- Review
- Open access
- Published:
The clinical application of artificial intelligence in cancer precision treatment
Journal of Translational Medicine volume 23, Article number: 120 (2025)
Abstract
Background
Artificial intelligence has made significant contributions to oncology through the availability of high-dimensional datasets and advances in computing and deep learning. Cancer precision medicine aims to optimize therapeutic outcomes and reduce side effects for individual cancer patients. However, a comprehensive review describing the impact of artificial intelligence on cancer precision medicine is lacking.
Observations
By collecting and integrating large volumes of data and applying it to clinical tasks across various algorithms and models, artificial intelligence plays a significant role in cancer precision medicine. Here, we describe the general principles of artificial intelligence, including machine learning and deep learning. We further summarize the latest developments in artificial intelligence applications in cancer precision medicine. In tumor precision treatment, artificial intelligence plays a crucial role in individualizing both conventional and emerging therapies. In specific fields, including target prediction, targeted drug generation, immunotherapy response prediction, neoantigen prediction, and identification of long non-coding RNA, artificial intelligence offers promising perspectives. Finally, we outline the current challenges and ethical issues in the field.
Conclusions
Recent clinical studies demonstrate that artificial intelligence is involved in cancer precision medicine and has the potential to benefit cancer healthcare, particularly by optimizing conventional therapies, emerging targeted therapies, and individual immunotherapies. This review aims to provide valuable resources to clinicians and researchers and encourage further investigation in this field.
McCarthy J et al. coined “artificial intelligence” at the initiation of the 1956 Dartmouth summer research project, establishing the foundation of this field. This term, broadly defined as ‘the approach to creating intelligent machines,’ was based on the hypothesis that machines could emulate various aspects of learning and intelligence [1]. Briefly speaking, Artificial intelligence (AI) refers to a branch of computer science that emulates human intelligent behavior and addresses challenges such as reasoning, knowledge representation, automated planning, natural language processing, machine perception, robotics, etc [2,3,4]. AI predominantly encompasses machine learning and deep learning, although these three terms are sometimes used synonymously. Emerging as a subtype of AI, machine learning focuses on utilizing computational algorithms to identify patterns within data and fit predictive models to it [5]. Machine learning can be categorized as supervised learning, unsupervised learning, semi-supervised learning and reinforcement learning. Supervised learning uses human-labelled data for training and predicts outcomes through classification or regression. On the contrary, data-driven unsupervised learning are used for clustering or dimension reduction by training on unlabeled data. Semi-supervised learning is in between, using both labelled and unlabeled data for training [6]. Finally, reinforcement learning is a specific method that compares against a pre-defined goal in iterative interactions, corresponding to the generation of rewards or penalties, and is performed repeatedly [7, 8]. Each of these methods can be subdivided into different algorithms that are used individually or integrated. Deep learning is a subset of machine learning that typically does not require feature extraction. Its end-to-end learning capabilities significantly enhance the processing of natural raw data, a capability not present in traditional machine learning [9]. (Fig. 1) Focusing on deep neural networks (DNNs), deep learning uses algorithms composed of multiple hidden processing layers to integrate extensive datasets and explore complex relationships, greatly impacting fields including image identification, speech recognition, object detection, and natural language processing, especially when involving biomedicine [10].
The concise overview of artificial intelligence and its associated algorithms. Artificial intelligence encompasses machine learning, which includes deep learning. The terms may be used interchangeably despite distinct characteristics. Algorithms serve as the fundamental basis for artificial intelligence and are classified into various categories
According to GLOBOCAN 2020, it is expected that there will be 28.4 million cancer cases globally in 2040. Besides powerful advocacy for cancer prevention, optimizing cancer management is crucial in the current context. Obama launched the Precision Medicine Initiative as a revolutionary strategy for tailored cancer care in his 2015 State of the Union address. Since then, the world has been a shift towards cancer precision medicine (CPM) - individualized cancer care that places at its core the unique characteristics of patients [11, 12], including clinical records, health history, lifestyle, genome, epigenome, transcriptome, proteome, metabolome, medical image, histopathologic feature and new data streams that may develop as oncology research progresses [13, 14]. Diverse patient-specific data is experiencing explosive growth with medical technology advances and is constantly in flux during a patient’s trajectory, which requires significant expertise and time for collation and utilization. Thus, the use of AI techniques has become progressively ubiquitous due to their ability to enhance efficiency and reproducibility through automation [5]. Furthermore, a single type of data is extremely limited in providing a comprehensive view of a tumor. AI is driving the development of multimodal data integration in oncology and enhancing the precision of its predictive models by combining complementary information from different modalities [13]. The integration of highly heterogeneous data can be effectively managed and analyzed by deep learning neural networks due to their capacity to include diverse raw data types and flexibility in data processing [15].
This review focused on the latest AI applications for CPM, particularly in the fields of optimizing conventional therapy, target prediction, drug selection, and personalized immunotherapy. (Fig. 2) Moreover, we discuss the current limitations and prospects in translating AI to clinical practice and present potential solutions to bridge the gap between AI and real-world medical practice.
The comprehensive flowchart illustrating the application of artificial intelligence in precision cancer treatment. With various algorithms at its core, artificial intelligence is gradually being involved in the field of oncology. Individual characteristics data, generated from the whole process of cancer management, can be seamlessly integrated and efficiently utilized by artificial intelligence systems to facilitate accurate precision diagnoses and subsequently advance the efficacy of precision treatment strategies
Optimizing conventional therapy with AI techniques
Conventional treatments, such as surgery, radiotherapy and chemotherapy are still widely used in solid tumor management. To meet the demand for better outcomes, less risk, and more economical cost, achieving accuracy and individualization of conventional therapies is crucial. AI models have the ability to process, analyze and integrate datasets efficiently, allowing each aspect in conventional therapeutic areas to change from relatively uniform processes to individualized solutions. (Fig. 3)
Artificial intelligence makes conventional cancer treatment more individualized. AI is revolutionizing conventional cancer treatments such as surgery, radiotherapy, and chemotherapy. It enables greater individualization throughout the entire treatment process, including pre-treatment decision-making, real-time management during treatment, and post-treatment care. AI approaches maximize the effectiveness of traditional treatment methods within the framework of precision medicine, enhancing therapeutic outcomes while minimizing side effects
AI in perioperative decision-making
Through AI algorithms, analyzing complex datasets supports accurate risk prediction, personalized surgical strategies and real-time adjustments, and leads to improved surgical outcomes and fewer complications. The combination of artificial intelligence and advanced imaging technologies continues to advance surgical precision and patient management [16]. Thus, AI has become an essential tool in the whole perioperative processes, including preoperative planning, intraoperative guidance, as well as postoperative care [17].
Preoperative planning
AI enhances preoperative strategies by providing surgeons with comprehensive insights into patient risk factors, tumor characteristics, and expected surgical outcomes. Especially, Lymph node metastasis (LNM) is critical in determining whether surgery is appropriate for patients with solid tumors. Machine learning significantly optimizes the decision-making for operation by predicting LNM [18]. In colorectal cancer, Song JH et al. used a deep learning model with H&E-stained endoscopic resection specimens to predict LNM. Compared to traditional methods using clinical and/or pathological features, this model performed better in predicting LNM for stage-T1 (AUC = 0.764 in validation set ). It substantially reducing unnecessary additional surgeries compared to current guidelines (67.4% vs. 82.5%) [19]. Similarly in other digestive system cancers, researchers combined AI algorithms with other data modalities, including clinical variables [20, 21], histopathological images [22], and radiomics features [23,24,25], enhancing the prediction of LNM. Moreover, in solid cancers of other systems, AI models also show great potential to achieve the same goal, such as thyroid [26], lung [27], breast [28], and endometrial tumors [29]. For example, in a multicenter retrospective study, researchers proposed an AI-based model of outstanding performance that demonstrated high accuracy in predicting LNM in bladder cancer (AUC ranged from 0.978 to 0.998 in 5 validation sets). This diagnostic model outperformed senior pathologists in diagnostic sensitivity (0.983 vs. 0.947) [30].
Intraoperative guidance
After personalizing the surgical plan, AI has also become a transformative approach in intraoperative care, which can significantly enhance precision, efficiency, and decision-making during surgical procedures. Firstly, AI provides real-time diagnosis and decision support through detection and localization using single dataset or multimodal datasets [31]. Based on digital pathology data, Sendín-Martín M et al. developed an automated approach using a deep learning algorithm with ex vivo confocal microscopy for rapid detection of basal cell carcinoma during Mohs surgery (AUC = 0.94 in validation set) [32]. Based on imaging data, in neuro-oncology, AI models have revealed its potential in detecting tumor boundaries [33, 34]. A deep learning model was used to identify the eloquent cortex from rs-fMRI connectivity before surgery [35], while an AI technique combining CNN and NIR-II fluorescence imaging (named FL-CNN) was similarly reported to enhance surgical safety by detecting glioma boundaries [36]. Besides using single data, real-time surgical decision support systems use multimodal AI to integrate patient-specific data to create dynamic dashboards that guide surgeons at critical moments [37]. These systems enhance situational awareness and inform decisions such as resection margins, biopsy necessity, or anatomical navigation. For example, Sturgeon, a patient-independent transfer-learning neural network, using rapid nanopore sequencing, allows rapid access to sparse methylation profiles during surgery, enabling accurate diagnosis of most samples within 40 min of starting sequencing [38].
In addition, AI-driven surgical robots are at the forefront of development [39]. They combine computer vision with machine learning algorithms to perform complex tasks such as tissue dissection, suture placement and real-time anatomical recognition [40,41,42]. These systems improve accuracy and reduce human error, and are particularly beneficial in minimally invasive surgery where precision is critical. Recently, Hani J Marcus et al. have presented the IDEAL framework for surgical robotics, providing guidelines for AI-driven surgical robots in clinical translational phases [43].
Moreover, predictive modelling has become an important tool, which assist in monitoring vital signs and physiological parameters during surgery. For example, the Hypotension Prediction Index System, which predicts the onset of intraoperative hypotension, enabling timely intervention and reduction of complications [44]. By analyzing time-series data from intraoperative monitoring systems, AI models– particularly one model based on recurrent neural networks (RNNs) - can also predict other complications such as hypoxia and excessive blood loss.(AUC = 0.94 in validation set of approximately 25000 patients) [45].
Furthermore, intraoperative AI has advantages in addressing non-technical aspects of surgery, such as optimizing surgical teamwork and promoting surgical skills education. An important development in improving surgical collaboration is the creation of an AI-powered mentor designed to detect how well a surgical team’s mental frameworks are aligned, which could be vital for surgical outcomes [46]. Furthermore, Various non-skill data streams including physiological metrics like heart rate variability, staffing levels and equipment availability could be integrated into a system designed for automation and optimization. Common intraoperative challenges such as fatigue, communication breakdowns and turnover issues, and equipment shortages could be solved through this kind of AI-system [47, 48]. In terms of surgical education, traditional surgical education lack effective assessment of feedbacks of trainees and consumes a longer period of time [49]. Reliable feedback from automated analysis of data makes AI methods uniquely suited to training surgical skills [50]. Rafal Kocielnik et al. proposed a collaborative human-machine refinement process that uses unsupervised machine learning algorithms to discover categories of feedback in the surgical record that significantly enhance the prediction of behavioral change in trainees. This facilitates modification of trainee behaviors and more efficient skill acquisition [51].
Postoperative care and recovery
After surgery is conducted, AI holds promising potential in postoperative management by predicting complications, tracking recovery, and providing personalized follow-up. This approach reduces readmission rates, improves patient outcomes, and optimizes healthcare resources allocation. For example, to better predict major postoperative complications in patients undergoing cytoreductive surgery, Deng H et al. utilized US Hyperthermic Intraperitoneal Chemotherapy Collaborative Database to create an explainable machine learning model (AUC = 0.75 in validation set) [52], which proved more accurate than the published MLR model (AUC = 0.54). After other types of tumor surgery, Hassan AM et al. developed a random forest model for predicting mastectomy skin flap necrosis (AUC = 0.70 in validation set) [53]. In measuring cosmetic outcomes, Kim DY et al. evaluated them after reconstructed breast surgery, using a generative adversarial network [54].
AI in individual radiotherapy
Besides surgery, radiotherapy remains one of the primary treatment modalities for cancer. The processes of radiotherapy generate various kinds of data, including clinical variables, imaging data, biological samples, planning parameters, and machine data. AI approaches show advantages in integrating these data to support tasks such as accurate contouring and dose prediction, thereby enhancing individualized radiotherapy [55, 56].
Tumor contouring
Tumor contouring is usually the first step of radiotherapy procedure. Traditionally, contouring of gross tumor volume, clinical target volume and organs at risk have been performed manually by radiation oncologists, relying highly on expertise, which results in unavoidable intra-observer viability and significant time consumption. AI-mediated multistep integrated radiation therapy workflow has shown promising results in a single center [57]. Especially, deep-learning based systems has shown remarkable progress in this field [58]. A 3D U-Net, base on deep learning methods, has automated and standardized the process of segmentation, delineating tumor boundaries with high precision [59]. Additionally, Shi F et al. generated RTP-net, a deep-learning model for radiotherapy planning, showing high accuracy with average Dice score of 0.95 [60]. To compare AI-led and oncologist-led methods, Jordan W et al. evaluated deep learning based auto-segmented contours trained by a single oncologist and expert contours created by multiple oncologists, concluding that the former performed accurately in organs at risk and provided significant time savings [61].
Dose optimizing
Precision-dose radiotherapy helps to maximize the effect on tumors while sparing healthy tissue. In terms of dose predicting, an individualized auto-planning system is urgently needed, as current radiotherapy operators often determine the dose of radiation based on standard protocols, adjusting them iteratively to coordinate doses in different regions. The method of iterative trials limits the efficiency and precision of radiotherapy [62]. Systems based on AI algorithms has made considerable progress in accurately making dose prediction [63, 64]. For example, Florian M et al. generated a generative adversarial network model to predict dose distributions inside unknown phantoms accurately, with potential applications in novel radiotherapy techniques requiring high accuracy, such as synchrotron X-ray microbeam radiation therapy [65]. Furthermore, the combination of radiomics data and dosimetry data has been widely used in response and toxicity prediction. For example, Jin C et al. presented a multi-task deep learning approach that allows solid tumor radiotherapy response prediction [66]. The imaging-based model, integrated with blood-based tumor markers, substantially improves prediction accuracy with AUC = 0.97 in validation set.
AI in personalized chemotherapy
While chemotherapy is a fundamental treatment for many cancers, it is often associated with severe side effects and variations in patient response. AI has become an invaluable tool for optimizing chemotherapy in therapy planning and drug utilization.
Chemotherapy planning
AI methods were primarily used for decision-making in chemotherapy of different cancers. In gastric cancer, Sundar R et al. used a genomic-based random forest model to guide the selection of patients with gastric cancer who would benefit from paclitaxel [67]. In colorectal cancer, ssing deep learning and H&E-stained tissue sections, DoMore-v1-CRC provided a clinical decision support system that stratified patients with stage II and III colorectal cancer with favorable prognosis, helping to avoid unnecessary adjuvant chemotherapy [68]. Similar results were obtained when a machine learning framework was used to predict the response of neoadjuvant chemotherapy in muscle-invasive bladder cancer [69]. In ovarian cancer, a 2021 study showed that AI models trained on gene expression data could predict the efficacy of platinum-based chemotherapy with more than 80% accuracy [70]. These predictive models help oncologists to avoid ineffective treatments, minimize the risk of unnecessary toxicity, and select the best beneficiaries.
Optimizing drug efficacy
Even if the potential beneficiaries of chemotherapy can be identified, drug resistance largely limits the application of conventional chemotherapy. Machine learning can be used to accurately predict patients’ resistance to chemotherapy drugs, potentially mitigating this issue. Sasaki K et al. integrated machine learning algorithms with CT images to predict neoadjuvant chemotherapy resistance in patients with advanced gastric cancer. The integrated model demonstrated better results than current clinical models (AUC > 0.752 in validation sets from three centers) [71]. The latest developments in AI methodologies and tools have also enhanced our understanding of cancer multidrug resistance. In response, Fu L et al. presented a new viewpoint on repurposing non-oncology small-molecule drugs as an attractive approach to improve cancer therapy [72].
AI in targeted treatment
Focusing on specific molecular targets involved in cancer progression, targeted therapy has become one of the most popular treatments in CPM [73]. Nevertheless, due to the limited understanding of carcinogenesis, obstacles remain in the development of targeted therapies, especially in the fields of target prediction and drug selection. Implementing the entire process from target identification to targeted drug therapy is challenging based on a single technique or single-omics data. Therefore, AI holds considerable potential to effectively integrate multiple datasets and explore interactions between genes and gene products involved in tumorigenesis, thereby advancing the clinical application of targeted drugs [74]. (Fig. 4)
Artificial intelligence involves throughout the targeted therapy process. AI plays an important role throughout the entire process of targeted therapy. From the identification of therapeutic targets and the development and utilization of targeted drugs to the precise delivery of these drugs, AI demonstrates significant potential at every stage of the targeted therapy workflow
Prediction of emerging targets with AI
Precisely predicting drugs is the basis of targeted therapy. AI-driven algorithms primarily predict new druggable targets by combining omics data. Using nonparametric random forest to analysis genomics data, Zare A et al. reported an inflammatory breast cancer (IBC)-specific gene signature (G59) to show the molecular differences between IBC and non-IBC patients, paving the path for discovering therapeutic targets at the genomic level [75]. In addition, the combination of machine learning models and mechanistic information could help to demonstrate signal transduction network heterogeneity and identify potential therapeutic target. For example, Pham TH et al. integrated a machine-learning framework with chemicogenomics and transcriptomics data to identify YAP/TAZ dependency across cancers and propose a potential therapeutic target in Hippo pathway dysregulation [76]. Moreover, integrating multi-omics data, Xiao Y et al. used machine learning methods based on the polar metabolome and lipidome analysis to distinguish triple-negative breast tumors into two prognostic metabolomics subgroups (C2 and C3) [77]. They subsequently revealed that N-acetyl-aspartyl-glutamate is a critical tumor-promoting metabolite and might exist as a potential therapeutic target for high-risk C2 and C3 tumors.
AI in targeted drugs
After identifying potential therapeutic targets, AI could participate form drug generation to selection to utilization. With AI models, it is easier to generate and validate drugs that interact most effectively. Furthermore, drug repurposing and combination therapies can be realized through powerful analytical capabilities of AI, which are also of vital importance combating potential drug resistance and minimizing side effects.
Drug generation
For traditional drug generation, the industry and reasearch have often focused on the chemical aspects of compound generation while overlooking the biological consequences. In the stage of drug discovery, aberrant m6A regulators have emerged as popular drug targets in recent years. AI-assisted synthesis of FTO inhibitors and METTL3 inhibitors has greater advantages over natural compounds [78]. Using deep learning and molecular dynamics simulation-based drug screening, Zhang H et al. identified UM-164 as a potential TIPE2 inhibitor [79]. Despite only focusing on chemical aspects, following machine-learning-based virtual screening, surface plasmon resonance, molecular docking, and pharmacokinetic analyses identified two potential inhibitors of anti-apoptotic members of the Bcl-2 family in solid tumors [80]. In addition, knowledge of drug mode of action (MoA) is an important part of developing anti-cancer drugs. Mohamad Saoud et al. have successfully predicted MoA of novel drug candidates in prostate cancer cells using metabolomics data combined with machine learning, and the predicted results of MoA based on prostate cancer cell therapy was successfully validated in breast cancer and Ewing’s sarcoma [81].
Drug selection
Turing from drug generation to practical clinics, drug selection is essential for individualized therapy. Based primarily on the chemical information of drugs, Su R et al. proposed a deep learning network named Siamese Response Deep Factorization Machines to directly rank the drugs for helping each patient receive the most effective drugs [82]. Additionally, at the single-cell sequencing data level, Chen J et al. developed a deep transfer learning model called scDEAL to predict and select drugs [83]. Other investigators have also obtained the goal with using AI to integrate data from different modalities. For example, Liu X et al. predicted drug response to guide anti-cancer drug selection by GraphCDR, a contrastive learning neural network based on multi-omics profiles and chemical structure of drugs [84].
Drug utilization
In the area of targeted drug utilization, AI contributes to drugs combination, repurposing, and delivery. Multi-targeted drugs combination is an emerging theme due to the increased understanding of intra-tumor heterogeneity, which is an important attempt in precision medicine. However, identifying multi-targets and selecting co-suppressive patient-specific therapies is difficult, as it is challenging in terms of both efficacy and toxicity. Researchers have proposed the AI-driven model scTherapy in conjunction with single-cell transcriptomics data to priorities multi-targeted therapeutic options for tumor patients. In a pan-cancer analysis of five cancer types, 19% of the therapeutic regimens generated by the system were patient-specific [85]. Using multi-omics data, Li X et al. also integrated cancer informatics algorithms and machine learning methods to develop a system called REFLECT [86]. It could optimize therapeutic benefits by choosing proper drug combinations. To improve drug repurposing, Cui C et al. integrated drug-exposure expression profiles and drug-drug links, using a graph neural network to develop a breast tumor drug repurposing strategy [87]. In addition, given the heterogeneity of delivery among drugs, using AI to enhance nanomedicine design for improved drug delivery efficiency represents a promising development [88, 89].
AI in personalized immunotherapy
Cancer immunotherapy represents a transformative approach in cancer treatment, restoring the normal antitumor immune response to control and eliminate tumors by restarting the tumor-immune cycle [90]. However, the effectiveness of immunotherapy varies significantly among patients. With integrating multimodal data, AI has emerged as a key tool to better understand tumor microenvironment (TME) and tumor immunology. Specially, AI models help to predict patient responses to immunotherapy, guide the discovery of neoantigens and tumor vaccines, and understand new concerns in tumor immunity such as LncRNA. (Fig. 5)
Artificial intelligence’s promising role in personalized immunotherapy. AI holds significant promise in personalized immunotherapy by contributing to key aspects of cancer immunotherapy, including predicting immune responses and generating tumor vaccines. This means that AI not only enhances the precision of widely used immunotherapeutic approaches, such as immune checkpoint inhibitors, but also exhibits great potential in the development of future individualized immunotherapy strategies
AI in predicting immunotherapy response
Immune checkpoint inhibitors(ICIs) are the most widely used approach in the field of tumor immunotherapy, and predicting its biomarkers is extremely important to enable personalized immunotherapy. Analyzing genomics, transcriptomics, epigenomics, radiomics and digital pathology data derived from high-throughput sequencing, AI is promising in predicting biomarkers for immunotherapy, including the most common biomarkers for ICIs, novel biomarkers, and predictive tools to be mined from TME [91, 92].
Through prediction of common biomarkers of ICIs
PD-L1 is one of the most important biomarkers in ICIs. Assessment of PD-L1 is crucial for treatment stratification and predicting immunotherapy outcomes. Combining digital pathological images [93, 94] or radiomics data [95, 96], AI models have significantly enhanced the ability to predict PD-L1 expression in solid tumors. For example, in non-small cell lung cancer, Choi S et al. developed an analyzer for PD-L1 tumor proportion score using whole-slide images [97]. Comparing the accuracy of manual evaluation and AI-assisted evaluation, they found that AI assistance increased the overall concordance rate among pathologists to 90.2% (compared to 81.4% for manual evaluation) with statistical significance (P < 0.001). In terms of multimodal data integration, Vanguri R S et al. also integrated AI and pathology with CT images, demonstrating an AUC of 0.80 in validation set for their model in predicting ICIs response in lung tumor [98].
In addition to PD-L1, other biomarkers are also potentially useful for predicting ICIs response with the aid of AI algorithms [99]. For example, Gong X et al. developed a machine-learning model and demonstrated that HLA gene expression is able to predict the immune subtypes of patients receiving ICIs [100]. Tumor mutational burden is another key feature that has been extensively studied. To evaluate tumor mutational burden and predict response to immune checkpoint blockade, researchers respectively combined AI algorithms with transcriptomics [101], radiogenomics [102], and digital pathology [103,104,105], demonstrating potential clinical benefits.
Through prediction of novel biomarkers
Many novel biomarkers have been confirmed with the assistance of AI [92]. Through genomics data, loss of heterozygosity status in human leukocyte antigen and genomic intra-tumor heterogeneity were identified to be associated with ICIs efficacy by Wang et al. with ML-based methods in non-small-cell lung cancer patients [106]. Using epigenomic data, artificial intelligence-driven approaches have identified a number of DNA methylation indicators as biomarkers for ICIs in different solid tumors [107, 108]. Through radiomics data, Dercle et al. used random forest algorithm to analysis 575 patients with melanoma treated with ICIs in KEYNOTE-002 and KEYNOTE-006 trials. They identified a radiomic biomarker composed of volumetric growth (absolute tumor volume difference), tumor volume, quantitative representation of tumor spatial heterogeneity, and quantitative representation of tumor edge phenotype, achieving precisely predicting OS (AUC = 0.92) [109].
The application of AI to integrate multimodal data to predict biomarkers has been a hot topic in recent years [110]. Through AI-driven multi-modal data integration, researchers have proposed a number of unique classifications to be used as biomarkers for immunotherapy, some of which have achieved good results [111]. For example, Shen et al. used ML-based methods to establish an immune cell phenotype with three classifications based on data from multiple sources, and identified that the LAG-3 + CD8 + T-cell population can be a novel biomarker for poorer OS and PFS in patients with melanoma and uroepithelial carcinoma(OS: P < 0.001; PFS: P = 0.004) [112]. Similarly, in bladder cancer, Shuai Ren et al. used the Graph Neural Networks model to integrate multi-omics data to generate a simple linear scoring model, responseScore, to predict immunotherapy response and identify key pathways. The model had an AUC of 0.839 in the validation set [113]. However, it is important to notice that multi-data integration brings higher challenges on the quality of input data. In addition, the validation datasets of these published multi-modal data integration AI systems are limited, so the evaluation of these AI procedures needs to be cautious.
Through exploration of TME
TME is the living space surrounding the tumor cells, including various types of cells, factors and matrix. AI techniques can accurately predict the status of TME and improve understanding of factors impacting immunotherapy efficacy. Along with the study of TME, some biomarkers, including neutrophil-lymphocyte ratio and tumor stemness showed potential predictive effects. With the help of support vector machine classifiers and random forests analysis, Raman spectroscopy shows potential for predicting response to ICIs in TME [114].
The tumor immune microenvironment (TIME), which represents the immune context of TME, has been proven to be associated with carcinogenesis, tumor progression, and identification of potential therapeutic targets [115, 116]. Tumor-infiltrating immune cells in TIME were widely researched. Based on biomarkers of tumor-infiltrating lymphocytes (TILs), Park S et al. used AI models and whole-slide images to predict the efficacy of ICIs and defined three immune phenotypes for predicting tumor response [117]. Similarly, Ao Z et al. developed a TILs score analyzer with clinical data and machine-learning algorithms. Using this analyzer, they evaluated the efficacy of drugs and selected an epigenetic drug called LSD1i which could enhance the therapeutic benefit of ICIs [118]. Furthermore, understanding T cell exhaustion (TEX) heterogeneity also helps to evaluate immunotherapies. Zhang Z et al. developed a machine learning-based gene signature to model the hierarchical TEX stages and optimize immunotherapy [119]. Similarly, Using unsupervised deep learning algorithms and analyzing spatial transcriptomics data, Chia-Kuei Mo et al. increased understanding of TME and proposed enhanced markers of immune depletion [120].
Using AI to target new tumor antigens
The search for tumor antigens as well as the developing of tumor vaccines are emerging strategies in immunotherapy. Somatic cancer mutations expressed especially in cancer cells, and these gene products could become neoantigens after breaking down and be targeted. Recognized by CD4 + or CD8 + T cells without being subject to central tolerance, neoantigens have great potential as targets for T cell-based immunotherapies [121]. As neoantigens are mostly unique for each patient, this therapeutic method is inherently personalized.
Neoantigen identification
Neoantigens are essentially the products of gene mutations in tumor cells. The development of AI techniques has promoted the discovery of mutations by analyzing high-throughput sequencing data [122,123,124]. Based on single data modality, by processing and analyzing transcriptome data, a machine-learning computational pipeline named EasyFuse developed by Weber D et al. to could help to detect cancer-associated gene fusions, potential sources of highly immunogenic neoantigens [125]. Integrating multimodal omics data including genomics, transcriptomics and proteomics data, the researchers developed an advanced ML-driven computational framework program called NeoDisc. This procedure was shown to outperform conventional methods in accurately prioritizing immunogenic neoantigens [126].
Prediction of MHC-antigen binding
Since neoantigens are presented on major histocompatibility complex (MHC) before bonding T cells, predicting peptide-MHC binding, is as crucial as predicting neoantigens and important in generating tumor vaccines [127]. Using label-agnostic protein sequence data, a transfer learning model called MHCRoBERTa was employed to address this task [128]. Using machine learning methods, the integrated model called Anthem combined mass spectrometry with machine learning, and particularly predicted HLA-I binding [129]. Similarly, Haodong Xu et al.so generated a DL-based system called ImmuneApp to predict HLA-I binding [130]. For the accurate prediction of HLA-II binding, Racle J et al. also realized it with a machine-learning framework [131].
Designing of personalized tumor vaccines
By simulating interactions with the immune system, AI contributes to the design and optimization of vaccines. It helps guide researchers in selecting the most promising personalized vaccines for further development and improves the efficiency and effectiveness of the vaccine development process [132]. Powderly JD et al. and Xu Y et al. designed mRNA vaccines and peptide vaccines with the help of AI, respectively, and conducted phase I clinical trials in different cancer areas [133, 134]. In addition, adjuvants play a crucial role as an essential component in the development of effective cancer vaccines that enhance the body’s immune response to cancer cells and improve the effectiveness of cancer vaccines [135]. AI is involved in the development of tumor vaccine adjuvants in five ways: preliminary design, virtual screening, property prediction, and reuse [136]. For example, Sajjad Haider et al. successfully predicted three potent CXCL12 inhibitors that could potentially act as adjuvants using a ligand-based virtual screening tool [137].
Promoting the use of long non-coding RNA in immunotherapy with AI
Long non-coding RNA (lncRNA), which regulates transcription, epigenetic modifications, and other post-transcriptional mechanisms, is closely associated with cancer immunity regulation and the TME [138]. AI helps to elucidate its various mechanisms and its role in tumor immunotherapy. It is documented in several types of cancer that lncRNA interacts with RNA-binding protein, and the use of machine learning methods revealed details of the mechanism and the potential of RNA-based therapeutics [139]. Given the importance of TILs in immunotherapy mentioned above, Zhou M et al. and Zhang N et al. respectively explored the tumor-infiltrating immune cell-associated lncRNA in low-grade glioma and tumor-infiltrating B lymphocytes [140]. Integrating machine learning algorithms and clinical profiles, they found that the tumor-infiltrating immune cell-associated lnc signature could select potential beneficiaries of immunotherapy. Liu Z et al. used an integrative procedure of machine learning to validate the clinical significance of lncRNA in colorectal tumors [141]. They constructed a consensus immune-related lncRNA signature to predict clinical outcomes, finding that the low-risk group benefited more from bevacizumab, whereas the high-risk group was better suited for fluorouracil-based chemotherapy. A consensus machine learning-derived lncRNA signature developed by Liu Z et al. reached a similar conclusion [142]. It was even further found to play a role in tumor recurrence prediction.
Limitations and challenges of AI in oncology
While AI has been widely studied in tumor management and has potentially shown a game-changing impact in this field, barriers remain in translating algorithms and research into clinical applications that require further discussion and investigation.
Quality and bias
Data bias could exist at every step of the process for AI applications [143, 144]. The greatest source of data used for training AI models is electronic health records from clinical activities, but unstructured and inconsistent records limit their usability, necessitating structured data organization [145]. Moreover, while data heterogeneity across institutions is common, evaluating external cohorts is crucial. Although more studies recently use training data from multiple centers, the representativeness of the target is still limited by biases that cannot be eliminated in gender, ethnicity, research interest, funding etc., potentially resulting in poor generalization of models and even increasing of health-care discrimination [146].
Interpretability and transparency
AI algorithms, especially black-box deep-learning models, often generate outputs without explaining the intermediate steps, resulting in a lack of interpretability [147]. This limitation in intelligibility challenges the collaboration between different clinicians or between AI systems and clinicians, partially limiting their real-world medical utility [148]. It is crucial for clinicians to understand AI models and relate them to human medical knowledge. In the era of precision oncology, this contributes to improved diagnostic accuracy and efficiency [149]. Biological network models in natural systems have the potential to enhance the cognition of how AI system works and improve its transparency [150]. Meanwhile, greater involvement of clinicians in data collection, bias mitigation, and technology application is needed to establish an efficient human-computer hybrid system rather than applying AI in isolation [151].
Security and privacy
Data security and privacy are also being challenged in the age of AI. Many AI systems, especially those connected to networks, face potential security risks, including malicious hacking that could compromise patient data privacy. Data breaches could lead to severe consequences, including the possibility of algorithm tampering to favor certain stakeholders. To address these risks, robust security measures, along with comprehensive laws and regulations, are essential for ensuring the safe deployment of AI applications.
Regulation and ethics
The regulatory framework for AI in healthcare is still evolving. Although some AI tools have been approved for clinical use by regulatory bodies like the FDA, the process of approving AI algorithms remains complex and time-consuming. The major ethical problem in this field is determining who should take responsibility for AI decision-making. Undeniably, clinicians providing medical advice, manufacturers of AI products, employers of clinicians, and health systems are all stakeholders in AI applications and should be held accountable if AI leads to suboptimal or adverse outcomes [152]. Under the current liability system, clinicians may hesitate to use AI especially when facing its opaqueness. Therefore, specialized adjudication systems together with special compensation systems funded by relevant people’s taxes or fees might be able to replace the traditional liability system in medical disputes involving AI [152].
Future prospects and conclusions
AI in oncology holds great prospects and has the potential to transform cancer treatment by enabling more precise, personalized and efficient interventions. Firstly, understanding the cancer biology will always be a focus and a challenge in the field of oncology, and integrating and analyzing an increasing number of multimodal datasets is the most effective means of addressing this issue. As the field of AI develops, it is becoming more and more capable of integrating data through ever-refined algorithms [153, 154]. Besides today’s most commonly used datasets from modern medical activities, including genomics data, imaging genomics data, TME-based data, and electronic health records, it is even possible to integrate potentially exploitable datasets such as facial and tongue visual diagnosis data generated from traditional Chinese medicine diagnosis and treatment [155]. This could better reveal the complex patterns of tumor behavior and treatment response and facilitate personalized tumor therapy. It is worth noting that most of the existing data used to train AI systems are derived from clinical studies. While this somewhat reduces bias, it undoubtedly makes AI systems less externally accessible. Therefore, compared to the limited amount of data originating from clinical studies, the larger and more complex real-world data is “an ocean” of lesser-explored data waiting to be integrated and utilized by AI [156]. In addition, the impact of AI on equality in oncology goes in both directions [157]. The ideal AI system is instead one of the best ways to address equality in oncology if its impact on health equality is considered in the early stages of its development, including integrating different datasets, addressing algorithmic bias, ensuring fair validation across different demographic and geographic contexts, and prioritizing fair outcomes. This is mainly in terms of removing implicit bias from clinicians’ subjective assessments [158]. AI provides tools to reduce disparities in cancer care by increasing access to diagnostics, treatments, and clinical trials across populations. In short, individualized patient information is used as the most important basis for decision-making. At the same time, AI has the potential to tailor tools for resource-limited settings, such as portable imaging devices and simplified diagnostic approaches, from which lower-income populations can still have access to advanced cancer treatments. Finally, the value of large-scale language models (such as ChatGPT and systems based on it) in oncology remains to be developed [154]. They can provide simplified communication and knowledge acquisition, enhance opportunities for patient engagement, and thus improve the efficiency of the entire cancer care process. In conclusion, it is undeniable that AI algorithms have great potential in tumor treatment, and promote the management of tumors in the era of precision medicine. However, gaps remain between AI models and their clinical implementation. With research fervor in this area extremely high, However, gaps remain between AI models and their clinical implementation. Large-scale prospective clinical trials are expected, and in particular, AI-driven cancer precision medicine should not be pursued cautiously to maintain cost-effectiveness. Future research should focus on improving security and interpretability to enhance coordination between AI and clinicians, making AI more applicable.
Data availability
Not applicable.
References
McCarthy J, Minsky ML, Rochester N, Shannon CE. A proposal for the Dartmouth Summer Research Project on Artificial Intelligence, August 31, 1955. AI Magazine. 2006;27(4):12. https://doi.org/10.1609/aimag.v27i4.1904.
Hamet P, Tremblay J. Artificial intelligence in medicine. Metab Clin Exp. 2017;36–s40. https://doi.org/10.1016/j.metabol.2017.01.011. 69s:S.
Bera K, Schalper KA, Rimm DL, Velcheti V, Madabhushi A. Artificial intelligence in digital pathology - new tools for diagnosis and precision oncology. Nat Reviews Clin Oncol. 2019;16(11):703–15. https://doi.org/10.1038/s41571-019-0252-y.
Loftus TJ, Shickel B, Ozrazgat-Baslanti T, Ren Y, Glicksberg BS, Cao J, et al. Artificial intelligence-enabled decision support in nephrology. Nat Rev Nephrol. 2022;18(7):452–65. https://doi.org/10.1038/s41581-022-00562-3.
Greener JG, Kandathil SM, Moffat L, Jones DT. A guide to machine learning for biologists. Nat Rev Mol Cell Biol. 2022;23(1):40–55. https://doi.org/10.1038/s41580-021-00407-0.
Moore A, Shim H, Zhu J, Gong M. Semi-supervised learning under General Causal models. IEEE Trans Neural Networks Learn Syst. 2024;Pp. https://doi.org/10.1109/tnnls.2024.3392750.
Theodosiou AA, Read RC. Artificial intelligence, machine learning and deep learning: potential resources for the infection clinician. J Infect. 2023;87(4):287–94. https://doi.org/10.1016/j.jinf.2023.07.006.
Jiang Y, Luo J, Huang D, Liu Y, Li DD. Machine learning advances in Microbiology: a review of methods and applications. Front Microbiol. 2022;13:925454. https://doi.org/10.3389/fmicb.2022.925454.
LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015;521(7553):436–44. https://doi.org/10.1038/nature14539.
Wainberg M, Merico D, Delong A, Frey BJ. Deep learning in biomedicine. Nat Biotechnol. 2018;36(9):829–38. https://doi.org/10.1038/nbt.4233.
Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372(9):793–5. https://doi.org/10.1056/NEJMp1500523.
Ashley EA. The precision medicine initiative: a new national effort. JAMA. 2015;313(21):2119–20. https://doi.org/10.1001/jama.2015.3595.
Lipkova J, Chen RJ, Chen B, Lu MY, Barbieri M, Shao D, et al. Artificial intelligence for multimodal data integration in oncology. Cancer Cell. 2022;40(10):1095–110. https://doi.org/10.1016/j.ccell.2022.09.012.
Kehl KL, Jee J, Pichotta K, Paul MA, Trukhanov P, Fong C, et al. Shareable artificial intelligence to extract cancer outcomes from electronic health records for precision oncology research. Nat Commun. 2024;15(1):9787. https://doi.org/10.1038/s41467-024-54071-x.
Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM, et al. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017;542(7639):115–8. https://doi.org/10.1038/nature21056.
Hashimoto DA, Rosman G, Rus D, Meireles OR. Artificial Intelligence in surgery: promises and perils. Ann Surg. 2018;268(1):70–6. https://doi.org/10.1097/sla.0000000000002693.
Varghese C, Harrison EM, O’Grady G, Topol EJ. Artificial intelligence in surgery. Nat Med. 2024;30(5):1257–68. https://doi.org/10.1038/s41591-024-02970-3.
Farrokhian N, Holcomb AJ, Dimon E, Karadaghy O, Ward C, Whiteford E, et al. Development and Validation of Machine Learning Models for Predicting Occult Nodal Metastasis in early-stage oral cavity squamous cell carcinoma. JAMA Netw open. 2022;5(4):e227226. https://doi.org/10.1001/jamanetworkopen.2022.7226.
Song JH, Hong Y, Kim ER, Kim SH, Sohn I. Utility of artificial intelligence with deep learning of hematoxylin and eosin-stained whole slide images to predict lymph node metastasis in T1 colorectal cancer using endoscopically resected specimens; prediction of lymph node metastasis in T1 colorectal cancer. J Gastroenterol. 2022;57(9):654–66. https://doi.org/10.1007/s00535-022-01894-4.
Ichimasa K, Nakahara K, Kudo SE, Misawa M, Bretthauer M, Shimada S, et al. Novel resect and analysis approach for T2 colorectal cancer with use of artificial intelligence. Gastrointest Endosc. 2022. https://doi.org/10.1016/j.gie.2022.04.1305.
Guan X, Yu G, Zhang W, Wen R, Wei R, Jiao S, et al. An easy-to-use artificial intelligence preoperative lymph node metastasis predictor (LN-MASTER) in rectal cancer based on a privacy-preserving computing platform: multicenter retrospective cohort study. Int J Surg (London England). 2023;109(3):255–65. https://doi.org/10.1097/js9.0000000000000067.
Brockmoeller S, Echle A, Ghaffari Laleh N, Eiholm S, Malmstrøm ML, Plato Kuhlmann T, et al. Deep learning identifies inflamed fat as a risk factor for lymph node metastasis in early colorectal cancer. J Pathol. 2022;256(3):269–81. https://doi.org/10.1002/path.5831.
Zhang Y, Yuan N, Zhang Z, Du J, Wang T, Liu B, et al. Unsupervised domain selective graph convolutional network for preoperative prediction of lymph node metastasis in gastric cancer. Med Image Anal. 2022;79:102467doi. https://doi.org/10.1016/j.media.2022.102467.
An C, Li D, Li S, Li W, Tong T, Liu L, et al. Deep learning radiomics of dual-energy computed tomography for predicting lymph node metastases of pancreatic ductal adenocarcinoma. Eur J Nucl Med Mol Imaging. 2022;49(4):1187–99. https://doi.org/10.1007/s00259-021-05573-z.
Bian Y, Zheng Z, Fang X, Jiang H, Zhu M, Yu J, et al. Artificial Intelligence To Predict Lymph Node Metastasis at CT in Pancreatic Ductal Adenocarcinoma. Radiology. 2023;306(1):160–9. https://doi.org/10.1148/radiol.220329.
Liu Y, Lai F, Lin B, Gu Y, Chen L, Chen G, et al. Deep learning to predict cervical lymph node metastasis from intraoperative frozen section of tumour in papillary thyroid carcinoma: a multicentre diagnostic study. EClinicalMedicine. 2023;60:102007doi. https://doi.org/10.1016/j.eclinm.2023.102007.
Wang YW, Chen CJ, Huang HC, Wang TC, Chen HM, Shih JY, et al. Dual energy CT image prediction on primary tumor of lung cancer for nodal metastasis using deep learning. Comput Med Imaging Graphics: Official J Comput Med Imaging Soc. 2021;91:101935. https://doi.org/10.1016/j.compmedimag.2021.101935.
You C, Shen Y, Sun S, Zhou J, Li J, Su G, et al. Artificial intelligence in breast imaging: current situation and clinical challenges. Explor (Beijing China). 2023;3(5):20230007. https://doi.org/10.1002/exp.20230007.
Yang LY, Siow TY, Lin YC, Wu RC, Lu HY, Chiang HJ, et al. Computer-aided segmentation and Machine Learning of Integrated Clinical and Diffusion-Weighted Imaging parameters for Predicting Lymph Node Metastasis in Endometrial Cancer. Cancers. 2021;13(6). https://doi.org/10.3390/cancers13061406.
Wu S, Hong G, Xu A, Zeng H, Chen X, Wang Y, et al. Artificial intelligence-based model for lymph node metastases detection on whole slide images in bladder cancer: a retrospective, multicentre, diagnostic study. Lancet Oncol. 2023;24(4):360–70. https://doi.org/10.1016/s1470-2045(23)00061-x.
Li H, Han Z, Wu H, Musaev ER, Lin Y, Li S, et al. Artificial intelligence in surgery: evolution, trends, and future directions. Int J Surg (London England). 2024. https://doi.org/10.1097/js9.0000000000002159.
Sendín-Martín M, Lara-Caro M, Harris U, Moronta M, Rossi A, Lee E, et al. Classification of basal cell carcinoma in Ex Vivo Confocal Microscopy Images from freshly excised tissues using a deep learning algorithm. J Invest Dermatol. 2022;142(5):1291–9. https://doi.org/10.1016/j.jid.2021.09.029.
Pati S, Baid U, Edwards B, Sheller M, Wang SH, Reina GA, et al. Federated learning enables big data for rare cancer boundary detection. Nat Commun. 2022;13(1):7346. https://doi.org/10.1038/s41467-022-33407-5.
Khan DZ, Valetopoulou A, Das A, Hanrahan JG, Williams SC, Bano S, et al. Artificial intelligence assisted operative anatomy recognition in endoscopic pituitary surgery. NPJ Digit Med. 2024;7(1):314. https://doi.org/10.1038/s41746-024-01273-8.
Nandakumar N, Manzoor K, Agarwal S, Pillai JJ, Gujar SK, Sair HI, et al. Automated eloquent cortex localization in brain tumor patients using multi-task graph neural networks. Med Image Anal. 2021;74:102203. https://doi.org/10.1016/j.media.2021.102203.
Shen B, Zhang Z, Shi X, Cao C, Zhang Z, Hu Z, et al. Real-time intraoperative glioma diagnosis using fluorescence imaging and deep convolutional neural networks. Eur J Nucl Med Mol Imaging. 2021;48(11):3482–92. https://doi.org/10.1007/s00259-021-05326-y.
Mascagni P, Alapatt D, Lapergola A, Vardazaryan A, Mazellier JP, Dallemagne B, et al. Early-stage clinical evaluation of real-time artificial intelligence assistance for laparoscopic cholecystectomy. Br J Surg. 2024;111(1). https://doi.org/10.1093/bjs/znad353.
Vermeulen C, Pagès-Gallego M, Kester L, Kranendonk MEG, Wesseling P, Verburg N, et al. Ultra-fast deep-learned CNS tumour classification during surgery. Nature. 2023;622(7984):842–9. https://doi.org/10.1038/s41586-023-06615-2.
Yip M, Salcudean S, Goldberg K, Althoefer K, Menciassi A, Opfermann JD, et al. Artificial intelligence meets medical robotics. Sci (New York NY). 2023;381(6654):141–6. https://doi.org/10.1126/science.adj3312.
Luongo F, Hakim R, Nguyen JH, Anandkumar A, Hung AJ. Deep learning-based computer vision to recognize and classify suturing gestures in robot-assisted surgery. Surgery. 2021;169(5):1240–4. https://doi.org/10.1016/j.surg.2020.08.016.
Lavanchy JL, Zindel J, Kirtac K, Twick I, Hosgor E, Candinas D, et al. Automation of surgical skill assessment using a three-stage machine learning algorithm. Sci Rep. 2021;11(1):5197. https://doi.org/10.1038/s41598-021-84295-6.
Takeuchi M, Kawakubo H, Saito K, Maeda Y, Matsuda S, Fukuda K, et al. Automated Surgical-Phase Recognition for Robot-assisted minimally invasive esophagectomy using Artificial Intelligence. Ann Surg Oncol. 2022;29(11):6847–55. https://doi.org/10.1245/s10434-022-11996-1.
Marcus HJ, Ramirez PT, Khan DZ, Layard Horsfall H, Hanrahan JG, Williams SC, et al. The IDEAL framework for surgical robotics: development, comparative evaluation and long-term monitoring. Nat Med. 2024;30(1):61–75. https://doi.org/10.1038/s41591-023-02732-7.
Hatib F, Jian Z, Buddi S, Lee C, Settels J, Sibert K, et al. Machine-learning Algorithm to Predict Hypotension based on high-fidelity arterial pressure Waveform Analysis. Anesthesiology. 2018;129(4):663–74. https://doi.org/10.1097/aln.0000000000002300.
Wiens J, Guttag J, Horvitz EJJMLR. Patient risk stratification with time-varying parameters: a multitask learning approach. 2016;17(79):1–23.
Seo S, Kennedy-Metz LR, Zenati MA, Shah JA, Dias RD, Unhelkar VV. Towards an AI Coach to Infer Team Mental Model Alignment in Healthcare. IEEE Conference on Cognitive and Computational Aspects of Situation Management (CogSIMA). 2021;2021:39–44.https://doi.org/10.1109/cogsima51574.2021.9475925
Agha RA, Fowler AJ, Sevdalis N. The role of non-technical skills in surgery. Annals of medicine and surgery (2012). 2015;4(4):422-7.https://doi.org/10.1016/j.amsu.2015.10.006
Gillespie BM, Harbeck E, Kang E, Steel C, Fairweather N, Chaboyer W. Correlates of non-technical skills in surgery: a prospective study. BMJ open. 2017;7(1):e014480. https://doi.org/10.1136/bmjopen-2016-014480.
Zhao LY, Zhang WH, Sun Y, Chen XZ, Yang K, Liu K, et al. Learning curve for gastric cancer patients with laparoscopy-assisted distal gastrectomy: 6-year experience from a single institution in western China. Medicine. 2016;95(37):e4875. https://doi.org/10.1097/md.0000000000004875.
Goodman ED, Patel KK, Zhang Y, Locke W, Kennedy CJ, Mehrotra R, et al. Analyzing Surgical technique in Diverse Open Surgical videos with Multitask Machine Learning. JAMA Surg. 2024;159(2):185–92. https://doi.org/10.1001/jamasurg.2023.6262.
Kocielnik R, Yang CH, Ma R, Cen SY, Wong EY, Chu TN, et al. Human AI collaboration for unsupervised categorization of live surgical feedback. NPJ Digit Med. 2024;7(1):372. https://doi.org/10.1038/s41746-024-01383-3.
Deng H, Eftekhari Z, Carlin C, Veerapong J, Fournier KF, Johnston FM, et al. Development and validation of an explainable machine learning model for major complications after cytoreductive surgery. JAMA Netw open. 2022;5(5):e2212930. https://doi.org/10.1001/jamanetworkopen.2022.12930.
Hassan AM, Biaggi AP, Asaad M, Andejani DF, Liu J, Offodile AC 2, et al. Development and Assessment of Machine Learning Models for Individualized Risk Assessment of Mastectomy skin flap necrosis. Ann Surg. 2022. https://doi.org/10.1097/sla.0000000000005386.
Kim DY, Lee SJ, Kim EK, Kang E, Heo CY, Jeong JH, et al. Feasibility of anomaly score detected with deep learning in irradiated breast cancer patients with reconstruction. NPJ Digit Med. 2022;5(1):125. https://doi.org/10.1038/s41746-022-00671-0.
Mak RH, Endres MG, Paik JH, Sergeev RA, Aerts H, Williams CL, et al. Use of Crowd Innovation to develop an Artificial Intelligence-based solution for Radiation Therapy Targeting. JAMA Oncol. 2019;5(5):654–61. https://doi.org/10.1001/jamaoncol.2019.0159.
Xue S, Gafita A, Dong C, Zhao Y, Tetteh G, Menze BH, et al. Application of machine learning to pretherapeutically estimate dosimetry in men with advanced prostate cancer treated with (177)Lu-PSMA I&T therapy. Eur J Nucl Med Mol Imaging. 2022. https://doi.org/10.1007/s00259-022-05883-w.
Yang YX, Yang X, Jiang XB, Lin L, Wang GY, Sun WZ, et al. Artificial Intelligence-Empowered Multistep Integrated Radiation Therapy Workflow for Nasopharyngeal Carcinoma. Int J Radiat Oncol Biol Phys. 2024. https://doi.org/10.1016/j.ijrobp.2024.11.096.
Ding J, Zhang Y, Amjad A, Xu J, Thill D, Li XA. Automatic contour refinement for Deep Learning Auto-segmentation of Complex organs in MRI-guided adaptive Radiation Therapy. Adv Radiation Oncol. 2022;7(5):100968. https://doi.org/10.1016/j.adro.2022.100968.
Isensee F, Jaeger PF, Kohl SAA, Petersen J, Maier-Hein KH. nnU-Net: a self-configuring method for deep learning-based biomedical image segmentation. Nat Methods. 2021;18(2):203–11. https://doi.org/10.1038/s41592-020-01008-z.
Shi F, Hu W, Wu J, Han M, Wang J, Zhang W, et al. Deep learning empowered volume delineation of whole-body organs-at-risk for accelerated radiotherapy. Nat Commun. 2022;13(1):6566. https://doi.org/10.1038/s41467-022-34257-x.
Wong J, Fong A, McVicar N, Smith S, Giambattista J, Wells D, et al. Comparing deep learning-based auto-segmentation of organs at risk and clinical target volumes to expert inter-observer variability in radiotherapy planning. Radiotherapy Oncology: J Eur Soc Therapeutic Radiol Oncol. 2020;144:152–8. https://doi.org/10.1016/j.radonc.2019.10.019.
Moore KL, Schmidt R, Moiseenko V, Olsen LA, Tan J, Xiao Y, et al. Quantifying unnecessary normal tissue complication risks due to Suboptimal Planning: a secondary study of RTOG 0126. Int J Radiat Oncol Biol Phys. 2015;92(2):228–35. https://doi.org/10.1016/j.ijrobp.2015.01.046.
Zhang J, Liu S, Yan H, Li T, Mao R, Liu J. Predicting Voxel-level dose distributions for esophageal radiotherapy using densely connected network with dilated convolutions. Phys Med Biol. 2020;65(20):205013. https://doi.org/10.1088/1361-6560/aba87b.
Chen X, Men K, Zhu J, Yang B, Li M, Liu Z, et al. DVHnet: a deep learning-based prediction of patient-specific dose volume histograms for radiotherapy planning. Med Phys. 2021;48(6):2705–13. https://doi.org/10.1002/mp.14758.
Mentzel F, Kröninger K, Lerch M, Nackenhorst O, Paino J, Rosenfeld A, et al. Fast and accurate dose predictions for novel radiotherapy treatments in heterogeneous phantoms using conditional 3D-UNet generative adversarial networks. Med Phys. 2022;49(5):3389–404. https://doi.org/10.1002/mp.15555.
Jin C, Yu H, Ke J, Ding P, Yi Y, Jiang X, et al. Predicting treatment response from longitudinal images using multi-task deep learning. Nat Commun. 2021;12(1):1851. https://doi.org/10.1038/s41467-021-22188-y.
Sundar R, Barr Kumarakulasinghe N, Huak Chan Y, Yoshida K, Yoshikawa T, Miyagi Y, et al. Machine-learning model derived gene signature predictive of paclitaxel survival benefit in gastric cancer: results from the randomised phase III SAMIT trial. Gut. 2022;71(4):676–85. https://doi.org/10.1136/gutjnl-2021-324060.
Kleppe A, Skrede OJ, De Raedt S, Hveem TS, Askautrud HA, Jacobsen JE, et al. A clinical decision support system optimising adjuvant chemotherapy for colorectal cancers by integrating deep learning and pathological staging markers: a development and validation study. Lancet Oncol. 2022. https://doi.org/10.1016/s1470-2045(22)00391-6.
Mi H, Bivalacqua TJ, Kates M, Seiler R, Black PC, Popel AS, et al. Predictive models of response to neoadjuvant chemotherapy in muscle-invasive bladder cancer using nuclear morphology and tissue architecture. Cell Rep Med. 2021;2(9):100382doi. https://doi.org/10.1016/j.xcrm.2021.100382.
Crispin-Ortuzar M, Woitek R, Reinius MAV, Moore E, Beer L, Bura V, et al. Integrated radiogenomics models predict response to neoadjuvant chemotherapy in high grade serous ovarian cancer. Nat Commun. 2023;14(1):6756. https://doi.org/10.1038/s41467-023-41820-7.
Zhang J, Cui Y, Wei K, Li Z, Li D, Song R, et al. Deep learning predicts resistance to neoadjuvant chemotherapy for locally advanced gastric cancer: a multicenter study. Gastric cancer: Official J Int Gastric Cancer Association Japanese Gastric Cancer Association. 2022;25(6):1050–9. https://doi.org/10.1007/s10120-022-01328-3.
Fu L, Jin W, Zhang J, Zhu L, Lu J, Zhen Y, et al. Repurposing non-oncology small-molecule drugs to improve cancer therapy: current situation and future directions. Acta Pharm Sinica B. 2022;12(2):532–57. https://doi.org/10.1016/j.apsb.2021.09.006.
You Y, Lai X, Pan Y, Zheng H, Vera J, Liu S, et al. Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Therapy. 2022;7(1):156. https://doi.org/10.1038/s41392-022-00994-0.
Cai Z, Apolinário S, Baião AR, Pacini C, Sousa MD, Vinga S, et al. Synthetic augmentation of cancer cell line multi-omic datasets using unsupervised deep learning. Nat Commun. 2024;15(1):10390. https://doi.org/10.1038/s41467-024-54771-4.
Zare A, Postovit LM, Githaka JM. Robust inflammatory breast cancer gene signature using nonparametric random forest analysis. Breast cancer Research: BCR. 2021;23(1):92. https://doi.org/10.1186/s13058-021-01467-y.
Pham TH, Hagenbeek TJ, Lee HJ, Li J, Rose CM, Lin E, et al. Machine-learning and Chemicogenomics Approach Defines and predicts cross-talk of Hippo and MAPK pathways. Cancer Discov. 2021;11(3):778–93. https://doi.org/10.1158/2159-8290.Cd-20-0706.
Xiao Y, Ma D, Yang YS, Yang F, Ding JH, Gong Y, et al. Comprehensive metabolomics expands precision medicine for triple-negative breast cancer. Cell Res. 2022;32(5):477–90. https://doi.org/10.1038/s41422-022-00614-0.
Deng LJ, Deng WQ, Fan SR, Chen MF, Qi M, Lyu WY, et al. m6A modification: recent advances, anticancer targeted drug discovery and beyond. Mol Cancer. 2022;21(1):52. https://doi.org/10.1186/s12943-022-01510-2.
Zhang H, Li J, Saravanan KM, Wu H, Wang Z, Wu D, et al. An Integrated Deep Learning and Molecular Dynamics Simulation-based Screening Pipeline identifies inhibitors of a New Cancer Drug Target TIPE2. Front Pharmacol. 2021;12:772296. https://doi.org/10.3389/fphar.2021.772296.
Valentini E, D’Aguanno S, Di Martile M, Montesano C, Ferraresi V, Patsilinakos A, et al. Targeting the anti-apoptotic Bcl-2 family proteins: machine learning virtual screening and biological evaluation of new small molecules. Theranostics. 2022;12(5):2427–44. https://doi.org/10.7150/thno.64233.
Saoud M, Grau J, Rennert R, Mueller T, Yousefi M, Davari MD, et al. Advancing Anticancer Drug Discovery: leveraging Metabolomics and Machine Learning for Mode of Action Prediction by Pattern Recognition. Adv Sci (Weinheim Baden-Wurttemberg Germany). 2024;11(47):e2404085. https://doi.org/10.1002/advs.202404085.
Su R, Huang Y, Zhang DG, Xiao G, Wei L. SRDFM: siamese response deep factorization machine to improve anti-cancer drug recommendation. Brief Bioinform. 2022;23(2). https://doi.org/10.1093/bib/bbab534.
Chen J, Wang X, Ma A, Wang QE, Liu B, Li L, et al. Deep transfer learning of cancer drug responses by integrating bulk and single-cell RNA-seq data. Nat Commun. 2022;13(1):6494. https://doi.org/10.1038/s41467-022-34277-7.
Liu X, Song C, Huang F, Fu H, Xiao W, Zhang W. GraphCDR: a graph neural network method with contrastive learning for cancer drug response prediction. Brief Bioinform. 2022;23(1). https://doi.org/10.1093/bib/bbab457.
Ianevski A, Nader K, Driva K, Senkowski W, Bulanova D, Moyano-Galceran L, et al. Single-cell transcriptomes identify patient-tailored therapies for selective co-inhibition of cancer clones. Nat Commun. 2024;15(1):8579. https://doi.org/10.1038/s41467-024-52980-5.
Li X, Dowling EK, Yan G, Dereli Z, Bozorgui B, Imanirad P, et al. Precision Combination therapies based on recurrent oncogenic coalterations. Cancer Discov. 2022;12(6):1542–59. https://doi.org/10.1158/2159-8290.Cd-21-0832.
Cui C, Ding X, Wang D, Chen L, Xiao F, Xu T, et al. Drug repurposing against breast cancer by integrating drug-exposure expression profiles and drug-drug links based on graph neural network. Bioinf (Oxford England). 2021;37(18):2930–7. https://doi.org/10.1093/bioinformatics/btab191.
Jiang L, Jiang C, Yu X, Fu R, Jin S, Liu X. DeepTTA: a transformer-based model for predicting cancer drug response. Brief Bioinform. 2022;23(3). https://doi.org/10.1093/bib/bbac100.
Sun L, Liu H, Ye Y, Lei Y, Islam R, Tan S, et al. Smart nanoparticles for cancer therapy. Signal Transduct Target Therapy. 2023;8(1):418. https://doi.org/10.1038/s41392-023-01642-x.
Xu Z, Wang X, Zeng S, Ren X, Yan Y, Gong Z. Applying artificial intelligence for cancer immunotherapy. Acta Pharm Sinica B. 2021;11(11):3393–405. https://doi.org/10.1016/j.apsb.2021.02.007.
Addala V, Newell F, Pearson JV, Redwood A, Robinson BW, Creaney J, et al. Computational immunogenomic approaches to predict response to cancer immunotherapies. Nat Reviews Clin Oncol. 2024;21(1):28–46. https://doi.org/10.1038/s41571-023-00830-6.
Prelaj A, Miskovic V, Zanitti M, Trovo F, Genova C, Viscardi G, et al. Artificial intelligence for predictive biomarker discovery in immuno-oncology: a systematic review. Annals Oncology: Official J Eur Soc Med Oncol. 2024;35(1):29–65. https://doi.org/10.1016/j.annonc.2023.10.125.
Hondelink LM, Hüyük M, Postmus PE, Smit V, Blom S, von der Thüsen JH, et al. Development and validation of a supervised deep learning algorithm for automated whole-slide programmed death-ligand 1 tumour proportion score assessment in non-small cell lung cancer. Histopathology. 2022;80(4):635–47. https://doi.org/10.1111/his.14571.
Shamai G, Livne A, Polónia A, Sabo E, Cretu A, Bar-Sela G, et al. Deep learning-based image analysis predicts PD-L1 status from H&E-stained histopathology images in breast cancer. Nat Commun. 2022;13(1):6753. https://doi.org/10.1038/s41467-022-34275-9.
Mu W, Jiang L, Shi Y, Tunali I, Gray JE, Katsoulakis E, et al. Non-invasive measurement of PD-L1 status and prediction of immunotherapy response using deep learning of PET/CT images. J Immunother Cancer. 2021;9(6). https://doi.org/10.1136/jitc-2020-002118.
Saad MB, Hong L, Aminu M, Vokes NI, Chen P, Salehjahromi M, et al. Predicting benefit from immune checkpoint inhibitors in patients with non-small-cell lung cancer by CT-based ensemble deep learning: a retrospective study. Lancet Digit Health. 2023;5(7):e404–20. https://doi.org/10.1016/s2589-7500(23)00082-1.
Choi S, Cho SI, Ma M, Park S, Pereira S, Aum BJ et al. Artificial intelligence-powered programmed death ligand 1 analyser reduces interobserver variation in tumour proportion score for non-small cell lung cancer with better prediction of immunotherapy response. European journal of cancer (Oxford, England: 1990). 2022;170:17–26.https://doi.org/10.1016/j.ejca.2022.04.011
Vanguri RS, Luo J, Aukerman AT, Egger JV, Fong CJ, Horvat N, et al. Multimodal integration of radiology, pathology and genomics for prediction of response to PD-(L)1 blockade in patients with non-small cell lung cancer. Nat cancer. 2022;3(10):1151–64. https://doi.org/10.1038/s43018-022-00416-8.
Cheong JH, Wang SC, Park S, Porembka MR, Christie AL, Kim H, et al. Development and validation of a prognostic and predictive 32-gene signature for gastric cancer. Nat Commun. 2022;13(1):774. https://doi.org/10.1038/s41467-022-28437-y.
Gong X, Karchin R, Pan-Cancer HLA. Gene-mediated Tumor Immunogenicity and Immune Evasion. Mol cancer Research: MCR. 2022;20(8):1272–83. https://doi.org/10.1158/1541-7786.Mcr-21-0886.
Katzir R, Rudberg N, Yizhak K. Estimating tumor mutational burden from RNA-sequencing without a matched-normal sample. Nat Commun. 2022;13(1):3092. https://doi.org/10.1038/s41467-022-30753-2.
Hoshino I, Yokota H, Iwatate Y, Mori Y, Kuwayama N, Ishige F, et al. Prediction of the differences in tumor mutation burden between primary and metastatic lesions by radiogenomics. Cancer Sci. 2022;113(1):229–39. https://doi.org/10.1111/cas.15173.
Shimada Y, Okuda S, Watanabe Y, Tajima Y, Nagahashi M, Ichikawa H, et al. Histopathological characteristics and artificial intelligence for predicting tumor mutational burden-high colorectal cancer. J Gastroenterol. 2021;56(6):547–59. https://doi.org/10.1007/s00535-021-01789-w.
Wang CW, Firdi NP, Lee YC, Chu TC, Muzakky H, Liu TC, et al. Deep learning for endometrial cancer subtyping and predicting tumor mutational burden from histopathological slides. NPJ Precision Oncol. 2024;8(1):287. https://doi.org/10.1038/s41698-024-00766-9.
Wang CW, Liu TC, Lai PJ, Muzakky H, Wang YC, Yu MH, et al. Ensemble transformer-based multiple instance learning to predict pathological subtypes and tumor mutational burden from histopathological whole slide images of endometrial and colorectal cancer. Med Image Anal. 2025;99:103372doi. https://doi.org/10.1016/j.media.2024.103372.
Wang L, Zhang H, Pan C, Yi J, Cui X, Li N, et al. Predicting durable responses to Immune checkpoint inhibitors in Non-small-cell Lung Cancer using a multi-feature model. Front Immunol. 2022;13:829634. https://doi.org/10.3389/fimmu.2022.829634.
Filipski K, Scherer M, Zeiner KN, Bucher A, Kleemann J, Jurmeister P, et al. DNA methylation-based prediction of response to immune checkpoint inhibition in metastatic melanoma. J Immunother Cancer. 2021;9(7). https://doi.org/10.1136/jitc-2020-002226.
Pan X, Zhang C, Wang J, Wang P, Gao Y, Shang S, et al. Epigenome signature as an immunophenotype indicator prompts durable clinical immunotherapy benefits in lung adenocarcinoma. Brief Bioinform. 2022;23(1). https://doi.org/10.1093/bib/bbab481.
Dercle L, Zhao B, Gönen M, Moskowitz CS, Firas A, Beylergil V, et al. Early readout on overall survival of patients with melanoma treated with immunotherapy using a novel imaging analysis. JAMA Oncol. 2022;8(3):385–92. https://doi.org/10.1001/jamaoncol.2021.6818.
Steyaert S, Pizurica M, Nagaraj D, Khandelwal P, Hernandez-Boussard T, Gentles AJ, et al. Multimodal data fusion for cancer biomarker discovery with deep learning. Nat Mach Intell. 2023;5(4):351–62. https://doi.org/10.1038/s42256-023-00633-5.
Boehm KM, Khosravi P, Vanguri R, Gao J, Shah SP. Harnessing multimodal data integration to advance precision oncology. Nat Rev Cancer. 2022;22(2):114–26. https://doi.org/10.1038/s41568-021-00408-3.
Shen R, Postow MA, Adamow M, Arora A, Hannum M, Maher C, et al. LAG-3 expression on peripheral blood cells identifies patients with poorer outcomes after immune checkpoint blockade. Sci Transl Med. 2021;13(608). https://doi.org/10.1126/scitranslmed.abf5107.
Ren S, Lu Y, Zhang G, Xie K, Chen D, Cai X, et al. Integration of graph neural networks and multi-omics analysis identify the predictive factor and key gene for immunotherapy response and prognosis of bladder cancer. J Translational Med. 2024;22(1):1141. https://doi.org/10.1186/s12967-024-05976-0.
Paidi SK, Rodriguez Troncoso J, Raj P, Monterroso Diaz P, Ivers JD, Lee DE, et al. Raman Spectroscopy and Machine Learning reveals early Tumor Microenvironmental Changes Induced by Immunotherapy. Cancer Res. 2021;81(22):5745–55. https://doi.org/10.1158/0008-5472.Can-21-1438.
Xu Y, Su GH, Ma D, Xiao Y, Shao ZM, Jiang YZ. Technological advances in cancer immunity: from immunogenomics to single-cell analysis and artificial intelligence. Signal Transduct Target Therapy. 2021;6(1):312. https://doi.org/10.1038/s41392-021-00729-7.
Tang T, Huang X, Zhang G, Hong Z, Bai X, Liang T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct Target Therapy. 2021;6(1):72. https://doi.org/10.1038/s41392-020-00449-4.
Park S, Ock CY, Kim H, Pereira S, Park S, Ma M, et al. Artificial Intelligence-powered spatial analysis of Tumor-infiltrating lymphocytes as complementary biomarker for Immune Checkpoint Inhibition in Non-small-cell Lung Cancer. J Clin Oncology: Official J Am Soc Clin Oncol. 2022;40(17):1916–28. https://doi.org/10.1200/jco.21.02010.
Ao Z, Cai H, Wu Z, Hu L, Nunez A, Zhou Z, et al. Microfluidics guided by deep learning for cancer immunotherapy screening. Proc Natl Acad Sci USA. 2022;119(46):e2214569119. https://doi.org/10.1073/pnas.2214569119.
Zhang Z, Chen L, Chen H, Zhao J, Li K, Sun J, et al. Pan-cancer landscape of T-cell exhaustion heterogeneity within the tumor microenvironment revealed a progressive roadmap of hierarchical dysfunction associated with prognosis and therapeutic efficacy. EBioMedicine. 2022;83:104207doi. https://doi.org/10.1016/j.ebiom.2022.104207.
Mo CK, Liu J, Chen S, Storrs E, Targino da Costa ALN, Houston A, et al. Tumour evolution and microenvironment interactions in 2D and 3D space. Nature. 2024;634(8036):1178–86. https://doi.org/10.1038/s41586-024-08087-4.
Parkhurst MR, Robbins PF, Tran E, Prickett TD, Gartner JJ, Jia L, et al. Unique neoantigens arise from somatic mutations in patients with gastrointestinal cancers. Cancer Discov. 2019;9(8):1022–35. https://doi.org/10.1158/2159-8290.Cd-18-1494.
Madadi Y, Monavarfeshani A, Chen H, Stamer WD, Williams RW, Yousefi S. Artificial Intelligence models for cell type and subtype identification based on single-cell RNA sequencing data in Vision Science. IEEE/ACM Trans Comput Biol Bioinf. 2023;20(5):2837–52. https://doi.org/10.1109/tcbb.2023.3284795.
Li Y, Wu X, Fang D, Luo Y. Informing immunotherapy with multi-omics driven machine learning. NPJ Digit Med. 2024;7(1):67. https://doi.org/10.1038/s41746-024-01043-6.
Pounraj S, Chen S, Ma L, Mazzieri R, Dolcetti R, Rehm BHA. Targeting Tumor heterogeneity with Neoantigen-Based Cancer vaccines. Cancer Res. 2024;84(3):353–63. https://doi.org/10.1158/0008-5472.Can-23-2042.
Weber D, Ibn-Salem J, Sorn P, Suchan M, Holtsträter C, Lahrmann U, et al. Accurate detection of tumor-specific gene fusions reveals strongly immunogenic personal neo-antigens. Nat Biotechnol. 2022;40(8):1276–84. https://doi.org/10.1038/s41587-022-01247-9.
Huber F, Arnaud M, Stevenson BJ, Michaux J, Benedetti F, Thevenet J, et al. A comprehensive proteogenomic pipeline for neoantigen discovery to advance personalized cancer immunotherapy. Nat Biotechnol. 2024. https://doi.org/10.1038/s41587-024-02420-y.
Kumar A, Dixit S, Srinivasan K, Vincent MD. Personalized cancer vaccine design using AI-powered technologies. Front Immunol. 2024;15:1357217. https://doi.org/10.3389/fimmu.2024.1357217.
Wang F, Wang H, Wang L, Lu H, Qiu S, Zang T, et al. MHCRoBERTa: pan-specific peptide-MHC class I binding prediction through transfer learning with label-agnostic protein sequences. Brief Bioinform. 2022;23(3). https://doi.org/10.1093/bib/bbab595.
Mei S, Li F, Xiang D, Ayala R, Faridi P, Webb GI, et al. Anthem: a user customised tool for fast and accurate prediction of binding between peptides and HLA class I molecules. Brief Bioinform. 2021;22(5). https://doi.org/10.1093/bib/bbaa415.
Xu H, Hu R, Dong X, Kuang L, Zhang W, Tu C, et al. ImmuneApp for HLA-I epitope prediction and immunopeptidome analysis. Nat Commun. 2024;15(1):8926. https://doi.org/10.1038/s41467-024-53296-0.
Racle J, Guillaume P, Schmidt J, Michaux J, Larabi A, Lau K, et al. Machine learning predictions of MHC-II specificities reveal alternative binding mode of class II epitopes. Immunity. 2023;56(6):1359–75. https://doi.org/10.1016/j.immuni.2023.03.009.
Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2018;18(3):168–82. https://doi.org/10.1038/nri.2017.131.
Powderly JD, Sullivan RJ, Gutierrez M, Khattak A, Thomas SS, Jimeno A et al. Phase 1/2 study of mRNA-4359 administered alone and in combination with immune checkpoint blockade in adult participants with advanced solid tumors. 2023;41(16_suppl):TPS2676–TPS https://doi.org/10.1200/JCO.2023.41.16_suppl.TPS2676
Xu Y. Deep neural networks for QSAR. Methods in molecular biology. (Clifton NJ). 2022;2390:233–60. https://doi.org/10.1007/978-1-0716-1787-8_10.
Lin YJ, Zimmermann J, Schülke S. Novel adjuvants in allergen-specific immunotherapy: where do we stand? Front Immunol. 2024;15:1348305. https://doi.org/10.3389/fimmu.2024.1348305.
Zhang WY, Zheng XL, Coghi PS, Chen JH, Dong BJ, Fan XX. Revolutionizing adjuvant development: harnessing AI for next-generation cancer vaccines. Front Immunol. 2024;15:1438030. https://doi.org/10.3389/fimmu.2024.1438030.
Haider S, Barakat A, Ul-Haq Z. Discovery of potential chemical probe as inhibitors of CXCL12 using ligand-based virtual screening and Molecular Dynamic Simulation. Molecules. 2020;25(20). https://doi.org/10.3390/molecules25204829.
Yao RW, Wang Y, Chen LL. Cellular functions of long noncoding RNAs. Nat Cell Biol. 2019;21(5):542–51. https://doi.org/10.1038/s41556-019-0311-8.
Shaath H, Vishnubalaji R, Elango R, Kardousha A, Islam Z, Qureshi R, et al. Long non-coding RNA and RNA-binding protein interactions in cancer: experimental and machine learning approaches. Sem Cancer Biol. 2022. https://doi.org/10.1016/j.semcancer.2022.05.013.
Zhou M, Zhang Z, Bao S, Hou P, Yan C, Su J, et al. Computational recognition of lncRNA signature of tumor-infiltrating B lymphocytes with potential implications in prognosis and immunotherapy of bladder cancer. Brief Bioinform. 2021;22(3). https://doi.org/10.1093/bib/bbaa047.
Liu Z, Liu L, Weng S, Guo C, Dang Q, Xu H, et al. Machine learning-based integration develops an immune-derived lncRNA signature for improving outcomes in colorectal cancer. Nat Commun. 2022;13(1):816. https://doi.org/10.1038/s41467-022-28421-6.
Liu Z, Guo C, Dang Q, Wang L, Liu L, Weng S, et al. Integrative analysis from multi-center studies identities a consensus machine learning-derived lncRNA signature for stage II/III colorectal cancer. EBioMedicine. 2022;75:103750doi. https://doi.org/10.1016/j.ebiom.2021.103750.
Vaidya A, Chen RJ, Williamson DFK, Song AH, Jaume G, Yang Y, et al. Demographic bias in misdiagnosis by computational pathology models. Nat Med. 2024;30(4):1174–90. https://doi.org/10.1038/s41591-024-02885-z.
Alderman JE, Palmer J, Laws E, McCradden MD, Ordish J, Ghassemi M, et al. Tackling algorithmic bias and promoting transparency in health datasets: the STANDING together consensus recommendations. Lancet Digit Health. 2025;7(1):e64–88. https://doi.org/10.1016/s2589-7500(24)00224-3.
Landi I, Glicksberg BS, Lee HC, Cherng S, Landi G, Danieletto M, et al. Deep representation learning of electronic health records to unlock patient stratification at scale. NPJ Digit Med. 2020;3:96. https://doi.org/10.1038/s41746-020-0301-z.
Zou J, Schiebinger L. AI can be sexist and racist - it’s time to make it fair. Nature. 2018;559(7714):324–6. https://doi.org/10.1038/d41586-018-05707-8.
Nensa F, Demircioglu A, Rischpler C. Artificial Intelligence in Nuclear Medicine. Journal of nuclear medicine: official publication. Soc Nuclear Med. 2019;60(Suppl 2):s29–37. https://doi.org/10.2967/jnumed.118.220590.
Banegas-Luna AJ, Peña-García J, Iftene A, Guadagni F, Ferroni P, Scarpato N, et al. Towards the Interpretability of Machine Learning Predictions for Medical Applications Targeting Personalised therapies: a Cancer Case Survey. Int J Mol Sci. 2021;22(9). https://doi.org/10.3390/ijms22094394.
Huang Z, Yang E, Shen J, Gratzinger D, Eyerer F, Liang B, et al. A pathologist-AI collaboration framework for enhancing diagnostic accuracies and efficiencies. Nat Biomedical Eng. 2024. https://doi.org/10.1038/s41551-024-01223-5.
Camacho DM, Collins KM, Powers RK, Costello JC, Collins JJ. Next-generation machine learning for Biological Networks. Cell. 2018;173(7):1581–92. https://doi.org/10.1016/j.cell.2018.05.015.
Zakhem GA, Fakhoury JW, Motosko CC, Ho RS. Characterizing the role of dermatologists in developing artificial intelligence for assessment of skin cancer. J Am Acad Dermatol. 2021;85(6):1544–56. https://doi.org/10.1016/j.jaad.2020.01.028.
Maliha G, Gerke S, Cohen IG, Parikh RB. Artificial Intelligence and Liability in Medicine: Balancing Safety and Innovation. Milbank Q. 2021;99(3):629–47. https://doi.org/10.1111/1468-0009.12504.
Fan JL, Nazaret A, Azizi E. A thousand and one tumors: the promise of AI for cancer biology. Nat Methods. 2024;21(8):1403–6. https://doi.org/10.1038/s41592-024-02364-w.
Zhang C, Xu J, Tang R, Yang J, Wang W, Yu X, et al. Novel research and future prospects of artificial intelligence in cancer diagnosis and treatment. J Hematol Oncol. 2023;16(1):114. https://doi.org/10.1186/s13045-023-01514-5.
Zhang P, Zhang Q, Li S. Advancing Cancer Prevention through an AI-Based integration of traditional and western medicine. Cancer Discov. 2024;14(11):2033–6. https://doi.org/10.1158/2159-8290.Cd-24-0832.
Jee J, Fong C, Pichotta K, Tran TN, Luthra A, Waters M, et al. Automated real-world data integration improves cancer outcome prediction. Nature. 2024;636(8043):728–36. https://doi.org/10.1038/s41586-024-08167-5.
Viswanathan VS, Parmar V, Madabhushi A. Towards equitable AI in oncology. Nat Reviews Clin Oncol. 2024;21(8):628–37. https://doi.org/10.1038/s41571-024-00909-8.
Mehari M, Sibih Y, Dada A, Chang SM, Wen PY, Molinaro AM, et al. Enhancing neuro-oncology care through equity-driven applications of artificial intelligence. Neurooncology. 2024;26(11):1951–63. https://doi.org/10.1093/neuonc/noae127.
Acknowledgements
This work was supported by the National Natural Science Foundation of China (No.82104212, Recipient: Peixin Du). The views expressed are those of the authors and not necessarily those of the NSF. We apologize for not being able to cite all the publications related to this topic due to space constraints of the journal.
Funding
The National Natural Science Foundation of China, No.82104212.
Author information
Authors and Affiliations
Contributions
JW contributed in conceptualization, methodology, writing of the original draft. ZZ and ZL contributed in writing and review of the original draft. GL, SZ, CL, SH and SW contributed in visualization and editing of the draft. LZ contributed in conceptualization, supervision and project administration.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have 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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Wang, J., Zeng, Z., Li, Z. et al. The clinical application of artificial intelligence in cancer precision treatment. J Transl Med 23, 120 (2025). https://doi.org/10.1186/s12967-025-06139-5
Received:
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1186/s12967-025-06139-5