Preprint
Review

This version is not peer-reviewed.

Artificial Intelligence in Oncology: Redefining Cancer Diagnosis and Therapy Through Data-Driven Precision

Submitted:

03 November 2025

Posted:

04 November 2025

You are already at the latest version

Abstract
Cancer remains one of the foremost challenges in global health, distinguished by profound biological complexity, heterogeneity, and therapeutic resistance. Over recent decades, oncology has transitioned from broadly cytotoxic regimens toward molecularly targeted and immune-based therapies; nevertheless, substantial clinical gaps persist. The emergence of artificial intelligence (AI) represents a transformative advance in this continuum, offering unprecedented capacity to integrate and interpret multidimensional biomedical data. AI-driven methodologies now play pivotal roles in early cancer detection, histopathological and radiomic assessment, biomarker discovery, and the prediction of therapeutic response. Furthermore, AI is accelerating drug discovery and enabling adaptive, patient-specific treatment strategies that transcend conventional evidence-based paradigms. This review synthesizes recent progress at the interface of AI and oncology, highlighting how machine learning, deep learning, and computational modeling are reshaping diagnostic precision, therapeutic optimization, and clinical decision-making. We also examine persisting challenges, including data heterogeneity, algorithmic transparency, and ethical implementation in real-world settings. Integrating computational intelligence with precision medicine heralds a future of cancer care that is predictive, preventive, and personalized—where human expertise and AI converge to redefine the boundaries of cancer treatment.
Keywords: 
;  ;  ;  ;  

1. Introduction: The Convergence of Oncology and Artificial Intelligence

Cancer remains a leading cause of morbidity and mortality worldwide, accounting for nearly 10 million deaths annually [1]. Despite remarkable advances in early detection, molecular profiling, and targeted therapy, the global cancer burden continues to rise, underscoring the need for more efficient diagnostic and therapeutic strategies [2]. The biological complexity and heterogeneity of tumors—driven by genomic instability, epigenetic alterations, and dynamic tumor–microenvironment interactions—pose significant challenges to conventional oncology paradigms [3].
The past two decades have witnessed an unprecedented expansion in the generation of biomedical data. High-throughput sequencing, digital pathology, radiomics, and multi-omics approaches now produce vast datasets that far exceed human interpretive capacity [4]. This data deluge has catalyzed the integration of Artificial Intelligence (AI) and machine learning (ML) into oncology, ushering in a new era of data-driven precision medicine [5].
AI, encompassing machine learning, deep learning, and natural language processing, offers powerful tools to analyze complex, high-dimensional datasets, identify subtle patterns, and derive predictive insights that support clinical decision-making [6]. Deep neural networks, in particular, have demonstrated human-comparable or even superior performance in diagnostic imaging, histopathology, and genomic variant interpretation [7,8]. By bridging the gap between molecular data and clinical phenotypes, AI has begun to transform the landscape of cancer diagnosis, prognosis, and treatment optimization.
Recent years have also seen the expansion of AI applications beyond diagnostics into therapeutic development and patient management. AI-driven drug discovery pipelines, virtual screening, and biomarker prediction models are accelerating the identification of novel anticancer agents [9,10]. Furthermore, integration of AI with clinical decision-support systems promises to enhance precision oncology by predicting treatment responses, minimizing adverse effects, and enabling adaptive, real-time interventions [11].
This review aims to provide a comprehensive overview of the role of Artificial Intelligence in cancer diagnosis and treatment. It begins with a summary of the biological and clinical challenges that define cancer heterogeneity, followed by an outline of evolving treatment modalities. We then explore the rapid advances in AI-driven diagnostics, drug discovery, and therapeutic monitoring, before discussing translational applications, regulatory challenges, and ethical considerations. Finally, we highlight emerging trends and future directions poised to shape the next generation of intelligent oncology.

2. Cancer Biology and Diagnostic Challenges

Cancer represents a constellation of diseases characterized by uncontrolled cell growth, genomic instability, and progressive acquisition of traits that confer survival advantages [12]. The modern understanding of cancer has evolved from a simplistic view of clonal expansion to a multidimensional model encompassing genetic, epigenetic, metabolic, and immune components [13]. Hanahan’s updated Hallmarks of Cancer framework captures this complexity, emphasizing the dynamic interplay between tumor cells and their microenvironment, including immune cells, fibroblasts, and extracellular matrix components [14].
A fundamental challenge in oncology lies in the heterogeneity of tumors—both intertumoral (across patients) and intratumoral (within the same tumor mass) [15]. This heterogeneity influences tumor progression, therapeutic response, and disease recurrence, rendering “one-size-fits-all” treatment strategies largely ineffective [16]. The rise of next-generation sequencing (NGS), single-cell omics, and spatial transcriptomics has revealed vast molecular diversity within cancers, yet translating these data into clinically actionable insights remains a daunting task [17].
From a diagnostic standpoint, early and accurate detection of malignancy is paramount to improving patient outcomes. However, several bottlenecks persist across conventional diagnostic modalities. Imaging techniques such as computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) are indispensable for tumor localization and staging, yet they often rely on the subjective interpretation of radiologists, introducing variability and potential oversight [18]. Similarly, histopathological examination—the gold standard for cancer diagnosis—demands extensive expertise and can be limited by sampling errors and inter-observer discrepancies [19].
The integration of molecular biomarkers into diagnostics, including circulating tumor DNA (ctDNA), microRNAs, and exosomal cargo, has enhanced detection sensitivity and specificity [20,21]. Nevertheless, identifying robust, reproducible biomarkers remains challenging due to tumor evolution, dynamic expression changes, and patient-to-patient variability [22].
Moreover, the rapid accumulation of multi-dimensional data from genomics, transcriptomics, proteomics, and imaging has far exceeded the analytic capacity of traditional statistical tools. Conventional bioinformatics pipelines often struggle to capture complex, nonlinear relationships inherent in biological systems [23]. This analytical gap underscores the urgent need for computational intelligence capable of handling massive datasets and uncovering hidden patterns that may inform diagnosis, prognosis, and therapeutic decision-making.
In this context, Artificial Intelligence (AI) emerges as a transformative solution. By leveraging deep learning, pattern recognition, and predictive analytics, AI systems can interpret complex biological and clinical data at unprecedented scale and accuracy [24]. The subsequent sections of this review discuss how AI is being harnessed to overcome diagnostic bottlenecks, redefine cancer phenotyping, and enable precision-guided treatment strategies across oncology.

3. Evolution of Cancer Treatment Modalities

The history of cancer therapy reflects a continuous evolution from empirical approaches to highly targeted interventions. The earliest modalities—surgery, radiotherapy, and chemotherapy—formed the foundational pillars of cancer management, yet each carries inherent limitations in specificity, toxicity, and resistance [25]. Over the past two decades, the advent of molecular oncology has catalyzed a paradigm shift toward precision-guided and immune-based therapies, transforming patient outcomes across multiple cancer types [26].

3.1. Surgery and Radiotherapy: The Classical Foundations

Surgical resection remains the most definitive curative option for localized malignancies, often complemented by adjuvant or neoadjuvant therapies [27]. Advances in imaging, intraoperative navigation, and minimally invasive techniques such as robotic-assisted surgery have significantly improved surgical precision and postoperative recovery [28].
Radiotherapy, first applied clinically in the early 20th century, has evolved through major technological innovations—including intensity-modulated radiation therapy (IMRT), stereotactic body radiation therapy (SBRT), and proton therapy—allowing for precise dose delivery while sparing normal tissue [29]. Nonetheless, radiation-induced toxicity and resistance remain key clinical challenges, necessitating better prediction of radiosensitivity and tumor response [30].

3.2. Chemotherapy: The Era of Cytotoxic Agents

The mid-20th century introduced systemic chemotherapy, marking a major breakthrough in treating metastatic disease [31]. However, conventional cytotoxic drugs lack tumor specificity, often resulting in off-target effects and cumulative toxicity [32]. Moreover, genetic and phenotypic heterogeneity among tumors drives intrinsic or acquired chemoresistance, limiting long-term efficacy [33]. The need to overcome these limitations led to the emergence of molecularly targeted therapies.

3.3. Targeted Therapy and the Rise of Molecular Oncology

Targeted therapies revolutionized oncology by exploiting specific genetic or signaling aberrations that drive tumor progression. The approval of imatinib for chronic myeloid leukemia and trastuzumab for HER2-positive breast cancer established the proof-of-concept for precision medicine [34,35]. Despite initial successes, targeted therapies frequently encounter resistance due to tumor evolution, pathway redundancy, and compensatory signaling mechanisms [36]. These challenges highlight the necessity for integrative approaches that account for the dynamic complexity of cancer biology.

3.4. Immunotherapy: Reawakening the Host Immune System

The emergence of cancer immunotherapy represents one of the most transformative advances in modern oncology. Immune checkpoint inhibitors (e.g., anti–PD-1/PD-L1, anti–CTLA-4 antibodies) and CAR-T cell therapies have achieved durable responses in previously refractory malignancies [37,38]. Nevertheless, only a subset of patients derives lasting benefit, and immune-related adverse events remain significant [39]. Understanding the molecular and cellular determinants of immune responsiveness continues to be a key research priority.

3.5. Toward Data-Guided and Adaptive Therapy

The integration of high-throughput molecular profiling, systems biology, and computational modeling has given rise to data-guided oncology. These technologies enable real-time assessment of tumor evolution, resistance mechanisms, and therapeutic efficacy [40]. Artificial Intelligence (AI) now plays a pivotal role in this evolution—enhancing imaging interpretation, predicting treatment responses, and optimizing drug combinations [41]. This convergence of clinical oncology with computational intelligence marks the transition from reactive treatment paradigms to adaptive, precision-guided cancer care.
The next section explores how technological innovations, including multi-omics integration, digital pathology, and radiomics, are generating the data ecosystems that underpin AI-driven advances in cancer diagnosis and therapy.

4. Technological Advancements Reshaping Cancer Therapy

The last decade has witnessed a technological revolution that is fundamentally transforming oncology. The integration of high-throughput molecular profiling, advanced imaging, and computational analytics has enabled a more holistic understanding of cancer biology and therapeutic response. These technologies are generating the data ecosystems that fuel Artificial Intelligence (AI)–driven discoveries and clinical applications.

4.1. Multi-Omics and Systems-Level Understanding

Advances in next-generation sequencing (NGS) and multi-omics platforms—including genomics, transcriptomics, proteomics, metabolomics, and epigenomics—have uncovered the intricate molecular networks driving tumorigenesis [42]. By capturing the complexity of cancer at multiple biological layers, multi-omics approaches enable a systems-level understanding of tumor behavior, resistance, and progression [43]. Integration of these datasets through computational frameworks facilitates the identification of novel biomarkers and therapeutic targets [44].
Single-cell sequencing has further deepened our appreciation of tumor heterogeneity, revealing distinct cellular subpopulations that influence disease evolution and therapeutic resistance [45]. Spatial transcriptomics and multiplexed imaging technologies now allow the mapping of gene expression in situ, preserving the spatial architecture of the tumor microenvironment [46]. These data-rich platforms are critical for training AI models that can learn multidimensional relationships between genotype, phenotype, and clinical outcome.

4.2. Radiomics and Imaging Analytics

Radiomics—the extraction of high-dimensional quantitative features from medical images—has emerged as a powerful approach for linking imaging phenotypes with molecular characteristics [47]. Combined with AI, radiomics enables non-invasive tumor characterization, prediction of treatment response, and detection of minimal residual disease [48]. Deep learning algorithms applied to radiology can surpass human performance in specific diagnostic tasks, reducing inter-observer variability and facilitating earlier detection [49].
Moreover, the fusion of imaging data with genomic and clinical parameters—known as radiogenomics—offers a powerful paradigm for precision oncology [50]. By correlating image-derived signatures with molecular and transcriptomic data, AI-driven radiogenomic models can predict aggressiveness, treatment sensitivity, and patient survival more accurately than traditional metrics.

4.3. Digital Pathology and Computational Histopathology

The digital transformation of pathology is another major technological leap. Whole-slide imaging and high-resolution scanners have enabled the digitization of histopathological slides, creating vast repositories for computational analysis [51]. Deep convolutional neural networks (CNNs) can now automatically identify tumor regions, grade malignancies, and predict molecular alterations directly from histological images [52]. Integration of pathology with genomics—termed pathomics—further enhances diagnostic precision by linking morphological patterns to underlying genetic aberrations [53].Such AI-powered pathology tools not only increase diagnostic efficiency but also reduce human variability, opening the door to real-time decision support in clinical workflows.

4.4. Organoids, CRISPR, and High-Content Screening

Emerging experimental models are revolutionizing functional cancer research. Three-dimensional (3D) organoid cultures derived from patient tumors recapitulate native tissue architecture and heterogeneity, serving as powerful platforms for drug screening and personalized therapy testing [54]. When coupled with CRISPR-based genome editing, these models allow systematic interrogation of gene function and drug resistance mechanisms [55]. High-content imaging and automated phenotypic screening, enhanced by AI-based image analysis, now enable the rapid identification of drug-response signatures and synergistic drug combinations [56].

4.5. The Convergence Toward Data-Driven Oncology

The integration of these diverse technologies—multi-omics, radiomics, pathomics, and functional genomics—creates an unprecedented volume and variety of biological data. However, the complexity and dimensionality of such datasets pose major analytical challenges. Traditional statistical tools fall short in uncovering nonlinear, multivariate relationships among biological systems [57]. Here, AI and machine learning offer transformative potential: enabling the discovery of hidden patterns, predicting clinical outcomes, and informing rational therapeutic design [58].
Collectively, these technological advancements have laid the groundwork for the next phase of precision oncology—one in which AI acts as the analytical bridge between complex biological data and actionable clinical insights. The following sections delve deeper into the role of AI in cancer diagnosis, therapy optimization, and drug discovery.

5. Artificial Intelligence in Cancer Diagnosis

Accurate and early diagnosis remains the cornerstone of effective cancer management. However, conventional diagnostic modalities often face limitations in sensitivity, specificity, and interpretive consistency. The integration of Artificial Intelligence (AI) into diagnostic workflows has ushered in a transformative era, enabling automated, scalable, and highly precise interpretation of complex biomedical data. Through the use of machine learning (ML), deep learning (DL), and computer vision, AI systems can uncover subtle and multidimensional diagnostic cues beyond the perceptual capacity of human experts [59].

5.1. AI in Radiology and Medical Imaging

Medical imaging has been one of the earliest and most successful domains of AI implementation in oncology. Deep learning algorithms, particularly convolutional neural networks (CNNs), have demonstrated expert-level performance in detecting and classifying tumors from radiographic images such as CT, MRI, mammography, and PET [60]. AI-based image analysis allows automated tumor segmentation, lesion characterization, and quantification of radiomic features, significantly improving diagnostic accuracy and reproducibility [61].
For example, AI-assisted mammography systems have achieved diagnostic accuracies comparable to radiologists in breast cancer screening, while reducing false positives and interpretation time [62]. In lung and brain cancers, deep learning models have been employed for early nodule detection, grading, and recurrence prediction [63,64]. Moreover, AI-powered radiogenomic models can integrate imaging phenotypes with molecular data to predict tumor genotype and treatment response non-invasively [65].
Beyond detection, AI is increasingly used in predictive imaging—forecasting disease progression, therapy response, and survival outcomes using longitudinal imaging datasets [66]. This shift from descriptive to predictive imaging marks a significant advancement toward personalized radiology and adaptive clinical decision-making.

5.2. AI in Pathology and Histological Analysis

Pathology has undergone a profound transformation with the advent of digital slide scanning and computational image analysis. Whole-slide imaging (WSI) has enabled the creation of vast digital repositories, providing the foundation for training AI algorithms to recognize morphological patterns associated with cancer [67].
Deep learning–based models can identify tumor regions, quantify mitotic figures, assess tumor–stroma ratio, and even predict molecular alterations such as microsatellite instability or mutational burden directly from hematoxylin and eosin (H&E) images [68,69]. For instance, Campanella et al. demonstrated that deep neural networks trained on gigapixel histopathology slides could achieve near-perfect classification performance across prostate, basal cell, and breast cancers [70].
Furthermore, AI-assisted pathology enhances diagnostic reproducibility by minimizing observer-dependent variability [71]. Integration of histological, genomic, and clinical features through pathomics enables a deeper understanding of tumor biology, facilitating both automated diagnosis and prognostic stratification [72].

5.3. AI in Genomics and Multi-Omics Integration

In the era of precision oncology, genomic and transcriptomic profiling have become integral to diagnosis and treatment selection. However, the scale and complexity of multi-omics data pose significant analytical challenges. AI models—particularly deep neural networks and ensemble learning—have demonstrated exceptional capability in identifying driver mutations, classifying tumor subtypes, and predicting therapeutic targets from raw sequencing data [73,74].
AI algorithms have been employed for variant calling, copy-number analysis, and gene–drug interaction prediction, substantially improving the interpretive efficiency of genomic workflows [75]. Moreover, integrative AI frameworks that combine genomics, transcriptomics, proteomics, and metabolomics data are being developed to construct comprehensive molecular signatures for cancer diagnosis and patient stratification [76].
These approaches not only accelerate biomarker discovery but also bridge the gap between genotype and phenotype—enabling molecular diagnostics that are faster, more accurate, and clinically actionable.

5.4. Clinical Decision Support Systems (CDSS) and Diagnostic AI

AI-driven Clinical Decision Support Systems (CDSS) are increasingly being integrated into clinical practice to aid oncologists in diagnostic and prognostic decisions. Such systems aggregate data from electronic health records (EHRs), imaging, pathology, and genomics to provide real-time diagnostic recommendations [77].
Notable examples include IBM Watson for Oncology, which demonstrated potential for evidence-based treatment matching, and PathAI, a platform improving diagnostic accuracy in digital pathology [78,79]. Although some early commercial tools faced challenges in clinical scalability, ongoing refinements are enhancing their interpretability, transparency, and clinical trustworthiness [80].
Explainable AI (XAI) methods now allow clinicians to visualize decision pathways, improving confidence in algorithmic outputs [81]. As diagnostic AI continues to mature, its seamless integration with clinician expertise is expected to enhance diagnostic precision, reduce time-to-diagnosis, and ultimately improve patient outcomes.

5.5. The Paradigm of AI-Enabled Precision Diagnostics

The collective application of AI across imaging, pathology, and genomics represents a paradigm shift from descriptive diagnostics toward predictive and integrative oncology. By combining multi-modal data, AI enables a more nuanced understanding of tumor heterogeneity, disease trajectory, and therapeutic vulnerability [82].
This convergence lays the groundwork for precision diagnostics—where each patient’s unique biological and clinical profile informs individualized care strategies. As AI-driven models evolve, they promise to transform diagnostic medicine from pattern recognition to holistic, data-integrated prediction.
The next section explores how these diagnostic innovations are being extended into AI-enabled therapeutic development, including drug discovery, treatment optimization, and precision medicine applications.

6. Artificial Intelligence in Cancer Treatment and Drug Discovery

The integration of Artificial Intelligence (AI) into oncology has moved beyond diagnostics to revolutionize therapeutic development, optimization, and personalization. By enabling rapid data analysis, drug response prediction, and treatment planning, AI now supports every stage of cancer therapy—from drug discovery to clinical decision-making. This convergence of computational intelligence and biomedical science is accelerating the shift toward precision oncology, where therapy is tailored to the unique molecular and clinical profile of each patient [83].

6.1. AI in Drug Discovery and Development

Drug discovery is traditionally a time-consuming and costly process, often spanning over a decade and exceeding billions of dollars per successful candidate [84]. AI has emerged as a transformative catalyst in this domain, drastically reducing the time and cost associated with target identification, compound screening, and optimization.
Machine learning algorithms can analyze vast chemical libraries to predict molecular properties, binding affinities, and toxicity profiles [85]. Deep learning models such as generative adversarial networks (GANs) and reinforcement learning (RL) are now capable of de novo molecular design—creating entirely new compounds optimized for potency, selectivity, and pharmacokinetics [86].
Several breakthroughs illustrate this paradigm shift. For example, Insilico Medicine employed generative AI to design a novel fibrosis drug candidate in just 46 days—a process that typically takes years [87]. Similarly, Atomwise’s convolutional neural networks have identified potential small molecules for leukemia and glioblastoma by screening over 10 million compounds in silico [88].
Beyond molecule generation, AI-driven platforms like DeepChem and ChemBERTa use transfer learning to predict drug-likeness and synergy, accelerating preclinical validation [89]. Collectively, these advancements have established AI as a pivotal enabler of rational drug design and repurposing, expediting the translation of computational predictions into clinical therapeutics.

6.2. AI in Precision Oncology and Therapy Response Prediction

Precision oncology relies on matching treatments to the molecular characteristics of a tumor. AI models can integrate genomic, transcriptomic, imaging, and clinical data to predict a patient’s likely response to specific therapies [90].
Supervised learning algorithms have been trained on multi-omics datasets to classify responders versus non-responders to targeted agents such as EGFR inhibitors or immune checkpoint blockade therapies [91]. Deep neural networks can also infer drug sensitivity directly from tumor gene expression or mutational landscapes, outperforming conventional biomarkers in predictive accuracy [92].
In radiation oncology, AI-based dose optimization systems can predict optimal dose distributions and organ-at-risk constraints, improving treatment precision and minimizing toxicity [93]. Similarly, reinforcement learning models are being explored to personalize chemotherapy schedules and adaptive radiotherapy protocols based on patient-specific response patterns [94].

6.3. AI in Immunotherapy and Combination Treatment Design

Immunotherapy represents one of the most promising yet unpredictable cancer treatment modalities. Despite remarkable success in some cancers, response rates remain limited, with many patients exhibiting primary or acquired resistance. AI offers tools to decode the complex tumor–immune interactions underlying this variability [95].
Deep learning models can predict neoantigen immunogenicity, optimize vaccine design, and identify immune checkpoints associated with therapeutic response [96]. For instance, AI-based analysis of tumor-infiltrating lymphocytes (TILs) and spatial immune profiling has been used to stratify responders to PD-1/PD-L1 blockade [97].
Additionally, AI is instrumental in rational combination therapy design. By simulating drug–drug and drug–immune interactions, ML algorithms can propose synergistic regimens that maximize efficacy while minimizing adverse effects [98]. Platforms such as BenevolentAI and DeepMind are pioneering the use of knowledge graphs and reinforcement learning to identify optimal drug combinations for refractory tumors [99].

6.4. AI-Guided Radiotherapy, Surgery, and Treatment Planning

AI is also redefining interventional oncology. In radiotherapy, deep learning models assist in automated contouring, organ segmentation, and dose prediction, significantly improving planning efficiency [100]. AI-based predictive models can forecast radiation-induced toxicity, enabling adaptive treatment plans that balance tumor control with tissue preservation [101].
In surgical oncology, computer vision and robotic-assisted systems powered by AI enhance intraoperative precision. Real-time image-guided navigation and augmented reality overlays help surgeons delineate tumor margins and avoid critical structures [102]. Robotic platforms such as the da Vinci Surgical System are now incorporating AI algorithms for motion optimization and skill assessment [103].
Furthermore, digital twins—virtual patient models integrating biological, imaging, and clinical data—are being developed to simulate treatment outcomes and optimize therapy sequences [104]. These virtual frameworks represent a new frontier in personalized cancer management.

6.5. AI in Clinical Decision Support and Treatment Optimization

AI-enabled Clinical Decision Support Systems (CDSS) synthesize patient-specific data to recommend evidence-based therapies, predict treatment efficacy, and flag potential adverse effects [105]. These systems bridge the gap between genomic insights and clinical actionability, empowering oncologists to make data-driven decisions in real time.
For example, Watson for Oncology demonstrated potential in aligning therapeutic recommendations with clinical guidelines in over 90% of breast and colorectal cancer cases [106]. Similarly, adaptive learning systems continuously update their predictions using post-treatment outcomes, enhancing accuracy and personalization over time [107].
As interpretability remains a key concern, emerging frameworks in explainable AI (XAI) are improving clinical trust by elucidating the rationale behind model outputs [108]. The synergy of AI with clinician expertise heralds a new era of intelligent, evidence-informed oncology care.

6.6. Challenges and Ethical Considerations in AI-Guided Therapy

Despite its transformative potential, several challenges impede the full-scale clinical translation of AI in cancer therapy. These include data heterogeneity, algorithmic bias, lack of transparency, and regulatory hurdles [109]. Models trained on limited or non-representative datasets may underperform across populations, risking inequitable care outcomes [110].
Moreover, integrating AI systems into clinical workflows demands interoperability with existing health information systems, standardized validation protocols, and continuous post-deployment monitoring [111]. Ethical considerations—such as patient consent, data privacy, and algorithm accountability—must be rigorously addressed to ensure responsible and equitable implementation [112].

7. Future Prospects and the Road Ahead

The convergence of Artificial Intelligence (AI) and oncology has already begun to redefine how cancer is detected, classified, and treated. Yet, the true transformative potential of AI in oncology lies ahead, as emerging technologies enable deeper biological insight, cross-modal data integration, and real-time clinical decision-making. The future of AI in cancer care will be shaped by advancements in computational models, ethical frameworks, and interdisciplinary collaboration across biomedical and data sciences [113].

7.1. Toward Multimodal and Integrative AI Systems

A major frontier in AI oncology involves the integration of heterogeneous data streams—radiological, histopathological, genomic, proteomic, and clinical—into multimodal AI frameworks [114]. Unlike traditional single-modality models, these systems can synthesize complementary information to construct a holistic view of tumor biology and patient status.
Recent advances in transformer architectures and cross-attention models have facilitated the fusion of imaging and omics data, enabling predictions of molecular signatures directly from radiological or histological inputs [115]. Initiatives such as The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA) provide vast open datasets that support training of such integrative systems. The future direction points toward real-time clinical models that can continuously learn and adapt, offering predictive insights during diagnosis and therapy.
Ultimately, multimodal AI will transform precision oncology from reactive to anticipatory care—identifying high-risk individuals, predicting relapse, and guiding treatment modification long before clinical manifestation.

7.2. Federated Learning and Data Privacy

While AI’s success depends on large, diverse datasets, concerns over patient privacy and data security often limit data sharing across institutions. Federated learning (FL) offers a compelling solution by enabling collaborative model training without transferring sensitive patient data [116]. Each participating institution trains a model locally, sharing only model parameters—not raw data—with a central aggregator.
Federated frameworks such as TensorFlow Federated and NVIDIA Clara have demonstrated feasibility in multi-center medical imaging and genomics studies, achieving performance comparable to centrally trained models [117]. This paradigm not only preserves privacy but also enhances model generalizability by incorporating diverse patient populations.
The adoption of FL is expected to play a pivotal role in building global, ethically compliant AI ecosystems that balance innovation with patient confidentiality and data sovereignty.

7.3. Explainable, Transparent, and Trustworthy AI

Clinical adoption of AI in oncology requires more than technical accuracy—it demands trust. One of the biggest barriers remains the “black box” nature of deep learning models. Efforts in Explainable AI (XAI) aim to address this by making model predictions interpretable and transparent to clinicians [118].
Techniques such as saliency maps, attention visualization, and surrogate modeling allow users to understand the rationale behind an AI’s decision, facilitating validation and accountability [119]. Moreover, embedding human-in-the-loop paradigms ensures that AI systems augment rather than replace clinical judgment. Regulatory bodies, including the U.S. FDA and European Medicines Agency, are increasingly prioritizing model interpretability and post-deployment auditing as prerequisites for clinical approval [120].
Trustworthy AI is thus not a technical afterthought but a cornerstone for sustainable integration into precision medicine.

7.4. AI-Driven Clinical Trials and Real-World Validation

The next decade will witness a shift from retrospective validation to AI-driven prospective clinical trials. Adaptive trial designs leveraging AI can optimize patient selection, dose escalation, and endpoint determination, improving efficiency and reducing costs [121].
Furthermore, the integration of real-world data (RWD)—from electronic health records, wearable devices, and patient-reported outcomes—will enable continuous learning systems capable of updating models as new evidence emerges [122]. Initiatives like Project Data Sphere and Cancer Moonshot are paving the way for such adaptive infrastructures.
AI’s role in clinical trials will thus extend beyond analysis to dynamic orchestration, bridging the gap between research and bedside implementation.

7.5. Ethical, Regulatory, and Societal Considerations

The rapid deployment of AI in oncology brings forth pressing ethical and regulatory challenges. Ensuring equity, accountability, and transparency in algorithmic decision-making is essential to prevent bias and safeguard patient welfare [123].
Global consensus is emerging around frameworks such as the EU AI Act and the WHO Guidance on Ethics and Governance of AI for Health, which emphasize fairness, explainability, and human oversight [124]. Collaborative efforts among clinicians, ethicists, and data scientists are required to establish ethical benchmarks for data governance, model validation, and post-market surveillance [125].
Beyond regulation, there is a moral imperative to democratize AI access—ensuring that resource-limited regions also benefit from these technological advancements in cancer care.

7.6. The Vision Ahead: Human–AI Synergy in Oncology

The ultimate promise of AI lies not in replacing oncologists but in augmenting human expertise. As computational systems evolve, their ability to process vast datasets and identify hidden patterns will complement the clinician’s contextual understanding and empathy.
Future oncology practice will likely operate through hybrid intelligence, where human judgment and machine precision coexist synergistically [126]. AI-assisted decision-making will become an invisible backbone of cancer care—powering diagnostics, therapy optimization, and population-level prevention strategies.
In the coming decade, the field stands poised to transcend data-driven prediction toward biologically interpretable, ethically aligned, and patient-centered AI. Realizing this vision will require sustained collaboration between academia, industry, and healthcare systems—uniting computational innovation with clinical compassion to usher in a new era of intelligent oncology.

8. Conclusion and Future Outlook

Cancer remains one of the most complex and adaptive diseases known to medicine. Over the past century, our understanding of its molecular foundations has transformed treatment from broadly cytotoxic approaches to targeted and immune-based therapies. Yet, despite these advances, challenges such as therapeutic resistance, tumor heterogeneity, and treatment-induced toxicity continue to limit long-term success.
The emergence of artificial intelligence represents a defining milestone in this ongoing evolution. By harnessing the power of computational learning, AI enables the extraction of clinically meaningful insights from vast and diverse datasets — spanning genomics, radiology, pathology, and patient records. These technologies are rapidly enhancing diagnostic precision, predicting therapeutic responses, and identifying novel drug candidates with unprecedented accuracy and speed.
Beyond its technical capabilities, AI is also reshaping how we conceptualize cancer treatment — moving from a reactive to a predictive and adaptive discipline. Integrating AI with multi-omics profiling, digital pathology, and real-time clinical data promises a future where therapy is not only personalized but dynamically optimized for each patient’s molecular and physiological profile. This vision would ensure that each cancer patient's treatment evolves in response to the tumor's changes, leading to more effective and sustained outcomes.
However, realizing this vision requires careful navigation of ethical, regulatory, and logistical challenges. Transparency, interpretability, and equitable access must remain central to the development and implementation of AI-driven healthcare systems. Ensuring the privacy and security of patient data, addressing biases in AI algorithms, and mitigating disparities in access to advanced treatments will be essential to creating a fair and inclusive healthcare system. The ultimate goal is not to replace human expertise but to amplify it — creating a synergistic partnership between clinicians and intelligent systems that can anticipate disease evolution, refine treatment choices, and improve patient survival.
As we look to the future, the fusion of biological discovery with computational intelligence will redefine the boundaries of oncology. In the next decade, advancements in AI, along with emerging technologies like quantum computing, will accelerate our ability to decode the complexities of cancer even further. By embracing this convergence, we move closer to a future where cancer care is truly data-driven, personalized, and curative rather than palliative — transforming cancer from a formidable adversary into a manageable condition, guided by the precision of artificial intelligence.

Authors Contribution

Conceptualization: VBSK, RP, JM Original manuscript drafting: AKM, SRA Proofread and Edit: AKM

Acknowledgment:

ICMR, Govt. of India
KSCSTE, Govt. of Kerala, India

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide. CA Cancer J Clin.
  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2023. CA Cancer J Clin.
  3. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell.
  4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell.
  5. Hanahan, D. Hallmarks of cancer: new dimensions. Cell.
  6. Sung H, Ferlay J, Siegel RL, et al. CA Cancer J Clin.
  7. Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers.
  8. Miller KD, Nogueira L, Devasia T, et al. CA Cancer J Clin.
  9. Ferlay J, Colombet M, Soerjomataram I, et al. Int J Cancer. 1941.
  10. Vineis P, Wild CP. Global cancer patterns: causes and prevention. Lancet. 9916.
  11. Marquart J, Chen EY, Prasad V. Estimation of the percentage of US patients with cancer who benefit from genome-driven oncology. JAMA Oncol. 1093.
  12. Hanahan D, Weinberg RA. Cell.
  13. Garraway LA, Lander ES. Lessons from the cancer genome. Cell.
  14. Hanahan, D. Cell. 2022;185(2):278–298.
  15. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Med.
  16. Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic. Nat Rev Clin Oncol.
  17. Kinker GS, Greenwald AC, Tal R, et al. Pan-cancer single-cell RNA-seq identifies recurring programs of cellular heterogeneity. Nat Genet.
  18. Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than pictures, they are data. Radiology.
  19. Komura D, Ishikawa S. Machine learning methods for histopathological image analysis. Cancer Sci. 1295.
  20. Wan JC, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer.
  21. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 6478.
  22. Hayes, DF. Biomarker validation and testing. Nat Rev Clin Oncol.
  23. Zou J, Huss M, Abid A, et al. A primer on deep learning in genomics. Nat Rev Genet.
  24. Esteva A, et al. Nat Med.
  25. DeVita VT, Chu E. A history of cancer chemotherapy. Cancer Res. 8643.
  26. Chabner BA, Roberts TG. Chemotherapy and the war on cancer. Nat Rev Cancer.
  27. O’Connor, R. Targeted therapy in cancer—a shift from the ‘one-size-fits-all’ approach. Clin Med Insights Oncol.
  28. Druker BJ, et al. Efficacy and safety of a specific inhibitor of the BCR–ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 1031.
  29. Slamon DJ, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer. N Engl J Med.
  30. Motzer RJ, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 1803.
  31. Robert C, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2521.
  32. Couzin-Frankel, J. Cancer immunotherapy. Science. 6165. [Google Scholar]
  33. Forde PM, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 1976.
  34. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 6382.
  35. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 6230.
  36. Maude SL, et al. Tisagenlecleucel in children with relapsed or refractory acute lymphoblastic leukemia. N Engl J Med.
  37. Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR–Cas9. Science. 6213.
  38. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol. 1974.
  39. Zhang J, et al. Personalized medicine: genetic variations and drug response. Pharmacol Ther.
  40. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution. Cell.
  41. Schork, NJ. Personalized medicine: time for one-person trials. Nature. 7549. [Google Scholar]
  42. Dienstmann R, Rodon J, Barretina J, Tabernero J. Genomic medicine frontier in human solid tumors. Nat Rev Clin Oncol.
  43. Bedard PL, Hansen AR, Ratain MJ, Siu LL. Nat Rev Clin Oncol.
  44. Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 7782.
  45. O’Neil BH, Wallmark JM, Lorente D, et al. Safety and antitumor activity of the anti–PD-L1 antibody durvalumab. J Clin Oncol. 2121.
  46. Mullard, A. FDA approves 100th monoclonal antibody product. Nat Rev Drug Discov.
  47. Subramanian I, Verma S, Kumar S, Jere A, Anamika K. Multi-omics data integration, interpretation, and its application. Bioinform Biol Insights. 1177.
  48. Lander, ES. The arrival of a revolution in cancer therapy. Nature. 7782. [Google Scholar]
  49. LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 7553.
  50. Esteva A, et al. Nature. 7639.
  51. Topol, EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med.
  52. Rajpurkar P, et al. CheXNet: Radiologist-level pneumonia detection on chest X-rays with deep learning. arXiv preprint. arXiv:1711.05225.
  53. Cireşan DC, Giusti A, Gambardella LM, Schmidhuber J. Deep neural networks segment neuronal membranes. NIPS. 2860.
  54. Litjens G, et al. A survey on deep learning in medical image analysis. Med Image Anal.
  55. Thrall JH, et al. Artificial intelligence and machine learning in radiology. Radiology.
  56. Lundervold AS, Lundervold A. An overview of deep learning in medical imaging. Front Public Health.
  57. Hinton, G. Deep learning—a technology with the potential to transform healthcare. Nat Med. 1299. [Google Scholar]
  58. He J, Baxter SL, Xu J, Xu J, Zhou X, Zhang K. The practical implementation of artificial intelligence technologies in medicine. Nat Med.
  59. Erickson BJ, Korfiatis P, Akkus Z, Kline TL. Clin Radiol.
  60. Esteva A, Kuprel B, Novoa RA, et al. Nature. 7639.
  61. Gillies RJ, Kinahan PE, Hricak H. Radiology.
  62. McKinney SM, et al. Nature. 7788.
  63. Ardila D, et al. Nat Med.
  64. Choi Y, et al. Eur Radiol. 1152.
  65. Kickingereder P, et al. Radiology.
  66. Yip SSF, Aerts HJWL. Nat Rev Clin Oncol.
  67. Madabhushi A, Lee G. Annu Rev Biomed Eng.
  68. Lu MY, Chen TY, Williamson DFK, et al. Nat Cancer.
  69. Coudray N, et al. Nat Med. 1559.
  70. Campanella G, et al. Nat Med. 1301.
  71. Bulten W, et al. Lancet Oncol.
  72. Echle A, et al. Nat Rev Clin Oncol.
  73. Libbrecht MW, Noble WS. Nat Rev Genet.
  74. Angermueller C, Pärnamaa T, Parts L, Stegle O. Mol Syst Biol.
  75. Poplin R, et al. Nat Biotechnol.
  76. Wang D, et al. Nat Commun. 2822.
  77. Shortliffe EH, Sepúlveda MJ. JAMA. 2199.
  78. Somashekhar SP, et al. Ann Oncol.
  79. Steiner DF, et al. JAMA Oncol.
  80. Kelly CJ, et al. Nat Med.
  81. Holzinger A, et al. Nat Rev Cancer.
  82. Topol, EJ. Nat Med. 2019;25(1):44–56.
  83. Topol, EJ. Nat Med. 2019;25(1):44–56.
  84. Paul SM, et al. Nat Rev Drug Discov.
  85. Chen H, Engkvist O, Wang Y, Olivecrona M, Blaschke T. Drug Discov Today. 1241.
  86. Zhavoronkov A, et al. Nat Biotechnol. 1038.
  87. Zhavoronkov A, et al. Nat Biotechnol.
  88. Wallach I, Dzamba M, Heifets A. J Chem Inf Model.
  89. Ramsundar B, et al. ACS Cent Sci.
  90. Kourou K, et al. Comput Struct Biotechnol J.
  91. Menden MP, et al. Nature. 7766.
  92. Chiu YC, et al. Brief Bioinform. 2211.
  93. Bibault JE, et al. Lancet Oncol.
  94. Cazaux M, et al. Nat Mach Intell.
  95. Schmidt, C. Nature. 2017;552(7685):S67–S69.
  96. Tran E, Robbins PF, Rosenberg SA. Science. 6366.
  97. Saltz J, et al. Nat Commun. 5456.
  98. Stokes JM, et al. Cell.
  99. Rodríguez-Mier P, et al. Drug Discov Today. 1036.
  100. Men K, et al. Med Phys.
  101. Thompson RF, et al. Int J Radiat Oncol Biol Phys. 1119.
  102. Hashimoto DA, et al. Nat Med. 1459.
  103. Peters BS, et al. BMJ Surg Interv Health Technol. 0000.
  104. Bruynseels K, Santoni de Sio F, van den Hoven J. Front Genet.
  105. Shortliffe EH, Sepúlveda MJ. JAMA. 2199.
  106. Somashekhar SP, et al. Ann Oncol.
  107. Senders JT, et al. Nat Rev Clin Oncol.
  108. Holzinger A, et al. Nat Rev Cancer.
  109. Wiens J, et al. Nat Med.
  110. Parikh RB, Teeple S, Navathe AS. JAMA. 1827.
  111. Liu X, Faes L, Kale AU, et al. Lancet Digit Health.
  112. Vayena E, Blasimme A, Cohen IG. Nat Mach Intell.
  113. Topol, EJ. Nat Med. 2019;25(1):44–56.
  114. Zhou T, Ruan S, Canu S. IEEE Trans Med Imaging. 3431.
  115. Kaul C, et al. Nat Biomed Eng.
  116. Esteva A, et al. Nature. 7787.
  117. Holzinger A, et al. Nat Rev Cancer.
  118. O’Connor JP, Aboagye EO, Adams JE, et al. Nat Rev Clin Oncol.
  119. Jiang F, Jiang Y, Zhi H, et al. J Thorac Dis.
  120. Marcus HJ, Hughes-Hallett A, Marcus AP, et al. Nat Mach Intell.
  121. Rajpurkar P, Chen E, Banerjee O, Topol EJ. Nat Med.
  122. Liu X, Faes L, Kale AU, et al. Lancet Digit Health.
  123. Vellido, A. Front Artif Intell. 2019;2:13.
  124. Wiens J, et al. Nat Med.
  125. Holzinger A, et al. Nat Rev Cancer.
  126. Topol, EJ. Nat Med. 4: 2019;25(1), 2019. [Google Scholar]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated