Submitted:
28 May 2025
Posted:
29 May 2025
You are already at the latest version
Abstract
Keywords:
1. Introduction to Integrating Artificial Intelligence with Genomic and Biomarker Data for Personalized Therapeutics
1.1. Background: The Evolution of Therapeutics
1.2. Problem Statement: Bridging Data Complexity to Actionable Therapeutics
- Data Complexity and Volume: Genomic and multi-omics data are inherently high-dimensional, noisy, and heterogeneous. Integrating these disparate data types (e.g., DNA sequences, RNA expression levels, protein profiles, clinical notes, imaging data) and extracting meaningful, actionable insights is beyond human cognitive capacity.
- Identifying Subtle Patterns: The molecular underpinnings of disease and individual drug responses are often characterized by subtle, complex interactions and non-linear relationships within this vast data. Traditional statistical methods may struggle to identify these nuanced patterns, leading to missed opportunities for therapeutic personalization.
- Predicting Therapeutic Response: Accurately predicting how an individual patient will respond to a specific therapy, or identifying those at high risk for adverse drug reactions, requires sophisticated predictive models that can learn from diverse patient cohorts and integrate multi-modal biological information.
- Translational Gap: A significant gap exists between basic scientific discoveries (often driven by genomic and biomarker research) and their translation into effective clinical treatments. This translational bottleneck is often due to the difficulty in identifying viable drug targets, optimizing lead compounds, and stratifying patients for clinical trials.
- The Imperative for Human Values and Oversight: As AI systems become more sophisticated in guiding therapeutic decisions, concerns arise regarding data privacy, algorithmic bias, transparency, accountability, and the impact on the physician-patient relationship. Without careful integration and human oversight, AI-driven therapeutics risk exacerbating health inequities or eroding trust.
1.3. Proposed Focus/Thesis
1.4. Scope and Objectives
- Define the types of genomic and biomarker data that are most relevant and impactful for personalized therapeutics, and how they contribute to understanding disease and drug response.
- Outline specific AI applications across the entire therapeutic pipeline, demonstrating how AI leverages this data for target identification, drug discovery, patient stratification, treatment selection, and real-time monitoring.
- Identify and critically examine the key ethical challenges inherent in AI-driven personalized therapeutics, including data privacy, algorithmic bias, transparency, accountability, and informed consent.
- Analyze the broader societal implications and challenges, such as issues of equity and access, economic burdens, regulatory gaps, public trust, and workforce transformation.
- Propose concrete strategies and solutions for a human-centric integration, advocating for ethical AI by design, robust data governance, explainable AI, human-AI collaboration models, equitable access policies, and adaptive regulatory frameworks.
1.5. Structure of the Outline
- Chapter 2: Foundational Concepts: Personalized Therapeutics, Genomics, Biomarkers, and AI will define personalized therapeutics and detail the types of genomic and biomarker data, along with key AI paradigms.
- Chapter 3: AI Applications in Personalized Therapeutics Across the Pipeline will elaborate on specific AI applications from target identification to dose optimization.
- Chapter 4: Ethical Challenges in AI-Driven Personalized Therapeutics will delve into specific ethical dilemmas concerning data, algorithms, transparency, consent, and the physician-patient relationship.
- Chapter 5: Societal Challenges and Implications will broaden the discussion to examine the wider societal impacts, including issues of equity, economic considerations, regulatory gaps, public trust, and workforce transformation.
- Chapter 6: Strategies for a Human-Centric Integration will propose concrete strategies and solutions to address the identified challenges.
- Chapter 7: Conclusion will summarize the key findings and provide a final outlook on the future of AI-driven personalized therapeutics.
2. Foundational Concepts: Personalized Therapeutics, Genomics, Biomarkers, and AI
2.1. Defining Personalized Therapeutics
- Maximizing Efficacy: By selecting treatments that are most likely to be effective for a specific patient, based on their molecular profile, thereby improving clinical outcomes.
- Minimizing Adverse Effects: By identifying patients who are genetically predisposed to severe side effects from certain drugs, allowing clinicians to choose safer alternatives.
- Optimizing Dosage: Adjusting drug dosages based on an individual's metabolism, genetic variants, and real-time physiological responses to achieve optimal therapeutic levels.
- Prognostic and Predictive Value: Utilizing individual data to predict disease progression and response to therapy, guiding proactive treatment adjustments.
2.2. Genomic Data in Therapeutics
2.2.1. Germline vs. Somatic Mutations
- Germline Mutations: These are genetic alterations inherited from parents and are present in virtually every cell of the body. They can predispose individuals to certain diseases (e.g., BRCA1/2 mutations for breast cancer) or influence drug metabolism (e.g., CYP450 enzyme variants).
- Somatic Mutations: These are genetic alterations acquired during a person's lifetime and are typically confined to specific cells or tissues (e.g., tumor cells). In oncology, somatic mutations are crucial for identifying actionable drug targets (e.g., EGFR mutations in lung cancer, BRAF mutations in melanoma).
2.2.2. Pharmacogenomics (PGx): Predicting Drug Response Based on Genetic Variants
- Definition: Pharmacogenomics is the study of how an individual's genes affect their response to drugs. It investigates genetic variations that influence drug absorption, distribution, metabolism, and excretion (ADME), as well as drug targets and immune responses.
- Application: PGx data can predict whether a patient will respond to a particular drug, require a higher or lower dose, or be at increased risk of adverse drug reactions. For example, variants in the CYP2D6 gene can affect how quickly antidepressants or opioids are metabolized.
2.2.3. Disease Susceptibility Genes and Prognostic Markers
- Disease Susceptibility: Identifying genetic variants that increase an individual's risk of developing a particular disease (e.g., APOE4 for Alzheimer's disease). While not directly therapeutic, this information can guide preventive strategies.
- Prognostic Markers: Genetic or genomic signatures that predict the likely course or outcome of a disease, independent of treatment. For instance, certain gene expression profiles in cancer can indicate a more aggressive tumor.
2.3. Biomarker Data in Therapeutics
2.3.1. Types of Biomarkers (Molecular, Imaging, Physiological, Clinical)
- Molecular Biomarkers: Measurable molecules (e.g., proteins, metabolites, nucleic acids) that indicate a biological process, pathogenic process, or pharmacologic response to a therapeutic intervention. Examples include PSA for prostate cancer, HER2 expression for breast cancer, or circulating tumor DNA (ctDNA).
- Imaging Biomarkers: Quantifiable features extracted from medical images (e.g., tumor size changes in CT scans, metabolic activity in PET scans) that reflect disease status or treatment response.
- Physiological Biomarkers: Measurements of bodily functions (e.g., blood pressure, heart rate variability, ECG readings) often collected from wearable devices, indicating health status or drug effects.
- Clinical Biomarkers: Observable signs, symptoms, or laboratory test results (e.g., blood glucose levels, kidney function tests) used in routine clinical practice.
2.3.2. Role in Diagnosis, Prognosis, and Predictive Response to Therapy
- Diagnostic: Identifying the presence of a disease (e.g., troponin for heart attack).
- Prognostic: Predicting the likely course or outcome of a disease (e.g., certain gene expression signatures in cancer).
- Predictive: Identifying patients who are most likely to respond to a specific therapy (e.g., PD-L1 expression for immunotherapy).
2.3.3. Multi-Omics Data Integration (Proteomics, Metabolomics, Epigenomics)
- Proteomics: The large-scale study of proteins, providing insights into protein expression levels, modifications, and interactions, which are often direct mediators of disease and drug action.
- Metabolomics: The study of small molecules (metabolites) involved in metabolic pathways, reflecting the physiological state of a cell or organism and its response to environmental factors or drugs.
- Epigenomics: The study of epigenetic modifications (e.g., DNA methylation, histone modification) that influence gene expression without altering the underlying DNA sequence, providing another layer of regulatory information.
- Integration: Combining these “omics” datasets with genomic and clinical data provides a more comprehensive and dynamic understanding of disease biology and individual drug response, enabling more precise therapeutic interventions.
2.4. Key AI Paradigms for Therapeutic Advancement
2.4.1. Machine Learning (Supervised, Unsupervised, Reinforcement Learning)
- Supervised Learning: Training models on labeled datasets (e.g., genomic profiles paired with known drug responses) to predict outcomes. Used for classification (e.g., responder vs. non-responder) and regression (e.g., predicting optimal drug dose).
- Unsupervised Learning: Discovering hidden patterns or structures in unlabeled data. Used for patient stratification (clustering patients into subgroups with similar molecular profiles) or identifying novel biomarkers.
- Reinforcement Learning: Training agents to make sequential decisions in an environment to maximize a reward. Can be applied to optimize drug dosing strategies over time based on patient feedback and biomarker changes.
2.4.2. Deep Learning (CNNs, RNNs, Graph Neural Networks for Molecular Structures)
- Deep Learning: A subset of ML using neural networks with multiple layers, capable of learning complex hierarchical representations directly from raw data.
- Convolutional Neural Networks (CNNs): Excellent for image analysis (e.g., analyzing pathology slides or medical images to identify biomarkers) and sequence data (e.g., identifying patterns in genomic sequences).
- Recurrent Neural Networks (RNNs): Suited for sequential data like time-series patient data (e.g., continuous monitoring from wearables) or long genomic sequences.
- Graph Neural Networks (GNNs): Emerging for analyzing molecular structures (e.g., drug compounds, protein structures) and biological networks (e.g., gene interaction networks), predicting properties and interactions.
2.4.3. Natural Language Processing (NLP) for Unstructured Clinical Data
- NLP: Enables computers to understand, interpret, and generate human language.
- Application: Crucial for extracting valuable, nuanced information from unstructured clinical notes, pathology reports, and scientific literature within EHRs, which often contain critical details about patient history, symptoms, and treatment responses not captured in structured fields.
2.4.4. Causal Inference and Explainable AI (XAI)
- Causal Inference: Techniques that aim to determine cause-and-effect relationships from observational data, rather than just correlations.
- Application: Essential for understanding why a particular genomic variant leads to a specific drug response, or why a drug is effective in a certain patient subgroup, moving beyond mere prediction to mechanistic understanding.
- Explainable AI (XAI): Focuses on making AI models' decisions interpretable and understandable to humans.
- Application: Crucial for building trust among clinicians and patients, enabling clinicians to critically evaluate AI-driven therapeutic recommendations, and facilitating the debugging of AI errors. XAI can highlight which genomic variants or biomarkers were most influential in an AI's treatment recommendation.
3. AI Applications in Personalized Therapeutics Across the Pipeline
3.1. Target Identification and Validation
3.1.1. AI-Driven Analysis of Genomic and Proteomic Data to Identify Novel Drug Targets
- AI Role: AI algorithms, particularly machine learning and deep learning models, can analyze vast datasets of genomic (e.g., gene expression, mutations), proteomic (e.g., protein-protein interactions, protein abundance), and metabolomic data from healthy and diseased individuals. They identify genes or proteins that are differentially expressed, mutated, or dysregulated in disease states, suggesting them as potential therapeutic targets. Graph Neural Networks (GNNs) are increasingly used to model complex biological networks and identify key nodes.
- Impact: Accelerates the identification of novel, disease-relevant targets, moving beyond well-trodden pathways and potentially uncovering entirely new therapeutic avenues.
3.1.2. Predicting Drug-Target Interactions and Binding Affinities
- AI Role: AI models can predict how well a potential drug molecule will bind to a specific protein target. This involves analyzing the chemical structure of compounds and the 3D structure of proteins, using techniques like deep learning for molecular representations.
- Impact: Speeds up the early discovery phase by computationally prioritizing compounds with high predicted binding affinity, reducing the need for extensive wet-lab screening.
3.2. Drug Discovery and Lead Optimization
3.2.1. De Novo Drug Design Using Generative AI (e.g., GANs, VAEs)
- AI Role: Generative AI models, such as Generative Adversarial Networks (GANs) and Variational Autoencoders (VAEs), can learn the chemical space of known drugs and then generate entirely new molecular structures with desired properties (e.g., high binding affinity, low toxicity).
- Impact: Moves beyond screening existing compound libraries to creating novel chemical entities, significantly expanding the potential therapeutic landscape.
3.2.2. Virtual Screening and Molecular Docking
- AI Role: AI algorithms can rapidly screen millions or billions of virtual compounds against a target protein to identify potential hits. Deep learning models can predict molecular properties and interactions much faster than traditional computational chemistry methods.
- Impact: Dramatically reduces the time and cost of lead identification by prioritizing the most promising compounds for experimental validation.
3.2.3. Predicting ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) Properties
- AI Role: Machine learning models are trained on large datasets of compounds with known ADMET profiles to predict these crucial properties for new drug candidates. This helps in identifying compounds that are likely to fail in later stages due to poor pharmacokinetics or toxicity.
- Impact: Improves the success rate of drug candidates by filtering out problematic molecules early in the development pipeline, saving significant resources.
3.3. Drug Repurposing and Combination Therapies
3.3.1. AI-Driven Identification of Existing Drugs for New Indications Based on Molecular Profiles
- AI Role: AI can analyze vast databases of drug-target interactions, disease pathways, genomic signatures, and clinical trial data to identify existing drugs that might be effective for new diseases. For example, an AI might find that a drug approved for an autoimmune disease affects a pathway implicated in a specific cancer subtype.
- Impact: Accelerates the development of new treatments by leveraging drugs with known safety profiles, significantly reducing development time and cost.
3.3.2. Predicting Synergistic Drug Combinations for Complex Diseases
- AI Role: For complex diseases like cancer or neurodegenerative disorders, combination therapies are often more effective. AI can analyze multi-omics data and drug interaction networks to predict synergistic drug combinations that yield better outcomes than individual drugs.
- Impact: Leads to more effective and personalized treatment regimens, especially for diseases that are resistant to monotherapy.
3.4. Patient Stratification and Treatment Selection
3.4.1. AI-Driven Clustering of Patients Based on Multi-Modal Data for Clinical Trial Enrollment
- AI Role: Unsupervised learning algorithms can cluster patients into molecularly distinct subgroups based on their genomic, proteomic, and clinical data. This allows for more precise patient selection for clinical trials, ensuring that participants are more likely to respond to the investigational therapy.
- Impact: Increases the success rate of clinical trials, reduces trial duration, and accelerates regulatory approval for personalized therapies.
3.4.2. Predicting Individual Patient Response to Specific Therapies (e.g., Oncology, Autoimmune Diseases)
- AI Role: Supervised learning models are trained on patient data (genomic variants, biomarker levels, clinical history) and their known responses to various therapies. These models can then predict the likelihood of an individual patient responding to a specific drug or class of drugs.
- Impact: Enables clinicians to select the most effective therapy for a given patient from the outset, avoiding ineffective treatments and their associated side effects and costs. This is particularly transformative in oncology, where targeted therapies are matched to specific tumor mutations.
3.4.3. Identifying Patients at High Risk for Adverse Drug Reactions
- AI Role: By analyzing pharmacogenomic data (e.g., CYP450 variants), clinical history, and comorbidities, AI can predict which patients are at increased risk of experiencing severe adverse drug reactions to specific medications.
- Impact: Enhances patient safety by allowing clinicians to choose alternative drugs or adjust dosages for high-risk individuals, preventing potentially life-threatening side effects.
3.5. Real-time Monitoring and Dose Optimization
3.5.1. AI Analysis of Wearable and Sensor Data for Continuous Patient Monitoring
- AI Role: AI algorithms can continuously analyze physiological data from wearable devices (e.g., heart rate, sleep patterns, activity levels, glucose levels) to monitor a patient's response to therapy and detect early signs of adverse effects or disease progression.
- Impact: Enables proactive intervention, allows for timely adjustments to treatment, and provides a more dynamic and personalized approach to long-term patient management.
3.5.2. Dynamic Dose Adjustment Based on Individual Response and Biomarker Changes
- AI Role: Reinforcement learning or predictive models can recommend dynamic adjustments to drug dosages based on real-time biomarker levels, patient-reported outcomes, and physiological data, aiming to maintain optimal therapeutic levels while minimizing toxicity.
- Impact: Optimizes treatment efficacy and safety over the course of therapy, adapting to individual patient needs as their condition or metabolism changes.
3.6. Clinical Trial Design and Optimization
3.6.1. AI-Driven Patient Recruitment and Retention
- AI Role: AI can analyze EHRs and genomic databases to identify eligible patients for clinical trials more efficiently, matching them to trials based on specific inclusion/exclusion criteria and molecular profiles. It can also predict patient dropout rates.
- Impact: Accelerates patient enrollment, reduces trial costs, and improves the representativeness of trial cohorts.
3.6.2. Predicting Trial Outcomes and Optimizing Trial Parameters
- AI Role: AI models can analyze historical clinical trial data, real-world evidence, and genomic/biomarker data to predict the likelihood of success for a new drug in a clinical trial. They can also suggest optimal trial designs (e.g., sample size, endpoints, duration).
- Impact: Increases the efficiency of drug development by guiding resource allocation to the most promising candidates and optimizing trial designs for better outcomes.
4. Ethical Challenges in AI-Driven Personalized Therapeutics
4.1. Data Privacy and Security
4.1.1. Extreme Sensitivity of Genomic and Health Data
- Challenge: Genomic data is unique in its immutability and its implications not only for the individual but also for their biological relatives. When combined with detailed health records, lifestyle information, and real-time physiological data, it creates an incredibly rich, yet highly sensitive and permanent, digital profile of an individual. The sheer volume and interconnectedness of this data increase the attack surface for cyber threats.
- Impact: A single data breach could expose deeply personal and potentially stigmatizing information (e.g., genetic predisposition to incurable diseases, mental health conditions, family medical history), leading to discrimination in employment, insurance, or social contexts.
4.1.2. Risks of Re-Identification and Data Breaches
- Challenge: Even with sophisticated anonymization or de-identification techniques, the unique nature of genomic data, combined with other demographic or clinical data points, can make re-identification of individuals possible, especially with advanced AI techniques designed for pattern recognition. The more data points collected and linked, the higher the risk.
- Impact: Undermines patient trust and privacy guarantees, potentially deterring individuals from participating in personalized therapeutics initiatives or sharing their data for research, thereby hindering scientific progress.
4.1.3. Consent for Data Use (Broad vs. Specific, Dynamic Consent)
- Challenge: Traditional informed consent models, often designed for specific clinical procedures or research studies, are frequently inadequate for the dynamic, evolving, and broad data use required by AI in personalized therapeutics. Obtaining specific consent for every potential future use of genomic data (e.g., for drug discovery, for new therapeutic predictions) is impractical, while broad consent raises concerns about true informedness and patient autonomy.
- Impact: Patients may not fully understand how their immutable genomic data will be used by AI, leading to a perceived erosion of autonomy and control over their personal information.
- Mitigation: Exploring “dynamic consent” models where patients can actively manage their data sharing preferences over time, or tiered consent approaches that allow for different levels of data access and use (e.g., for direct care, for research, for commercial development).
4.2. Algorithmic Bias and Fairness
4.2.1. Bias in Training Data (Underrepresentation of Diverse Populations)
- Challenge: Historical genomic and clinical datasets often suffer from a lack of diversity, being predominantly composed of data from specific demographic groups (e.g., populations of European descent). If AI models for personalized therapeutics are trained on such skewed data, they may not perform accurately or reliably for underrepresented minority groups.
- Impact: Leads to suboptimal treatment recommendations, missed opportunities for effective therapies, or increased risk of adverse drug reactions for certain populations, thereby exacerbating existing health inequities. For example, a drug response prediction model trained primarily on European genomic data might fail to accurately predict responses in African or Asian populations due to genetic variations not present in the training set.
4.2.2. Disparate Therapeutic Outcomes for Underserved Groups
- Challenge: Even if not intentionally programmed, AI algorithms can inadvertently lead to discriminatory therapeutic outcomes if their predictions disproportionately affect certain groups. For instance, an AI might recommend a less effective or more toxic treatment for a particular racial group if the training data was biased against that group.
- Impact: Undermines the principle of justice and can lead to a two-tiered healthcare system where some groups receive inferior or less personalized care.
4.2.3. Generalizability of Models Across Varied Clinical Contexts
- Challenge: An AI model developed and validated in one clinical setting or population (e.g., a major academic medical center with specific treatment protocols) may perform poorly when deployed in a different context (e.g., a rural clinic, a population with different environmental exposures, or variations in standard of care).
- Impact: Limits the widespread applicability and trustworthiness of AI tools, particularly in global health contexts, and can lead to erroneous therapeutic decisions.
4.3. Transparency, Interpretability, and Accountability
- Challenge: Many powerful AI models make predictions about drug response or optimal treatment paths based on intricate internal computations that are not easily decipherable by humans. Clinicians receive a recommendation but may not understand why the AI arrived at that specific therapeutic choice, especially when it involves complex genomic and biomarker interactions.
- Impact: Hinders clinician trust and adoption, makes it difficult to detect and debug AI errors, and complicates legal and ethical accountability. It also prevents clinicians from exercising their independent judgment and explaining decisions to patients.
4.3.2. Need for Explainable AI (XAI) for Clinical Trust and Legal Liability
- Challenge: For AI to be responsibly integrated into personalized therapeutics, clinicians need explanations that are understandable, relevant, and actionable. XAI aims to provide insights into an AI model's reasoning, highlighting the specific genomic variants, biomarkers, or clinical features that most influenced its therapeutic recommendation.
- Impact: Improves clinician comprehension, facilitates critical evaluation of AI recommendations, and is crucial for building trust. It also provides a basis for legal and ethical accountability, allowing for post-hoc analysis of AI decisions.
4.3.3. Defining Accountability for AI-Related Therapeutic Errors
- Challenge: If an AI-driven personalized therapeutic system recommends a suboptimal treatment or an incorrect dosage that leads to patient harm, determining legal and ethical accountability is profoundly complex. Is it the AI developer, the healthcare institution, the prescribing physician, or the patient who consented to its use? Existing legal frameworks are often ill-equipped to address this.
- Impact: Creates ambiguity in responsibility, potentially leading to legal disputes, hindering the adoption of AI due to liability concerns, and ultimately impacting patient safety.
4.4. Informed Consent and Patient Autonomy
4.4.1. Complexity of Explaining AI-Driven Therapeutic Decisions
- Challenge: Explaining the intricacies of genomic sequencing, multi-omics data analysis, probabilistic AI predictions, and their direct implications for therapeutic choices to patients in an understandable and comprehensive manner is a significant communication challenge. Patients need to grasp why a specific drug is recommended for them based on their unique molecular profile, and the associated risks and benefits.
- Impact: Patients may not fully grasp the implications of their data being used by AI for therapeutic decisions, potentially leading to less than truly informed consent and a feeling of disempowerment.
4.4.2. Understanding Risks/Benefits of Genetic Information in Treatment Choice
- Challenge: Personalized therapeutics often involves generating and interpreting genetic information that can reveal predispositions to future diseases (beyond the current illness) or implications for family members. Patients must understand these broader risks and benefits when consenting to genetic testing for therapeutic guidance.
- Impact: Can lead to anxiety, distress, or unintended consequences if patients are not adequately prepared for or educated about the full scope of genetic insights.
4.4.3. Right to Not Know Genetic Predispositions Affecting Treatment
- Challenge: Some patients may prefer not to know about certain genetic predispositions (e.g., for incurable adult-onset conditions), even if that information could potentially influence future treatment options. AI systems, by their nature, may uncover and utilize such information for their recommendations.
- Impact: Challenges the patient's right to choose what information they receive about their own health, potentially forcing knowledge upon them.
4.5. Physician-Patient Relationship and Clinical Judgment
4.5.1. Impact on Physician Autonomy and Potential for Automation Bias
- Challenge: If AI systems provide strong, data-driven therapeutic recommendations, there is a concern that physicians might become overly reliant on these suggestions, potentially diminishing their own critical thinking, clinical judgment, and ability to synthesize information from diverse sources (including non-quantifiable patient context). This is known as “automation bias.”
- Impact: Could lead to a “deskilling” phenomenon where human expertise in therapeutic decision-making atrophies, or a shift in responsibility from the human clinician to the AI, even if the AI is not legally accountable.
4.5.2. Deskilling of Human Expertise in Therapeutic Decision-Making
- Challenge: As AI automates complex analytical tasks related to genomic and biomarker interpretation for therapeutic selection, there is a risk that human clinicians may gradually lose proficiency in performing intricate diagnostic reasoning or therapeutic planning independently.
- Impact: Could create an over-dependency on AI, making healthcare vulnerable if AI systems fail, are unavailable, or provide erroneous recommendations.
4.5.3. Maintaining Trust and Empathy in AI-Augmented Consultations
- Challenge: The introduction of an AI “third party” into the therapeutic decision-making process can alter communication patterns. Patients may question whether the treatment recommendation is truly from their doctor or a machine. Explaining AI-generated insights effectively and empathetically to patients is crucial for maintaining trust and the humanistic aspect of medicine.
- Impact: Could reduce the personal connection between physician and patient, potentially leading to patient dissatisfaction or resistance to recommended therapies.
5. Societal Challenges and Implications
5.1. Equity and Access to Personalized Therapeutics
5.1.1. High Cost of AI-Driven Diagnostics and Targeted Therapies
- Challenge: The technologies underpinning personalized therapeutics—such as comprehensive genomic sequencing, advanced multi-omics profiling, AI-powered predictive analytics, and the development of highly targeted, often orphan, drugs—can be significantly more expensive than traditional “one-size-fits-all” approaches.
- Impact: Increases overall healthcare expenditures, potentially straining national healthcare budgets and leading to difficult decisions about resource rationing. Without robust public health policies, these innovations may remain inaccessible to large segments of the population.
5.1.2. Socioeconomic Disparities and the Digital Divide
- Challenge: Access to AI-driven personalized therapeutics often requires access to advanced digital infrastructure (reliable internet, specialized computing power, secure data platforms) and digital literacy. Populations in rural areas, low-income communities, or those with limited technological access may be excluded from these advancements.
- Impact: Creates a new form of healthcare inequality, where the benefits of cutting-edge medicine are disproportionately available to affluent and digitally connected segments of society.
5.1.3. Risk of Widening Health Disparities Globally
- Challenge: The implementation of AI-driven personalized therapeutics in high-income countries could inadvertently divert research funding, talent, and resources away from addressing more fundamental public health challenges prevalent in low- and middle-income countries (LMICs). Furthermore, the cost and infrastructure requirements mean LMICs may be left behind.
- Impact: Exacerbates global health disparities, potentially creating a new form of medical colonialism where advanced technologies are concentrated in wealthy nations, while basic healthcare needs remain unmet elsewhere.
5.2. Economic and Healthcare System Burden
5.2.1. Sustainability of Personalized Therapeutic Costs within Healthcare Systems
- Challenge: While personalized therapeutics promises to be more effective, its high upfront costs for diagnostics and targeted drugs pose a significant financial burden on healthcare systems globally. The long-term cost-effectiveness, though potentially positive, is not always immediately apparent or easily quantifiable.
- Impact: Leads to difficult decisions about resource allocation, potential rationing of advanced care, and increased pressure on public and private healthcare budgets.
5.2.2. Reimbursement Models and Insurance Coverage for AI-Driven PM
- Challenge: Existing reimbursement models are often not designed to accommodate the complex, multi-modal, and continuously evolving nature of AI-driven personalized therapeutics. Determining how to value and pay for AI-assisted diagnostic insights, predictive risk assessments, and highly specific personalized treatments is a significant hurdle.
- Impact: Creates uncertainty for healthcare providers and developers, potentially hindering adoption or leading to inequitable access if services are not adequately covered by insurance or public health programs.
5.2.3. Commercialization and Profit Motives vs. Public Health Imperatives
- Challenge: The commercialization of AI-driven personalized therapeutic technologies by private companies introduces profit motives that may sometimes conflict with broader public health goals, such as equitable access, data sharing for research, or affordability. Proprietary algorithms and data sets can create monopolies.
- Impact: Could lead to proprietary data silos, inflated costs for essential services, and a focus on developing therapies for diseases that promise higher financial returns, rather than those with the greatest public health burden or affecting smaller patient populations.
5.3. Regulatory and Governance Gaps
5.3.1. Pace of Innovation vs. Speed of Regulatory Adaptation
- Challenge: AI algorithms are constantly evolving, with new models and updates being deployed frequently. Traditional regulatory approval processes (e.g., for drugs or medical devices) are often slow, static, and designed for fixed products, not continuously learning AI systems.
- Impact: Creates a bottleneck for the deployment of beneficial AI innovations or, conversely, allows unvalidated or risky AI tools to enter the market without adequate oversight.
5.3.2. Validation and Oversight for Continuously Learning AI Therapeutic Tools
- Challenge: Some AI models are designed to continuously learn and adapt as they encounter new data in clinical practice, potentially changing their behavior post-deployment. Regulating such “living” algorithms, whose performance can drift over time, poses a unique challenge to traditional static approval processes.
- Impact: Raises complex questions about when re-approval is needed, how to monitor ongoing performance, and how to ensure safety and efficacy over the long term, particularly for therapeutic recommendations.
5.3.3. International Harmonization of Standards for Global Deployment
- Challenge: Different countries and regions have disparate regulatory requirements and ethical guidelines for AI in healthcare, especially for complex personalized therapeutic applications. This creates barriers to global research collaboration, product development, and market access.
- Impact: Hinders the widespread adoption of beneficial AI-driven personalized therapeutic solutions and increases development costs for companies operating internationally.
5.4. Public Trust and Acceptance
5.4.1. Public Perception and Misinformation About AI in Treatment
- Challenge: Public understanding of AI is often shaped by sensationalized media portrayals or science fiction, leading to unrealistic expectations or unfounded fears about “robot doctors” or AI making life-or-death decisions autonomously. Misinformation about AI's capabilities or risks can spread rapidly.
- Impact: Creates skepticism, resistance to adoption of AI-driven therapies, or unrealistic demands from patients who may not understand AI's limitations.
5.4.2. Concerns About Genetic Discrimination and Data Misuse
- Challenge: Public apprehension exists regarding the potential for genetic information, revealed by personalized therapeutics, to be used for discriminatory purposes (e.g., by employers, insurance companies) or for non-medical surveillance.
- Impact: Undermines willingness to share genomic and health data, which is essential for training robust AI models and advancing personalized therapeutics.
5.4.3. Building and Maintaining Confidence in AI-Driven Therapies
- Challenge: Building public confidence requires transparent communication about AI's benefits and limitations, clear ethical guidelines, demonstrable commitment to privacy and fairness, and a track record of safe and effective deployment.
- Impact: Without sustained public trust, even the most innovative AI solutions may fail to achieve widespread adoption and impact, limiting their potential to improve health outcomes.
5.5. Workforce Transformation and Education
5.5.1. New Skill Requirements for Clinicians and Researchers
- Challenge: Clinicians will need to develop new competencies in data literacy, AI interpretation, genomic counseling, and human-AI collaboration. Researchers will need skills in AI model development, multi-omics data analysis, and ethical considerations.
- Impact: A workforce unprepared for AI integration can lead to suboptimal use of tools, errors, or resistance to adoption, potentially creating a gap between technological capability and clinical readiness.
5.5.2. Adapting Medical and AI Education Curricula
- Challenge: Current medical education curricula are not adequately preparing future physicians for an AI-augmented practice rooted in personalized therapeutics. Similarly, AI education often lacks specific modules on the nuances of healthcare data, ethics, and clinical application.
- Impact: Creates a knowledge gap between emerging technologies and the practical skills required for effective and ethical deployment.
5.5.3. Ethical Training for AI Developers in Therapeutics
- Challenge: AI developers and data scientists working in personalized therapeutics need specific training in medical ethics, patient privacy, the unique sensitivities of genomic data, and the potential for algorithmic bias in life-altering therapeutic contexts, beyond general AI ethics.
- Impact: Lack of such specialized training can lead to the development of AI systems that inadvertently violate ethical principles, exacerbate health disparities, or compromise patient safety.
6. Strategies for a Human-Centric Integration
6.1. Ethical AI by Design
6.1.1. Embedding Ethical Principles into AI Development Lifecycle
- Strategy: Implement “ethics-by-design” principles, ensuring that ethical considerations (e.g., fairness, privacy, transparency, accountability) are integrated at every stage of AI development, from problem definition and data collection to algorithm selection, model training, validation, and deployment. This includes conducting ethical impact assessments.
- Impact: Proactively identifies and mitigates potential ethical harms, reducing the likelihood of unintended negative consequences and building safeguards into the technology itself.
6.1.2. Multi-Stakeholder Co-Creation of Ethical Guidelines
- Strategy: Foster inclusive, ongoing dialogues and collaborations among all relevant stakeholders—including AI developers, clinicians, patients, ethicists, legal experts, and policymakers—to co-create and refine ethical guidelines. This ensures that guidelines are comprehensive, practical, and reflect diverse perspectives.
- Impact: Leads to more robust, widely accepted, and clinically relevant ethical principles that are grounded in real-world experiences and foster shared responsibility.
6.2. Robust Data Governance and Privacy-Preserving AI
6.2.1. Stronger Data Protection Laws and Secure Infrastructure
- Strategy: Enact and rigorously enforce comprehensive data protection laws that specifically address the unique challenges of genomic and multi-omics data in the AI era. Simultaneously, invest in state-of-the-art cybersecurity infrastructure, encryption protocols, and access controls to protect vast repositories of sensitive health data from breaches and unauthorized access.
- Impact: Provides a strong legal and technical foundation for protecting patient privacy and deterring misuse, thereby building trust.
6.2.2. Federated Learning and Homomorphic Encryption for Genomic Data
-
Strategy: Promote and invest in research and implementation of privacy-preserving AI techniques.
- ○
- Federated Learning: Allows AI models to be trained on decentralized, sensitive patient data located at various institutions without the raw data ever leaving its source. Only model updates are shared.
- ○
- Homomorphic Encryption: Enables computation on encrypted data, meaning data can be processed by AI without ever being decrypted, offering maximal privacy.
- Impact: Addresses critical data privacy concerns, enables collaborative AI model development across institutions without centralizing sensitive patient information, and facilitates access to larger, more diverse datasets for robust model training.
6.2.3. Innovative Consent Models (Dynamic, Tiered)
-
Strategy: Implement flexible and granular consent mechanisms that empower patients to have greater control over how their health data is used for AI development and personalized therapeutics.
- ○
- Dynamic Consent: Allows patients to actively manage and update their data sharing preferences over time.
- ○
- Tiered Consent: Offers different levels of data access and use (e.g., for direct care, for specific research, for broad research, for commercial development).
- Impact: Enhances patient autonomy, fosters transparency, and builds trust by providing greater control and understanding of data utilization.
6.3. Prioritizing Explainable AI (XAI) and Interpretability
6.3.1. Research and Development of Clinically Actionable XAI Techniques
- Strategy: Prioritize research and development into XAI techniques that provide understandable, relevant, and actionable insights into AI model reasoning for clinicians. This includes methods like saliency maps (highlighting influential data points), feature importance scores, counterfactual explanations (what would have changed the recommendation), and rule-based explanations.
- Impact: Empowers clinicians to critically evaluate AI therapeutic recommendations, understand underlying molecular rationales, identify potential biases, and communicate effectively with patients, thereby improving adoption and safety.
6.3.2. Standardized Communication of AI Rationale and Confidence
- Strategy: Develop standardized guidelines and user interface designs for clearly communicating AI system capabilities, limitations, the rationale behind its recommendations, and its level of confidence (e.g., probability scores, uncertainty estimates) to both clinicians and patients. Avoid overstating AI's abilities or presenting it as infallible.
- Impact: Sets realistic expectations, prevents over-reliance or automation bias, and fosters informed, shared decision-making.
6.4. Fostering Human-AI Collaboration Models
6.4.1. AI as a Decision Support System, Not a Replacement
- Strategy: Design and implement AI systems explicitly as decision support tools that augment, rather than replace, human clinical judgment. AI should provide insights, flag anomalies, and offer recommendations, with the final therapeutic decision resting with the human clinician.
- Impact: Preserves physician autonomy, maintains ethical accountability, and leverages the unique strengths of both human intuition/empathy and AI's analytical power.
6.4.2. Interactive AI Interfaces for Clinician Exploration
- Strategy: Develop intuitive and interactive AI interfaces that allow clinicians to explore the AI's reasoning, input additional patient information, test “what-if” scenarios, or adjust parameters to see how the AI's recommendations change.
- Impact: Fosters a dynamic partnership, encourages critical thinking, and allows clinicians to integrate AI insights into their holistic understanding of the patient.
6.4.3. Training Programs for Effective Human-AI Teaming
- Strategy: Implement comprehensive training programs for healthcare professionals on how to effectively collaborate with AI systems. This includes understanding AI's strengths and weaknesses, recognizing automation bias, and developing skills in interpreting AI outputs and communicating them to patients.
- Impact: Ensures a prepared workforce that can maximize the benefits of AI while mitigating its risks.
6.5. Ensuring Equitable Access and Addressing Bias
6.5.1. Inclusive Data Collection and Model Validation Across Diverse Populations
- Strategy: Mandate and incentivize the proactive collection of diverse and representative datasets for AI training, actively seeking data from underrepresented racial, ethnic, socioeconomic, and geographic groups. Rigorous multi-site validation studies across varied clinical contexts are essential.
- Impact: Reduces algorithmic bias, improves the generalizability and fairness of AI models, and ensures that personalized therapeutic recommendations are accurate and effective for all populations.
6.5.2. Policy Interventions for Affordability and Resource Allocation
- Strategy: Develop and implement policies that ensure equitable access to AI-driven personalized therapeutic technologies. This could include universal healthcare coverage for PM services, subsidies for low-income patients, tiered pricing models, and strategic investment in infrastructure for underserved areas.
- Impact: Prevents the creation of a two-tiered healthcare system and promotes health equity by making advanced care accessible to all who can benefit.
6.5.3. Continuous Auditing for Algorithmic Fairness
- Strategy: Implement continuous monitoring and auditing mechanisms for algorithmic fairness at every stage of the AI lifecycle, from data acquisition and model development to deployment and post-market surveillance. This involves using fairness metrics and independent audits.
- Impact: Ensures ongoing fairness, allows for timely detection and correction of any emerging biases, and builds sustained trust in AI systems.
6.6. Adaptive Regulatory and Policy Frameworks
6.6.1. Agile Pathways for AI-driven Therapeutics
- Strategy: Develop regulatory pathways that are flexible enough to accommodate the rapid evolution and continuous learning nature of AI models, while still ensuring rigorous validation of safety and efficacy. This might involve adaptive approval processes, real-world evidence integration, and post-market surveillance requirements.
- Impact: Facilitates timely market access for beneficial innovations while maintaining high safety standards and preventing unnecessary delays.
6.6.2. Clear Accountability and Liability Models
- Strategy: Establish explicit legal and ethical accountability frameworks that clearly define responsibilities in cases of AI-related therapeutic errors or harms. This may involve shared liability models, specific guidelines for developers regarding model transparency and robustness, and clear responsibilities for clinicians in their use of AI tools.
- Impact: Provides clarity for all stakeholders, encourages responsible development, and ensures recourse for patients, thereby fostering trust and safe adoption.
6.6.3. International Collaboration for Global Standards
- Strategy: Promote global cooperation and multi-lateral dialogues among regulatory bodies, ethical committees, and industry stakeholders to harmonize standards for AI in personalized therapeutics.
- Impact: Reduces regulatory fragmentation, facilitates cross-border research and development, accelerates the global adoption of safe and effective AI-driven personalized therapeutic solutions, and addresses global health equity.
7. Conclusions
7.1. Recapitulation of AI's Transformative Potential in Personalized Therapeutics
- Accelerating Drug Discovery: Through de novo design, virtual screening, and ADMET prediction, AI streamlines the identification and optimization of novel therapeutic compounds.
- Enhancing Drug Repurposing: AI efficiently identifies new indications for existing drugs and predicts synergistic combination therapies for complex diseases.
- Precise Patient Stratification: AI clusters patients based on multi-modal data, enabling more effective clinical trial enrollment and targeted treatment selection.
- Optimizing Treatment Selection: AI predicts individual patient response to specific therapies and identifies risks for adverse drug reactions, ensuring the right treatment for the right patient.
- Dynamic Therapeutic Management: AI analyzes real-time monitoring data to enable continuous dose optimization and proactive intervention.
7.2. Reiteration of the Urgency to Address Ethical and Societal Challenges
- Data Privacy and Security: The extreme sensitivity of genomic and multi-omics data necessitates robust protection against re-identification and breaches, alongside innovative and transparent consent mechanisms.
- Algorithmic Bias and Fairness: The critical risk of AI models perpetuating or exacerbating existing health inequities through biased training data, leading to disparate therapeutic outcomes for underserved populations, demands constant vigilance and mitigation.
- Transparency, Interpretability, and Accountability: The “black box” problem of complex AI models challenges clinician trust, hinders error analysis, and complicates legal and ethical accountability for AI-generated therapeutic errors.
- Informed Consent and Patient Autonomy: Communicating complex AI-driven insights to patients and ensuring truly informed, uncoerced consent for data use and therapeutic decisions remains a significant hurdle.
- Physician-Patient Relationship: Concerns about deskilling, automation bias, and the potential erosion of trust and empathy in AI-augmented consultations require careful management.
- Equity and Access: The high cost and technological requirements of personalized therapeutics threaten to widen health disparities, creating a two-tiered system where advanced care is only available to a privileged few.
- Economic Implications: Sustainable reimbursement models and balancing commercial interests with public health imperatives are vital for broad adoption.
- Regulatory and Governance Gaps: The need for agile, harmonized, and continuously evolving regulatory frameworks to oversee AI's dynamic nature is paramount.
- Public Trust and Workforce Transformation: Building public confidence and preparing a healthcare workforce equipped with new AI literacy and collaborative skills are essential for successful integration.
7.3. Emphasizing the Indispensable Role of a Human-Centric Framework
- Develop comprehensive ethical frameworks: Integrating “ethics-by-design” principles from the outset of AI development.
- Establish robust and adaptive regulatory policies: Balancing innovation with safety, efficacy, and equity.
- Invest in Explainable AI (XAI) and privacy-preserving technologies: Fostering transparency, interpretability, and protecting sensitive data.
- Promote equitable access: Through policy interventions, inclusive data collection, and affordability initiatives.
- Transform education and training: Preparing both healthcare professionals and AI developers for this new era of human-AI collaboration.
- Foster public dialogue and trust: Through transparent communication and engagement.
7.4. Final Outlook: Towards Responsible, Equitable, and Effective AI-Driven Personalized Therapeutics
References
- Hossan, K. M. R. , Rahman, M. H., & Hossain, M. D. HUMAN-CENTERED AI IN HEALTHCARE: BRIDGING SMART SYSTEMS AND PERSONALIZED MEDICINE FOR COMPASSIONATE CARE.
- Hossain, M. D. , Rahman, M. H., & Hossan, K. M. R. (2025). Artificial Intelligence in healthcare: Transformative applications, ethical challenges, and future directions in medical diagnostics and personalized medicine.
- Kim, J.W.; Khan, A.U.; Banerjee, I. Systematic Review of Hybrid Vision Transformer Architectures for Radiological Image Analysis. J. Imaging Informatics Med. 2025, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Springenberg, M.; Frommholz, A.; Wenzel, M.; Weicken, E.; Ma, J.; Strodthoff, N. From modern CNNs to vision transformers: Assessing the performance, robustness, and classification strategies of deep learning models in histopathology. Med Image Anal. 2023, 87, 102809. [Google Scholar] [CrossRef] [PubMed]
- Atabansi, C.C.; Nie, J.; Liu, H.; Song, Q.; Yan, L.; Zhou, X. A survey of Transformer applications for histopathological image analysis: New developments and future directions. Biomed. Eng. Online 2023, 22, 1–38. [Google Scholar] [CrossRef] [PubMed]
- Sharma, R. R. , Sungheetha, A., Tiwari, M., Pindoo, I. A., Ellappan, V., & Pradeep, G. G. S. (2025, May). Comparative Analysis of Vision Transformer and CNN Architectures in Medical Image Classification. In International Conference on Sustainability Innovation in Computing and Engineering (ICSICE 2024) (pp. 1343-1355). Atlantis Press.
- Patil, P. R. (2025). Deep Learning Revolution in Skin Cancer Diagnosis with Hybrid Transformer-CNN Architectures. Vidhyayana-An International Multidisciplinary Peer-Reviewed E-Journal-ISSN 2454-8596, 10(si4).
- Shobayo, O.; Saatchi, R. Developments in Deep Learning Artificial Neural Network Techniques for Medical Image Analysis and Interpretation. Diagnostics 2025, 15, 1072. [Google Scholar] [CrossRef] [PubMed]
- Karthik, R. , Thalanki, V., & Yadav, P. (2023, December). Deep Learning-Based Histopathological Analysis for Colon Cancer Diagnosis: A Comparative Study of CNN and Transformer Models with Image Preprocessing Techniques. In International Conference on Intelligent Systems Design and Applications (pp. 90-101). Cham: Springer Nature Switzerland.
- Xu, H.; Xu, Q.; Cong, F.; Kang, J.; Han, C.; Liu, Z.; Madabhushi, A.; Lu, C. Vision Transformers for Computational Histopathology. IEEE Rev. Biomed. Eng. 2023, 17, 63–79. [Google Scholar] [CrossRef] [PubMed]
- Singh, S. (2024). Computer-aided diagnosis of thoracic diseases in chest X-rays using hybrid cnn-transformer architecture. arXiv preprint arXiv:2404.11843.
- Fu, B.; Zhang, M.; He, J.; Cao, Y.; Guo, Y.; Wang, R. StoHisNet: A hybrid multi-classification model with CNN and Transformer for gastric pathology images. Comput. Methods Programs Biomed. 2022, 221, 106924. [Google Scholar] [CrossRef] [PubMed]
- Bougourzi, F.; Dornaika, F.; Distante, C.; Taleb-Ahmed, A. D-TrAttUnet: Toward hybrid CNN-transformer architecture for generic and subtle segmentation in medical images. Comput. Biol. Med. 2024, 176, 108590. [Google Scholar] [CrossRef] [PubMed]
- Islam, M. T. , Rahman, M. A., Mazumder, M. T. R., & Shourov, S. H. (2024). COMPARATIVE ANALYSIS OF NEURAL NETWORK ARCHITECTURES FOR MEDICAL IMAGE CLASSIFICATION: EVALUATING PERFORMANCE ACROSS DIVERSE MODELS. American Journal of Advanced Technology and Engineering Solutions, 4(01), 01-42.
- Vanitha, K.; Manimaran, A.; Chokkanathan, K.; Anitha, K.; Mahesh, T.R.; Kumar, V.V.; Vivekananda, G.N. Attention-Based Feature Fusion With External Attention Transformers for Breast Cancer Histopathology Analysis. IEEE Access 2024, 12, 126296–126312. [Google Scholar] [CrossRef]
- Borji, A.; Kronreif, G.; Angermayr, B.; Hatamikia, S. Advanced hybrid deep learning model for enhanced evaluation of osteosarcoma histopathology images. Front. Med. 2025, 12, 1555907. [Google Scholar] [CrossRef] [PubMed]
- Aburass, S.; Dorgham, O.; Al Shaqsi, J.; Abu Rumman, M.; Al-Kadi, O. Vision Transformers in Medical Imaging: a Comprehensive Review of Advancements and Applications Across Multiple Diseases. J. Imaging Informatics Med. 2025. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Yang, S.; Zhang, J.; Wang, M.; Zhang, J.; Yang, W.; Huang, J.; Han, X. Transformer-based unsupervised contrastive learning for histopathological image classification. Med Image Anal. 2022, 81, 102559. [Google Scholar] [CrossRef] [PubMed]
- Xia, K.; Wang, J. Recent advances of Transformers in medical image analysis: A comprehensive review. MedComm – Futur. Med. 2023, 2. [Google Scholar] [CrossRef]
- Gupta, S.; Dubey, A.K.; Singh, R.; Kalra, M.K.; Abraham, A.; Kumari, V.; Laird, J.R.; Al-Maini, M.; Gupta, N.; Singh, I.; et al. Four Transformer-Based Deep Learning Classifiers Embedded with an Attention U-Net-Based Lung Segmenter and Layer-Wise Relevance Propagation-Based Heatmaps for COVID-19 X-ray Scans. Diagnostics 2024, 14, 1534. [Google Scholar] [CrossRef] [PubMed]
- Henry, E. U., Emebob, O., & Omonhinmin, C. A. (2022). Vision transformers in medical imaging: A review. arXiv preprint arXiv:2211.10043.
- Manjunatha, A. , & Mahendra, G. (2024, December). TransNet: A Hybrid Deep Learning Architecture Combining CNNs and Transformers for Enhanced Medical Image Segmentation. In 2024 International Conference on Computing and Intelligent Reality Technologies (ICCIRT) (pp. 221-225). IEEE.
- Reza, S. M. , Hasnath, A. B., Roy, A., Rahman, A., & Faruk, A. B. (2024). Analysis of transformer and CNN based approaches for classifying renal abnormality from image data (Doctoral dissertation, Brac University).
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).