Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Radiol. May 28, 2025; 17(5): 108011
Published online May 28, 2025. doi: 10.4329/wjr.v17.i5.108011
Harnessing artificial intelligence to address immune response heterogeneity in low-dose radiation therapy
Jing-Qi Zeng, Yi-Wei Gao, Xiao-Bin Jia, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, China
ORCID number: Jing-Qi Zeng (0000-0003-3210-7315); Xiao-Bin Jia (0000-0003-0471-8258).
Author contributions: Zeng JQ conceptualized the editorial, reviewed the literature, and drafted the manuscript; Gao YW contributed to the literature review, assisted in drafting sections related to emerging technologies, and provided editorial feedback; Jia XB provided critical insights, revised the content for scientific accuracy, and contributed to the discussion on artificial intelligence applications. All authors finalized the manuscript and approved the submitted version.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Jing-Qi Zeng, Academic Fellow, Postdoc, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing 211198, Jiangsu Province, China. zjingqi@163.com
Received: April 3, 2025
Revised: April 12, 2025
Accepted: May 8, 2025
Published online: May 28, 2025
Processing time: 53 Days and 19.2 Hours

Abstract

Low-dose radiation therapy has emerged as a promising modality for cancer treatment because of its ability to stimulate antitumor immune responses while minimizing damage to healthy tissues. However, the significant heterogeneity in immune responses among patients complicates its clinical application, hindering outcome prediction and treatment personalization. Artificial intelligence (AI) offers a transformative solution by integrating multidimensional data such as immunomics, radiomics, and clinical features to decode complex immune patterns and predict individual therapeutic outcomes. This editorial explored the potential of AI to address immune response heterogeneity in low-dose radiation therapy and proposed an AI-driven framework for precision immunotherapy. While promising, challenges, including data standardization, model interpretability, and clinical validation, must be overcome to ensure successful integration into oncological practice.

Key Words: Low-dose radiation; Immune response; Heterogeneity; Artificial intelligence; Precision medicine; Immunotherapy; Radiomics

Core Tip: Artificial intelligence (AI) is revolutionizing low-dose radiation therapy by addressing immune response heterogeneity in patients with cancer. By integrating multidimensional datasets such as immunomics, radiomics, and clinical profiles, AI employs advanced machine learning to decode complex immune patterns and predict individualized therapeutic outcomes. This enables tailored treatment strategies that enhance antitumor efficacy while minimizing side effects. Thus, AI paves the way for precision oncology and enables customized treatments for each patient’s unique biological signature.



INTRODUCTION

Low-dose radiation (LDR) has emerged as a promising modality for cancer therapy because of its ability to modulate immune responses while minimizing damage to healthy tissues. Research has shown that LDR can enhance antitumor immunity by activating immune cells, such as natural killer cells and T cells, offering a complementary approach to immunotherapy[1]. A bibliometric analysis by Wang et al[2] highlighted the growing interest in this field, with annual publications on LDR-induced immune responses remaining steady at approximately 30 papers per year between 2004 and 2013, followed by a marked increase in the subsequent decade, culminating in a total of 1244 publications by 2023. Key research hotspots include the integration of LDR with immune checkpoint inhibitors (e.g., ipilimumab) and stereotactic body radiotherapy, reflecting its potential in combination therapies.

Despite these advances, the heterogeneity of immune responses to LDR across patients remains a critical challenge. Studies have revealed significant variability in immune cell activation, with factors such as genetic background, tumor microenvironment, and radiation parameters influencing therapeutic outcomes[3,4]. This variability complicates the prediction of therapeutic efficacy and hinders personalization of LDR-based treatments. Traditional statistical approaches, which often rely on population averages, fail to account for individual differences and limit the precision of clinical applications.

Artificial intelligence (AI) offers a transformative solution by leveraging its capacity to process complex multidimensional data. With applications in immunomics, radiomics, and clinical outcome prediction, AI can decode the heterogeneity of LDR-induced immune responses, paving the way for precision medicine[5,6]. This editorial examined immune response variability, highlighted potential AI solutions, and outlined future directions for its integration into LDR therapy.

THE CHALLENGE OF IMMUNE RESPONSE HETEROGENEITY IN LDR THERAPY

LDR therapy offers significant potential in cancer treatment owing to its immunostimulatory effects. However, the heterogeneity of patient responses presents a formidable barrier to its widespread adoption. Research shows that LDR activates immune components variably. For example, natural killer cell activation differs widely across individuals based on factors such as radiation dose and baseline immune status[7]. In contrast, T cell responses, including CD4+ and CD8+ subsets, are modulated by tumor type, genetic predispositions, and the local inflammatory milieu[8,9]. This variability results in inconsistent therapeutic outcomes, as some patients exhibit robust antitumor immunity post-LDR, whereas others experience minimal effects or in rare cases tumor progression due to immunosuppressive shifts[10]. Consequently, these disparities emphasize the need for personalized strategies in LDR therapy.

The implications of this heterogeneity extend beyond inconsistent efficacy and pose significant risks to treatment success. Without personalized assessments, patients may receive suboptimal LDR doses, reducing therapeutic benefits and potentially compromising outcomes[11]. Moreover, heterogeneous immune responses can undermine the effectiveness of LDR when used in combination therapies such as immune checkpoint inhibitors, as evidenced by variable results in trials combining LDR with anti-PD-1 agents[12]. Current research methods, which predominantly rely on cohort-level data, struggle to capture these individual variations, limiting their ability to predict and tailor treatment effectively. Thus, innovative tools are urgently required to enhance the precision and personalization of LDR therapies.

AI: A PARADIGM SHIFT IN LDR THERAPY

AI is poised to revolutionize LDR therapy by addressing the limitations of traditional approaches through its capacity to process and analyze vast, complex datasets. This capability enables AI to identify patterns in immune response heterogeneity that conventional methods overlook, thereby offering a pathway to overcome the unpredictability of patient outcomes in LDR treatment. For instance, a systematic review by AlOsaimi et al[13] demonstrated the efficacy of AI models in identifying prognostic and predictive biomarkers in lung cancer, underscoring their potential to decipher tumor-immune interactions and support personalized treatment.

By integrating multidimensional biological signals, such as cytokine profiles, immune cell dynamics, and tumor microenvironment features, AI can uncover patient-specific immune signatures that inform tailored therapeutic strategies[14]. For instance, studies have shown that AI models can correlate T cell infiltration levels with LDR outcomes, accurately identifying responders and enhancing treatment precision[15]. Beyond pattern recognition, AI facilitates individualized outcome predictions and real-time adjustments, marking a significant shift toward precision oncology. This transformative potential positions AI as a cornerstone for optimizing the clinical efficacy of LDR therapy. Ultimately, AI integration could help translate empirical insights into actionable, patient-specific treatments.

Emerging evidence indicates that immune responses to LDR vary considerably across tumor types, necessitating tumor-specific AI models. For example, in non-small cell lung cancer, PD-L1 expression and the spatial distribution of tumor-infiltrating lymphocytes have been shown to correlate with response to immunotherapy. A machine learning-based histological analysis of tumor-infiltrating lymphocytes can aid in identifying responders and refining LDR strategies guided by AI[16]. In colorectal cancer, immune evasion is frequently associated with microsatellite instability and low antigenicity in microsatellite-stable tumors. AI algorithms are being used to stratify these subtypes and optimize immunostimulatory interventions in combination with radiotherapy[17]. Renal cell carcinoma, particularly the clear cell subtype, is characterized by dense immune infiltration and angiogenesis. AI-based profiling of genomic and immune features enables prediction of response to immunotherapy and may guide LDR application in this vascularized tumor context[18]. Incorporating such distinctions into AI frameworks enhances predictive power and aligns LDR applications with tumor-specific biology.

AI holds strong potential to enhance LDR therapy by predicting immune responses, guiding treatment stratification, and enabling adaptive planning. Machine learning techniques can analyze baseline biomarkers, such as immune cell densities and cytokine levels, to forecast outcomes and identify responders with high precision[19]. AI also facilitates real-time monitoring of immune dynamics using data from wearable sensors or liquid biopsies, enabling adjustments in radiation dose or immunotherapy timing based on patient feedback[20]. Despite recent advances, several obstacles remain, including the scarcity of LDR-specific datasets and the difficulty in interpreting complex AI models[21]. Furthermore, clinical validation through prospective multicenter trials is essential to ensure the reliability and generalizability of AI-assisted decision-making[22]. The ongoing integration of radiomics, single-cell profiling, and explainable AI tools is expected to strengthen the role of AI in delivering precision LDR therapy.

CONCLUSION

The heterogeneity of immune responses to LDR therapy presents a significant challenge for its optimization in cancer treatment. AI offers a compelling solution by integrating multidimensional data to enable individualized outcome prediction and adaptive therapeutic strategies. Although obstacles such as data availability and model interpretability remain, the integration of AI with LDR nonetheless holds transformative potential for advancing precision oncology. Future efforts should prioritize standardized data collection, interpretable AI frameworks, and robust clinical trials to bridge the gap between research and practice. Harnessing AI could ultimately transform LDR therapy into a cornerstone of personalized cancer care.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Radiology, nuclear medicine and medical imaging

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade A

Creativity or Innovation: Grade A

Scientific Significance: Grade B

P-Reviewer: Ampollini L S-Editor: Liu H L-Editor: Filipodia P-Editor: Yu HG

References
1.  Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y, Sun Z. Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis. 2023;14:679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
2.  Wang SY, Wu JX, An X, Yuan Z, Ren YF, Yu XF, Tian XD, Wei W. Structural and temporal dynamics analysis on immune response in low-dose radiation: History, research hotspots and emerging trends. World J Radiol. 2025;17:101636.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (1)]
3.  Harary PM, Rajaram S, Chen MS, Hori YS, Park DJ, Chang SD. Genomic predictors of radiation response: recent progress towards personalized radiotherapy for brain metastases. Cell Death Discov. 2024;10:501.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
4.  Jia Q, Wang A, Yuan Y, Zhu B, Long H. Heterogeneity of the tumor immune microenvironment and its clinical relevance. Exp Hematol Oncol. 2022;11:24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 110]  [Article Influence: 36.7]  [Reference Citation Analysis (0)]
5.  Lee TF, Chang CH, Chi CH, Liu YH, Shao JC, Hsieh YW, Yang PY, Tseng CD, Chiu CL, Hu YC, Lin YW, Chao PJ, Lee SH, Yeh SA. Utilizing radiomics and dosiomics with AI for precision prediction of radiation dermatitis in breast cancer patients. BMC Cancer. 2024;24:965.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
6.  Upadhaya T, Chetty IJ, McKenzie EM, Bagher-Ebadian H, Atkins KM. Application of CT-based foundational artificial intelligence and radiomics models for prediction of survival for lung cancer patients treated on the NRG/RTOG 0617 clinical trial. BJR Open. 2024;6:tzae038.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
7.  Gao L, Zhang A. Low-dose radiotherapy effects the progression of anti-tumor response. Transl Oncol. 2023;35:101710.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
8.  Lumniczky K, Impens N, Armengol G, Candéias S, Georgakilas AG, Hornhardt S, Martin OA, Rödel F, Schaue D. Low dose ionizing radiation effects on the immune system. Environ Int. 2021;149:106212.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 50]  [Cited by in RCA: 98]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
9.  Takashima ME, Berg TJ, Morris ZS. The Effects of Radiation Dose Heterogeneity on the Tumor Microenvironment and Anti-Tumor Immunity. Semin Radiat Oncol. 2024;34:262-271.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
10.  Wang Z, Wang L, Zhang Q, Xu Y, Zhang C. Long-term outcome and antitumor immune activation response in prostate cancer treated with low-dose-rate brachytherapy. Medicine (Baltimore). 2024;103:e40574.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
11.  Enei Y, Urabe F, Miki K, Suzuki H, Iwatani K, Tashiro K, Tsuzuki S, Furuta A, Sasaki T, Sato S, Takahashi H, Aoki M, Kimura T. Effect of Adequate Local Radiation Dose on Oncological Outcomes in Localized Prostate Cancer Patients Treated With Low-dose-rate Brachytherapy. Anticancer Res. 2023;43:3159-3166.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
12.  Bergeron P, Dos Santos M, Sitterle L, Tarlet G, Lavigne J, Liu W, Gerbé de Thoré M, Clémenson C, Meziani L, Schott C, Mazzaschi G, Berthelot K, Benadjaoud MA, Milliat F, Deutsch E, Mondini M. Non-homogenous intratumor ionizing radiation doses synergize with PD1 and CXCR2 blockade. Nat Commun. 2024;15:8845.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
13.  AlOsaimi HM, Alshilash AM, Al-Saif LK, Bosbait JM, Albeladi RS, Almutairi DR, Alhazzaa AA, Alluqmani TA, Al Qahtani SM, Almohammadi SA, Alamri RA, Alkurdi AA, Aljohani WK, Alraddadi RH, Alshammari MK. AI models for the identification of prognostic and predictive biomarkers in lung cancer: a systematic review and meta-analysis. Front Oncol. 2025;15:1424647.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
14.  Corti C, Cobanaj M, Dee EC, Criscitiello C, Tolaney SM, Celi LA, Curigliano G. Artificial intelligence in cancer research and precision medicine: Applications, limitations and priorities to drive transformation in the delivery of equitable and unbiased care. Cancer Treat Rev. 2023;112:102498.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
15.  Cui S, Hope A, Dilling TJ, Dawson LA, Ten Haken R, El Naqa I. Artificial Intelligence for Outcome Modeling in Radiotherapy. Semin Radiat Oncol. 2022;32:351-364.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
16.  Rakaee M, Adib E, Ricciuti B, Sholl LM, Shi W, Alessi JV, Cortellini A, Fulgenzi CAM, Viola P, Pinato DJ, Hashemi S, Bahce I, Houda I, Ulas EB, Radonic T, Väyrynen JP, Richardsen E, Jamaly S, Andersen S, Donnem T, Awad MM, Kwiatkowski DJ. Association of Machine Learning-Based Assessment of Tumor-Infiltrating Lymphocytes on Standard Histologic Images With Outcomes of Immunotherapy in Patients With NSCLC. JAMA Oncol. 2023;9:51-60.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 25]  [Cited by in RCA: 64]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
17.  Hildebrand LA, Pierce CJ, Dennis M, Paracha M, Maoz A. Artificial Intelligence for Histology-Based Detection of Microsatellite Instability and Prediction of Response to Immunotherapy in Colorectal Cancer. Cancers (Basel). 2021;13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 41]  [Cited by in RCA: 54]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
18.  Wang J, Tu W, Qiu J, Wang D. Predicting prognosis and immunotherapeutic response of clear cell renal cell carcinoma. Front Pharmacol. 2022;13:984080.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
19.  Mueller AN, Morrisey S, Miller HA, Hu X, Kumar R, Ngo PT, Yan J, Frieboes HB. Prediction of lung cancer immunotherapy response via machine learning analysis of immune cell lineage and surface markers. Cancer Biomark. 2022;34:681-692.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
20.  Birla M, Rajan, Roy PG, Gupta I, Malik PS. Integrating Artificial Intelligence-Driven Wearable Technology in Oncology Decision-Making: A Narrative Review. Oncology. 2025;103:69-82.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
21.  Wang H, Fu T, Du Y, Gao W, Huang K, Liu Z, Chandak P, Liu S, Van Katwyk P, Deac A, Anandkumar A, Bergen K, Gomes CP, Ho S, Kohli P, Lasenby J, Leskovec J, Liu TY, Manrai A, Marks D, Ramsundar B, Song L, Sun J, Tang J, Veličković P, Welling M, Zhang L, Coley CW, Bengio Y, Zitnik M. Scientific discovery in the age of artificial intelligence. Nature. 2023;620:47-60.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 270]  [Article Influence: 135.0]  [Reference Citation Analysis (0)]
22.  Kann BH, Hosny A, Aerts HJWL. Artificial intelligence for clinical oncology. Cancer Cell. 2021;39:916-927.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 170]  [Cited by in RCA: 160]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]