Radiogenomics and genomics-guided radiotherapy, including tumor radiosensitivity and dose personalization

Radiotherapy and Genomic Interactions

Recent studies have highlighted the intricate relationship between radiotherapy and genomic interactions, particularly focusing on how specific genomic signatures can predict treatment responses. For instance, the PORTOS signature was validated in two major clinical trials, revealing that patients with higher PORTOS scores experienced significant benefits from dose escalation in prostate cancer treatments (ref: Dal Pra doi.org/10.1016/j.annonc.2025.01.017/). Additionally, a phase Ib trial investigated the combination of olaparib, a PARP inhibitor, with radiotherapy in soft-tissue sarcoma, establishing a recommended phase II dose of 100 mg olaparib twice daily, which may enhance therapeutic efficacy (ref: Sargos doi.org/10.1016/j.annonc.2025.01.016/). The role of immunotherapy in conjunction with radiotherapy was further explored in a phase 2 trial where atezolizumab, an anti-PD-L1 antibody, was administered post-chemoradiotherapy in esophageal squamous cell carcinoma, resulting in a notable complete response rate of 42.1% (ref: Bando doi.org/10.1038/s43018-025-00918-1/). These findings underscore the potential of integrating genomic profiling with therapeutic strategies to optimize patient outcomes. Moreover, innovative approaches to enhance radiotherapy's effectiveness have emerged, such as the use of manganese-coordinated chitosan microparticles that activate the cGAS-STING pathway, thereby boosting anti-tumor immunity (ref: Zhang doi.org/10.1002/adma.202418583/). Another study introduced a magnetic ion-generator that simultaneously increases DNA damage and reduces immunosuppression, effectively overcoming radioresistance (ref: Zhang doi.org/10.1002/adma.202406378/). Collectively, these studies illustrate a multifaceted approach to improving radiotherapy through genomic insights and novel therapeutic combinations, paving the way for more personalized cancer treatment strategies.

Immunotherapy and Tumor Microenvironment

The interplay between immunotherapy and the tumor microenvironment has become a focal point in understanding cancer treatment resistance. A significant finding is the role of the lactate receptor HCAR1 in colorectal cancer, which drives the recruitment of immunosuppressive PMN-MDSCs, thereby contributing to the failure of immunotherapy in many patients (ref: He doi.org/10.1038/s41590-024-02068-5/). This highlights the need for strategies that can counteract these immunosuppressive mechanisms. In a novel approach, researchers have utilized ionizable lipid nanoparticles to deliver mRNA encoding a damage-suppressor protein from tardigrades, which has shown promise in radioprotection of healthy tissues during radiation therapy (ref: Kirtane doi.org/10.1038/s41551-025-01360-5/). This innovative method aims to mitigate the side effects of radiation while preserving the efficacy of cancer treatments. Furthermore, the development of a PD-L1 siRNA-loaded boron nanoparticle represents a targeted strategy to enhance the synergistic effects of radiotherapy and immunotherapy by minimizing damage to effector immune cells (ref: Deng doi.org/10.1002/adma.202419418/). The study of STING's interaction with poly(ADP-ribose) (PAR) has also revealed its critical role in promoting apoptosis following ionizing radiation, suggesting that enhancing STING signaling could improve therapeutic outcomes (ref: Sun doi.org/10.1038/s41418-025-01457-z/). These findings collectively emphasize the importance of understanding and manipulating the tumor microenvironment to enhance the efficacy of immunotherapy and radiotherapy.

Mechanisms of Radioresistance

Understanding the mechanisms underlying radioresistance is crucial for improving cancer treatment outcomes. Recent research has identified several pathways and factors contributing to this phenomenon. For example, the study on METTL3-mediated m6A modification of SLC7A11 demonstrated that increased stability of this transcript inhibits ferroptosis, thereby enhancing radioresistance in nasopharyngeal carcinoma (ref: Dai doi.org/10.7150/ijbs.100518/). This highlights the role of RNA modifications in regulating cellular responses to radiation. Additionally, RSK2's involvement in the phosphorylation and degradation of UBE2O has been shown to inhibit hepatocellular carcinoma growth and its resistance to radiotherapy, suggesting that targeting this pathway may enhance treatment efficacy (ref: Huang doi.org/10.1016/j.canlet.2025.217558/). Moreover, the concurrent inhibition of the RAS-MAPK pathway alongside PIKfyve has emerged as a promising therapeutic strategy for pancreatic cancer, indicating that metabolic pathways are intricately linked to radioresistance (ref: DeLiberty doi.org/10.1158/0008-5472.CAN-24-1757/). The exploration of cellular senescence and epithelial-mesenchymal transition (EMT) alterations has also revealed distinct patterns of disease relapse in locally advanced cervical cancer, emphasizing the need for tailored approaches based on individual tumor characteristics (ref: Zhang doi.org/10.1002/advs.202412574/). These insights into the molecular mechanisms of radioresistance provide a foundation for developing more effective therapeutic strategies.

Nanotechnology in Cancer Therapy

Nanotechnology has emerged as a transformative approach in cancer therapy, particularly in enhancing the efficacy of radiotherapy and targeted drug delivery. Recent advancements include the development of glutathione and transglutaminase-responsive Janus gold nanorods, which serve as photoacoustic imaging-guided agents for radiotherapy and chemodynamic therapy (ref: Zhang doi.org/10.1016/j.jconrel.2025.02.026/). These nanorods leverage their strong X-ray attenuation properties to improve tumor imaging and treatment precision. Additionally, a novel pyroptosis radiosensitizer has been introduced, which targets hypoxic tumor cells by inducing cell death through the activation of gasdermin C, thereby enhancing the effectiveness of radiotherapy (ref: Gu doi.org/10.1002/smll.202409594/). Furthermore, the application of boron neutron capture therapy (BNCT) using PD-L1 siRNA-loaded boron nanoparticles has shown potential in selectively targeting tumor cells while sparing normal tissues, addressing a significant challenge in conventional radiotherapy (ref: Deng doi.org/10.1002/adma.202419418/). These innovations underscore the versatility of nanotechnology in overcoming barriers associated with traditional cancer therapies, paving the way for more effective and personalized treatment modalities.

Biomarkers and Predictive Models

The identification of biomarkers and predictive models is crucial for personalizing cancer treatment and improving patient outcomes. Recent studies have focused on the development of biomarkers for radioresistance, particularly in head and neck cancer, where mutations in NFE2L2/KEAP1/CUL3 have been correlated with local recurrence and disease-free survival (ref: Rao doi.org/10.1158/1078-0432.CCR-24-4223/). This highlights the potential for using genetic profiling to tailor treatment strategies. Additionally, the use of manganese-coordinated chitosan microparticles has been shown to enhance the immunogenicity of radiotherapy by activating the cGAS-STING pathway, suggesting a novel approach to boost anti-tumor immunity (ref: Zhang doi.org/10.1002/adma.202418583/). Moreover, the study of UV-induced reorganization of the 3D genome has provided insights into the mechanisms of DNA damage response, emphasizing the importance of genomic architecture in mediating repair processes (ref: Kaya doi.org/10.1038/s41467-024-55724-7/). The role of STING in promoting apoptosis following acute ionizing radiation further underscores its potential as a biomarker for predicting treatment responses (ref: Sun doi.org/10.1038/s41418-025-01457-z/). Collectively, these findings illustrate the critical role of biomarkers in guiding therapeutic decisions and enhancing the efficacy of cancer treatments.

Cellular Mechanisms and Pathways

Recent research has elucidated various cellular mechanisms and pathways that play pivotal roles in cancer treatment responses, particularly in the context of radiotherapy and immunotherapy. The validation of the PORTOS signature in prostate cancer patients has demonstrated its predictive power for treatment outcomes, indicating that patients with higher PORTOS scores benefit significantly from dose escalation (ref: Dal Pra doi.org/10.1016/j.annonc.2025.01.017/). This genomic insight is crucial for personalizing treatment strategies. Additionally, the study on the interaction between STING and PAR has revealed its importance in promoting apoptosis following DNA damage from ionizing radiation, suggesting that enhancing STING signaling could improve therapeutic efficacy (ref: Sun doi.org/10.1038/s41418-025-01457-z/). Moreover, the investigation into how cancer cells evade ferroptosis through fatty acid binding proteins has highlighted a mechanism by which tumors resist immune-mediated cell death, emphasizing the need for strategies that can overcome these protective pathways (ref: Freitas-Cortez doi.org/10.1186/s12943-024-02198-2/). The role of EZH2 in suppressing IR-induced ferroptosis by forming a co-repressor complex with HIF-1α further underscores the complexity of cellular responses to radiation and the potential for targeting these pathways to enhance radiosensitivity (ref: Pan doi.org/10.1038/s41418-025-01451-5/). These findings collectively enhance our understanding of the cellular mechanisms underlying cancer treatment responses and highlight potential therapeutic targets.

Clinical Trials and Therapeutic Strategies

Clinical trials continue to play a crucial role in advancing cancer treatment strategies, particularly in the integration of novel therapies with existing modalities. A phase Ib study evaluating the combination of olaparib with radiotherapy in soft-tissue sarcoma has established a recommended phase II dose of 100 mg olaparib twice daily, indicating a promising avenue for enhancing treatment efficacy (ref: Sargos doi.org/10.1016/j.annonc.2025.01.016/). Additionally, the EPOC1802 trial assessed the efficacy of atezolizumab following definitive chemoradiotherapy in patients with unresectable locally advanced esophageal squamous cell carcinoma, reporting a complete response rate of 42.1%, which underscores the potential of combining immunotherapy with conventional treatments (ref: Bando doi.org/10.1038/s43018-025-00918-1/). Furthermore, innovative strategies such as the use of manganese-coordinated chitosan microparticles to activate the cGAS-STING pathway have shown promise in potentiating the immunogenicity of radiotherapy, suggesting a novel approach to enhance treatment outcomes (ref: Zhang doi.org/10.1002/adma.202418583/). The development of a magnetic ion-generator that augments DNA damage while diminishing immunosuppression also represents a significant advancement in overcoming radioresistance (ref: Zhang doi.org/10.1002/adma.202406378/). These clinical findings highlight the importance of integrating novel therapeutic strategies to improve patient outcomes and personalize cancer treatment.

Metabolic Reprogramming in Cancer

Metabolic reprogramming has emerged as a critical factor in cancer progression and treatment resistance. Recent studies have demonstrated that alterations in metabolic pathways can significantly influence tumor behavior and response to therapies. For instance, the loss of TRAF3 in glioblastoma has been shown to protect cells from lipid peroxidation and immune elimination, indicating that dysregulated lipid metabolism plays a key role in tumor survival (ref: Zeng doi.org/10.1172/JCI178550/). This finding underscores the importance of targeting metabolic pathways to enhance therapeutic efficacy. Additionally, the study of mitochondrial DNA damage following radiation exposure has revealed transgenerational genomic instability, highlighting the long-term effects of radiation on cellular metabolism and genetic integrity (ref: Seino doi.org/10.1016/j.envint.2025.109315/). The role of EZH2 in suppressing IR-induced ferroptosis by forming a co-repressor complex with HIF-1α further emphasizes the interplay between metabolism and treatment responses, suggesting that targeting metabolic regulators could enhance radiosensitivity (ref: Pan doi.org/10.1038/s41418-025-01451-5/). These insights into metabolic reprogramming provide a foundation for developing innovative therapeutic strategies that address the metabolic vulnerabilities of cancer cells.

Key Highlights

Disclaimer: This is an AI-generated summarization. Please refer to the cited articles before making any clinical or scientific decisions.