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

Radiotherapy Mechanisms and Resistance

Recent studies have elucidated various mechanisms by which radiotherapy (RT) induces cell death and overcomes tumor resistance. One significant finding is that RT promotes cuproptosis, a copper-dependent cell death mechanism, in cancer cells. This process is independent of traditional apoptosis and ferroptosis pathways. The mechanism involves RT elevating mitochondrial copper levels through the upregulation of copper transporter 1 (CTR1) and depleting mitochondrial glutathione, a known copper chelator, leading to the depletion of lipoylated proteins and iron-sulfur cluster proteins, which are hallmarks of cuproptosis (ref: Lei doi.org/10.1016/j.ccell.2025.03.031/). Additionally, the conversion of Ku80 from K568 crotonylation to SUMOylation has been shown to facilitate DNA non-homologous end joining, enhancing cancer radioresistance by promoting the assembly of the DNA-PK complex necessary for DNA repair (ref: Zhao doi.org/10.1038/s41392-025-02210-1/). Furthermore, GLS2 inhibition has been identified as a synergistic approach with copper to reprogram the TCA cycle, enhancing radiosensitivity in esophageal cancer (ref: Jing doi.org/10.1186/s40164-025-00653-4/). These findings collectively highlight the multifaceted nature of RT and its interactions with cellular metabolism and DNA repair mechanisms, suggesting potential therapeutic targets to enhance treatment efficacy. Moreover, the impact of the tumor microenvironment on treatment outcomes has been explored, particularly in melanoma, where stromal lipid species from adipocytes influence cancer cell metabolism and metastatic behavior. High oxidative phosphorylation (OXPHOS) in melanoma cells was linked to their tropism for specific organs, indicating that metabolic adaptations in response to the tumor microenvironment can dictate metastatic patterns (ref: Gurung doi.org/10.1016/j.ccell.2025.04.001/). Additionally, the disruption of DNA methylation in non-neoplastic tissues surrounding tumors has been implicated in neuroinflammation following targeted CNS radiotherapy, suggesting that epigenetic changes may contribute to the adverse effects of treatment (ref: Millner doi.org/10.1093/brain/). Overall, these studies underscore the complexity of radiotherapy responses and the need for integrated approaches to overcome resistance and improve patient outcomes.

Genomic and Molecular Insights in Cancer

The exploration of genomic and molecular factors in cancer treatment has yielded promising insights, particularly regarding the management of mismatch repair-deficient tumors. A study demonstrated that neoadjuvant PD-1 blockade resulted in a high clinical complete response rate, allowing a significant proportion of patients with early-stage dMMR solid tumors to opt for nonoperative management (ref: Cercek doi.org/10.1056/NEJMoa2404512/). This highlights the potential of immunotherapy to facilitate organ preservation in select patient populations. In the realm of lung cancer, the efficacy of sacituzumab tirapazamine in advanced non-small-cell lung cancer (NSCLC) was evaluated, revealing encouraging activity in patients with or without EGFR mutations, despite previous phase 3 trials showing limited success for TROP2-ADCs (ref: Zhao doi.org/10.1038/s41591-025-03638-2/). These findings emphasize the need for tailored therapeutic strategies based on genetic profiles. Moreover, the integration of artificial intelligence in predicting treatment outcomes has emerged as a significant advancement. A multimodal AI-derived biomarker was developed to predict the benefits of long-term androgen deprivation therapy in high-risk prostate cancer patients, utilizing clinical data and digital pathology images from multiple trials (ref: Armstrong doi.org/10.1200/JCO.24.00365/). This approach underscores the potential of AI in enhancing precision medicine. Additionally, the combination of ctDNA analysis and radiomics for dynamic risk assessment in localized lung cancer has shown promise in predicting progression-free survival, further illustrating the utility of integrating genomic data with imaging modalities (ref: Moding doi.org/10.1158/2159-8290.CD-24-1704/). Collectively, these studies highlight the evolving landscape of cancer genomics and the importance of personalized approaches in improving treatment efficacy.

Immunotherapy and Tumor Microenvironment

The interplay between immunotherapy and the tumor microenvironment (TME) has become a focal point in cancer research, particularly in understanding how tumors evade immune responses. A study revealed that tumor-derived erythropoietin acts as an immunosuppressive switch, influencing the immunotype of hepatocellular carcinoma (HCC). The presence of EPO in the TME was shown to dictate the immune landscape, suggesting that targeting this pathway could enhance the efficacy of immunotherapies (ref: Chiu doi.org/10.1126/science.adr3026/). Furthermore, the challenges faced by CAR T cell therapies in solid tumors were addressed, with findings indicating that tumors can upregulate the secretion of extracellular vesicles carrying tumor antigens, leading to fratricide among CAR T cells. This highlights the need for strategies to overcome this resistance mechanism to improve treatment outcomes (ref: Zhong doi.org/10.1038/s43018-025-00949-8/). Additionally, innovative approaches combining microbial photosynthetic oxygenation with radiotherapy have been explored to induce pyroptosis in cancer cells, addressing the issue of tumor hypoxia, which often limits the effectiveness of RT (ref: Li doi.org/10.1002/adma.202503138/). The prevention of efferocytosis by terminating MerTK recycling on tumor-associated macrophages has also been proposed as a strategy to enhance antitumor immunity, although the transient nature of this effect raises questions about its long-term efficacy (ref: Huang doi.org/10.1021/jacs.5c05640/). These findings collectively underscore the complexity of the TME in shaping immune responses and the necessity for multifaceted therapeutic strategies that can effectively modulate the immune landscape to improve cancer treatment.

Artificial Intelligence and Biomarkers in Cancer Treatment

The integration of artificial intelligence (AI) in cancer treatment has shown significant promise in enhancing predictive capabilities and personalizing therapy. A notable advancement is the development of a multimodal AI-derived biomarker aimed at predicting the benefits of long-term androgen deprivation therapy in high-risk prostate cancer patients. This biomarker was trained using clinical data and digital pathology images from multiple phase III trials, demonstrating the potential of AI to guide treatment decisions and optimize patient outcomes (ref: Armstrong doi.org/10.1200/JCO.24.00365/). Additionally, the combination of circulating tumor DNA (ctDNA) analysis with radiomics has emerged as a powerful tool for dynamic risk assessment in localized lung cancer, providing real-time insights into treatment response and progression-free survival (ref: Moding doi.org/10.1158/2159-8290.CD-24-1704/). Moreover, the development of functionally tunable star-shaped multivalent crRNAs for CRISPR/Cas editing systems represents a significant leap in genetic engineering, allowing for precise spatial and temporal control of gene editing (ref: Chen doi.org/10.1002/anie.202506527/). This innovation could enhance therapeutic applications of CRISPR technology, particularly in cancer treatment. Furthermore, the exploration of microbial photosynthetic oxygenation combined with radiotherapy to induce pyroptosis in cancer cells addresses the challenge of tumor hypoxia, potentially improving treatment efficacy (ref: Li doi.org/10.1002/adma.202503138/). Collectively, these advancements highlight the transformative role of AI and biomarker integration in refining cancer treatment strategies and improving patient outcomes.

Nanotechnology in Cancer Therapy

Nanotechnology has emerged as a transformative approach in cancer therapy, particularly in enhancing the efficacy of radiotherapy and overcoming treatment resistance. One innovative strategy involves the development of camouflaged nanozymes designed to trigger ferroptosis, a form of regulated cell death, in breast cancer cells. These nanozymes, coated with cancer cell membranes, enhance the effectiveness of radioimmunotherapy by promoting oxidative stress within tumor cells, thereby improving therapeutic outcomes (ref: Qiao doi.org/10.1002/advs.202417370/). Additionally, nanoscale coordination polymer particles have been engineered to induce cellular senescence and enhance immune responses against tumors, demonstrating the potential of nanotechnology to not only target cancer cells directly but also to modulate the immune landscape (ref: Zhen doi.org/10.1016/j.biomaterials.2025.123355/). Furthermore, the development of tumor-specific activated polymeric nanotuners aims to disrupt the feedback cycle between hypoxia and apoptosis evasion, which often limits the effectiveness of radiotherapy. These nanotuners are designed to enhance the apoptotic response in tumor cells, thereby improving radiotherapy outcomes (ref: Hou doi.org/10.1016/j.biomaterials.2025.123361/). The role of METTL3 in promoting an immunosuppressive microenvironment in bladder cancer has also been highlighted, suggesting that targeting metabolic pathways could synergize with immunotherapy to enhance treatment efficacy (ref: Tong doi.org/10.1136/jitc-2024-011108/). Overall, these studies underscore the potential of nanotechnology to revolutionize cancer therapy by improving drug delivery, enhancing therapeutic efficacy, and modulating the tumor microenvironment.

Cancer Genomics and Personalized Medicine

The field of cancer genomics and personalized medicine is rapidly evolving, with significant advancements in understanding the genetic underpinnings of cancer and tailoring treatments accordingly. A pivotal study demonstrated that patients with mismatch repair-deficient tumors achieved high rates of clinical complete response when treated with neoadjuvant PD-1 blockade, allowing many to opt for nonoperative management. This finding underscores the potential of immunotherapy to preserve organ function in select patient populations (ref: Cercek doi.org/10.1056/NEJMoa2404512/). In the context of lung cancer, the efficacy of sacituzumab tirapazamine was evaluated in advanced non-small-cell lung cancer patients, revealing promising results in those with and without EGFR mutations, despite previous phase 3 trials showing limited success for TROP2-ADCs (ref: Zhao doi.org/10.1038/s41591-025-03638-2/). Moreover, the integration of artificial intelligence in predicting treatment outcomes has emerged as a significant advancement. A multimodal AI-derived biomarker was developed to predict the benefits of long-term androgen deprivation therapy in high-risk prostate cancer patients, utilizing clinical data and digital pathology images from multiple trials (ref: Armstrong doi.org/10.1200/JCO.24.00365/). This approach underscores the potential of AI in enhancing precision medicine. Additionally, the combination of ctDNA analysis and radiomics for dynamic risk assessment in localized lung cancer has shown promise in predicting progression-free survival, further illustrating the utility of integrating genomic data with imaging modalities (ref: Moding doi.org/10.1158/2159-8290.CD-24-1704/). Collectively, these studies highlight the evolving landscape of cancer genomics and the importance of personalized approaches in improving treatment efficacy.

Clinical Trials and Treatment Efficacy

Clinical trials remain a cornerstone in evaluating the efficacy of novel cancer therapies and understanding their impact on patient outcomes. Recent studies have highlighted the role of METTL3 in promoting an immunosuppressive microenvironment in bladder cancer, demonstrating that targeting this pathway can enhance responses to immunotherapy. The synergistic effect of METTL3 with anti-PD-1 treatment was confirmed in both orthotopic and ectopic models, suggesting that metabolic modulation could improve therapeutic efficacy (ref: Tong doi.org/10.1136/jitc-2024-011108/). Additionally, the development of photoactivatable immunomodulator polyprodrugs has shown promise in boosting antitumor immunity when combined with STING agonists and IDO inhibitors, indicating a novel approach to enhance immunotherapy outcomes (ref: Huang doi.org/10.7150/thno.107774/). Moreover, the integration of ctDNA analysis and radiomics for dynamic risk assessment in localized lung cancer has demonstrated significant prognostic value, providing insights into treatment response and progression-free survival (ref: Moding doi.org/10.1158/2159-8290.CD-24-1704/). This innovative approach highlights the potential of combining genomic and imaging data to tailor treatment strategies. Furthermore, the exploration of GLS2 inhibition in esophageal cancer has revealed its role in enhancing radiosensitivity, suggesting that metabolic reprogramming could be a viable strategy to improve treatment outcomes (ref: Jing doi.org/10.1186/s40164-025-00653-4/). Collectively, these findings underscore the importance of clinical trials in advancing our understanding of cancer therapies and their potential to improve patient outcomes.

Metabolic Reprogramming in Cancer

Metabolic reprogramming has emerged as a critical area of research in cancer, with studies revealing how cancer cells adapt their metabolism to support growth and evade therapy. One significant finding is that GLS2 inhibition can synergize with copper to reprogram the TCA cycle, enhancing radiosensitivity in esophageal squamous cell carcinoma. This study demonstrated that knocking down GLS2 not only suppressed cell proliferation but also augmented sensitivity to radiotherapy, highlighting the potential of targeting metabolic pathways to improve treatment outcomes (ref: Jing doi.org/10.1186/s40164-025-00653-4/). Additionally, research has shown that oxidative phosphorylation (OXPHOS) plays a crucial role in the energy metabolism of chemoresistant triple-negative breast cancer cells. These cells preferentially utilize OXPHOS over glycolysis, suggesting that OXPHOS inhibitors could be effective in overcoming chemoresistance (ref: Uslu doi.org/10.1016/j.redox.2025.103637/). Furthermore, the role of METTL3 in promoting an immunosuppressive microenvironment in bladder cancer has been linked to m6A-dependent regulation of chemokines, indicating that metabolic reprogramming can influence immune responses and treatment efficacy (ref: Tong doi.org/10.1136/jitc-2024-011108/). Collectively, these studies underscore the importance of understanding metabolic adaptations in cancer cells and their implications for therapeutic strategies.

Key Highlights

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