initiating T-cell priming. (4) Immunotherapy reduces T-cell exhaustion and promotes lymphocyte movement to secondary tumor sites. (5) Low-dose radiation, on the other hand, affects the tumor stroma, enhancing the infiltration of natural killer (NK) cells and T cells into secondary tumor sites. This leads to better immune recognition of tumor cells and continuous tumor cell destruction with ongoing antigen release
A novel approach called the Radscopal Technique has been proposed to mitigate these drawbacks. This strategy combines HDRT at the primary tumor site with LDRT targeted at secondary metastatic sites. LDRT enhances the immune-stimulatory effects of HDRT by modulating the TME [24]. Specifically, it promotes the conversion of tumor-associated macrophages (TAMs) from the immunosuppressive M2 phenotype to the pro-inflammatory M1 phenotype, increases chemokine production, recruits T and NK cells, and downregulates TGF-β and Treg-mediated immunosuppression [3, 24]. LDRT can synergize with immunotherapies at various stages, amplifying their efficacy (Fig. 3). By optimizing the TME and immune cell recruitment, this combination strategy holds promise for improving outcomes in metastatic cancers.
Notably, LDRT-led polarization of M1 macrophages results in the secretion of cytokines/chemokines such as IL-12, IFN-γ, and RANTES, which recruit effector T cells and promote normalization of the tumor vasculature. Our recent findings build upon Klug et al.‘s findings, reinforcing that LDRT promotes a phenotypic shift of tumor-associated macrophages (TAMs) from the immunosuppressive M2 state to the antitumor M1 phenotype [15]. This macrophage polarization facilitates the recruitment of CD4 + T cells and NK cells while simultaneously downregulating TGF-β, a key inhibitory cytokine [3]. Expanding on this, we conducted proteomic analyses to examine the TME-specific changes induced by LDRT, revealing elevated levels of Granzyme B, MIP1α, and CD137 (4-1BB) within tumor-infiltrating CD4 + T cells, signifying enhanced activation and effector functionality. These findings underscore the multifaceted role of LDRT in modifying the tumor immune microenvironment [29]. Further support for these insights was identified in a randomized phase II trial evaluating the combination of PD-L1 and CTLA-4 inhibitors with either LDRT (0.5 Gy per fraction) or hypofractionated radiation (HFRT, 3 fractions of 24 Gy) in patients with metastatic colorectal cancer. Both radiation regimens were found to modulate local immune responses and systemic immunogenicity, highlighting the potential of LDRT in synergizing with checkpoint inhibitors [30]. Moreover, preclinical studies demonstrated that applying LDRT to murine tumors enhances T-cell infiltration and amplifies the response to combination immunotherapy driven by interferon-dependent mechanisms. Together, these findings illustrate the profound immunomodulatory effects of LDRT, emphasizing its value as a complementary approach to existing cancer therapies [17].
The potential of LDRT has gained considerable attention in treating tumors that are classified as immune-cold or immune-desert environments, such as liver metastasis or uveal melanoma [21]. Recent studies have highlighted the promising combination of LDRT with ICI, a strategy to overcome the immunosuppressive tumor microenvironment (TME) and improve therapeutic outcomes [3]. LDRT not only alters the physical characteristics of the tumor by reducing intratumoral pressure, but it also facilitates better drug delivery and retention within the tumor, increasing the effectiveness of subsequent therapies [31]. Further enhancing this strategy, the Radscopal Method, which involves delivering LDRT to secondary metastatic sites while applying high-dose radiation therapy (HDRT) to the primary tumor, significantly amplifies the systemic antitumor immune response [24]. This combination approach has improved treatment outcomes by expanding the therapeutic scope of radiation and immunotherapy. Additionally, LDRT, in conjunction with low-dose targeted radionuclide therapy, sensitizes previously immunologically cold tumors to ICI, resulting in a previously unattainable level of responsiveness [3, 24, 32, 33]. Together, these findings underscore the importance of LDRT in reshaping the TME to enhance local tumor control and drive systemic antitumor immunity. The combination of LDRT with immunotherapies presents a promising avenue for improving the treatment of difficult-to-target, immune-resistant tumors [34]. Figure 3 illustrates how immunotherapies interact with the modulated TME to boost immune responses.
Illustration of the mechanism-based application of immunotherapies with LDRT. The release of tumor-associated antigens by high-dose radiation therapy (HDRT) or chemotherapy initiates T-cell priming, further amplified by enhanced T-cell infiltration to metastatic sites using low-dose radiation therapy (LDRT). At this stage, combining immunotherapies with LDRT can be particularly beneficial. The release of chemokines, stromal modulation, and further boosting of T cell priming enhance the effects of ICI (anti-PD1 plus anti-CTLA-4). Additionally, the increased infiltration of T and NK cells helps harness the synergistic effects of adoptive cell therapy (ACT) when combined with LDRT. Moreover, LDRT can convert M2 tumor-associated macrophages (TAMs) to the antitumor M1 type. This also explains the positive abscopal antitumor immune responses observed at metastatic sites when using LDRT combined with immunotherapies
Combination of LDRT and immunotherapies in metastatic melanoma
LDRT has demonstrated significant potential to alter the tumor microenvironment and enhance immune responses. As a result, extensive preclinical and clinical studies have highlighted the benefits of combining LDRT with immunotherapy. The integration of LDRT with treatments like checkpoint inhibitors and cell therapy offers substantial promise in cancer management, primarily due to LDRT’s capacity to strengthen the function of effector cells. Table 1 lists clinical and preclinical studies utilizing LDRT and immunotherapy.
LDRT with checkpoint inhibitors(s)
Immune checkpoint inhibition has significantly improved overall survival rates among melanoma patients. Nonetheless, challenges arise in cases of tumor recurrence following therapy, instances of resistance, and particularly in patients with immunologically cold uveal melanoma. Liver metastases exacerbate the challenge by significantly diminishing immunotherapeutic efficacy, principally due to the minimal immune response elicited in such cases [42]. Remarkably, a stage-IV melanoma patient demonstrated a complete response following LDRT to liver metastases, having undergone pretreatment with T-cell therapy [21]. Preclinical studies utilizing mouse models of melanoma and other cancers have revealed significant tumor control when combining LDRT with ICI, surpassing the efficacy of either treatment alone or in one such study using a B16 melanoma mouse model, LDRT treatment (50 cGy administered twice daily to a total dose of 500 cGy) notably enhanced the immunological response to PDL1 blockade. This dual therapy approach substantially decreased tumor growth, improved overall survival, and led to a remarkable 40% increase in complete response rate [38]. Furthermore, in a preclinical study involving an immunologically cold cutaneous melanoma B78 cell model derived from murine skin cancer B16 cells, flank tumors showed elimination when treated with an ISV regimen of radiation (12 Gy × 1 F), immune-cytokine, and anti-CTLA-4. However, this regimen had minimal impact on intracranial tumors. Interestingly, mice subjected to a precisely timed low-dose whole-brain radiation treatment (WBRT, 4 Gy × 1 F) demonstrated significantly enhanced survival rates and improved control over metastatic tumors [22]. This indicates that LDRT can sensitize an otherwise immunologically resistant tumor to ICB therapy. Low-dose targeted radionuclide therapy (TRT), specifically with 90Y-NM600, has been shown to alter the response of ICB in immunologically cold syngeneic B78 melanoma tumors. Notably, low-dose TRT has been reported to enhance NK cell activity significantly and increase tumor-infiltrating myeloid cells, resulting in an elevated ratio of CD8+ to suppressor T regulatory cells [34]. Neither checkpoint blockade nor LDRT alone significantly impacted melanoma tumor growth. However, combining both treatments led to a dramatic reduction in tumor growth rate, prolonged survival, and a 40% complete response rate in mice receiving dual therapy [17]. Notably, PD-L1 blockade alone enhanced the proliferation, antigen experience, activation, and cytotoxicity of circulating and tumor-resident CD8+ T-cells. In contrast, LDRT alone increased the prevalence of antigen-experienced circulating CD8+ T-cells and activated tumor-resident CD8+ T-cells but had no significant effect on their proliferation in either compartment [38]. These findings demonstrate that while neither LDRT nor checkpoint blockade is sufficient as a standalone treatment, their combination holds significant potential to sensitize immunologically resistant tumors to immunotherapy, offering a promising strategy for clinical application.
LDRT combination with CAR-T cell infusion
The emergence of chimeric antigen receptor (CAR) engineering has propelled the advancement of potent cellular therapies for cancer. Referred to as “living drugs,” these modified immune effector cells have entered clinical practice. While CAR-T cell-based treatments have shown significant clinical success, logistical challenges and associated toxicities remain recognized limitations [5]. These engineered immune cells hold therapeutic promise for cancer-related conditions and inflammatory and infectious diseases [43]. CAR-T cell therapies have transformed the management of hematological malignancies like leukemia and lymphoma. However, effectively treating solid tumors with CAR-T cells remains a significant challenge in the field, with attempts to surmount these obstacles yielding limited success thus far. Numerous preclinical studies in melanoma models have explored CAR T cell approaches targeting various tumor antigens including CD16 [44], HER2 [45], GP100-HLA-A2 complex [46], CD126 [47], VEGFR-2 [48], CD20 & MCSP [49], MCSP [50], gp100 and MCSP [51], GD2 [52], GD3 [53], B7-H3 [54], and CD70 [55]. Likewise, several clinical trials have been undertaken in melanoma, employing CAR-T cells directed against various target tumor antigens, including VEGFR2 [56], GD2 in uveal melanoma [57], cMet [58], hCD70 [59], gp100 [60], NY-ESO-1 [61] IL13Ralpha2 [62], B7H3, and Bispecific B7H3xCD19 [63].
We recently reported a compelling finding that LDRT enhances CAR-T cell infiltration into the solid tumor microenvironment, leading to improved tumor control and extended survival. This approach presents a promising strategy to address the challenges associated with CAR-T therapy in solid tumors [64]. While high-dose radiotherapy leads to cellular detrimental effects, including apoptosis, low-dose radiation (LDR) has shown promise in benefiting patients by stimulating an anti-tumor immune response and addressing antigen escape [65,66,67]. Reports emphasize the synergistic potential of RT and CAR-T cell therapy in controlling tumor growth [68]. In a leukemia model, LDRT alters the tumor-intrinsic transcriptional state in a computable “Death Receptor Score,” which reflects a tumor’s innate sensitivity to CAR-T cells. This Death Receptor Score transiently increases with low-dose total-tumor irradiation (TTI), and independently correlates with outcomes in banked samples from a series of patients with ALL [68]. Furthermore, Weiss et al. showed that a subtherapeutic dose of local RT in combination with CAR-T cells targeting NKG2D showed synergy against mouse glioma by promoting CAR-T cell migration and enhancing their cytolytic function [69]. As illustrated in Fig. 3, multiple tumor models demonstrate that RT induces the release of chemokines such as CXCL9, CXCL10, and CXCL11, which enhance T-cell trafficking. Moreover, RT upregulates adhesion molecules like intracellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) on tumor blood vessels, facilitating CAR-T cell infiltration into the tumor microenvironment. RT also reshapes the immune landscape within the tumor microenvironment (TME) by altering immune cell composition and increasing the expression of immune-stimulatory cytokines, further enhancing the functional activity of effector T cells. RT-induced release of the chemokine IL-8 was co-opted to drive the migration, antitumor efficacy, and persistence of CAR-T cells modified to express the cognate IL-8 receptors CXCR1 and CXCR2 in murine models of human glioma, breast, and pancreatic cancer [70]. Taken together, these effects render a more favorable TME that amplifies the efficacy of CAR-T therapy, as shown in Fig. 4.
Building on these findings, combining LDRT with CAR-T cell therapy offers significant potential for improving tumor response and modifying the TME to support immune cell activity. This combination strategy may also reduce treatment-related toxicities, including autologous graft-versus-host disease (GvHD), cytokine release syndrome, and Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS), while enhancing the overall effectiveness of CAR-T therapy. Such an approach could lower the required therapeutic doses, addressing challenges such as manufacturing time (typically 2–4 weeks for autologous CAR-T therapy) and high costs.
Despite these promising implications, no reports have explored the use of LDRT and CAR-T synergy in melanoma models. Our review highlights this critical research gap, encouraging melanoma researchers to investigate this novel combination in preclinical and clinical settings. By doing so, this area of research could unlock new therapeutic avenues and significantly improve outcomes for melanoma patients.
Radiotherapy amplifies the effectiveness of CAR-T cell therapy in combination treatments. (1,2) Radiation-induced IFN-γ triggers the secretion of chemokines such as CXCL9, CXCL10, and CXCL11, which help guide CAR-T cells to the tumor site. (3) Reducing barriers within the tumor microenvironment (TME), facilitates CAR-T cell infiltration. (4) Radiation also increases the expression of ICAM-1 and VCAM-1 on tumor blood vessels, further aiding CAR-T cell infiltration. (5) Radiotherapy shifts macrophages in the TME from the M2 to the M1 phenotype. (6) Additionally, radiation decreases the presence of tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC). (7) By increasing the expression of proinflammatory cytokines, radiation transforms the TME from an immunosuppressive “cold” state to a more immune-active “hot” state. (8) This enhances the function of infiltrating CAR-T cells and supports the expansion of CAR-T cells. This figure was generated using BioRender.com
LDRT combination with CAR-NK cell infusion
There is increasing interest in exploring alternative immune effector cell types for CAR therapy. Among these, natural killer (NK) cells stand out as a promising choice due to their innate cytotoxic capability and independence from antigen presentation in the MHC pathway. NK cells are integral to the innate immune system and can directly target and kill tumors, contributing to immunosurveillance and antitumor immune responses. Their innate cytotoxic potential against malignancies can be augmented by incorporating CAR technology into NK cells. As observed in Fig. 5, radiation therapy enhances the activity, infiltration, and antitumor efficacy of NK cells within the tumor microenvironment. RT promotes NK cell expansion and activation, increasing cytotoxic potential and cytokine secretion (e.g., IFN-γ and TNF-α) [71]. Ionizing radiation therapy (RT) induces the secretion of chemokines, notably interleukin-8 (IL-8). In ex vivo human pancreatic cancer specimens, RT-induced IL-8 effectively recruits unmodified natural killer (NK) cells, demonstrating a synergistic potential for combining radiation therapy with NK cell-based treatments to enhance cancer therapy outcomes [72]. NK cell therapy provides a significant advantage by functioning independently of major histocompatibility complex (MHC) antigen presentation, positioning it as a promising allogeneic treatment option. Unlike allogeneic CAR-T therapy, which is associated with graft-versus-host disease (GVHD), CAR-NK therapy exhibits minimal toxicity and eliminates the risk of GVHD. This feature eliminates the requirement for human leukocyte antigen (HLA) matching, positioning CAR-NK therapy as a safer and more accessible option for immunotherapy [73, 74].
So far, LDRT has shown positive effects on NK cells in melanoma tumor microenvironment. For example, radiofrequency radiation (RFR) has been found to reprogram the TIME into an antitumor phenotype in pulmonary metastatic melanoma, promoting the active transformation of tumor-infiltrating CD8+ T and NK cells [75]. Furthermore, low-dose radiation (75 to 150 mGy) has significantly enhanced the expansion and secretion of effector proteins such as IFN-γ and TNF-α from NK cells [76, 77]. Similarly, studies involving tumor-bearing rats subjected to low-dose total-body irradiation (0.1–0.2 Gy) reported significantly reduced tumor metastases and increased NK cell cytolytic activity [78, 79]. Despite these findings, no research currently examines the combination of LDRT and CAR-NK therapy in melanoma models. This represents a notable scientific gap, and our review encourages melanoma researchers to explore this promising combination in appropriate preclinical models.
The impact of radiation therapy on NK and CAR-NK cell activity, infiltration, and antitumor cytolytic ability. Radiation therapy is key in enhancing the activity and infiltration of NK and CAR-NK cells within the tumor microenvironment. The timing of radiotherapy mediates several crucial effects: (1) Radiation promotes the expansion of natural killer (NK) cells, boosting their cytotoxic potential. (2) It activates NK cells, stimulating the secretion of effector cytokines like IFN-γ and TNF-α, strengthening their antitumor responses. (3) Radiotherapy reduces tumor burden, facilitating the infiltration of CAR-NK cells into the tumor site. (4) Radiation-induced lymphopenia creates additional space for CAR-NK cell expansion, enhancing their therapeutic efficacy. (5) Radiation also induces the release of chemokines, which attract CAR-NK cells to the tumor, optimizing their localization and activity. The synergy between radiation therapy and CAR-NK cell therapy underscores the potential of this combined approach for cancer treatment
LDRT in combination with cytokines
The synergistic effects of RT and cytokine treatment have gained increasing attention for their ability to enhance the antitumor immune response. Combining LDRT with cytokine therapy significantly enhances antitumor responses [80, 81]. LDRT amplifies the immune-stimulating effects of cytokines, boosting innate immunity and promoting the production of proinflammatory cytokines, thereby strengthening the overall antitumor immune response (Fig. 6). RT serves as a primer for the tumor microenvironment, preparing it for subsequent immunotherapies, including cytokine therapy [82]. Preclinical and clinical studies have demonstrated the potential of combining RT with various cytokines, such as IL-12 and IL-2, to amplify antitumor immune responses [83,84,85].
A phase 2 clinical trial by Bulgareli and Piccinini (2021) evaluated the combination of RT and IL-2 in patients with metastatic melanoma and renal cell carcinoma. Patients received three daily doses of 6–12 Gy of radiation alongside IL-2, administered via continuous infusion over 72 h and repeated every three weeks for a total of 4 cycles. The study found that 15.7% of patients had a partial response, and 36.8% exhibited stable disease, resulting in a disease control rate of 52.6%. These findings suggest that combination therapy could be a promising option for patients with advanced melanoma, especially those undergoing standard treatments [2]. Recent advances have also explored the delivery of immunocytokines [86], such as NHS-IL2, which combines the third-generation tumor necrosis therapy antibody NHS76 with IL-2. This engineered cytokine has shown promise in selectively activating high-affinity IL-2 receptors. In animal models, Heuvel et al. demonstrated that combining NHS-IL2 with low-dose RT and cisplatin led to significant tumor regression (80-100%). In a phase-IB clinical trial for metastatic non-small cell lung carcinoma patients, the combination of local irradiation (4 Gy x 5) and escalating doses of NHS-IL2 (0.15 mg/kg, 0.30 mg/kg, and 0.45 mg/kg) administered intravenously over three consecutive days every three weeks resulted in long-term survival in two patients, one of whom achieved long-term tumor control. Although adverse events like fatigue, anorexia, rash, and thyroid dysfunction were reported, these results support the efficacy of combining low-dose radiation with NHS-IL2 in enhancing antitumor responses [83]. In a murine melanoma model, an in-situ vaccination (ISV) regimen combining RT, intratumoral injection of immune-cytokine (anti-GD2 antibody fused to IL-2), and the ICI anti-CTLA-4 demonstrated strong efficacy in eliminating peripheral flank tumors. However, its effects on intracranial tumors were more modest [85].
IL-12 is critical for Th1 differentiation, CD4 + T cell reactivation, CD8+ T cell and NK cell cytotoxicity, and survival [87]. However, its clinical application is limited by dose-limiting toxicities, with a maximum tolerated dose of rhIL-12 reported as 500 ng/kg in a phase I trial [88]. A fusion of IL-12 (NHS-IL12), heterodimers with DNA/histone-binding antibodies (NHS76), targets neoplastic lesions by binding DNA fragments in histones [89]. In mouse models, NHS-IL12 shows superior antitumor effects compared to recombinant IL-12, mainly via CD8+ T cells. Its efficacy is enhanced with tumor vaccines, radiotherapy, or chemotherapy [90]. While a phase I trial confirmed its safety at doses up to 16.8 µg/kg, no significant antitumor effects were observed in patients [91]. Developing genetically engineered cytokines holds significant potential for advancing antitumor therapies, particularly for aggressive cancers like metastatic melanoma.
These findings underscore the promise of combining low-dose radiotherapy with immunotherapy, highlighting the potential for improved outcomes in treating advanced melanoma and other cancers.
Combining various cytokines with low-dose radiotherapy (LDRT) demonstrates significant direct antitumor potential. LDRT enhances the immune-modulating effects of cytokines by amplifying their ability to stimulate the immune system. This approach directly boosts innate immunity and promotes the production of antitumor proinflammatory cytokines, strengthening the overall antitumor response
Tumor infiltrating lymphocyte therapy combination with LDRT
In a groundbreaking development, the FDA recently approved the first-ever tumor-infiltrating lymphocyte (TIL) therapy, Iovance’s lifileucel, for advanced melanoma. This approval represents a pivotal moment in cancer treatment, signaling a transformative step forward in managing solid tumors. TIL therapy, once overshadowed by approaches such as ICI and chimeric antigen receptor (CAR) T-cell therapies, is experiencing a renaissance. Figure 7 shows the isolation of TILs from a metastatic lesion followed by their expansion. With renewed focus and proven efficacy, TIL therapy’s relevance in treating advanced melanoma is gaining momentum, meriting in-depth exploration of its potential [92]. Adoptive cell therapy using TILs has demonstrated remarkable promise in metastatic melanoma. Clinical responses have been observed in checkpoint inhibitor-naïve and -refractory cutaneous melanoma cases. Notably, uveal melanoma (UM)—a subtype historically resistant to ICI—has shown encouraging clinical results with TIL therapy in early studies. For instance, a phase II trial of lifileucel in advanced melanoma patients who had previously undergone ICI and targeted therapy (BRAF, MEK inhibitors) reported durable and significant responses, reaffirming the potential of TILs even in heavily pretreated individuals [93]. These trials underscore lifileucel’s potential as a one-time treatment for patients with limited therapeutic options, particularly in cases of ICI-refractory disease [94].
Despite the advancements brought by ICIs and targeted therapies, nearly half of patients with advanced melanoma fail to achieve durable responses. This gap has propelled interest in TIL therapy as an alternative. Phase I/II trials of TILs have shown notable clinical efficacy, with phase III data revealing superior progression-free survival (25.8 months) compared to ipilimumab-treated patients (18.9 months) [95]. In contrast to CAR T-cell therapies, which face significant barriers in solid tumors, TILs appear uniquely suited for such malignancies. A phase III randomized clinical trial for metastatic melanoma further highlighted the potential of TIL therapy, with findings from the Netherlands Cancer Institute demonstrating a 50% response rate among treated patients and durable complete responses. These findings also underscored the critical role of neoantigen-specific T-cell reactivity, laying the groundwork for larger multicenter trials [96].
TIL therapy’s efficacy has also been explored in uveal melanoma, a historically challenging subtype to treat. A phase II trial targeting metastatic uveal melanoma achieved objective tumor regression in 35% of patients, with one complete response and six partial responses [17]. Notably, some responders had prior ICI-refractory disease, highlighting TIL therapy’s potential even in heavily pretreated populations [97]. Parallel innovations at MD Anderson Cancer Center have optimized TIL harvesting and expansion using a three-signal culture method involving TCR, 4-1BB stimulation, and high-dose IL-2. This strategy achieved a disease control rate of 66% (22% partial response and 44% stable disease), further advancing TIL therapy’s efficacy [98].
Beyond ex vivo innovations, the tumor microenvironment itself has been manipulated to improve TIL therapy outcomes. Radiation therapy (RT), chemotherapy (CT), and combined chemo-radiation therapy (CRT) have been shown to increase CD3 + and CD8+ TIL density in treated tumors significantly. High pretreatment TIL levels have been positively associated with prolonged disease-free survival (DFS) and overall survival, highlighting the synergistic effects of these modalities [4].
Recent innovations in LDRT have provided additional insights into how TIL therapy can be enhanced. LDRT has improved ex vivo TIL expansion while increasing the antitumor activity of adoptively transferred T cells. Combining TIL therapy with radiation represents a promising strategy to boost response rates, particularly in recurrent or metastatic melanoma [99].
The FDA approval of lifileucel marks a new era in TIL therapy, validating its potential as a viable treatment for advanced melanoma. TIL therapy’s resurgence, supported by clinical trial data and innovations in combination therapies, highlights its role in filling critical gaps left by ICIs and CAR T-cell therapies. The integration of TIL therapy with radiation or other adjuncts not only offers a means to enhance efficacy but also provides a pathway to address the unmet needs of refractory and metastatic disease. As ongoing studies refine this approach, TIL therapy may become a cornerstone of immuno-oncology, offering renewed hope for patients battling advanced melanoma.
Combining oncolytic virus therapy with LDRT
Oncolytic viruses (OVs) represent an innovative therapeutic avenue for targeting metastatic sites in advanced melanoma, such as brain metastases in cutaneous melanoma and liver metastases in uveal melanoma. These engineered viruses selectively infect and lyse tumor cells while simultaneously eliciting immune-mediated tumor regression [6]. Talimogene laherparepvec (T-VEC), the first FDA-approved oncolytic virus, exemplifies this approach. T-VEC is a modified herpes simplex virus engineered to express granulocyte-macrophage colony-stimulating factor (GM-CSF), enhancing immune cell recruitment and tumor destruction. Its approval for unresectable cutaneous melanoma is based on improved response rates compared to GM-CSF alone [100]. Figure 8 illustrates how combining OVs and LDRT may amplify these effects, offering a promising therapeutic synergy.
Numerous oncolytic viruses are under investigation in solid tumors, including melanoma, employing diverse viral vectors and transgenes. Adenovirus 5 and herpes simplex virus (HSV)-based OVs are commonly administered intratumorally and are often engineered with GM-CSF to recruit dendritic cells (DCs) and NK cells [101, 102]. In a phase I clinical trial, the vesicular stomatitis virus (VSV) expressing interferon beta (IFN-β) and tyrosinase-related protein 1 (TYRP1) demonstrated safety and induced dose-dependent immunogenicity and T-cell responses in uveal melanoma [6]. Similarly, the enteric cytopathic human orphan virus type 7 (ECHO-7), also known as Rigvir, exhibited preclinical cytolytic activity against human uveal melanoma cells [103]. A phase 1 trial investigated the intravenous administration of oncolytic adenovirus ICOVIR-5 in patients with cutaneous and uveal melanoma [104]. These findings highlight the versatility and therapeutic potential of OVs in melanoma treatment.
Preclinical studies further support the efficacy of combining OVs with radiotherapy. For example, in an anti-PD-1 refractory melanoma mouse model, combining oncolytic virus particles (OncoVECmGMCSF/mT-VEC) with radiation therapy (6 Gy / 1 fraction) significantly reduced tumor growth and improved survival. This strategy transformed immunologically “cold” tumors into “hot” ones, relying on CD8+ T cell-dependent and IL-1α-dependent mechanisms. A notable clinical case involved a PD-1-refractory patient with cutaneous squamous cell carcinoma who achieved prolonged disease control with a combination of OV, radiation therapy, and ICI [39]. Such evidence underscores the transformative potential of OVs in enhancing immune responses and overcoming resistance mechanisms.
Oncolytic viruses are also being engineered to disrupt DNA damage repair (DDR), rendering tumors more susceptible to radiation therapy. For instance, adenovirus E4orf6 protein inhibits DNA repair pathways, increasing tumor radiosensitivity [105]. The vaccinia virus GLV-1h153 has shown promise in triple-negative breast cancer (TNBC) by increasing intratumoral radionuclide accumulation and enhancing radiation therapy efficacy [41]. Other genetically modified OVs express proteins, such as the sodium iodide symporter (NIS), which selectively facilitates radionuclide uptake, improving radiotherapy precision and safety [106,107,108] (Fig. 8). For example, a vaccinia virus expressing NIS combined with 131I radiation therapy significantly suppressed prostate carcinoma growth compared to either treatment alone [40]. These innovative approaches illustrate the versatility of OVs in improving radiotherapy outcomes. An oncolytic NDV expressing an anti-CTLA4 antibody acted as a radio-enhancing agent, synergizing with standard radiation to enhance tumor repression [109].
Despite advancements in immune checkpoint inhibitors, metastatic uveal melanoma (mUM) continues to exhibit limited responses due to its low tumor mutational burden (TMB) and reduced PD-L1 expression, which impairs neoantigen recognition by tumor-specific T cells [110,111,112]. Tebentafusp, an immune-mobilizing monoclonal T-cell receptor (TCR) therapeutic, represents a significant breakthrough for mUM, targeting HLA-A0201 and gp100 antigens with a median overall survival of 15.3 months [113]. However, many mUM patients are HLA-A0201 negative and ineligible for this therapy, underscoring the urgent need for alternative treatments. Oncolytic viruses present a promising immunotherapeutic option for these patients, offering a novel mechanism to stimulate anti-tumor immunity and potentially overcome resistance to existing therapies. Integrating oncolytic viruses with radiation therapy and immunotherapy has the potential to revolutionize melanoma treatment. These strategies aim to address the unmet needs of metastatic uveal melanoma patients, providing hope for improved outcomes through innovative and combinatorial approaches.
Combination of Tumor Treating fields (TTFs) with LDRT
Tumor Treating Fields (TTFields) represent a promising loco-regional treatment approach that utilizes low-intensity alternating electric fields (1–3 V/cm) with frequencies ranging from 100 to 400 kHz. These electric fields are delivered transdermally to tumors using two transducer arrays activated sequentially every second. This activation induces directional shifts in the electric field targeting the tumor. TTFields provide a non-invasive cancer therapy by disrupting cellular structures in metastatic tumors. The optimal alternating current (AC) frequency is tumor-specific, with glioblastoma, for instance, showing maximum benefit at 200 kHz [114]. The directionality of the applied electric field influences the efficacy of TTFields. Initial hypotheses suggested that TTFields acted on polarizable intracellular structures to disrupt mitosis (Fig. 9). Subsequent studies have confirmed this, demonstrating that the treatment’s effectiveness depends on the direction and amplitude of the electric field [114, 115]. This suggests that the efficacy of TTFields, as observed by subcellular structures, including its amplitude of effect, may depend on the direction of the imposed electric field. Kirson and colleagues further hypothesized that periodically changing the field direction would enhance efficacy, validated for direction-change intervals of 1 and 2 Hz. The mechanism of action of TTFields includes inducing antimitotic cell cycle arrest, triggering autophagy and endoplasmic reticulum (ER) stress in cancer cells, and causing DNA damage and replication stress. Notably, TTFields also stimulate antitumor immune responses. Tumors treated with TTFields exhibit increased infiltration of immune cells, including CD45 + cells, CD4+, and CD8 + lymphocytes, creating an immunologically “hot” tumor microenvironment compared to untreated “cold” tumors [116, 117].
Studies have highlighted a remarkable synergy between TTFields and ionizing RT. For instance, TTFields combined with RT effectively suppress cell migration and invasion in glioblastoma multiforme (GBM) cells by inhibiting proteins such as MMP-9 and vimentin [118]. These findings underscore TTFields’ potential to enhance RT outcomes by inhibiting double-stranded DNA damage repair, inducing mitotic catastrophe, and reducing tumor cell survival (Fig. 9) [119]. Based on this synergy, Novocure conducts clinical trials using TTFields in metastatic uveal melanoma (mUM). One such trial (NCT05004025) evaluates the NovoTTF-2001 device in combination with ICI, nivolumab, and ipilimumab. As many uveal melanoma cases recur in the liver, array patches are strategically placed on the abdomen and worn for at least 18 h daily. TTFields have already demonstrated efficacy in brain tumors, including glioblastoma multiforme, with FDA approval at 200 kHz for newly diagnosed and recurrent GBM and 150 kHz for malignant pleural mesothelioma.
In glioblastoma, pilot studies assessing the concurrent use of TTFields with RT and temozolomide (TMZ) in newly diagnosed patients have demonstrated safety and feasibility [120]. This combination significantly enhances therapeutic outcomes, making TTFields one of the most innovative and advanced modalities to complement radiation therapy in metastatic melanoma.
Tumor Treating Fields (TTFs) disrupt tumor mitosis and engage other mechanisms, including a potent synergistic antitumor immune response when combined with radiation therapy. The combination of TTFs and low-dose radiation therapy (LDRT) holds significant promise but requires detailed investigation. This area of research offers considerable potential for further exploration and development
Exploring the potential integration of cancer vaccines and low-dose radiation therapy (LDRT) in metastatic melanoma
Combining low-dose radiation therapy and cancer vaccines: a promising strategy
Integrating radiation therapy, particularly LDRT, with cancer vaccines offers a promising new approach for managing metastatic solid tumors [22, 121,122,123,124,125,126]. Cancer vaccines, a form of immunotherapy, aim to educate the immune system to recognize and eliminate abnormal tumor cells. However, unlike vaccines for infectious diseases, the development of cancer vaccines is inherently complex. The challenges stem from the similarity between cancer and normal cells and the heterogeneity of tumor-specific antigens, which vary significantly between individuals [127, 128]. Cancer vaccines aim to stimulate anti-tumor immune responses by delivering tumor-associated antigens in various forms, including tumor cells, viruses, antigen-presenting cells (APCs) or dendritic cells, peptides, DNA, and RNA. Their primary goal is to overcome the immune-suppressive tumor environment and elicit robust cellular and humoral immune responses. These vaccines are classified into four main platforms based on their preparation methods: cell-based, virus-based, peptide-based, and nucleic acid-based vaccines [129, 130] (Fig. 10). Dendritic cell (DC) vaccines have shown significant efficacy in clinical trials. Nucleic acid vaccines, such as DNA and mRNA vaccines, have also gained traction due to their ability to encode tumor-associated antigens, initiate selective tumor responses, and generate robust immune activation [131,132,133,134].
Radiation therapy as an in-situ vaccine
RT has been demonstrated to act as an in-situ vaccine by triggering systemic immune responses against localized tumors. This phenomenon, known as the abscopal effect, has been observed across various tumor types, including melanoma, non-small cell lung cancer, renal cell carcinoma, and hepatocellular carcinoma [110]. Through mechanisms such as enhanced antigen presentation and T-cell activation, RT destroys cancer cells locally and primes the immune system to target distant metastatic lesions [135, 136]. Indeed, mounting evidence supports the synergistic effects of RT and immunotherapies. These immune-modulating effects have made RT an attractive partner for cancer vaccines, with evidence suggesting that the combination significantly improves clinical outcomes, including disease-free survival and overall survival, across diverse cancer types [137,138,139]. Thus, radiation therapy’s ability to induce a tumor-specific immune response has led to its recognition as an in-situ vaccine.
Enhancing cancer vaccines with Low-dose radiation therapy
The combination of LDRT and cancer vaccines is increasingly recognized for its potential to amplify tumor-specific immune responses (Fig. 10). For instance, in metastatic melanoma models, LDRT applied to the brain, combined with in-situ vaccines and anti-CTLA-4 therapy, transformed immunologically “cold” tumors into “hot” tumors, characterized by increased immune cell infiltration [22]. Similarly, combining mRNA-based vaccines with radiation therapy has demonstrated robust synergistic anti-tumor effects, including delayed tumor growth and complete tumor eradication in some cases. Gene expression analyses revealed increased antigen presentation, immune cell adhesion, innate immune system activation, significant downregulation of tumor-associated factors, and upregulation of tumor suppressors [140].
Radiation priming of immune responses
Studies by Lugade et al. have highlighted the role of RT in priming immune responses. In murine melanoma models, RT increased antigen-presenting cells and IFN-γ-secreting T cells, enhancing tumor-specific CD8+ T-cell infiltration. Further investigation ha shown that RT can re-sensitize tumors resistant to anti-PD-1 therapy by increasing type I interferon secretion and MHC class I expression [141, 142]. A notable example includes a study combining RT with SD-101, a TLR9 agonist, which successfully converted “cold” tumors into T-cell-inflamed, “hot” tumors, thereby overcoming checkpoint blockade resistance [125, 143].
LDRT and dendritic cell vaccines: a synergistic approach
Emerging evidence suggests that exposing dendritic cells (DCs) to LDRT enhances their immunogenic potential during vaccine preparation. In preclinical models, LDRT-exposed DCs demonstrated superior migratory capacity, increased T-cell proliferation induction, and heightened cytotoxic T lymphocyte (CTL) activity. Mice treated with DC vaccines exposed to LDRT exhibited improved survival, enhanced CTL infiltration into tumors, and increased tumor cell apoptosis. Upregulated serum levels of IFN-γ and interleukin-12 accompanied these effects [124]. Furthermore, LDRT has been shown to induce a hormetic effect, enhancing the activity of T cells and NK cells, and promoting the production of IL-12, a cytokine critical for Th1 differentiation and CTL activation [121,122,123, 144,145,146,147].
Moreover, a single exposure of DCs to LDRT at a dose of 0.2 Gy has been shown to significantly enhance the production of IL-12, a pivotal cytokine that drives the differentiation of naive T cells into Th1 cells and activates CD8 + CTLs. This increase in IL-12 production amplifies the DCs’ capacity to induce robust T-cell proliferation, thereby strengthening anti-tumor immune responses [148]. Additionally, another study demonstrated that exposure to LDRT at the same dose markedly improved the migratory ability of DCs, enabling them to reach lymphoid tissues and present antigens to T cells more effectively [149]. Together, these findings underscore the potential of integrating optimized doses of LDRT with advanced, specific cancer vaccines to enhance immune priming and activation. This combined approach holds great promise for overcoming the challenges associated with advanced melanoma, paving the way for more effective therapeutic strategies.
Conclusions
LDRT has emerged as a transformative approach in treating metastatic melanoma, marking a significant advancement in oncology. Extensive research has highlighted LDRT’s unique ability to remodel the tumor microenvironment, fostering conditions that support a robust immune response. By stimulating the infiltration, activation, and functionality of immune cells essential for tumor destruction, LDRT enhances the efficacy of immunotherapeutic strategies. When integrated with immunotherapies—such as ICI, CAR-T cell therapy, cytokine therapy, TIL therapy, oncolytic virus therapy, and cancer vaccines—LDRT exhibits remarkable synergistic effects. These combinations improve tumor control and patient survival and address critical challenges, including tumor recurrence and resistance to conventional treatments. Additionally, LDRT facilitates the conversion of immunologically “cold” tumors into “hot” tumors, significantly enhancing their responsiveness to immunotherapy.
Overall, LDRT offers a compelling avenue for advancing metastatic melanoma treatment, providing new hope for improved outcomes and expanding therapeutic options for patients battling this aggressive disease. Its potential to reshape the standard of care underscores the importance of further research and clinical exploration to unlock its full capabilities.
Future directions
The combination of LDRT and immunotherapy holds great promise in advancing the treatment landscape for metastatic melanoma. To fully harness its potential, several critical areas warrant further exploration. Optimizing treatment regimens is essential to maximize therapeutic efficacy while minimizing adverse effects. This optimization involves systematically investigating the sequencing, dosing schedules, and specific combinations of LDRT with various immunotherapeutic agents to determine the most effective protocols. Personalized treatment strategies are also paramount. Identifying reliable biomarkers to guide patient selection and tailor therapies to individual tumor profiles can significantly enhance treatment outcomes. Emerging combinations of LDRT with TTFs and oncolytic viruses represent exciting avenues. With their advanced antitumor mechanisms, these novel modalities demand comprehensive exploration to unlock their full therapeutic potential. Additionally, understanding and addressing mechanisms of treatment resistance remains a critical focus. Insights into resistance pathways can inform the development of innovative strategies to overcome these challenges and sustain durable responses. Beyond metastatic melanoma, LDRT’s applications are increasingly being considered for other cancers where immunotherapy has shown efficacy. Expanding research into integrating LDRT with targeted therapies, chemotherapy, and other novel approaches could reveal synergistic effects, broadening the spectrum of therapeutic possibilities. Integrating LDRT with immunotherapy marks a transformative shift in managing metastatic melanoma. However, realizing its full potential hinges on continued rigorous research, including well-designed preclinical studies and clinical trials. These efforts are crucial to refining therapeutic strategies, improving patient outcomes, and expanding the applicability of this innovative approach across diverse cancer types.
Data availability
Not applicable.
Abbreviations
- LDRT:
-
Low-Dose Radiation Therapy
- HDRT:
-
High-Dose Radiation Therapy
- ICIs:
-
Immune Checkpoint Inhibitors
- CAR-T:
-
Chimeric Antigen Receptor T cells
- TME:
-
Tumor Microenvironment
- TIL:
-
Tumor-Infiltrating Lymphocytes
- ISV:
-
In-Situ Vaccination
- WBRT:
-
Whole-Brain Radiation Therapy
- TTField:
-
Tumor Treating Fields
- TRT:
-
Targeted Radionuclide Therapy
- PD-L1:
-
Programmed Death-Ligand 1
- NK:
-
Natural Killer (cells)
- CD8+:
-
Cluster of Differentiation 8 Positive (T-cells)
- CTLA-4:
-
Cytotoxic T-Lymphocyte Antigen 4
- mUM:
-
Metastatic Uveal Melanoma
- HCC:
-
Hepatocellular Carcinoma
- VEGFA:
-
Vascular Endothelial Growth Factor A
- CT:
-
Chemotherapy
- CRT:
-
Chemo-Radiation Therapy
- PD:
-
Progressive Disease
- DFS:
-
Disease-Free Survival
- CXCL10/CXCR3:
-
Chemokine Ligand 10 / Chemokine Receptor 3
- GM-CSF:
-
Granulocyte-Macrophage Colony-Stimulating Factor
- OV:
-
Oncolytic Virus
- TCR:
-
T Cell Receptor
- CI:
-
Confidence Interval
- PD-1:
-
Programmed Cell Death Protein 1
- VEGFR:
-
Vascular Endothelial Growth Factor Receptor
- IL-12:
-
Interleukin 12
- ICAM-1:
-
Intercellular Adhesion Molecule 1
- VCAM-1:
-
Vascular Cell Adhesion Molecule 1
- IFN-γ:
-
Interferon Gamma
- ECM:
-
Extracellular Matrix
- MDSC:
-
Myeloid-Derived Suppressor Cells
- CAF:
-
Cancer-Associated Fibroblasts
- PD1/PD-L1:
-
Programmed Death 1 / Programmed Death Ligand 1
- MHC:
-
Major Histocompatibility Complex
References
Ali Z, Yousaf N, Larkin J. Melanoma epidemiology, biology and prognosis. EJC Suppl. 2013;11:81–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejcsup.2013.07.012.
Bulgarelli J, et al. Radiotherapy and High-Dose Interleukin-2: clinical and immunological results of a proof of Principle Study in Metastatic Melanoma and Renal Cell Carcinoma. Front Immunol. 2021;12:778459. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.778459.
Barsoumian HB, et al. Low-dose radiation treatment enhances systemic antitumor immune responses by overcoming the inhibitory stroma. J Immunother Cancer. 2020;8. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-000537.
Blach J, Wojas-Krawczyk K, Nicos M, Krawczyk P. Failure of immunotherapy-the molecular and immunological origin of immunotherapy resistance in lung cancer. Int J Mol Sci. 2021;22. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22169030.
Krantz BA, Dave N, Komatsubara KM, Marr BP, Carvajal RD. Uveal melanoma: epidemiology, etiology, and treatment of primary disease. Clin Ophthalmol. 2017;11:279–89. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/OPTH.S89591.
Smith KER, et al. A phase I oncolytic virus trial with vesicular stomatitis virus expressing human interferon beta and tyrosinase related protein 1 administered intratumorally and intravenously in uveal melanoma: safety, efficacy, and T cell responses. Front Immunol. 2023;14:1279387. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1279387.
Xu Y, et al. Epidemiological study of uveal melanoma from US surveillance, epidemiology, and end results program (2010–2015). J Ophthalmol. 2020;2020(3614039). https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2020/3614039.
Steininger J, et al. Systemic therapy of metastatic melanoma: on the road to cure. Cancers (Basel). 2021;13. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13061430.
Heppt MV, et al. Combined immune checkpoint blockade for metastatic uveal melanoma: a retrospective, multi-center study. J Immunother Cancer. 2019;7:299. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40425-019-0800-0.
Szeligo BM, Ivey AD, Boone BA. Poor response to checkpoint immunotherapy in uveal melanoma highlights the persistent need for innovative regional therapy approaches to manage liver metastases. Cancers (Basel). 2021;13. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13143426.
Wang MM, Coupland SE, Aittokallio T, Figueiredo CR. Resistance to immune checkpoint therapies by tumour-induced T-cell desertification and exclusion: key mechanisms, prognostication and new therapeutic opportunities. Br J Cancer. 2023;129:1212–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41416-023-02361-4.
Westcott PMK, et al. Low neoantigen expression and poor T-cell priming underlie early immune escape in colorectal cancer. Nat Cancer. 2021;2:1071–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s43018-021-00247-z.
Zhou X, et al. Mechanisms of tumor resistance to immune checkpoint blockade and combination strategies to overcome resistance. Front Immunol. 2022;13:915094. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.915094.
Li S, et al. Low-dose radiotherapy combined with dual PD-L1 and VEGFA blockade elicits antitumor response in hepatocellular carcinoma mediated by activated intratumoral CD8(+) exhausted-like T cells. Nat Commun. 2023;14:7709. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-43462-1.
Klug F, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013;24:589–602. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccr.2013.09.014.
Hanahan D. Hallmarks of Cancer: New dimensions. Cancer Discov. 2022;12:31–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2159-8290.CD-21-1059.
Herrera FG, et al. Low-dose Radiotherapy reverses Tumor Immune Desertification and Resistance to Immunotherapy. Cancer Discov. 2022;12:108–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2159-8290.CD-21-0003.
Zhou L, et al. Low-dose radiation therapy mobilizes antitumor immunity: New findings and future perspectives. Int J Cancer. 2024;154:1143–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ijc.34801.
Jin JY, et al. Higher radiation dose to the immune cells correlates with worse tumor control and overall survival in patients with stage III NSCLC: a secondary analysis of RTOG0617. Cancers (Basel). 2021;13. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers13246193.
Gough MJ, Crittenden MR. The paradox of radiation and T cells in tumors. Neoplasia. 2022;31:100808. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neo.2022.100808.
He K, et al. Novel use of low-dose Radiotherapy to modulate the Tumor Microenvironment of Liver metastases. Front Immunol. 2021;12:812210. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.812210.
Clark PA, et al. Low-dose Radiation potentiates the propagation of Anti-tumor immunity against Melanoma Tumor in the brain after in situ vaccination at a Tumor outside the brain. Radiat Res. 2021;195:522–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1667/RADE-20-00237.1.
Davis RJ, Van Waes C, Allen CT. Overcoming barriers to effective immunotherapy: MDSCs, TAMs, and Tregs as mediators of the immunosuppressive microenvironment in head and neck cancer. Oral Oncol. 2016;58:59–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.oraloncology.2016.05.002.
Barsoumian HB, et al. The RadScopal technique as an Immune adjuvant to treat Cancer. Immuno. 2023;3:74–85.
Gao L, Zhang A. Low-dose radiotherapy effects the progression of anti-tumor response. Transl Oncol. 2023;35:101710. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tranon.2023.101710.
Wang J, et al. Exploring low-dose radiotherapy to overcome radio-immunotherapy resistance. Biochim Biophys Acta Mol Basis Dis. 2023;1869:166789. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbadis.2023.166789.
Ozpiskin OM, Zhang L, Li JJ. Immune targets in the tumor microenvironment treated by radiotherapy. Theranostics. 2019;9:1215–31. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.32648.
Golden EB, Demaria S, Schiff PB, Chachoua A, Formenti SC. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol Res. 2013;1:365–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2326-6066.CIR-13-0115.
Patel RR, et al. Use of Multi-site Radiation Therapy for systemic Disease Control. Int J Radiat Oncol Biol Phys. 2021;109:352–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijrobp.2020.08.025.
Monjazeb AM, et al. A randomized trial of combined PD-L1 and CTLA-4 inhibition with targeted low-dose or Hypofractionated Radiation for patients with metastatic colorectal Cancer. Clin Cancer Res. 2021;27:2470–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-20-4632.
Barsoumian HB, et al. Radiation Therapy modulates Tumor Physical characteristics to reduce Intratumoral pressure and enhance Intratumoral Drug Delivery and Retention. Adv Radiat Oncol. 2023;8:101137. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.adro.2022.101137.
Savage T, Pandey S, Guha C. Postablation Modulation after single high-dose Radiation Therapy improves Tumor Control via enhanced immunomodulation. Clin Cancer Res. 2020;26:910–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-18-3518.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20052494.
Patel RB, et al. Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade. Sci Transl Med. 2021;13. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scitranslmed.abb3631.
Safwat A, Aggerholm N, Roitt I, Overgaard J, Hokland M. Low-dose total body irradiation augments the therapeutic effect of interleukin-2 in a mouse model for metastatic malignant melanoma. J Exp Ther Oncol. 2003;3:161–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1359-4117.2003.01093.x.
Safwat A, Aggerholm N, Roitt I, Overgaard J, Hokland M. Tumour burden and interleukin-2 dose affect the interaction between low-dose total body irradiation and interleukin 2. Eur J Cancer. 2004;40:1412–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejca.2004.01.037.
Jin SZ, Pan XN, Wu N, Jin GH, Liu SZ. Whole-body low dose irradiation promotes the efficacy of conventional radiotherapy for cancer and possible mechanisms. Dose Response. 2007;5:349–58. https://doiorg.publicaciones.saludcastillayleon.es/10.2203/dose-response.07-020.Jin.
Vezys V. L. N. S. B. N. P. A. S. A. L. C. N. C. T. W. N. Tumor-targeted low-dose radiation and immunotherapy in mouse models of melanoma. Int J Radiat Oncol Biol Phys. 2020;108:E556–7.
Jhawar SR, et al. Combination oncolytic virus, radiation therapy, and immune checkpoint inhibitor treatment in anti-PD-1-refractory cancer. J Immunother Cancer. 2023;11. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2023-006780.
Mansfield DC, et al. Oncolytic vaccinia virus as a vector for therapeutic sodium iodide symporter gene therapy in prostate cancer. Gene Ther. 2016;23:357–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/gt.2016.5.
Gholami S, et al. Vaccinia virus GLV-1h153 in combination with 131I shows increased efficiency in treating triple-negative breast cancer. FASEB J. 2014;28:676–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.13-237222.
Pelster MS, et al. Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: results from a single-arm phase II study. J Clin Oncol. 2021;39:599–607. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.20.00605.
Aghajanian H, Rurik JG, Epstein JA. CAR-based therapies: opportunities for immuno-medicine beyond cancer. Nat Metab. 2022;4:163–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42255-022-00537-5.
Rataj F, et al. High-affinity CD16-polymorphism and Fc-engineered antibodies enable activity of CD16-chimeric antigen receptor-modified T cells for cancer therapy. Br J Cancer. 2019;120:79–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41416-018-0341-1.
Forsberg EMV, et al. HER2 CAR-T cells eradicate uveal melanoma and T-cell therapy-resistant human melanoma in IL2 Transgenic NOD/SCID IL2 receptor knockout mice. Cancer Res. 2019;79:899–904. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-18-3158.
Zhang G, et al. Anti-melanoma activity of T cells redirected with a TCR-like chimeric antigen receptor. Sci Rep. 2014;4:3571. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep03571.
Mishra AK, Kemler I, Dingli D. Preclinical development of CD126 CAR-T cells with broad antitumor activity. Blood Cancer J. 2021;11:3. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41408-020-00405-z.
Chinnasamy D, et al. Simultaneous targeting of tumor antigens and the tumor vasculature using T lymphocyte transfer synergize to induce regression of established tumors in mice. Cancer Res. 2013;73:3371–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-12-3913.
Schmidt P, et al. Eradication of melanomas by targeted elimination of a minor subset of tumor cells. Proc Natl Acad Sci U S A. 2011;108:2474–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1009069108.
Harrer DC, et al. RNA-transfection of gamma/delta T cells with a chimeric antigen receptor or an alpha/beta T-cell receptor: a safer alternative to genetically engineered alpha/beta T cells for the immunotherapy of melanoma. BMC Cancer. 2017;17:551. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-017-3539-3.
Simon B, Harrer DC, Schuler-Thurner B, Schuler G, Uslu U. Arming T cells with a gp100-Specific TCR and a CSPG4-Specific CAR using combined DNA- and RNA-based receptor transfer. Cancers (Basel). 2019;11. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers11050696.
Gargett T, et al. GD2-specific CAR T cells undergo potent activation and deletion following Antigen Encounter but can be protected from activation-induced cell death by PD-1 blockade. Mol Ther. 2016;24:1135–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/mt.2016.63.
Lo AS, Ma Q, Liu DL, Junghans RP. Anti-GD3 chimeric sFv-CD28/T-cell receptor zeta designer T cells for treatment of metastatic melanoma and other neuroectodermal tumors. Clin Cancer Res. 2010;16:2769–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-10-0043.
Zhang Z, et al. B7-H3-Targeted CAR-T cells exhibit potent Antitumor effects on hematologic and solid tumors. Mol Ther Oncolytics. 2020;17:180–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.omto.2020.03.019.
Yang M, et al. Tandem CAR-T cells targeting CD70 and B7-H3 exhibit potent preclinical activity against multiple solid tumors. Theranostics. 2020;10:7622–34. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.43991.
Yu J, et al. Anti-GD2/4-1BB chimeric antigen receptor T cell therapy for the treatment of Chinese melanoma patients. J Hematol Oncol. 2018;11:1. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-017-0548-2.
Cramer SF. The origin of epidermal melanocytes. Implications for the histogenesis of nevi and melanomas. Arch Pathol Lab Med. 1991;115:115–9.
Matthews NH, Li WQ, Qureshi AA, Weinstock MA, Cho E. In: Ward WH, Farma JM, editors. Cutaneous melanoma: etiology and therapy. 2017.
Yamaguchi Y, Hearing VJ. Melanocytes and their diseases. Cold Spring Harb Perspect Med. 2014;4. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a017046.
Fioranelli M, et al. A new medical imaging technique for diagnosing dermatologic diseases: a clue to treatment choices. Dermatol Ther. 2020;33:e13405. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/dth.13405.
Sandru A, Voinea S, Panaitescu E, Blidaru A. Survival rates of patients with metastatic malignant melanoma. J Med Life. 2014;7:572–6.
Naidoo C, Kruger CA, Abrahamse H. Photodynamic therapy for metastatic melanoma treatment: a review. Technol Cancer Res Treat. 2018;17:1533033818791795. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1533033818791795.
Barden H, Levine S. Histochemical observations on rodent brain melanin. Brain Res Bull. 1983;10:847–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0361-9230(83)90218-6.
Puebla-Osorio N, et al. Enhanced tumor control and survival in preclinical models with adoptive cell therapy preceded by low-dose radiotherapy. Front Oncol. 2024;14:1407143. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2024.1407143.
Ina Y, Sakai K. Activation of immunological network by chronic low-dose-rate irradiation in wild-type mouse strains: analysis of immune cell populations and surface molecules. Int J Radiat Biol. 2005;81:721–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/09553000500519808.
Lacoste-Collin L, et al. Effect of continuous irradiation with a very low dose of gamma rays on life span and the immune system in SJL mice prone to B-cell lymphoma. Radiat Res. 2007;168:725–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1667/RR1007.1.
DeSelm C, et al. Low-dose Radiation Conditioning enables CAR T cells to mitigate Antigen escape. Mol Ther. 2018;26:2542–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ymthe.2018.09.008.
Kim AB, et al. Intrinsic tumor resistance to CAR T cells is a dynamic transcriptional state that is exploitable with low-dose radiation. Blood Adv. 2023;7:5396–408. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/bloodadvances.2022009543.
Weiss T, Weller M, Guckenberger M, Sentman CL, Roth P. NKG2D-Based CAR T cells and Radiotherapy Exert Synergistic Efficacy in Glioblastoma. Cancer Res. 2018;78:1031–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-17-1788.
Jin L, et al. CXCR1- or CXCR2-modified CAR T cells co-opt IL-8 for maximal antitumor efficacy in solid tumors. Nat Commun. 2019;10:4016. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-019-11869-4.
Boopathi E, Den RB, Thangavel C. Innate immune system in the context of radiation therapy for cancer. Cancers (Basel). 2023;15. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers15153972.
Walle T, et al. Radiotherapy orchestrates natural killer cell dependent antitumor immune responses through CXCL8. Sci Adv. 2022;8:eabh4050. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abh4050.
Li T, et al. CAR-NK cells for cancer immunotherapy: recent advances and future directions. Front Immunol. 2024;15:1361194. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2024.1361194.
Daher M, Melo Garcia L, Li Y, Rezvani K. CAR-NK cells: the next wave of cellular therapy for cancer. Clin Transl Immunol. 2021;10:e1274. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cti2.1274.
Jiao JZ, et al. Radiofrequency radiation reshapes tumor immune microenvironment into antitumor phenotype in pulmonary metastatic melanoma by inducing active transformation of tumor-infiltrating CD8(+) T and NK cells. Acta Pharmacol Sin. 2024;45:1492–505. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41401-024-01260-5.
Yang G, et al. Low-dose ionizing radiation induces direct activation of natural killer cells and provides a novel approach for adoptive cellular immunotherapy. Cancer Biother Radiopharm. 2014;29:428–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/cbr.2014.1702.
Sonn CH, et al. Augmentation of natural cytotoxicity by chronic low-dose ionizing radiation in murine natural killer cells primed by IL-2. J Radiat Res. 2012;53:823–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jrr/rrs037.
Cheda A, et al. Single low doses of X rays inhibit the development of experimental tumor metastases and trigger the activities of NK cells in mice. Radiat Res. 2004;161:335–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1667/rr3123.
Hashimoto S, et al. The suppression of metastases and the change in host immune response after low-dose total-body irradiation in tumor-bearing rats. Radiat Res. 1999;151:717–24.
Palata O, et al. Radiotherapy in Combination with Cytokine Treatment. Front Oncol. 2019;9:367. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2019.00367.
Zegers CM, et al. Radiotherapy combined with the immunocytokine L19-IL2 provides long-lasting antitumor effects. Clin Cancer Res. 2015;21:1151–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-14-2676.
Zhang C, Liang Z, Ma S, Liu X. Radiotherapy and Cytokine Storm: risk and mechanism. Front Oncol. 2021;11:670464. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2021.670464.
van den Heuvel MM, et al. NHS-IL2 combined with radiotherapy: preclinical rationale and phase Ib trial results in metastatic non-small cell lung cancer following first-line chemotherapy. J Transl Med. 2015;13:32. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-015-0397-0.
Yu CF, et al. Local Interleukin-12 treatment enhances the efficacy of radiation therapy by overcoming radiation-induced immune suppression. Int J Mol Sci. 2021;22. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms221810053.
Clark PA, et al. In situ vaccination at a peripheral tumor site augments response against melanoma brain metastases. J Immunother Cancer. 2020;8. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-000809.
Shi W, Liu N, Lu H. Advancements and challenges in immunocytokines: a new arsenal against cancer. Acta Pharm Sin B. 2024;14:4649–4664. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.apsb.2024.07.024
Liu J, et al. Interleukin-12: an update on its immunological activities, signaling and regulation of gene expression. Curr Immunol Rev. 2005;1:119–37. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1573395054065115.
Alatrash G, et al. Clinical and immunologic effects of subcutaneously administered interleukin-12 and interferon alfa-2b: phase I trial of patients with metastatic renal cell carcinoma or malignant melanoma. J Clin Oncol. 2004;22:2891–900. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.2004.10.045.
Fu Y, Tang R, Zhao X. Engineering cytokines for cancer immunotherapy: a systematic review. Front Immunol. 2023;14:1218082. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1218082.
Fallon J, et al. The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget. 2014;5:1869–84. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.1853.
Strauss J, et al. First-in-human phase I trial of a tumor-targeted cytokine (NHS-IL12) in subjects with metastatic solid tumors. Clin Cancer Res. 2019;25:99–109. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-18-1512.
Mullard A. Tumour-infiltrating lymphocyte cancer therapy nears FDA finish line. Nat Rev Drug Discov. 2024;23:3–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/d41573-023-00206-6.
Sarnaik AA, et al. Lifileucel, a tumor-infiltrating lymphocyte therapy, in metastatic melanoma. J Clin Oncol. 2021;39:2656–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.21.00612.
Chesney J, et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C-144-01 study. J Immunother Cancer. 2022;10. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2022-005755.
Rohaan MW, et al. Tumor-infiltrating lymphocyte therapy or Ipilimumab in Advanced Melanoma. N Engl J Med. 2022;387:2113–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa2210233.
van den Berg JH, et al. Tumor infiltrating lymphocytes (TIL) therapy in metastatic melanoma: boosting of neoantigen-specific T cell reactivity and long-term follow-up. J Immunother Cancer. 2020;8. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-000848.
Chandran SS, et al. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 2017;18:792–802. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S1470-2045(17)30251-6.
Patel SP, Forget MA, Kreidieh FY, Pelster M, Davies MA, Amaria RN, Gombos DS, Bernatchez C. Tumor infiltrating lymphocyte (TIL) harvest and ex vivo expansion from primary and metastatic (met) uveal melanoma (UM) tumors. J Clin Oncol. 2023;41. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.2023.41.16_suppl.95.
Obertopp N, Thomas A, Ali J, Blauvelt J, Hall A, Enderling H, Pilon-Thomas S. Utilizing radiation to improve adoptive cell therapy with tumor infiltrating lymphocytes. J Immunother Cancer. 2024;2024 12. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2024-ITOC10.55.
Andtbacka RH, et al. Talimogene Laherparepvec improves durable response rate in patients with Advanced Melanoma. J Clin Oncol. 2015;33:2780–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.2014.58.3377.
Macedo N, Miller DM, Haq R, Kaufman HL. Clinical landscape of oncolytic virus research in 2020. J Immunother Cancer. 2020;8. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-001486.
Cook J, et al. Clinical activity of single-dose systemic oncolytic VSV virotherapy in patients with relapsed refractory T-cell lymphoma. Blood Adv. 2022;6:3268–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/bloodadvances.2021006631.
Tilgase A, et al. Effect of oncolytic ECHO-7 virus strain Rigvir on uveal melanoma cell lines. BMC Res Notes. 2020;13:222. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-020-05068-4.
Garcia M, et al. A phase 1 trial of Oncolytic Adenovirus ICOVIR-5 administered intravenously to cutaneous and uveal melanoma patients. Hum Gene Ther. 2019;30:352–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/hum.2018.107.
Hart LS, et al. The adenovirus E4orf6 protein inhibits DNA double strand break repair and radiosensitizes human tumor cells in an E1B-55K-independent manner. J Biol Chem. 2005;280:1474–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M409934200.
Goel A, et al. Radioiodide imaging and radiovirotherapy of multiple myeloma using VSV(Delta51)-NIS, an attenuated vesicular stomatitis virus encoding the sodium iodide symporter gene. Blood. 2007;110:2342–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2007-01-065573.
Li H, Nakashima H, Decklever TD, Nace RA, Russell SJ. HSV-NIS, an oncolytic herpes simplex virus type 1 encoding human sodium iodide symporter for preclinical prostate cancer radiovirotherapy. Cancer Gene Ther. 2013;20:478–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cgt.2013.43.
Opyrchal M, et al. Effective radiovirotherapy for malignant gliomas by using oncolytic measles virus strains encoding the sodium iodide symporter (MV-NIS). Hum Gene Ther. 2012;23:419–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/hum.2011.158.
Vijayakumar G, Palese P, Goff PH. Oncolytic Newcastle Disease virus expressing a checkpoint inhibitor as a radioenhancing agent for murine melanoma. EBioMedicine. 2019;49:96–105. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebiom.2019.10.032.
Siva S, MacManus MP, Martin RF, Martin OA. Abscopal effects of radiation therapy: a clinical review for the radiobiologist. Cancer Lett. 2015;356:82–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.canlet.2013.09.018.
Hoefsmit EP, et al. Comprehensive analysis of cutaneous and uveal melanoma liver metastases. J Immunother Cancer. 2020;8. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-001501.
Bakhoum MF, Esmaeli B. Molecular characteristics of uveal melanoma: insights from the cancer genome atlas (TCGA) project. Cancers (Basel). 2019;11. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers11081061.
Nathan P, et al. Overall Survival Benefit with Tebentafusp in Metastatic Uveal Melanoma. N Engl J Med. 2021;385:1196–206. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa2103485.
Kirson ED, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci U S A. 2007;104:10152–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0702916104.
Giladi M, et al. Mitotic spindle disruption by Alternating Electric fields leads to Improper chromosome segregation and mitotic catastrophe in Cancer cells. Sci Rep. 2015;5:18046. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep18046.
Kirson ED, et al. Alternating electric fields (TTFields) inhibit metastatic spread of solid tumors to the lungs. Clin Exp Metastasis. 2009;26:633–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10585-009-9262-y.
van der Woude LL, Gorris MAJ, Halilovic A, Figdor CG, de Vries IJ. M. migrating into the Tumor: a Roadmap for T cells. Trends Cancer. 2017;3:797–808. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.trecan.2017.09.006.
Kim EH, et al. Biological effect of an alternating electric field on cell proliferation and synergistic antimitotic effect in combination with ionizing radiation. Oncotarget. 2016;7:62267–79. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.11407.
Giladi M, et al. Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cells. Radiat Oncol. 2017;12:206. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13014-017-0941-6.
Bokstein F, et al. Concurrent Tumor Treating Fields (TTFields) and Radiation Therapy for newly diagnosed glioblastoma: a prospective safety and feasibility study. Front Oncol. 2020;10:411. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2020.00411.
Pedersen AE, et al. Phenotypic and functional characterization of clinical grade dendritic cells generated from patients with advanced breast cancer for therapeutic vaccination. Scand J Immunol. 2005;61:147–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.0300-9475.2005.01531.x.
Hsieh CS, et al. Development of TH1 CD4 + T cells through IL-12 produced by Listeria-induced macrophages. Science. 1993;260:547–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.8097338.
Schmidt CS, Mescher MF. Peptide antigen priming of naive, but not memory, CD8 T cells requires a third signal that can be provided by IL-12. J Immunol. 2002;168:5521–9. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.168.11.5521.
Wang S, et al. Exposure to low-dose Radiation enhanced the Antitumor Effect of a dendritic cell vaccine. Dose Response. 2019;17:1559325819832144. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1559325819832144.
Zheng W, et al. Combination of radiotherapy and vaccination overcomes checkpoint blockade resistance. Oncotarget. 2016;7:43039–51. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.9915.
Cadena A, et al. Radiation and anti-cancer vaccines: a winning combination. Vaccines (Basel). 2018;6. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/vaccines6010009.
Liu D, Che X, Wang X, Ma C, Wu G. Tumor vaccines: unleashing the power of the immune system to fight cancer. Pharmaceuticals (Basel). 2023;16. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph16101384.
Gupta M, et al. Recent advances in cancer vaccines: challenges, achievements, and futuristic prospects. Vaccines (Basel). 2022;10. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/vaccines10122011.
Verma C, et al. Cancer vaccines in the immunotherapy era: promise and potential. Vaccines (Basel). 2023;11. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/vaccines11121783.
Liu J, et al. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15:28. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-022-01247-x.
Le DT, Pardoll DM, Jaffee EM. Cellular vaccine approaches. Cancer J. 2010;16:304–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/PPO.0b013e3181eb33d7.
Khong H, Overwijk WW. Adjuvants for peptide-based cancer vaccines. J Immunother Cancer. 2016;4:56. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40425-016-0160-y.
Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A, Chlichlia K. DNA vaccines to attack cancer: strategies for improving immunogenicity and efficacy. Pharmacol Ther. 2016;165:32–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2016.05.004.
Jou J, Harrington KJ, Zocca MB, Ehrnrooth E, Cohen EE. W. The changing Landscape of Therapeutic Cancer vaccines-Novel platforms and Neoantigen Identification. Clin Cancer Res. 2021;27:689–703. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-20-0245.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijrobp.2012.06.020.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijrobp.2003.09.012.
Kim HJ, Cantor H. CD4 T-cell subsets and tumor immunity: the helpful and the not-so-helpful. Cancer Immunol Res. 2014;2:91–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2326-6066.CIR-13-0216.
Seyedin SN, et al. Strategies for combining immunotherapy with radiation for anticancer therapy. Immunotherapy. 2015;7:967–80. https://doiorg.publicaciones.saludcastillayleon.es/10.2217/imt.15.65.
Demaria S, Golden EB, Formenti SC. Role of local Radiation Therapy in Cancer Immunotherapy. JAMA Oncol. 2015;1:1325–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jamaoncol.2015.2756.
Fotin-Mleczek M, et al. mRNA-based vaccines synergize with radiation therapy to eradicate established tumors. Radiat Oncol. 2014;9:180. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1748-717X-9-180.
Lugade AA, et al. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. 2005;174:7516–23. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.174.12.7516.
Wang X, et al. Suppression of type I IFN Signaling in Tumors mediates resistance to Anti-PD-1 treatment that can be overcome by Radiotherapy. Cancer Res. 2017;77:839–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-15-3142.
Levy R, Reagan PM, Friedberg JW, Bartlett NL, Gordon LI, Leung A, Peterkin J, Xing B, Coffman R, Janssen R, Candia A, Khodadoust M, Frank MJ, Long SR, Czerwinski DK, Chu M. SD-101, a novel class C CpG-Oligodeoxynucleotide (ODN) toll-like receptor 9 (TLR9) agonist, given with low dose radiation for untreated low grade B-cell lymphoma: interim results of a phase 1/2 Trial. Blood. 2016;128:2974. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.V128.22.2974.2974
Liu SZ. Cancer control related to stimulation of immunity by low-dose radiation. Dose Response. 2006;5:39–47. https://doiorg.publicaciones.saludcastillayleon.es/10.2203/dose-response.06-108.Liu.
Ren H, et al. Augmentation of innate immunity by low-dose irradiation. Cell Immunol. 2006;244:50–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cellimm.2007.02.009.
Pandey R, Shankar BS, Sharma D, Sainis KB. Low dose radiation induced immunomodulation: effect on macrophages and CD8+ T cells. Int J Radiat Biol. 2005;81:801–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/09553000500531886.
Gridley DS, et al. Low-dose, low-dose-rate proton radiation modulates CD4(+) T cell gene expression. Int J Radiat Biol. 2009;85:250–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/09553000902748609.
LI. PYQJFLRHC. W. X. effects of low dose X-ray irradiation on antigen presentation and IL-12 secretion in human dendritic cells in vitro. Chin J Radiological Med Prot. 2012;32:225–9.
Li X, Jiang Q, Li F, Lv J, Wang S, He R, Zou Y, Sun S, Wang C, Liu J. Effect of low dose X-ray irradiation on migration of human dendrite cells and underlying mechanism. Chin J Radiological Med Prot. 2013;33:476–9. https://doiorg.publicaciones.saludcastillayleon.es/10.3760/cma.j.issn.0254-5098.2013.05.005.
Acknowledgements
Use of Bio-render to generate all the figures in this manuscript.
Funding
This research was supported by the MDACC Institutional Research Grant (IRG) to NPO, and the Department of Radiation Oncology at The University of Texas MD Anderson Cancer Center, Houston, TX, USA. Additional support was provided by the Cancer Prevention and Research Institute of Texas (CPRIT) through the Texas Regional Excellence in Cancer (TREC) program under grant ID RP230446 to WL.
Author information
Authors and Affiliations
Contributions
ZR and NPO conceptualized the review and conducted the literature analysis. Together, they authored the manuscript. MK contributed to figure creation, while HB and YH formatted the figures. GSM provided critical feedback, streamlined the content, and added key references. CL, AH, and FM reviewed the final draft. JW, WL, and NPO conducted a comprehensive review of the manuscript. NPO led the coordination and finalization of the entire manuscript. All authors reviewed and approved the final version.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
JWW: Accuray (SAB, consulting), Alpine Immune Science (SAB, consulting, equity), Boehringer Ingelheim (SAB, consulting), Checkmate Pharmaceuticals (SAB, consulting, equity), China Medical Tribune (SAB, consulting), Genentech (SAB, consulting), GI Innovation (SAB, consulting), Kezar Life Sciences (consulting, SAB), Legion Healthcare Partners (SAB, consulting), Life Science Dynamic Limited (SAB, consulting), McKesson Corporation (SAB, consulting), Molecular Match (equity), Nanorobotix (SAB, consulting), OligoImmune (founder), Roche (SAB, consulting), Roche Molecular Systems (SAB, consulting), Nanobiotix (Research grant, Travel expenses, SAB), BMS (Research grant), Merck (Research grant), Varian (Research grant, Travel expenses, clinical sponsored research), Reflexion (Research grant, Travel expenses, Stock options, SAB), Hotspot Therapeutics (Research grant), Gilead (Research grant), Novocure (SAB), Oncoresponse (SAB, Stock options), Astra Zeneca (consultant, Research grant), Bayer Healthcare (Research grant), Kiromic (Research grant), Alkermes (Research grant, SAB), Artidis (Research grant), Sciclone (Research grant), Takeda (Research grant), Pebble Life Science (Research grant). MD Anderson has a trademark designation for RadScopalTM technique. The remaining authors declared no competing interests.
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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Rafiq, Z., Kang, M., Barsoumian, H.B. et al. Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects. J Exp Clin Cancer Res 44, 31 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13046-025-03281-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13046-025-03281-2