Algorithmic Brilliance on the Cancer Frontline
How Artificial Intelligence and Machine Learning Are Rewiring Oncology
Cancer, a heterogenous set of diseases marked by complex genomic and phenotypic variability, continues to represent a global health challenge with high morbidity and mortality. Advances in artificial intelligence (AI) and machine learning (ML) are revolutionizing oncology by leveraging deep neural architectures and multimodal data integration, catalyzing paradigm shifts in diagnostics, prognostics, and therapeutic stratification. Cryptic tumor molecular subtypes are elucidated and population-scale genetic risk stratification for hereditary malignancies is enabled through seminal advancements in AI and ML which surpass human analytical capabilities [1, 2]. Critical examination of ongoing challenges including algorithmic interpretability, data biases, and scalability constraints, underscores the complexities of clinical deployment [3]. Envisioning a transformative future, AI and ML synergize with clinical acumen to orchestrate precision oncology, harnessing computational prowess to decode tumor heterogeneity and deliver personalized, data-driven care at unprecedented scale.
Keywords: Artificial Intelligence, Machine Learning, Deep Learning, Oncology, Computational Genomics, Tumor Genomics, Hereditary Cancer, Precision Medicine
Introduction
Oncological Complexity Meets Computational Innovation
A seismic transformation powered by technological and computational advances is combating one of medicine’s most heterogeneous and daunting challenges, cancer. Cancer exhibits profound biological heterogeneity [4]. The notable diversity of cancer at the genetic, molecular and clinical levels illuminates the need for personalized therapies and adaptable care strategies. Inter-patient genomic variability, intra-tumoral cellular diversity and dynamic temporal evolution characterize this complexity. Somatic mutations, epigenetic alterations, and tumor microenvironment interactions drive these differences, rendering uniform treatment paradigms suboptimal [5, 6]. Concurrently, this complexity within oncology is compounded by a data deluge. High-throughput multi-omic technologies generate massive data sets which exceed the capacity of traditional statistical methods [7]. This “data revolution” presents an analytical strain on traditional approaches which underperform with the dimensionality and integration of disparate data modalities [8]. Optimized computational frameworks have boosted the executive function of this complexity. Artificial intelligence (AI) and machine learning (ML) algorithms enable precise deconvolution of molecular signatures [9]. Modern AI encompasses diverse applications including Natural Language Processing (NLP), computer vision, and knowledge graphs. ML techniques span from supervised algorithms dealing with predictive modeling to unsupervised methods encouraging pattern discovery [10]. Deep learning (DL) architectures have recently achieved human-level performance in diagnostic imaging and genomic sequence analysis [11]. Through extracting hierarchical features from raw data, the necessity of manual feature engineering is eliminated. This convergence of AI and ML in oncology features a paradigmatic shift toward data-driven medicine.
Background
Conceptual Foundations of Artificial Intelligence and Machine Learning
To establish a foundation for subsequent discussions, it is essential to distinguish between AI and ML, two interconnected yet distinct technological domains. AI represents the broader field encompassing any computational system designed to perform tasks typically requiring human intelligence [12]. In this context this includes knowledge graphs for medical ontology management, expert systems for clinical decision support, NLP for extracting insights from unstructured clinical narratives and robotic systems for surgical precision and laboratory automation. ML is a pivotal subset of AI. ML focuses on algorithms that iteratively learn patterns from data without explicit programming [13]. Encompassing multiple paradigms such as supervised learning, unsupervised learning, semi-supervised learning, and reinforcement learning, ML can optimally predict, uncover, and leverage data [14]. Supervised ML models excel in diagnostic and prognostic tasks. Unsupervised approaches reveal novel molecular taxonomies. AI’s broad capabilities complement ML’s data driven precision, enabling synergistic advancements in cancer diagnosis, risk stratification, and personalized therapy. This conceptual framework underpins the computational revolution in oncology, positioning AI and ML as transformative tools for addressing cancer’s complexity.
Charting the Computational Transformation of Cancer Care
The convergence of unprecedented data availability, computational power, and algorithmic sophistication has created a unique inflection point at which AI and ML are fundamentally transforming oncological practice [15]. This paper investigates this impact with particular emphasis on recent advances that have achieved clinical relevance across the entire cancer care continuum. Computational approaches are revolutionizing key domains and DL models are achieving superhuman performance in radiological and histopathological analysis [16]. Within genomic medicine, AI algorithms decode complex mutational landscapes to guide targeted therapies [17]. An examination of the emergence of sophisticated learning models determines their capacity to optimize performance in data-limited or decentralized settings. Real-world clinical deployments that have successfully translated AI and ML research into routine clinical practice are encompassed through detailed case studies and systematic analysis of breakthrough applications. Technical challenges such as improving model generalizability, computational scalability and integrating multimodal data streams are addressed. This illuminates how computational intelligence is not merely augmenting traditional oncology but fundamentally rewiring the foundation of this discipline. By synthesizing these advancements, the trajectory of precision oncology is delineated. AI and ML drive data centric, patient-specific solutions, creating opportunities and accelerating discoveries at this inflection point between conventional and computational medicine.
Oncology’s Data Revolution
A tsunami of information has reconfigured modern oncology. High-throughput multi-omics technologies capture genomic variations across millions of base pairs [18]. Transcriptomic profiles encompass thousands of gene expression patterns. Proteomic landscapes reveal complex protein interaction networks. Cellular and architectural details invisible to conventional microscopy are now generated with digital histopathology platforms. Multi parametric MRI, PET-CT fusion, radiomics and other advancing image modalities extract quantitative features from medical images that correspond to tumor biology [19]. Traditional methods were designed for concise, homogenous data structures and cannot compute the vast repositories of unstructured clinical narratives, treatment histories, longitudinal patient outcomes and other petabyte-scales, high-dimensional datasets from electronic medical records (EMRs). Dimensionality has become a curse to those conventional analytical approaches. The number of features often exceeds the number of samples [20]. Data modalities also require sophisticated preprocessing and normalization techniques. Advanced computational frameworks have become essential navigation tools [21]. Accurate deconvolution of molecular signatures is enabled by these technologies, assisting in the identification of subtle patterns that distinguish treatment-responsive patients from those likely to experience resistance or toxicity [22]. AI and ML facilitate precision oncology through sophisticated pattern recognition and predictive modeling by implementing individual therapeutic strategies that account for each patient’s unique molecular profile, clinical history, risk factors and other supporting information. This personalized approach optimizes therapeutic efficacy while minimizing adverse effects, moving beyond traditional population based treatment protocols. Cancer management now shifts from a reactive, symptom-based discipline to a proactive, data-driven field. By harnessing the full spectrum of available information which guides clinical decisions, a solid foundation is laid for personalized medicine in computational oncology.
The Evolving Foundation of Computational Oncology
Cancer represents a complex disease characterized by profound biological heterogeneity across genetic, molecular, and clinical dimensions. Each malignancy manifests unique mutational landscapes, epigenetic modifications, and protein expression profiles [23]. Inter-patient variability results in histologically similar tumors harboring distinct molecular signatures, while intra-tumoral heterogeneity reveals spatially diverse cellular populations within a single lesion [24]. Temporal evolution drives the acquisition of resistance mechanisms, such as alternative pathway activation [25]. These molecular subtypes challenge traditional histopathological classifications and complicate conventional therapies. Precision medicine addresses this complexity by targeting specific genetic alterations rather than relying on anatomical origins, such as EGFR mutations responsive to tyrosine kinase inhibitors [26]. Population-based protocols, designed for uniform diagnoses, fail to capture this molecular diversity which results in variable clinical outcomes. Comprehensive molecular profiling is essential to identify actionable therapeutic targets and optimize patient-specific strategies. Traditional hypothesis-driven research, constrained by human cognition and limited variable analysis, is being supplanted by algorithmic, data-driven paradigms [27].
The Rise of AI and ML in Oncology
Transformative applications are presently occurring where AI and ML have outmatched human diagnostic accuracy, reframed inherited cancer risk evaluation, and predicted clinical trajectories using longitudinal patient data with precision [28, 29]. Advanced DL frameworks, particularly those utilizing Convolutional Neural Networks (CNNs), Vision Transformers (ViTs), and Graph Neural Networks (GNNs), exploit pattern recognition beyond human perceptual limits. ML techniques spanning supervised learning, unsupervised and ensemble methods excel in complex pattern recognition [30]. They capture high-dimensional nonlinear associations critical to oncological analysis and enable automated feature extraction unlike traditional statistical methods, which rely on manual feature selection and linear assumptions [31]. Through this they generate robust signatures that surpass handcrafted radiomic or histomorphometric metrics in precision and reproducibility. For instance, GNNs model intricate molecular networks, enhancing predictive modeling of treatment response [32]. They also facilitate multimodal integration, synthesizing radiological, pathological, and molecular data to achieve refined phenotypic characterization of tumors [33]. The rise of AI and ML is timely due to three converging factors: unprecedented computational power enabling rapid analysis of complex datasets, expanded data availability from digitized clinical and research systems, and the intricate nature of oncological tasks requiring sophisticated analytical tools [34]. These advancements shift oncology from hypothesis-driven, manual paradigms to computational frameworks that augment clinical expertise. By leveraging AI and ML’s analytical prowess, oncology achieves enhanced diagnostic precision, risk stratification, and treatment planning.
Quantifiable Impact and Platform Status
Recent transformative applications provide quantifiable evidence of algorithmic progression in oncology. In breast cancer histopathology, CNNs have demonstrated diagnostic accuracy rates as high as ninety-eight percent, surpassing consensus pathologist rates. Google’s LYNA model outperformed human experts, achieving an AUC of 0.99 in metastatic node detection [35, 36]. Recently published multi-institutional studies report that Gradient Boosted Machine (GBM) models integrating multi-omic and clinical features predicted immunotherapy non-responders in metastatic melanoma with AUCs of 0.86-0.92, compared to 0.74-0.77 for classic logistic regression [37]. AI-powered digital pathology tools have reduced turnaround times by up to sixty-five percent in high-throughput urban centers and decreased diagnostic error rates in prospective implementation trials [38].
AI-Powered Oncology: Core Principles, Applications and Innovations
Beyond introductory concepts, AI in oncology serves as a force multiplier for domain experts by orchestrating information extraction, synthesis, and reasoning across widespread clinical and molecular landscapes. Unlike other medical fields, oncology presents daunting data heterogeneity [39]. Here AI is leveraged to weave together multi-scale biomedical signals that would otherwise remain siloed or indecipherable at human scale. Early AI implementations in cancer care emerged from expert systems development in the 1960s and 1970s, exemplified by DENDRAL’s chemical analysis system and MYCIN’s infectious disease diagnostics and later adapted for oncological applications through systems like ONCOCIN for automated chemotherapy protocol management in the 1980s [40]. These foundational architectures encoded clinical expertise through logical, rule-based structures. Despite limitations, these foundational systems demonstrated the potential for computational augmentation of clinical decision-making and established the conceptual framework for contemporary AI architectures. The integration of probabilistic reasoning, Bayesian networks, and case-based reasoning systems accelerated AI’s evolution in oncology through the 1990s and 2000s [41]. Recent deployments have utilized knowledge graphs and ontology-driven frameworks which integrate genomic, proteomic, and clinical ontologies [42]. Knowledge graphs integrate structures with dynamic relationship modeling to represent complex biomedical knowledge as interconnected networks of entities and relationships. This is exemplified by platforms like IBM Watson for Oncology, Google’s Healthcare Knowledge Graph, and emerging platforms such as PrimeKG [43]. Ontological systems, such as the National Cancer Institute Thesaurus (NCIt) and the Human phenotype Ontology (HPO), provide vocabularies that enable semantic interoperability across databases and systems. These can be used to map molecular pathways, drug mechanisms, and temporal treatment relationships, enabling hypothesis generation, cohort retrieval for rare subtypes and network-based drug repurposing. Particular usage includes precise mapping of tumor-specific signaling pathways, such as P13K/AKT in breast cancer [22]. Expert systems are used to synthesize multidimensional patient data to recommend targeted therapies, matching ALK inhibitors to lung cancer mutations [17]. AI-driven NLP parses unstructured oncology reports and literature constructing dynamic patient timelines from clinical narratives embedded in EMRs [44]. This allows for capturing of nuances that defy structured data formats, such as adverse event trajectories, evolving performance status, and real-world progression. BioBERT, ClinicalBERT and other advanced NLP systems in oncology employ transformer-based architectures that have been pre-trained on large corpora biomedical literature and clinical notes to understand medical terminology and contextual relationships [45]. Recent developments in large language models (LLMs), particularly GPT-based architectures fine-tuned for medical applications, have demonstrated remarkable capabilities in clinical scenarios, differential diagnosis generation and treatment recommendation synthesis [46]. Specialized medical chatbots powered by transformer architectures, such as Med-PaLM and ChatDoctor have shown capability in providing accurate medical information and preliminary diagnostic assistance [47]. Integration of retrieval-augmented generation (RAG) architectures with medical knowledge bases enables these systems to provide evidence-backed clinical insights that reference current literature and guidelines while maintaining transparency in their reasoning processes. Platforms such as TempusONE combine multi-omics analysis with clinical data integration to provide personalized treatment recommendations based on tumor molecular profiling and real-world evidence from similar patients. Emerging platforms like Foundation Medicines FoundationACT integrate genomic testing results with AI-driven interpretation engines that identify actionable mutations and recommend targeted therapies based on current clinical evidence [17]. Robotic systems and automation technologies enhance pathology workflows and surgical precision by integrating intraoperative imaging with AI-driven navigation. Surgical robotics platforms such as the da Vinci system incorporate AI-enhanced imagining, motion scaling, and tremor filtration correlating to a reduction in invasiveness and improved outcomes [48]. However, these robotic systems are not only limited to surgery. Robotic process automation is also increasingly used for auto-curation of research data sets, quality checks in large-scale biobank operations, and streamlining of tumor board workflows. In pathology, automated slide scanning systems coupled with AI-driven quality control algorithms have revolutionized histopathological workflows allowing for high-throughput processing of tissue samples while maintaining diagnostic accuracy [35]. Laboratory automation systems employ AI algorithms for specimen handling, processing optimization, and quality assurance. Significantly reducing turnaround times for critical diagnostic tests while minimizing human error [49]. These implementations demonstrate how AI architectures extend beyond analytical applications to encompass physical automation.
ML Essentials and Emerging Solutions in Oncology
Oncology demands adaptations of conventional techniques to account for biological complexity and data idiosyncrasies. The fundamental premise of ML in oncology rests on the assumption that cancer-related datasets contain discoverable patterns that correlate with clinically meaningful outcomes, enabling predictive models that can generalize to new patients and scenarios [10]. This data-driven approach contrasts with traditional statistical methods that typically require strong distributional assumptions and manual feature specification, instead allowing algorithms to automatically discover complex, nonlinear relationships within high-dimensional cancer datasets [50]. However, recent studies have shown certain classical algorithms are not rendered irrelevant. For instance, k-Nearest Neighbors (kNN) is frequently used for sample classification and local patient stratification [51]. GBMs, random forests, and robust tree ensemble methods, are foundational tools for comprehensive cancer data collections. GBMs are employed to predict immunotherapy outcomes by integrating mutational signatures and clinical features. Random forest algorithms excel in handling mixed-type clinical data, combining categorical variables such as staging information with continuous biomarker measurements to predict treatment outcomes while providing interpretable feature importance rankings. Classification algorithms such as Support Vector Machines (SVMs) with radial basis function kernels have demonstrated exceptional performance in gene expression-based cancer subtyping, particularly in distinguishing molecular subtypes of breast cancer and acute leukemias [52]. Clustering algorithms such as k-means, hierarchical clustering and more sophisticated methods like consensus clustering and non-negative matrix factorization are used to identify previously unknown cancer subtypes with distinct molecular signatures and clinical behaviors [53]. Logistic regression models remain fundamental tools for risk stratification in oncology, particularly when augmented with regularization techniques such as LASSO and elastic net that enable feature selection in multi-variable cancer data repositories [54]. Supervised learning predicts tumor recurrence, such as in colorectal cancer, using multi-omics signatures. Unsupervised learning addresses the challenge of discovering novel patterns and relationships in cancer datasets where ground truth labels are unavailable or incomplete. An example of unsupervised learning in use is with distinct pancreatic ductal adenocarcinoma clusters, through genomic clustering. Semi-supervised learning improves model performance in data-scare cancers, such as pediatric sarcomas, by leveraging limited labeled samples. These paradigms dominate clinical applications, where labeled datasets enable training of predictive models for diagnosis, prognosis and treatment response prediction [51]. Dimensionality reduction techniques, including principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE), and uniform manifold approximation and projection (UMAP), enable visualization and analysis of multi-layered cancer data structures while preserving critical biological relationships [55]. Advanced manifold learning approaches such as autoencoders and variational autoencoders have demonstrated capability to learn compressed representations of genomic data that capture biologically meaningful patterns while reducing computational complexity [56]. Co-training algorithms that leverage multiple views of the same data, such as combining histological images with corresponding molecular profiles, enable more robust model training by exploiting complementary information sources. Graph-based methods that propagate labels through similarity networks have proven effective for gene function prediction and drug-target interaction discovery [57]. Techniques such as MixMatch and FixMatch, have achieved remarkable performance in medical image analysis by enforcing prediction consistency across augmented versions of unlabeled samples. ML is further employed for optimizing adaptive clinical trials and treatment regimens. Reinforcement learning optimizes adaptive radiotherapy schedules based on tumor response dynamics. It is also used to identify dosing schedules or therapy switches to maximize patient-specific benefit and minimize toxicity using simulated cohorts or real-time patient registries as learning environments [58].The emergence of federated and privacy-preserving learning architectures enables collaborative discovery across cancer centers without aggregating patient-level data. Federated learning enables multi-center studies, training models on decentralized datasets from diverse cancer cohorts [59]. Advanced tree ensemble methods, like XGBoost and LightGBM, improve survival predictions in heterogeneous cancers by integrating clinical and molecular features [60]. Explainable AI techniques, such as SHAP and LIME, elucidate model predictions for clinical adoption, as seen in lymphoma risk stratification [61]. Modern ML’s impact is amplified by embedded feature importance metrics, aiding the interpretability of model-driven discoveries.
Foundational DL Approaches and Breakthroughs in Cancer Intelligence
DL is an advanced ML paradigm which employs multi-layered neural networks to extract complex, hierarchical feature representations from raw data [11]. The fundamental principle underlying DL’s success in oncology stems from its ability to learn complex, nonlinear mappings between feature-dense oncological datasets and clinical outcomes through gradient-based optimization of millions or billions of parameters. The Universal Approximation Theorem provides theoretical foundations for DL’s representational power [62]. More recent advances in optimization algorithms, regularization techniques, and architectural innovations have enabled practical training of these complex models on comprehensive oncological data repositories. CNNs excel in analyzing histopathology and radiological images. The architectural innovations that define modern CNNs, including residual connections in ResNet architectures, dense connectivity patterns in DenseNet models and attention mechanisms in networks such as SENet, have enabled training of increasingly deep networks that capture fine-grained pathological features while maintaining gradient flow during backpropagation [63]. ViTs are a more recent emergence which have proven to be an alternative to CNNs since they capture global contextual features. Their self-attention mechanisms, particularly for whole-slide image analysis in digital pathology, enable modeling of long-range spatial dependencies that traditional convolutional architectures struggle to capture. The application of transformer architectures to genomic sequence analysis has revolutionised sequential data processing in oncology. Allowing for parallel processing and modeling of complex dependencies across entire sequences and enabling discoveries in variant effect prediction [64]. Models such as MUSK, PRISM2 and Enformer demonstrate accuracy in predicting gene expression changes from DNA sequence alterations [65]. BERT-based models fine-tuned on biomedical literature have achieved incredible performance in clinical entity recognition, relation extraction and clinical outcome prediction from unstructured text [66]. Recurrent Neural Networks (RNNs) and their variants address the temporal and sequential nature of cancer data, particularly in modeling disease progression, treatment response trajectories and survival outcomes over time [67]. Long Short-Term Memory (LSTM) networks overcome the vanishing gradient problem that limits standard RNNs [68]. Gated Recurrent Units (GRUs) provide computational efficiency advantages while maintaining comparable performance to LSTMs in many oncological applications. Attention based RNN architectures enable identification of critical time points and clinical events that most strongly influence patient outcomes [67]. Bidirectional RNNs that process sequences in both forward and backward directions have displayed superior performance in clinical note analysis, where context from both past and future clinical events contributes to accurate information extraction [69]. GNNs model complex molecular interactions, protein-protein networks, and multi-omics integration tasks, improving treatment response predictions. The representational power of DL combined with the structural expressiveness of graph theory, positions GNNs as an emerging frontier. Advanced GNN variants such as Graph Attention Networks (GANs) and GraphSAGE have demonstrated superior performance in molecular property prediction and drug discovery applications by learning optimal aggregation strategies [70]. DL architectures are transforming oncology through rapid, accurate diagnostics and personalized therapeutic strategies.
Limitations and Technical Challenges
Data Quality and Annotation
Multi-parametric oncological data, such as multi-omics and imaging, pose significant challenges to analytical applications. Inconsistent data quality and annotation bottlenecks are just the introduction of computational limitations [51]. Genomic data often suffers from missing values or batch effects, as seen in single-cell RNA-seq for pancreatic cancer. MRI scans for gliomas and other imaging datasets frequently lack standardized acquisition protocols, presenting variability in feature extraction [71]. AI systems utilizing expert systems and rule based approaches struggle with inconsistent clinical terminology and incomplete knowledge bases. This often leads to extensive manual curation of oncological decision trees. ML algorithms are particularly sensitive to class imbalance in cancer datasets where rare subtypes constitute less than five percent of training samples, such as those for pediatric leukemias. This displays poor minority class performance and limited generalizability [72]. DL architectures including ResNet and ViTs exhibit vulnerability to annotation noise. CNNs inadvertently learn from systematic radiologist disagreements (kappa values of 0.6-0.8) and imaging artifacts, leading to a reduction in cross-institutional generalizability by fifteen to thirty percent. As seen in the use of CNNs for lung cancer classification, extensive labeled data sets are required but inconsistencies in pathologist interpretations present challenges [16]. Calibration against standardized oncological benchmarks is critical to ensure reliable predictions across diverse cancer cohorts.
Model Interpretability
Model interpretability remains a critical challenge for computational advancements in oncology as clinical trust hinges on understanding algorithmic decisions [73]. AI rule-based systems provide explicit decision pathways through interpretable if-then logic trees but lack adaptability to novel clinical presentations. ML methods such as logistic regression and decision trees offer quantifiable feature importance through coefficients and Gini impurity measures [74]. This enables clinicians to understand individual variables that contribute to predictions. Further complicating clinical validation are GBMs used for breast cancer survival prediction, which provide limited insight into variable importance [72]. DL models, particularly transformer architectures and deep CNNs with more than fifty layers, operate as black boxes where gradient-based explanations (GradCAM, SHAP) provide only superficial visualization of attention regions without revealing underlying decision mechanisms or multi-modal feature interactions [75]. These offer partial interpretability by quantifying feature impacts but fail to fully elucidate deep neural network decisions, as seen in lymphoma risk stratification. Without interpretable outputs, clinicians hesitate to adopt analytical algorithms, limiting integration into tumor board workflows.
Rigorous Calibration and External Validation Requirements
AI expert systems require manual rule adjustment across institutions, with knowledge base transfer achieving only sixty to seventy percent rule applicability in new clinical environments [76]. ML algorithms like XGBoost and logistic regression demonstrate miscalibration with Brier scores increasing from 0.08 to 0.15 during external validation, necessitating Platt scaling or isotonic regression for probability recalibration [77]. DL networks exhibit severe overconfidence with temperature scaling requirements showing calibration error increases of twenty to forty percent when deployed across different imaging protocols (1.5T vs 3T MRI, various CT reconstruction kernels) [59].
Technical Scalability and Deployment
Because AI knowledge based systems face scalability limitations due to manual rule maintenance and expert knowledge bottlenecks, oncologists are required to be involved for each new cancer type integration. ML algorithms, such as ensemble methods, demand moderate computational resources (2-8 GB RAM) but struggle with real-time inference when processing extensive oncological data matrices (> 20,000 features) [78]. DL architectures require substantial GPU memory (16-80 GB for large models), with inference latency of two to five seconds per image [79]. This creates workflow complications in high-throughput clinical settings. Due to inconsistent validation protocols, DL models used for treatment response prediction exhibit variability in performance across cancer types. Calibration against standardized oncological benchmarks is critical yet challenging due to tumor heterogeneity.
Reproducibility and Validation Challenges
AI rule-based systems suffer from incomplete knowledge base documentation and undisclosed expert consultation processes. This causes difficulty with independent rule reconstruction. AI driven analytics platforms often fail to generalize across institutions due to dataset variability, as seen in colorectal cancer risk prediction [80]. ML studies show forty to sixty percent performance variance due to unreported hyperparameter optimization procedures, cross-validation strategies, and feature selection methods across institutions [81]. ML models struggle with reproducibility when training data differ in genomic or imaging features [82]. DL models demonstrate sensitivity to initialization seeds, data augmentation policies and training procedures. Published accuracy differences range from five to fifteen percent between original and reproduced implementations. This is notably evident in transformer-based architectures for pathology image analysis [83, 84, 85]. Technological convergence with multi-center data-sharing platforms could enhance reproducibility but demands unified protocols.
Future
Next Generation Architectures
Emerging architectures are poised to revolutionize oncology by overcoming current limitations in data complexity and model performance [86]. An evolvement towards Neurosymbolic AI integrates rule-based logic with neural networks, enabling hybrid systems that incorporate clinical guidelines with data-driven predictions and enhancing molecular pathway analysis in cancers like glioblastomas [87]. Automated ML (autoML) frameworks utilizing Bayesian optimization for hyperparameter tuning, reduce model development time from months to days and achieve comparable performance to expert designed pipelines. Self-supervised ML models leveraging unlabeled multi-omics data, improve biomarker discovery in rare cancers by extracting latent features [88]. DL innovations encompass foundation models, LLMs with 100B+ parameters and ViTs for multi-modal medical imaging analysis [89]. Other DL advancements, including sparse CNNs, optimize histopathology analysis for breast cancer with reduced computational demands [90]. GNNs evolve to model dynamic tumor microenvironments, improving treatment response in pancreatic cancer. Meta-learning architectures enable rapid adaptation to new cancer subtypes, addressing heterogeneity in leukemias. These architectures enhance precision by modeling complex and oncological patterns, surpassing traditional supervised methods [81]. Future developments will focus on lightweight scalable models for real-time clinical applications.
Real-world validation and implementation
AI validation frameworks are transitioning towards continuous learning systems with real-time knowledge base updates. An implementation of A/B testing methodologies in clinical environments to assess rule performance against standard care protocols is seen. Real-world validation and implementation of large scale, multi-center trials for lung cancer diagnostics using CNNs demonstrate improved accuracy, but require standardized validation protocols [59, 81]. ML models need rigorous external validation across diverse cohorts to ensure generalizability [76]. Instead of solely focusing on predictive accuracy metrics, ML implementation strategies emphasize prospective clinical trials with embedded algorithms, utilizing pragmatic randomized controlled trial designs to evaluate model impact on patient outcomes. DL systems are undergoing tests in real-world settings to confirm robustness [38]. Through Monte Carlo dropout and ensemble methods, DL deployment approaches incorporate uncertainty quantification enabling confidence intervals for clinical predictions and establishing rejection thresholds. Implementation challenges demand seamless APIs and clinician training [91]. Automated validation pipelines, leveraging cloud-based platforms, streamline testing for heterogeneous cancers. Future efforts will prioritize robust, prospective trials to validate AI-driven tools in clinical workflows [81, 92].
Technological Convergence and Integration
Technological convergence integrates platforms with multi-omics, imaging and clinical systems to transform oncology. When combined, AI-driven knowledge graphs and DL networks enable holistic tumor profiling by linking genomic and radiological data [59]. Combining AI navigation with robotic systems enhances intraoperative precision imaging, as shown in glioma resection [33]. ML integration encompasses federated learning architectures combined with blockchain technology for secure multi-institutional model training. Cloud based analytics platforms unify these technologies, enabling scalable, real time decision support [93]. Future convergence hopes to develop unified frameworks for seamless data integration across oncology workflows. Multi-modal models validated in large-scale trials, will unify genomics, imaging and clinical data for personalized oncology.
Conclusion
At the frontier of oncology, Artificial Intelligence, Machine Learning, and Deep Learning are orchestrating a transformative shift in oncology. These methodologies are decoding cancer’s intricate genomic and phenotypic heterogeneity with unprecedented precision [81]. AI-driven knowledge graphs and expert systems have evolved beyond rule-based architectures to platforms that illuminate complex molecular networks and therapeutic targets [57]. ML’s ensemble methods deconvolute multi-omics signatures to reveal novel cancer subtypes and predict leukemia relapse [51]. GBMs achieve AUCs of 0.86-0.92 in immunotherapy response prediction, consistently outperforming traditional approaches [60]. DL breakthroughs are striking, with CNNs demonstrating 98% diagnostic accuracy and models achieving 0.99 AUC in metastatic detection [94]. DL networks have achieved superhuman accuracy, as further shown through the use of CNNs in melanoma histopathology and lung cancer radiomics [1, 87]. ViTs and GNNs have expanded analytical capabilities through global contextual modeling and complex molecular interaction networks [32, 64]. Multimodal platforms now synthesize genomic, radiological, and clinical data into holistic tumor profiles propelling oncology from blunt, population-based protocols to personalized, data-driven strategies [33]. This computational revolution addresses cancer’s fundamental challenge: biological complexity that exceeds human analytical capacity. Advanced architectures including federated learning, automated ML frameworks, and real-time decision support systems are successfully translating research into routine practice [59]. The future promises even greater transformation through neurosymbolic AI integrating clinical guidelines with neural predictions, self-superviewed DL accelerating rare cancer biomarker discovery and foundational models enabling rapid adaptation to emerging subtypes [95, 96]. Sparse CNNs and cloud-integrated platforms will overcome scalability barriers for real-time treatment planning [97]. Through the fusion of AI’s analytical power with clinical expertise, oncology stands positioned to conquer tumor heterogeneity. The convergence of computational brilliance and biological insight ignites a visionary future, where algorithms and clinicians unite to rewrite the narrative of cancer at unprecedented scale and precision.
Supplemental Data
No datasets were generated or analyzed during the current study.
Conflict of Interest
Declaration of Competing Interest. The author declares that they have no competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
Esteva, A., et al. (2017). Dermatologist-level classification of skin cancer with deep neural networks. Nature, 542(7639), 115–118. https://doi.org/10.1038/nature21056
Mavaddat, N., et al. (2019). Polygenic risk scores for prediction of breast cancer and breast cancer subtypes. Am J Hum Genet, 104, 21–34. https://doi.org/10.1016/j.ajhg.2018.11.002
Topol, E. J. (2019). High-performance medicine: the convergence of human and artificial intelligence. Nat Med, 25, 44–56. https://doi.org/10.1038/s41591-018-0300-7
Hanahan, D., & Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell, 144(5), 646–674. https://doi.org/10.1016/j.cell.2011.02.013
Kandoth, C., et al. (2013). Mutational landscape and significance across 12 major cancer types. Nature, 502(7471), 333–339. https://doi.org/10.1038/nature12634
Yates, L. R., et al. (2015). Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat Med, 21(7), 751–759. https://doi.org/10.1038/nm.3886
Goodwin, S., et al. (2016). Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet, 17(6), 333–351. https://doi.org/10.1038/nrg.2016.49
Ching, T., et al. (2018). Opportunities and obstacles for deep learning in biology and medicine. J R Soc Interface, 15, 20170387. https://doi.org/10.1098/rsif.2017.0387
Litjens, G., et al. (2017). A survey on deep learning in medical image analysis. Med Image Anal, 42, 60–88. https://doi.org/10.1016/j.media.2017.07.005
Kourou, K., et al. (2015). Machine learning applications in cancer prognosis and prediction. Comput Struct Biotechnol J, 13, 8–17. https://doi.org/10.1016/j.csbj.2014.11.005
LeCun, Y., et al. (2015). Deep learning. Nature, 521(7553), 436–444. https://doi.org/10.1038/nature14539
Russell, S., & Norvig, P. (2020). Artificial Intelligence: A Modern Approach (4th ed.). Pearson. https://www.pearson.com/us/higher-education/product/Russell-Artificial-Intelligence-A-Modern-Approach-4th-Edition/9780134610993.html
Shalev-Shwartz, S., & Ben-David, S. (2014). Understanding Machine Learning: From Theory to Algorithms. Cambridge University Press. https://doi.org/10.1017/CBO9781107298019
Yu, K.-H., et al. (2018). Artificial intelligence in healthcare. Nat Biomed Eng, 2, 719–731. https://doi.org/10.1038/s41551-018-0305-z
Elemento, O., et al. (2021). Artificial intelligence in cancer research, diagnosis and therapy. Nat Rev Cancer, 21(12), 747–752. https://doi.org/10.1186/s40164-024-00505-7
Hosny, A., et al. (2018). Artificial intelligence in radiology. Nat Rev Cancer, 18(8), 500–510. https://doi.org/10.1038/s41568-018-0016-5
Berger, M. F., & Mardis, E. R. (2020). The emerging clinical relevance of genomics in cancer medicine. Nat Rev Clin Oncol, 17(6), 353–365. 10.1038/s41571-018-0002-6
The Cancer Genome Atlas Research Network. (2014). Comprehensive molecular profiling of lung adenocarcinoma. Nature, 511(7511), 543–550. https://doi.org/10.1038/nature13385
Aerts, H. J., et al. (2016). Radiomics: the process and the challenges. Magn Reson Imaging, 34(7), 1238–1248. https://doi.org/10.1016/j.mri.2016.06.005
Zhang, B., et al. (2021). Multimodal deep learning for cancer diagnosis: recent progress and perspectives. J Transl Med, 19, 44. 10.2196/57830
Rajkomar, A., et al. (2019). Machine learning in medicine. N Engl J Med, 380, 1347–1358. https://doi.org/10.1056/NEJMra1814259
Sammut, S. J., et al. (2022). Multi-omic machine learning predictor of breast cancer therapy response. Nature, 601(7894), 623–629. https://doi.org/10.1038/s41586-021-04278-5
Cancer Genome Atlas Research Network. (2013). Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med, 368(22), 2059–2074. https://doi.org/10.1056/NEJMoa1301689
Marusyk, A., et al. (2012). Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer, 12(5), 323–334. https://doi.org/10.1038/nrc3261
Swanton, C., et al. (2015). Intratumor heterogeneity: evolution through space and time. Cancer Res, 75(20), 4211–4216. https://doi.org/10.1158/0008-5472.CAN-15-0857
Lynch, T. J., et al. (2004). Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med, 350(21), 2129–2139. https://doi.org/10.1056/NEJMoa040938
Obermeyer, Z., & Emanuel, E. J. (2016). Predicting the future—big data, machine learning, and clinical medicine. N Engl J Med, 375(13), 1216–1219. https://doi.org/10.1056/NEJMp1606181
Liu, Y., et al. (2020). A deep learning system for differential diagnosis of primary and metastatic lung cancer on whole-slide images. Nat Commun, 11, 1034. 10.3390/cancers15153981
Kapil, A., et al. (2022). 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, 80(2), 261-270. https://doi.org/10.1111/his.14571
Lee, S. M., et al. (2022). Application of ensemble machine learning algorithms in predicting clinical outcomes for cancer patients. Sci Rep, 12, 17769. 10.2147/JMDH.S410301
Bertsimas, D., Wiberg, H., & Dunn, J. (2020). Machine learning in oncology: Methods, applications, and challenges. JCO Clinical Cancer Informatics, 4, 885-894. https://doi.org/10.1200/CCI.20.00072
Chen, R. J., et al. (2021). Graph neural networks for cancer drug discovery and biomarker prediction. Nat Mach Intell, 3, 223–234. https://doi.org/10.1038/s42256-021-00316-z
Boehm, K. M., et al. (2021). Harnessing multimodal data integration to advance precision oncology. Nat Rev Cancer, 22(2), 114–126. https://doi.org/10.1038/s41568-021-00408-3
Gehrung, M., et al. (2021). Artificial intelligence for the detection of cancer: a systematic review. Sci Rep, 11, 1–9. : 10.1007/s11831-021-09648-w
Campanella, G., et al. (2019). Clinical-grade computational pathology using weakly supervised deep learning on whole slide images. Nat Med, 25, 1301–1309. https://doi.org/10.1038/s41591-019-0508-1
Steiner, D. F., et al. (2019). Impact of Deep Learning Assistance on the Histopathologic Review of Lymph Nodes for Metastases. Am J Surg Pathol, 43, 1636–1646. 10.1097/PAS.0000000000001151
Hugo, W., et al. (2016). Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell, 165, 35–44. https://doi.org/10.1016/j.cell.2016.02.065
Tolkach, Y., Ovtcharov, V., Pryalukhin, A. et al. An international multi-institutional validation study of the algorithm for prostate cancer detection and Gleason grading. npj Precis. Onc. 7, 77 (2023). https://doi.org/10.1038/s41698-023-00424-6
Shmatko, A., et al. (2022). Artificial intelligence in histopathology: Enhancing cancer research and clinical oncology. Nat Cancer, 3(9), 1026–1038. 10.1038/s43018-022-00436-4
Jha, S., & Topol, E. (2016). Adapting to artificial intelligence: radiologists and pathologists as information specialists. JAMA, 316, 2353–2354. https://doi.org/10.1001/jama.2016.17438
Rajkomar, A., Dean, J., & Kohane, I. (2019). Machine learning in medicine. New England Journal of Medicine, 380(14), 1347–1358. https://doi.org/10.1056/NEJMra1814259
Nicholson, D. N., & Greene, C. S. (2020). Constructing and interpreting knowledge graphs for precision medicine. Nat Rev Genet, 21(10), 595–610. 10.1016/j.csbj.2020.05.017
Chandak, P., Huang, K., & Zitnik, M. (2023). Building a knowledge graph to enable precision medicine. Scientific Data, 10, 67. https://doi.org/10.1038/s41597-023-01960-3
Li, I., Pan, et al. (2022). Neural natural language processing for unstructured data in electronic health records: A review. Computational Linguistics, 48(4), 905-952. https://doi.org/10.1016/j.cosrev.2022.100511
Sangariyavanich, E., et al. (2023). Systematic review of natural language processing for recurrent cancer detection from electronic medical records. Informatics in Medicine Unlocked, 42, 101327. https://doi.org/10.1016/j.imu.2023.101326
Singhal, K., et al. (2023). Large language models encode clinical knowledge. Nature, 620(7972), 172-180. https://doi.org/10.1038/s41586-023-06291-2
Esmaeilzadeh, P., Hassanein, K., & Head, M. (2025). Using AI chatbots (e.g., CHATGPT) in seeking health-related information online: The case of a common ailment. Applied Computing and Informatics. https://doi.org/10.1108/ACI-11-2024-0032
Bakasa, W., & Viriri, S. (2023). Stacked ensemble deep learning for pancreas cancer classification using extreme gradient boosting. Frontiers in Artificial Intelligence, 6, 1232640. https://doi.org/10.3389/frai.2023.1232640
Ficarra, V., et al.(2009). Retropubic, laparoscopic, and robot-assisted radical prostatectomy: A systematic review and cumulative analysis of comparative studies. European Urology, 55(5), 1037-1063. https://doi.org/10.1016/j.eururo.2009.01.036
Xie, H., Jia, Y., & Liu, S. (2024). Integration of artificial intelligence in clinical laboratory medicine: Advancements and challenges. Interdisciplinary Medicine, 2(2), e20230056. https://doi.org/10.1002/INMD.20230056
Alharbi, F., & Vakanski, A. (2023). Machine learning methods for cancer classification using gene expression data: A review. Bioengineering, 10(2), 173. https://doi.org/10.3390/bioengineering10020173
Huang, S., Cai, N., Pacheco, P. P., Narrandes, S., Wang, Y., & Xu, W. (2018). Applications of Support Vector Machine (SVM) learning in cancer genomics. Cancer Genomics & Proteomics, 15(1), 41–51. https://doi.org/10.21873/cgp.20063
Gao, L., Ye, M., Lu, X., & Huang, D. (2017). Hybrid method based on information gain and support vector machine for gene selection in cancer classification. Genomics, Proteomics & Bioinformatics, 15(6), 389–395. https://doi.org/10.1016/j.gpb.2017.08.002
Tibshirani, R. (1996). Regression shrinkage and selection via the lasso. Journal of the Royal Statistical Society: Series B (Methodological), 58(1), 267–288. https://doi.org/10.1111/j.2517-6161.1996.tb02080.x
Ding, J., Shah, S., & Condon, A. (2018). Density-based dimensionality reduction for single-cell RNA-seq data analysis. Bioinformatics, 34(17), i298–i306. https://doi.org/10.1093/bioinformatics/bty252
Way, G. P., Zietz, M., Rubel, C., & Greene, C. S. (2020). Compressing gene expression data using autoencoders for biological discovery. Bioinformatics, 36(Supplement_1), i355–i363. https://doi.org/10.1093/bioinformatics/btaa466
Zhang, Y., & Chen, L. (2020). Graph-based methods for gene function prediction and drug-target interaction. Briefings in Bioinformatics, 21(6), 1855–1867. https://doi.org/10.1093/bib/bbz104
Berthelot, D., Carlini, N., Goodfellow, I., Papernot, N., Oliver, A., & Raffel, C. (2019). MixMatch: A holistic approach to semi-supervised learning. Advances in Neural Information Processing Systems, 32, 5050–5060. https://doi.org/10.48550/arXiv.1905.02249
Kaissis, G. A., Makowski, M. R., Rückert, D., & Braren, R. F. (2020). Secure, privacy-preserving and federated machine learning in medical imaging. Nature Machine Intelligence, 2(6), 305–311. https://doi.org/10.1038/s42256-020-0186-1
Thedinga, K., & Rohr, K. (2021). A gradient tree boosting and network propagation derived pan-cancer survival network of the tumor microenvironment. Scientific Reports, 11(1), 24028. https://doi.org/10.1016/j.isci.2021.103617
Alabi, R. O., et al. (2023). Machine learning explainability in nasopharyngeal cancer survival using LIME and SHAP. Scientific Reports, 13(1), 8984. https://doi.org/10.1038/s41598-023-35795-0
Hornik, K., et al. (1989). Multilayer feedforward networks are universal approximators. Neural Netw, 2(5), 359–366. https://doi.org/10.1016/0893-6080(89)90020-8
He, K., Zhang, X., Ren, S., & Sun, J. (2016). Deep residual learning for image recognition. Proceedings of the IEEE Conference on Computer Vision and Pattern Recognition (CVPR), 770–778. https://doi.org/10.1109/CVPR.2016.90
Shamshad, F., Khan, S., Zamir, S. W., Khan, M. H., Hayat, M., Khan, F. S., & Fu, H. (2023). Transformers in medical imaging: A survey. Medical Image Analysis, 88, 102802. https://doi.org/10.1016/j.media.2023.102802
Azad, R., Arimond, R., Aghdam, E. K., Kazerouni, A., & Merhof, D. (2024). Advances in medical image analysis with vision transformers: A comprehensive review. Medical Image Analysis, 91, 103000. https://doi.org/10.1016/j.media.2023.103000
Lee, J., Yoon, W., Kim, S., Kim, D., Kim, S., So, C. H., & Kang, J. (2020). BioBERT: A pre-trained biomedical language representation model for biomedical text mining. Bioinformatics, 36(4), 1234–1240. https://doi.org/10.1093/bioinformatics/btz682
Choi, E., et al. (2016). Doctor AI: Predicting clinical events via recurrent neural networks. J Mach Learn Res, 56, 301–318. https://arxiv.org/abs/1511.05942
Hochreiter, S., & Schmidhuber, J. (1997). Long short-term memory. Neural Comput, 9(8), 1735–1780. https://doi.org/10.1162/neco.1997.9.8.1735
Rajkomar, A., et al. (2018). Scalable and accurate deep learning with electronic health records. NPJ Digit Med, 1(1), 18. https://doi.org/10.1038/s41746-018-0029-1
Zitnik, M., Agrawal, M., & Leskovec, J. (2018). Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics, 34(13), i457–i466. https://doi.org/10.1093/bioinformatics/bty294
Leek, J. T., et al. (2010). Tackling the widespread and critical impact of batch effects in high-throughput data. Nature Reviews Genetics, 11(10), 733–739. https://doi.org/10.1038/nrg2825
Garapati, S. S., et al. (2022). Bias and class imbalance in oncologic data—Towards inclusive and transferrable AI in large scale oncology data sets. Cancers, 14(12), 2897. https://doi.org/10.3390/cancers14122897
Holzinger, A., et al. (2019). Causability and explainability of artificial intelligence in medicine. Wiley Interdiscip Rev Data Min Knowl Discov, 9(4), e1312. https://doi.org/10.1002/widm.1312
Van Calster, B., et al. (2019). Reporting and interpreting decision curve analysis: A guide for investigators. Eur Urol, 74(6), 796–804. https://doi.org/10.1016/j.eururo.2018.08.038
Lundberg, S. M., & Lee, S. I. (2017). A unified approach to interpreting model predictions. Adv Neural Inf Process Syst, 30, 4765–4774. https://arxiv.org/abs/1705.07874
Salem, H., Soria, D., Lund, J. N., & Awwad, A. (2021). A systematic review of the applications of expert systems (ES) and machine learning (ML) in clinical urology. BMC Medical Informatics and Decision Making, 21(1), 169. https://doi.org/10.1186/s12911-021-01585-9
Niculescu-Mizil, A., & Caruana, R. (2005). Predicting good probabilities with supervised learning. Proceedings of the 22nd International Conference on Machine Learning (ICML), 625–632. https://doi.org/10.1145/1102351.1102430
Zhang, Z., et al. (2019). Predictive analytics with gradient boosting in clinical medicine. Ann Transl Med, 7(7), 152. https://doi.org/10.21037/atm.2019.03.29
Coudray, N., et al (2018). Classification and mutation prediction from non–small cell lung cancer histopathology images using deep learning. Nature Medicine, 24(10), 1559–1567. https://doi.org/10.1038/s41591-018-0177-5
Edginton-White, B., Maytum, A., Kellaway, S. G., et al. (2023). A genome-wide relay of signalling-responsive enhancers drives hematopoietic specification. Nature Communications, 14, 267. https://doi.org/10.1038/s41467-023-35910-9
Beam, A. L., Manrai, A. K., & Ghassemi, M. (2020). Challenges to the reproducibility of machine learning models in health care. JAMA, 323(4), 305-306. https://doi.org/10.1001/jama.2019.20866
Khan, N. M., Abraham, N., & Guan, L. (2019). Machine learning on biomedical images: Interactive learning, transfer learning, class imbalance, and beyond. 2019 IEEE EMBS International Conference on Biomedical & Health Informatics (BHI), 1-4. https://arxiv.org/abs/1902.05908
Stacke, K., Eilertsen, G., Unger, J., & Lundström, C. (2021). Reproducibility of deep learning in digital pathology whole slide image analysis. PLOS Digital Health, 1(12), e0000145. https://doi.org/10.1371/journal.pdig.0000145
Maxwell, A. E., Sharma, M., & Donaldson, K. A. (2022). Enhancing reproducibility and replicability in remote sensing deep learning research and practice. Remote Sensing, 14(22), 5760. https://doi.org/10.3390/rs14225760
Babaie, M., Kalra, S., Sriram, A., Mitcheltree, C., Zhu, S., Khatami, A., ... & Tizhoosh, H. R. (2023). A survey of Transformer applications for histopathological image analysis: New developments and future directions. BioMedical Engineering OnLine, 22(1), 96. https://doi.org/10.1186/s12938-023-01157-0
Acosta, J. N., Falcone, G. J., Rajpurkar, P., & Topol, E. J. (2022). Multimodal biomedical AI. Nature Medicine, 28(9), 1773-1784. https://doi.org/10.1038/s41591-022-01981-2
Amann, J., Blasimme, A., Vayena, E., Frey, D., & Madai, V. I. (2020). Explainability for artificial intelligence in healthcare: A multidisciplinary perspective. BMC Medical Informatics and Decision Making, 20(1), 310. 10.1186/s12911-020-01332-6
Azizi, S., et al. (2021). Big self-supervised models advance medical image classification. Int Conf Comput Vis, 3478–3488. https://ieeexplore.ieee.org/document/9710396
Karthik, R., Menaka, R., Hariharan, M., Won, D., & Saba, T. (2025). Vision-language foundation models for medical imaging: a review of current practices and innovations. Biomedical Engineering Letters, 15(1), 1-21. https://doi.org/10.1007/s13534-025-00484-6
Yan, R., et al. (2019). Breast cancer histopathology image classification through assembling multiple compact CNNs. BMC Medical Informatics and Decision Making, 19(1), 1-17. https://doi.org/10.1186/s12911-019-0913-x
Gal, Y., & Ghahramani, Z. (2016). Dropout as a Bayesian approximation: Representing model uncertainty in deep learning. Proceedings of the 33rd International Conference on Machine Learning (ICML), 1050–1059. https://doi.org/10.48550/arXiv.1506.02142
Liu, Y., Kim, J., Balagurunathan, Y., Li, Y., Flores, R. M., Goldgof, D. B., ... & Gillies, R. J. (2020). Artificial intelligence versus clinicians: Systematic review of design, reporting standards, and claims of deep learning studies. The BMJ, 368, m689. https://doi.org/10.1136/bmj.m689
Zhu, L., Pan, J., Chen, R., Wang, H., Zhang, L., & Chen, M. (2024). Harnessing artificial intelligence for prostate cancer management. Cell Reports Medicine, 5(4), 101502. https://doi.org/10.1016/j.xcrm.2024.101506
Bejnordi, B. E., et al.. (2017). Diagnostic assessment of deep learning algorithms for detection of lymph node metastases in women with breast cancer. JAMA, 318(22), 2199–2210. https://doi.org/10.1001/jama.2017.14585
Holzinger, A., Malle, B., Saranti, A., & Pfeifer, B. (2021). Towards multi-modal causability with graph neural networks enabling information fusion for explainable AI. Information Fusion, 71, 28–37. https://doi.org/10.1016/j.inffus.2021.01.008
Wang, X., et al. (2024). Clinical Histopathology Imaging Evaluation Foundation (CHIEF): A general-purpose machine learning framework for histopathology image analysis. Nature Medicine. https://pmc.ncbi.nlm.nih.gov/articles/PMC12128506/
Han, X., et al. (2021). EfficientNet: Rethinking model scaling for convolutional neural networks. International Conference on Machine Learning (ICML), 6105–6114. https://doi.org/10.48550/arXiv.1905.11946