Azza Moustafa Fahmy
Parasitology Department, Theodore Bilharz Research Institute, Cairo, Egypt
Correspondence to: azzafhmy@gmail.com

Additional information
- Ethical approval: N/a
- Consent: N/a
- Funding: No industry funding
- Conflicts of interest: N/a
- Author contribution: Azza Moustafa Fahmy – Conceptualization, Writing – original draft, review and editing
- Guarantor: Azza Moustafa Fahmy
- Provenance and peer-review:
Commissioned and externally peer-reviewed - Data availability statement: N/a
Keywords: Nanocarriers, Liposomal formulations, Enhanced permeability and Retention (epr), Stimuli-responsive systems, Blood-brain barrier.
Peer-review
Received: 18 December 2024
Revised: 19 January 2025
Accepted: 19 January 2025
Published: 31 January 2025
Abstract
With the help of nanotechnology, cancer treatment can be optimized while still keeping the level of side effects low. This includes recent advances that treat cancer on a molecular level, including mechanisms for targeting the delivery of therapeutic substances and, last, new intelligent delivery systems. Active targeting approaches by surface modification have increased tumor cell ingestion and decreased the required dose for treatment. Yet, enhanced permeability and retention, which is its capital effect, is functional as well, but at a very low impact, as studies show only 0.7% of the injected nanoparticles accumulate in the tumors. FDA-approved nanoformulations using liposomes and polymers exhibit significant clinical benefits owing to their improved pharmacokinetics and less toxicity.
Significant obstacles impede market entry, including low production scalability, complex regulatory frameworks, and development costs exceeding $2.5 billion. The existence of biological impediments, notably the blood-brain barrier and intricate tumor microenvironments, exacerbates the challenges of drug administration. The advances in artificial intelligence and smart delivery systems have enhanced the design and efficacy of nanocarriers. Machine learning algorithms can predict the success rate in drug delivery with 92% accuracy when the process is continuously monitored, which enables dynamic treatment adjustment. Along with these, quantum dots and DNA nanostructures represent new technologies offering more targeted and personalized approaches to cancer treatment. However, there are some hurdles in need of overcoming, such as current regulation and manufacturing.
Introduction
One out of six fatalities is attributed to cancer, which is a substantial global health burden. In 2020, there were an estimated 10.3 million deaths and 19.3 million new cases of cancer worldwide; 70% of the deaths occurred in low- and middle-income countries.1 The major factors contributing to this dismal outcome are a lack of early diagnosis methods and a shortage of therapies.2
Current Therapeutic Limitations
Traditional cancer treatments, including radiation and chemotherapy, provide numerous clinical challenges. Neuropathy, myelosuppression, alopecia, nephrotoxicity, and cardiotoxicity are dose-limiting adverse effects stemming from the drugs’ insufficient selectivity for neoplastic cells and the necessity for elevated dosages to attain therapeutic efficacy.3 The inadequate water solubility of numerous chemotherapeutic agents results in formulation challenges and adverse pharmacokinetic properties.4,5
Nanotechnology: An Innovative Approach
By improving therapeutic efficacy and minimizing side effects, nanotechnology has completely changed the way that cancer is treated.6 Various biological interactions are facilitated by the unique physicochemical properties that are imparted by manipulating materials at the nanoscale (1–100 nm).7 Nanocarriers’ substantial surface-area-to-volume ratios and nanoscale dimensions (less than 500 nm) enhance tumor targeting and drug pharmacokinetics.8
Targeting Mechanisms and Advanced Systems
There are two main active methods of focus in the field of cancer nanomedicine. According to Yao et al.,9 passive targeting depends on the enhanced permeability and retention (EPR), which is when nanoparticles accumulate in the tumor tissue because of compromised vasculature and poor lymphatic drainage. Chehelgerdi et al.7 define active targeting as a modification of nanocarrier surfaces through the use of certain ligands that attach to the receptors of cancer cells.
Hybrid Nanocarriers Integrating Passive and Active Targeting Mechanisms
Novel hybrid nanocarriers are suggested as a way to get around the drawbacks of individual targeting techniques. These nanocarriers combine passive targeting through the EPR effect with active targeting through surface modifications. These systems exploit the unique features of tumors, such as leaky vasculature combined with receptor-ligand interactions to improve specificity. For example, dual functional nanoparticles may be constructed with PEGylation to prolong circulation time and folate or peptide ligands for specific attachment to overexpressed tumor receptors.10,11 In addition, pH sensitive coatings or thermoresponsive liposomes such as ThermoDox are also stimuli-responsive materials for controlled drug release, where systemic toxicity is reduced through the release of drugs into cancer tumor sites.12,13 These hybrid approaches represent a hopeful strategy for improving therapeutic efficacy while reducing off-target effects (Figure 1).

The Scope of Review: This paper presents an important review of nanotechnology in targeted cancer therapy. It focuses on current targeting mechanisms, clinical outcomes, and nanocarrier systems. Some elements that the current approach to cancer therapy was addressing, but which nanotechnology does also, are drug resistance, systemic toxicity, and therapeutic efficiency.
Nanocarrier Systems: An effective and successful nanocarrier must (1) avoid the premature leak of therapeutic agents during circulation in the blood under physiological conditions; (2) possess targeting ability to accumulate at the tumor sites to provide a sufficient dose of therapeutic agents and reduce the severe side effects of therapeutic agents on healthy tissues; (3) show biocompatibility and exhibit biodegradability.14
Liposomal Formulations: Recently reported studies have identified that drug delivery is effectively achieved through lipid-bilayer liposomes. Due to their amphiphilic structure, they have the ability to conceal drugs inside them and carry them to the site of need while allowing controlled release.15 By 2023, liposomes were used in 15 FDA-approved medicines, making them the most successful nanocarrier system for treatment.16 They are widely used because of their high flexibility, low immunogenicity, and established efficacy.17 The liposomal formulations authorized by the FDA include Doxil C-130 (non-PEGylated liposomal daunorubicin) for multiple myeloid leukemia and ovarian cancer. These preparations have enhanced pharmacokinetics and reduced cardiotoxicity. They have also improved tumor targeting by the EPR effect. Their design in a lipid bilayer permits controlled drug release at tumor sites while protecting drugs from degradation during circulation.
Limitations:
- Low EPR Efficiency: The average size of liposomes of ~100 nm leads to liposomes being susceptible to hepatic and renal metabolism to excretion outside the body, leading to poor drug delivery efficiency with a total utilization rate of less than 0.7%, resulting in unfavorable treatment outcomes.18
- Limited Tumor Penetration: According to Sharifi et al., liposomes have been found to achieve only partial penetration into tumor cores. Such shallow penetration compromises their therapeutic efficiency.19
- Immunogenicity Risks: After a couple of uses, little to no bio-agents have an effect on the body; this is a side effect of accelerated blood clearance (ABC). A ligand that was once effective has now lost its potency due to ABC. This is an effectiveness risk caused by immunogenicity.20
Proposed Solutions:
- EPR Optimization: Passive targeting combined with nonoperation is one of the CNS treatment techniques. Anti-angiogenic agents are also used to passively target tumor sites and aid in the enhancement of EPR.19,21
- Active Targeting: Functionalize liposomes with ligands for receptor-mediated endocytosis to enhance selective uptake by cancer cells.22
- Stimuli-Responsive Systems: Stimuli-responsive drug delivery systems have been created so as to reduce the negative effects produced by lossy conventional chemotherapy, targeting over a specific range of times or spaces such as the tumor, but there are exogenous and endogenous stimuli used. The responsive nanocarrier utilizes internal biological stimuli such as pH, redox, and enzyme and external stimuli like temperature and ultrasound, to modulate drug release systems and deliver imaging and chemotherapeutic agents to help fight cancer.13 For example, develop thermoresponsive liposomes like ThermoDox that release drugs at specific temperatures to target tumors more precisely while minimizing systemic toxicity.14
Polymeric Nanoparticles
Active substances can be encapsulated within or on the surface of polymeric nanoparticles, which have a diameter ranging from 1 to 1000 nm. They have considerable promise for precise drug delivery in many disorders.23
Types and Characteristics: According to Thepa and Kim (2023), polymeric nanoparticles improve cancer therapy by facilitating regulated release, shielding medications from environmental influences, and increasing bioavailability.24 PEG-poly (lactic acid) formulations like Genexol PM, PEG-poly (aspartic acid) systems like NK105 and NK911, and albumin-based particles like Abraxane25 are the main polymeric nanocarrier systems.26
Clinical Progress: NK105 shows a 90-fold increase in plasma concentration over free paclitaxel. Genexol PM is approved for breast, lung, and ovarian cancers. CRLX101 is in Phase II trials for non-small-cell lung cancer.25 These carriers improve cancer treatment by offering adaptable structures, controlled release, and enhanced targeting while reducing systemic toxicity.
Limitations:
- Biocompatibility Issues: Some of the polymers are capable of inducing immunogenic responses or can exhibit chronic toxic effects, which can be a security problem in the long run.23
- Manufacturing Scalability: Batch reproducibility is often hard to sustain in these types of goods as the synthesis processes get quite complicated.27
- Restricted Tumor Infiltration: Physiological barriers present in the tumor microenvironment (TME) induce limited tumor infiltration similar to liposomes.26
Proposed Solutions:
- Biocompatible Polymers: Biodegradable polymers such as poly (lactic-co-glycolic acid) PLGA should be used to decrease toxicity, but not at the cost of effectiveness.23,28
- Scalable Manufacturing: Micromanufacturing explores the use of microfluidics technology to improve batch consistency on a larger scale.23
- Multifunctional Nanoparticles: Nanoparticles that are multifunctional: Combine nanoparticle-mediated drug delivery with imaging agents in polymeric nanoparticles to monitor the delivery of the drug through the tumor and its response.26
Smart Nanoparticles
Stimuli-Responsive Systems: Smart nanoparticles accurately distribute medications by responding to biological signals or environmental stimuli. By integrating many functionalities, they can enhance therapeutic efficacy while minimizing adverse effects.29
Multifunctional Capabilities: These designs combine diagnostic imaging with therapeutic functions for real-time monitoring and improved targeting mechanisms.30
Advanced Formulations of the Stimuli-Responsive Systems: Stimuli-responsive systems like ThermoDox release drugs at specific temperatures or pH levels to target tumors accurately while minimizing systemic toxicity.16,25 This revised section consolidates overlapping content on FDA approvals and clinical efficacy while maintaining a clear focus on the unique advantages of each nanocarrier system type.
Limitations:
- Complex Design Requirements: The formation of multifunctional smart nanoparticles with exact responsiveness is both expensive and technically demanding.13,29
- Challenges of the TME: The heterogeneous characteristics of the TME hinder activation at the specific site.13,29
- Obstacles to Regulation: The design of smart nanoparticles is more complex than that of simpler systems, complicating the regulatory approval process.31
Proposed Solutions:
- Artificial Intelligence (AI) Integration: AI can optimize nanoparticle design by predicting drug release patterns and improving targeting efficiency, achieving a prediction accuracy of 92%.32
- Microenvironment-Specific Triggers: Enhance targeted delivery by developing nanoparticles that respond to specific tumor characteristics like hypoxia or particular enzymes.22
- Standardized Protocols: Establishing uniform characterization processes for smart nanoparticles can facilitate regulatory pathways (Table 1).33
| Table 1: Comparative analysis of nanocarrier systems in cancer therapy. | |||
| Feature | Liposomes | Polymeric Nanoparticles | Smart Nanoparticles |
| Clinical Approvals | Doxil, Vyxeos | Genexol PM, NK105 | Limited clinical trials |
| Drug Release Mechanism | Passive (EPR effect) | Controlled release | Stimuli-responsive |
| Targeting Capability | Enhanced with PEGylation | Active targeting possible | High precision via stimuli |
| Challenges | Low EPR efficiency; ABC effect | Biocompatibility; scalability | Complex design; regulatory issues |
Fundamental Targeting Mechanisms
Cancer nanomedicine employs two primary targeting approaches: passive and active targeting, each exploiting distinct mechanisms for therapeutic delivery.
Passive Targeting
The EPR impact increases nanoparticle accumulation in tumor tissues by impairing lymphatic outflow and vasculature.10 The distinctive characteristics of tumor vasculature promoted nanoparticle extravasation, including compromised basement membranes and enlarged endothelial gaps (~1 μm).22 Rapid tumor proliferation induces vascular hyperpermeability by the overproduction of mediators, including bradykinin, nitric oxide, and VEGF (Figure 2).34

Clinical Translation and Enhancement
Despite its theoretical potential, EPR-mediated transport yields an accumulation in tumors that is less than double that of normal tissues.35 Tumor heterogeneity and high interstitial fluid pressure prevent core tumor penetration.10 A variety of enhancement strategies have emerged to optimize EPR-based delivery, including pharmacological therapies using vascular modulators, physical enhancement techniques such as heat and sonoporation, and methods for vascular normalization.22
Active Targeting
Active targeting improves passive systems via molecular recognition. Surface-modified nanocarriers with specific targeting moieties are employed to facilitate selective tumor cell adhesion.11 Targeting proteins, peptides, and antibodies against tumor-specific antigens by receptor-mediated endocytosis provides exact cellular interaction (Figure 3).22

Advanced Design Parameters
According to Suk et al.,36 surface engineering standards call for PEGylation for prolonged circulation, neutral surface charge to reduce nonspecific interactions, and exact control of nanocarrier size (10–100 nm) for maximum tumor penetration. Current developments comprise stimuli-responsive drug release procedures and multifunctional stages that participate in imaging and therapeutic functions. AI-driven design optimization has boosted targeting effectiveness through unconventional computational techniques.37 While additional optimization is essential to get past present restrictions, the blend of passive and active targeting mechanisms, assisted by new technology, offers a strategy for refining therapeutic effectiveness in the management of cancer.22
Biological Barriers in Cancer Nanomedicine
The successful delivery of nanotherapeutics encounters multiple physiological obstacles that significantly impact therapeutic efficacy.
Blood-Brain Barrier (BBB)
The delivery of nanotherapeutics for brain malignancies is considerably obstructed by the blood-brain barrier, which inhibits around 98% of small-molecule medications and practically all macromolecular therapies.38 Endothelial cells, active efflux transporters, and transport proteins that govern molecular access from this selective barrier. Brain tumors can compromise the integrity of the BBB, leading to a heterogeneous blood-tumor barrier with variable permeability; yet, substantial delivery problems persist because of nonuniform distribution and elevated interstitial pressure (Figure 4).24

TME
The TME is an intricate and dynamic unit, including stromal cells, immune cells, blood vessels, and extracellular matrix, which is closely related to the incidence and progress of cancers, as well as their treatment.39 The intricate and diverse characteristics of the cancer cell microenvironment present substantial challenges for nanoparticle-mediated drug delivery.40 Because of the uneven patterns of the vascular structure and the permeability of the arteries, the delivery of medication varies across different tumor sites. Within the tumor mass, various forms of extracellular matrix create physical barriers that hinder effective medication delivery.41 Tumor cells in various locations respond biologically to treatment differently due to their distinct microenvironmental contexts.42
Immune System Synergy
Substantial interactions between nanocarriers and the immune system profoundly influence their therapeutic efficacy. According to La-Beck et al.,43 these interactions cause the initiation of complement and the opportunity for false-allergic reactions. While interactions with immune cells in the TME can either enhance or decline therapeutic efficiency, the mononuclear phagocyte system quickly removes nanoparticles from the bloodstream. These immune interactions have an extensive influence on pharmacokinetics, biodistribution, and finally therapeutic benefits.44 These biological difficulties may be overcome thanks to recent improvements in surface modification techniques and nanocarrier design. To recover barrier diffusion while conserving therapeutic efficiency, surface engineering methods make use of specific targeted ligands and stealth features.45 The improvement of these delivery mechanisms requires a comprehensive examination of nanocarrier characteristics and a methodical measurement of their efficacy in biological systems.
Clinical Translation in Cancer Nanomedicine
There are a number of complex problems that must be fixed logically as cancer nanomedicine transfers from lab research to clinical use.
Regulatory Framework
Regulatory challenges stem from the unique physicochemical characteristics of nanomedicine formulations. As per Đorđević et al.,46 the FDA and EMA have stringent regulations for assessing nanoformulations, which include comprehensive physicochemical characterization and expert safety assessments. The frequent failure of appealing laboratory results to yield similar therapeutic efficacy in human subjects exacerbates the gap between preclinical and clinical outcomes, complicating the licensing procedure.47
Patient Selection and Stratification
Advanced methods of patient selection are required since the effectiveness of nanomedicine varies greatly depending on physiological and malignant parameters. Improving patient products now depends on the detection of certain biomarkers for forecasting treatment response.7 This modified approach requires the use of sophisticated analytical tools and a close inspection of the unique patient features that impact the efficiency of treatment.
Considerations in Manufacturing
Development costs can surpass $2.5 billion, and scale-up issues mostly impact complicated formulations that require numerous synthesis processes.48 Maintaining quality consistency in large-scale production is an ongoing challenge, and stability issues across the product lifecycle can undermine both physical integrity and therapeutic efficacy.7
Establishing Alternatives
Recent technological advancements suggest that these translation issues may be resolved. Innovative manufacturing methods and microfluidic technology are anticipated to resolve issues related to scalability and repeatability.7 AI has been integrated into design optimization and quality control measures to transcend the limitations of traditional inventions.30 These are several interrelated issues that will eventually connect lab successes with clinical efficacy if they are resolved by methodical approaches and continuous improvements in the clinical translation of cancer nanomedicine.
Current Challenges in Cancer Nanomedicine
The outcome of treatment is greatly impacted by a number of interrelated obstacles that must be overcome throughout the shift from laboratory development to clinical use.
Manufacturing Complexities
Scalability difficulties and strict chemistry manufacturing and controls supplies are the chief reasons for obstacles related to manufacturing.49 Large-scale manufacturing offerings face repeatability issues between batches, especially for complex structures that need several manufacturing procedures.50 Physical integrity and therapeutic efficiency are both wedged by product stability during the course of its lifecycle.7
Biological and Physiological Barriers
In clinical settings, there are major biological barriers to therapeutic efficacy. Suboptimal tumor penetration is caused by heterogeneous distribution within tumors and increased interstitial pressure.47 The effectiveness of the EPR effect varies depending on the type of cancer; studies show that drug delivery is less than twice as high in these organs as in normal ones.7
Long-Term Biocompatibility of Advanced Nanocarriers
Thorough review protocols on immunogenicity, bioaccumulation, and chronic toxicity should be conducted to ensure that future safety studies can ascertain the long-term safety of advanced nanocarriers. The efficiency of extending circulation time is ascertained by PEGylated nanoparticles, but these nanoparticles have disadvantages like ABC phenomena.7,38 Biodegradable coatings and extended biodistribution research are crucial to address oxidative stress and bioaccumulation in organs such as the liver and spleen, which are caused by metallic nanoparticles, including gold and iron oxide.6,51 Despite being programmable, DNA nanostructures entail chemical alterations to improve stability and reduce immunogenicity.52,53 Preclinical testing approaches, including prolonged in vitro cytotoxicity assays, in vivo biodistribution investigations, and advanced organ-on-a-chip models, are essential for evaluating long-term safety and efficacy. Tang et al. in 2021 and Meng et al. in 2024 demonstrated that researchers can alleviate potential adverse effects and improve the clinical use of nanomedicine by employing these strategies.38,51
Technical and Economic Restraints
Precise regulation of drug release kinetics in vivo is a significant technical challenge.7 The average tumor accumulation of only 0.7% nanoparticles indicates a markedly low targeting efficacy.47 Development costs frequently surpass $2.5 billion due to complex synthesizers, specialized supply requirements, and extensive characterization analysis.48
Scale-up Deliberations
The transition from laboratory to commercial production introduces additional complexities, including:54
- Implementation of GMP-compliant manufacturing methods
- Specialized facility requirements for nanomaterial handling
- Maintenance of consistent quality throughout scale-up.
Because of the need for small-batch production, specific customization, and complex characterization procedures, personalized nanomedicines put additional financial strain on the economy.46 Systematized approaches are vital to addressing these combined issues while preserving treatment success and financial sustainability if cancer nanomedicine is to continue effectively.
Future Perspectives
Incorporating AI and nanotechnology has the perspective of renovating cancer treatment by providing those new ways to adapt treatments.
Advanced Nanocarrier Systems
New designs for nanocarriers are redefining the possibilities of targeted medicine delivery. DNA nanostructures and quantum dots are promising platforms for cancers of the future.52 These new paradigms reduce the adverse effects on surrounding tissue while enhancing the delivery of water-insoluble drugs through programmable construction and precise control over the geometric scale and morphology of the drug complex. It has been largely documented that DNA nanostructures, when conjugated with functional entities such as proteins, peptides, and aptamers that are functional, are less sensitive to dipeptidase and have a longer circulation time in vivo (Figure 5).

Smart Nanoparticles and Stimuli-Responsive Systems
Cancers are being targeted with smart nanoparticles that can respond to the microenvironment, a new avenue in cancer nanomedicine.29 Such intelligent systems achieve spatial and temporal control over the site and time of drug delivery through a variety of stimuli, including pH, temperature, enzymes, magnetic fields, and other factors. For instance, ThermoDox and other thermoresponsive liposomes offer stimulated drug release at certain temperature ranges, which have been designed for the treatment of cancerous sites.12
AI-Enhanced Design and Optimization
Introducing AI informs a revolution in nanoparticle design and effectiveness projection. Targeting efficiency, physicochemical characteristics, and drug interactions with the nanoparticle can all be improved by machine learning flood of big data.44 Aided by AI analysis, the development pipeline is fast, as many disease-specific nanoparticle types are synthesized in a fraction of the time. Remarkably, some models have predicted up to 92% delivery efficiency for predicting models.7
Personalized Nanomedicine
Anyone may now obtain personalized cancer therapies thanks to the merging of AI and nanomedicine research. When applied to patient data, such as genetic information or tumor features, machine learning algorithms can assist in the creation of nanocarriers that are distinct to each individual.55 Overall, this strategy has a good chance of increasing treatment efficacy while decreasing adverse effects and enhancing medication effectiveness.
Multimodal Theranostic Platforms
The notion of advanced nanomedicine that integrates all traditional functions, including diagnosis, imaging, and therapy, into a single entity, is plausible. Nanoparticles engineered with theranostic properties will provide real-time modifications of pharmaceutical protocols and assessments of treatment efficacy.53 These pathways will provide the early detection of cancer biomarkers and the real-time evaluation of therapy efficacy by imaging agents and sensors integrated within nanocarriers.
Concrete Examples and Case Studies in Future Directions
A multitude of case studies illustrates the effective implementation of proposed solutions in both preclinical and clinical environments, hence confirming their practical efficacy. Microfluidic platforms have been employed to tackle manufacturing scale challenges by generating homogeneous, uniformly high-quality PLGA-based nanoparticles.54 Modular manufacturing techniques that combine synthesis purification and quality control have reduced expenses and improved rigidity.56 AI-driven predictive models have expedited regulatory approval processes by simplifying nanoparticle characterization and simulating drug release properties.57 Despite the promising imaging and therapeutic applications of quantum dots in cancer models, new technologies like DNA nanostructures show that tumor-bearing animals have longer circulation durations and less enzymatic breakdown.58 These cases show how cutting-edge methods can improve clinical translation while addressing major obstacles to nanomedicine’s progress.
Challenges and Future Directions
Before these technologies may be used in clinical situations, several problems still need to be fixed, despite the hopeful expansions. Manufacturing scalability, regulatory hurdles, cost reduction, emerging technologies, and multimodal theranostic platforms are important issues that necessitate care.48 Improved nanocarriers’ long-term safety and biocompatibility entail extensive study and clinical testing. Upcoming research endeavors should focus on:
Manufacturing Scalability
The production of nanocarriers for cancer nanomedicine is challenging to scale up while maintaining consistency and quality due to the complexity of formulations like liposomes and polymeric nanoparticles. This aids in waste minimization and enhances overall performance, which are major problems in scaling up.59,60 Furthermore, it has been indicated that modular production systems reduce the complexity of manufacturing liposomal formulations such as Doxil, hence cutting costs by up to 30% in comparison to normal batch production.56,59 Other authors suggested that microfluidics could enhance the percentage of PLGA nanoparticles with the aim of reducing the processing time by 20%.54 Also, modular production systems have been manifested to reduce the total time of liposomal Doxil experiments by nearly 40%.56 As we can see, the aforementioned advancements indicate the pursuit of advanced manufacturing systems for the rapid clinical translation of cancer nanomedicine while being cost- and scale-efficient.
Regulatory Hurdles
Regulatory frameworks for nanomedicine remain complex due to the unique physicochemical properties of nanoparticles and the lack of conventional characterization techniques, which often delay clinical translation. To get around these issues, it has been proposed that standardized methods for describing nanoparticles—such as size distribution, surface charge effect, drug-loading efficiency, and release kinetics—be created. This new guide will speed up the regulatory approval process and ensure consistency across investigations.46 Additionally, AI-driven predictive models may be integrated by modeling nanoparticle activity under various conditions, generating trustworthy data to support regulatory filings. For example, AI models have been used to predict medication release profiles and interactions of nanoparticles with biological systems to expedite the approval process.57,58
Cost and Proposed Solutions for Medical Nanomedicine in the Context of Emerging Healthcare Systems
Because of the complicated synthesis procedures and long clinical trials involved in developing nanomedicine, each formulation might cost over $2.5 billion. One solution that has been suggested is an AI-enhanced quality control system to maximize production by detecting flaws in real time, cutting waste, and guaranteeing constant quality throughout batches. Furthermore, by facilitating the manufacture of nanoparticles on a broad scale with little human involvement, automated manufacturing processes have shown the potential to reduce labor costs and increase efficiency. Microfluidics, for example, are perfect for scalable production because they reduce batch-to-batch variability and provide fine control over nanoparticle size and characteristics.56,59 These advances offer inexpensive ways to get past the financial hindrances that come with developing nanomedicine.
Emerging Technologies in Cancer Treatment
Thanks to their improved targeting capabilities and programmable designs, cutting-edge technologies like DNA nanostructures and quantum dots are completely changing the way that cancer is treated. By improving circulation times and reducing enzymatic degradation, DNA nanostructures with programmable forms, biocompatibility, and functionalization provide for precise control over drug delivery.61 Real-time monitoring of therapy effectiveness and improved treatment accuracy are made possible by the use of quantum dots in improved medication delivery.58,62
Multimodal Theranostic Platforms
The forthcoming individualized cancer treatment lies in the combination of imaging, treatments, and diagnostics on a single platform. Real-time treatment efficacy monitoring is made possible by theranostic nanoparticles, which combine therapeutic and imaging properties. For example, DNA nanostructures and quantum dots are being created for both therapeutic and dual imaging applications, providing improved treatment outcomes and precise tumor targeting.58,63 Furthermore, ThermoDox and other stimuli-responsive systems release medications in reaction to environmental cues like temperature or pH, guaranteeing accurate administration while reducing systemic toxicity.12,64 These developments demonstrate how multimodal theranostic platforms have the potential to transform cancer therapy and diagnosis by facilitating individualized and flexible treatment plans.
Ethical Considerations in AI-Driven Medicine
From predictive diagnosis and treatment plans to individualized care, AI is revolutionizing medicine. Given the significant ethical issues raised by the application of AI in healthcare, the technology must be implemented fairly and responsibly. These include, among other things, responsibility, model interpretability, and data privacy.65
Data Privacy
The dependency on AI systems with vast amounts of sensitive patient data, such as genetic data, imaging tests, and electronic health records, raises concerns over confidentiality violation and illegal access.66 Tang et al.38 found that these dangers may be reduced by using secure storage protocols, multifactor authentication, role-based access restrictions, and advanced encryption techniques. These measures preserve data both during transmission and storage. To this end, federated learning approaches have been developed to train AI systems on distributed datasets without sending sensitive data, hence reducing privacy concerns while keeping performance stable.7 Moreover, for ensuring compliance and protecting patient rights, privacy regulations such as HIPAA (USA) and GDPR (EU) must be followed. Transparency regarding the use of personal information in AI applications forms the very foundation of trust and patient autonomy.67
Model Explainability
Most algorithms in AI applications are “black boxes” that generate results without giving any explanations behind the decisions reached.68 Besides inhibiting the process of regulatory clearance, this lack of transparency erodes trust among patients and practitioners. With the incorporation of XAI frameworks that provide interpretable results using simple decision trees or visualizations, clinicians will have a better understanding of how specific features contribute to a prediction.69 The collaboration of medical experts, ethicists, and AI developers has resolved the challenges in a successful manner. For compliance with GDPR using different datasets, Google Health collaborated with radiologists on the development of an AI model for breast cancer detection that reduced false positives by 5.7% and false negatives by 9.4%.70 To address sections of HIPAA, a collaboration by Mayo Clinic and NVIDIA resorted to federated learning while maintaining patient privacy for medical imaging analysis.71 Continuous auditing of the AI models, which becomes necessary for interpretability and to remove bias, becomes difficult as the datasets evolve.72
Accountability
It is challenging to establish accountability in AI-driven healthcare because so many parties are engaged, including developers, clinicians, and institutions. According to Morley et al., errors in AI recommendations may cause substantial patient harm.73 Legal frameworks need to specify responsibilities to address this issue: developers should stringently test algorithms before their use; physicians must have the final say in decisions; and organizations should ensure that staff working with AI systems are adequately trained. Multidisciplinary experts on ethical oversight committees can review the implications of AI initiatives before deployment.73 In addition, liability insurance for developers and organizations using AI systems may cover anticipated damages due to failure or malfunction.74 Anomaly detection in AI recommendations requires real-time monitoring systems that will aid in taking corrective measures with speed.75
Ethical Challenges and Lessons from AI in Healthcare
The use of AI in health training has brought up several ethical issues despite the great potential for transformation. For example, a widely used algorithm employed by a major US healthcare system systematically overestimated how sick Black patients were compared to White patients with similar conditions because of biased training data, as reported by Obermeyer et al. in 2019.76 Such a case points to the need for interdisciplinary oversight from ethicists and medical experts in creating algorithms. A 2020 partnership between a digital enterprise and a healthcare organization was criticized for poor anonymization of patient data. The partnership brought into focus the need for transparency in the light of legislation on privacy.77 A different incident where an AI system used “irrelevant parts of the image,” such as ruler marks, to diagnose skin lesions in clinical trials provided incorrect diagnoses and, hence, incorrect treatment recommendations in the process.69 These examples further outline that, to do no harm, explainable AI frameworks are a necessity, together with continuous audits by interdisciplinary teams and strong ethical oversight (Figure 6).

Conclusion
Medication delivery methods based on nanoparticles have demonstrated the ability to overcome tumor disease-related medication resistance. The production of stimulus-responsive multifunctional nanocarriers, which may react to biological signals and other environmental stimuli, is what distinguishes this development and enables highly accurate spatiotemporal management of medicine delivery. Basic research into the discovery and clinical translation of nanomedicine has brought significant therapeutic benefits. Economically successful, clinically approved formulations, such as polymeric nanocarriers and liposomal doxorubicin, address some of the major shortcomings of classic chemotherapy. In comparison with conventional drug delivery, these nanoformulations generally show improved accumulation in tumors, reduced toxicity, and improved pharmacokinetics. Despite these successes, several key issues remain unaddressed:
- A very important problem that remains is the scalability of manufacturing at large-scale production while keeping quality consistent, especially for complex nanoformulations.
- The median accumulation of the administered nanoparticles in tumors is approximately 0.7%, which signals continuous failure in targeting efficiency. This, therefore, calls for more targeted surgery and also increased knowledge of tumor biology.
- With innovating nanomedicine technologies surrounded by complex regulatory frameworks that limit their clinical translational possibilities, there is a need for standardized nanoparticle characterization and quality assurance processes.
- Tailored techniques nevertheless raise questions about cost-effectiveness, particularly for bespoke nanomedicines that need complex manufacturing.
The combination of AI, novel delivery methods, and customized treatments can help overcome these obstacles. Some models can now administer essential medications with a 92% probability thanks to AI-optimized design. Advances in the areas of microfluidics and other manufacturing processes ease scalability and reproducibility concerns. Such techniques, when put together with AI-mediated tumor feature analysis, open new avenues toward very personalized cancer treatments. For instance, nanotechnology, in amalgamation with material science, biotechnology, and AI, could bring in new forms of cancer treatment in the future. Efficiency in overcoming the challenges of modernity and using technology for better and more affordable healthcare solutions is of pivotal essence. Patient-centered methodology, real-time monitoring, and versatile theranostic system development are crucial in realizing all the potentials of nanomedicine during its current development.
References
1 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin [Internet]. 2021;71(3):209-49. Available from: https://acsjournals.onlinelibrary.wiley.com/doi/10.3322/caac.21660
https://doi.org/10.3322/caac.21660
2 González-Moles MÁ, Aguilar-Ruiz M, Ramos-García P. Challenges in the early diagnosis of oral cancer, evidence gaps and strategies for improvement: A scoping review of systematic reviews. Cancers (Basel) [Internet]. 2022;14(19):4967. Available from: https://www.mdpi.com/2072-6694/14/19/4967
https://doi.org/10.3390/cancers14194967
3 Altun İ, Sonkaya A. The most common side effects experienced by patients were receiving first cycle of chemotherapy. Iran J Public Health [Internet]. 2018;47(8):1218-9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/30186799
4 Ioele G, Chieffallo M, Occhiuzzi MA, De Luca M, Garofalo A, Ragno G, et al. Anticancer drugs: Recent strategies to improve stability profile, pharmacokinetic and pharmacodynamic properties. Molecules [Internet]. 2022;27(17):5436. Available from: https://www.mdpi.com/1420-3049/27/17/5436
https://doi.org/10.3390/molecules27175436
5 Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release [Internet]. 2010;148(2):135-46. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0168365910007108
https://doi.org/10.1016/j.jconrel.2010.08.027
6 Yu Z, Gao L, Chen K, Zhang W, Zhang Q, Li Q, et al. Nanoparticles: A new approach to upgrade cancer diagnosis and treatment. Nanoscale Res Lett [Internet]. 2021;16(1):88. Available from: https://link.springer.com/10.1186/s11671-021-03489-z
https://doi.org/10.1186/s11671-021-03489-z
7 Chehelgerdi M, Chehelgerdi M, Allela OQB, Pecho RDC, Jayasankar N, Rao DP, et al. Progressing nanotechnology to improve targeted cancer treatment: Overcoming hurdles in its clinical implementation. Mol Cancer [Internet]. 2023;22(1):169. Available from: https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-023-01865-0
https://doi.org/10.1186/s12943-023-01865-0
8 Elumalai K, Srinivasan S, Shanmugam A. Review of the efficacy of nanoparticle-based drug delivery systems for cancer treatment. Biomed Technol [Internet]. 2024;5:109-22. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2949723X23000533
https://doi.org/10.1016/j.bmt.2023.09.001
9 Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, et al. Nanoparticle-based drug delivery in cancer therapy and its role in overcoming drug resistance. Front Mol Biosci [Internet]. 2020;7:193. Available from: https://www.frontiersin.org/article/10.3389/fmolb.2020.00193/full
https://doi.org/10.3389/fmolb.2020.00193
10 Wu J. The enhanced permeability and retention (EPR) effect: The significance of the concept and methods to enhance its application. J Pers Med [Internet]. 2021;11(8):771. Available from: https://www.mdpi.com/2075-4426/11/8/771
https://doi.org/10.3390/jpm11080771
11 Yoo J, Park C, Yi G, Lee D, Koo H. Active targeting strategies using biological ligands for nanoparticle drug delivery systems. Cancers (Basel) [Internet]. 2019;11(5):640. Available from: https://www.mdpi.com/2072-6694/11/5/640
https://doi.org/10.3390/cancers11050640
12 Amin M, Lammers T, ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev [Internet]. 2022;189:114503. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0169409X22003933
https://doi.org/10.1016/j.addr.2022.114503
13 Murugan B, Sagadevan S, Fatimah I, Oh WC, Motalib Hossain MA, Johan MR. Smart stimuli-responsive nanocarriers for the cancer therapy – nanomedicine. Nanotechnol Rev [Internet]. 2021;10(1):933-53. Available from: https://www.degruyter.com/document/doi/10.1515/ntrev-2021-0067/html
https://doi.org/10.1515/ntrev-2021-0067
14 Zhao X, Bai J, Yang W. Stimuli-responsive nanocarriers for therapeutic applications in cancer. Cancer Biol Med [Internet]. 2021;18(2):319-35. Available from: http://www.cancerbiomed.org/lookup/doi/10.20892/j.issn.2095-3941.2020.0496
https://doi.org/10.20892/j.issn.2095-3941.2020.0496
15 Hamad I, Harb AA, Bustanji Y. Liposome-based drug delivery systems in cancer research: An analysis of global landscape efforts and achievements. Pharmaceutics [Internet]. 2024;16(3):400. Available from: https://www.mdpi.com/1999-4923/16/3/400
https://doi.org/10.3390/pharmaceutics16030400
16 Gatto MS, Johnson MP, Najahi-Missaoui W. Targeted liposomal drug delivery: Overview of the current applications and challenges. Life [Internet]. 2024;14(6):672. Available from: https://www.mdpi.com/2075-1729/14/6/672
https://doi.org/10.3390/life14060672
17 Gu W, Andrews GP, Tian Y. Recent clinical successes in liposomal nanomedicines. Int J Drug Discov Pharmacol [Internet]. 2023;52-9. Available from: https://www.sciltp.com/journals/ijddp/2023/1/218
https://doi.org/10.53941/ijddp.0201009
18 Zhang D, Chen L, Zhao Y, Ni H, Quan Q, Ma J, et al. As1411-modified liposomes to enhance drug utilization and augment the anti-tumor efficacy. Cancer Nanotechnol [Internet]. 2024;15(1):25. Available from: https://cancer-nano.biomedcentral.com/articles/10.1186/s12645-024-00262-6
https://doi.org/10.1186/s12645-024-00262-6
19 Sharifi M, Cho WC, Ansariesfahani A, Tarharoudi R, Malekisarvar H, Sari S, et al. An updated review on EPR-based solid tumor targeting nanocarriers for cancer treatment. Cancers (Basel) [Internet]. 2022;14(12):2868. Available from: https://www.mdpi.com/2072-6694/14/12/2868
https://doi.org/10.3390/cancers14122868
20 Moosavian SA, Bianconi V, Pirro M, Sahebkar A. Challenges and pitfalls in the development of liposomal delivery systems for cancer therapy. Semin Cancer Biol [Internet]. 2021;69:337-48. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1044579X19300987
https://doi.org/10.1016/j.semcancer.2019.09.025
21 Park J, Choi Y, Chang H, Um W, Ryu JH, Kwon IC. Alliance with EPR effect: Combined strategies to improve the EPR effect in the tumor microenvironment. Theranostics [Internet]. 2019;9(26):8073-90. Available from: http://www.thno.org/v09p8073.htm
https://doi.org/10.7150/thno.37198
22 Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther [Internet]. 2023;8(1):293. Available from: https://www.nature.com/articles/s41392-023-01536-y
https://doi.org/10.1038/s41392-023-01536-y
23 Zielińska A, Carreiró F, Oliveira AM, Neves A, Pires B, Venkatesh DN, et al. Polymeric nanoparticles: Production, characterization, toxicology and ecotoxicology. Molecules [Internet]. 2020;25(16):3731. Available from: https://www.mdpi.com/1420-3049/25/16/3731
https://doi.org/10.3390/molecules25163731
24 Thapa RK, Kim JO. Nanomedicine-based commercial formulations: Current developments and future prospects. J Pharm Investig [Internet]. 2023;53(1):19-33. Available from: https://link.springer.com/10.1007/s40005-022-00607-6
https://doi.org/10.1007/s40005-022-00607-6
25 Hwang D, Ramsey JD, Kabanov AV. Polymeric micelles for the delivery of poorly soluble drugs: From nanoformulation to clinical approval. Adv Drug Deliv Rev [Internet]. 2020;156:80-118. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0169409X20301332
https://doi.org/10.1016/j.addr.2020.09.009
26 Xiao X, Teng F, Shi C, Chen J, Wu S, Wang B, et al. Polymeric nanoparticles-Promising carriers for cancer therapy. Front Bioeng Biotechnol [Internet]. 2022;10. Available from: https://www.frontiersin.org/articles/10.3389/fbioe.2022.1024143/full
https://doi.org/10.3389/fbioe.2022.1024143
27 Herdiana Y, Wathoni N, Shamsuddin S, Muchtaridi M. Scale-up polymeric-based nanoparticles drug delivery systems: Development and challenges. OpenNano [Internet]. 2022;7:100048. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2352952022000111
https://doi.org/10.1016/j.onano.2022.100048
28 Dhilip Kumar SS, Abrahamse H. Recent advances in the development of biocompatible nanocarriers and their cancer cell targeting efficiency in photodynamic therapy. Front Chem [Internet]. 2022;10. Available from: https://www.frontiersin.org/articles/10.3389/fchem.2022.969809/full
https://doi.org/10.3389/fchem.2022.969809
29 Sun L, Liu H, Ye Y, Lei Y, Islam R, Tan S, et al. Smart nanoparticles for cancer therapy. Signal Transduct Target Ther [Internet]. 2023;8(1):418. Available from: https://www.nature.com/articles/s41392-023-01642-x
https://doi.org/10.1038/s41392-023-01642-x
30 Kemp JA, Kwon YJ. Cancer nanotechnology: Current status and perspectives. Nano Converg [Internet]. 2021;8(1):34. Available from: https://nanoconvergencejournal.springeropen.com/articles/10.1186/s40580-021-00282-7
https://doi.org/10.1186/s40580-021-00282-7
31 Lombardo D, Kiselev MA, Caccamo MT. Smart nanoparticles for drug delivery application: Development of versatile nanocarrier platforms in biotechnology and nanomedicine. J Nanomater [Internet]. 2019;2019:1-26. Available from: https://www.hindawi.com/journals/jnm/2019/3702518/
https://doi.org/10.1155/2019/3702518
32 Wang J, Liu G, Zhou C, Cui X, Wang W, Wang J, et al. Application of artificial intelligence in cancer diagnosis and tumor nanomedicine. Nanoscale [Internet]. 2024;16(30):14213-46. Available from: https://xlink.rsc.org/?DOI=D4NR01832J
https://doi.org/10.1039/D4NR01832J
33 Souto EB, Blanco-Llamero C, Krambeck K, Kiran NS, Yashaswini C, Postwala H, et al. Regulatory insights into nanomedicine and gene vaccine innovation: Safety assessment, challenges, and regulatory perspectives. Acta Biomater [Internet]. 2024;180:1-17. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1742706124001831
https://doi.org/10.1016/j.actbio.2024.04.010
34 Subhan MA, Yalamarty SSK, Filipczak N, Parveen F, Torchilin VP. Recent advances in tumor targeting via EPR effect for cancer treatment. J Pers Med [Internet]. 2021;11(6):571. Available from: https://www.mdpi.com/2075-4426/11/6/571
https://doi.org/10.3390/jpm11060571
35 Nakamura Y, Mochida A, Choyke PL, Kobayashi H. Nanodrug delivery: Is the enhanced permeability and retention effect sufficient for curing cancer? Bioconjug Chem [Internet]. 2016;27(10):2225-38. Available from: https://pubs.acs.org/doi/10.1021/acs.bioconjchem.6b00437
https://doi.org/10.1021/acs.bioconjchem.6b00437
36 Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev [Internet]. 2016;99:28-51. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0169409X15002173
https://doi.org/10.1016/j.addr.2015.09.012
37 Garg P, Singhal G, Kulkarni P, Horne D, Salgia R, Singhal SS. Artificial intelligence-driven computational approaches in the development of anticancer drugs. Cancers (Basel) [Internet]. 2024;16(22):3884. Available from: https://www.mdpi.com/2072-6694/16/22/3884
https://doi.org/10.3390/cancers16223884
38 Tang L, Feng Y, Gao S, Mu Q, Liu C. Nanotherapeutics overcoming the blood-brain barrier for glioblastoma treatment. Front Pharmacol [Internet]. 2021;12:786700. Available from: https://www.frontiersin.org/articles/10.3389/fphar.2021.786700/full
https://doi.org/10.3389/fphar.2021.786700
39 Wang Y, Deng T, Liu X, Fang X, Mo Y, Xie N, et al. Smart nanoplatforms responding to the tumor microenvironment for precise drug delivery in cancer therapy. Int J Nanomedicine [Internet]. 2024;19:6253-77. Available from: https://www.dovepress.com/smart-nanoplatforms-responding-to-the-tumor-microenvironment-for-preci-peer-reviewed-fulltext-article-IJN
https://doi.org/10.2147/IJN.S459710
40 Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B [Internet]. 2020;10(11):2110-24. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2211383520305906
https://doi.org/10.1016/j.apsb.2020.05.008
41 Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, et al. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther [Internet]. 2021;6(1):153. Available from: https://www.nature.com/articles/s41392-021-00544-0
https://doi.org/10.1038/s41392-021-00544-0
42 Wu P, Han J, Gong Y, Liu C, Yu H, Xie N. Nanoparticle-based drug delivery systems targeting tumor microenvironment for cancer immunotherapy resistance: Current advances and applications. Pharmaceutics [Internet]. 2022;14(10):1990. Available from: https://www.mdpi.com/1999-4923/14/10/1990
https://doi.org/10.3390/pharmaceutics14101990
43 La-Beck NM, Islam MR, Markiewski MM. Nanoparticle-induced complement activation: Implications for cancer nanomedicine. Front Immunol [Internet]. 2021;11. Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2020.603039/full
https://doi.org/10.3389/fimmu.2020.603039
44 Fan YN, Zhao G, Zhang Y, Ye QN, Sun YQ, Shen S, et al. Progress in nanoparticle-based regulation of immune cells. Med Rev [Internet]. 2023;3(2):152-79. Available from: https://www.degruyter.com/document/doi/10.1515/mr-2022-0047/html
https://doi.org/10.1515/mr-2022-0047
45 Su S, M Kang P. Recent advances in nanocarrier-assisted therapeutics delivery systems. Pharmaceutics [Internet]. 2020;12(9):837. Available from: http://www.ncbi.nlm.nih.gov/pubmed/32882875
https://doi.org/10.3390/pharmaceutics12090837
46 Đorđević S, Gonzalez MM, Conejos-Sánchez I, Carreira B, Pozzi S, Acúrcio RC, et al. Current hurdles to the translation of nanomedicines from bench to the clinic. Drug Deliv Transl Res [Internet]. 2022;12(3):500-25. Available from: https://link.springer.com/10.1007/s13346-021-01024-2
https://doi.org/10.1007/s13346-021-01024-2
47 Anchordoquy TJ, Barenholz Y, Boraschi D, Chorny M, Decuzzi P, Dobrovolskaia MA, et al. Mechanisms and barriers in cancer nanomedicine: Addressing challenges, looking for solutions. ACS Nano [Internet]. 2017;11(1):12-8. Available from: https://pubs.acs.org/doi/10.1021/acsnano.6b08244
https://doi.org/10.1021/acsnano.6b08244
48 Bhatia SN, Chen X, Dobrovolskaia MA, Lammers T. Cancer nanomedicine. Nat Rev Cancer [Internet]. 2022;22(10):550-6. Available from: https://www.nature.com/articles/s41568-022-00496-9
https://doi.org/10.1038/s41568-022-00496-9
49 Zheng C, Li M, Ding J. Challenges and opportunities of nanomedicines in clinical translation. BIO Integr [Internet]. 2021;2(2). Available from: https://scienceopen.com/hosted-document?doi=10.15212/bioi-2021-0016
https://doi.org/10.15212/bioi-2021-0016
50 Moore S. The power of AI-generated nanoparticles in targeted drug delivery – latest nanotechnology news. Nano Magazine [Internet]. 2023 [cited 2024 Nov 30]. Available from: https://nano-magazine.com/news/2023/7/3/the-power-of-ai-generated-nanoparticles-in-targeted-drug-delivery
51 Meng YQ, Shi YN, Zhu YP, Liu YQ, Gu LW, Liu DD, et al. Recent trends in preparation and biomedical applications of iron oxide nanoparticles. J Nanobiotechnol [Internet]. 2024;22(1):24. Available from: https://jnanobiotechnology.biomedcentral.com/articles/10.1186/s12951-023-02235-0
https://doi.org/10.1186/s12951-023-02235-0
52 Yadav K, Gnanakani SPE, Sahu KK, Veni Chikkula CK, Vaddi PS, Srilakshmi S, et al. Nano revolution of DNA nanostructures redefining cancer therapeutics-A comprehensive review. Int J Biol Macromol [Internet]. 2024;274:133244. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0141813024040492
https://doi.org/10.1016/j.ijbiomac.2024.133244
53 Hosseini SM, Mohammadnejad J, Salamat S, Beiram Zadeh Z, Tanhaei M, Ramakrishna S. Theranostic polymeric nanoparticles as a new approach in cancer therapy and diagnosis: A review. Mater Today Chem [Internet]. 2023;29:101400. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2468519423000277
https://doi.org/10.1016/j.mtchem.2023.101400
54 Operti MC, Bernhardt A, Grimm S, Engel A, Figdor CG, Tagit O. PLGA-based nanomedicines manufacturing: Technologies overview and challenges in industrial scale-up. Int J Pharm [Internet]. 2021;605:120807. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0378517321006128
https://doi.org/10.1016/j.ijpharm.2021.120807
55 Nuhn L. Artificial intelligence assists nanoparticles to enter solid tumours. Nat Nanotechnol [Internet]. 2023;18(6):550-1. Available from: https://www.nature.com/articles/s41565-023-01382-7
https://doi.org/10.1038/s41565-023-01382-7
56 Fabozzi A, Della Sala F, di Gennaro M, Barretta M, Longobardo G, Solimando N, et al. Design of functional nanoparticles by microfluidic platforms as advanced drug delivery systems for cancer therapy. Lab Chip [Internet]. 2023;23(5):1389-409. Available from: https://xlink.rsc.org/?DOI=D2LC00933A
https://doi.org/10.1039/D2LC00933A
57 Gavas S, Quazi S, Karpiński TM. Nanoparticles for cancer therapy: Current progress and challenges. Nanoscale Res Lett [Internet]. 2021;16(1):173. Available from: https://link.springer.com/10.1186/s11671-021-03628-6
https://doi.org/10.1186/s11671-021-03628-6
58 Devi S, Kumar M, Tiwari A, Tiwari V, Kaushik D, Verma R, et al. Quantum dots: An emerging approach for cancer therapy. Front Mater [Internet]. 2022;8. Available from: https://www.frontiersin.org/articles/10.3389/fmats.2021.798440/full
https://doi.org/10.3389/fmats.2021.798440
59 Naghib SM, Mohammad-Jafari K. Microfluidics-mediated liposomal nanoparticles for cancer therapy: Recent developments on advanced devices and technologies. Curr Top Med Chem [Internet]. 2024;24(14):1185-211. Available from: https://www.eurekaselect.com/227510/article
https://doi.org/10.2174/0115680266286460240220073334
60 Bargahi N, Ghasemali S, Jahandar-Lashaki S, Nazari A. Recent advances for cancer detection and treatment by microfluidic technology, review and update. Biol Proced Online [Internet]. 2022;24(1):5. Available from: https://biologicalproceduresonline.biomedcentral.com/articles/10.1186/s12575-022- 00166-y
https://doi.org/10.1186/s12575-022-00166-y
61 Li L, Wang Z, Zhang X, Deng Y, Mu Y, Li J, et al. Recent advances in DNA nanomaterials for cancer diagnosis and treatment. Nanotechnol Rev [Internet]. 2023;12(1). Available from: https://www.degruyter.com/document/doi/10.1515/ntrev-2023-0135/html
https://doi.org/10.1515/ntrev-2023-0135
62 Khan MS, Sheikh A, Abourehab MAS, Gupta N, Kesharwani P. Understanding the theranostic potential of quantum dots in cancer management. Mater Today Commun [Internet]. 2023;36:106424. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2352492823011157
https://doi.org/10.1016/j.mtcomm.2023.106424
63 Sharma A, Vaswani P, Bhatia D. Revolutionizing cancer therapy using tetrahedral DNA nanostructures as intelligent drug delivery systems. Nanoscale Adv [Internet]. 2024;6(15):3714-32. Available from: https://xlink.rsc.org/?DOI=D4NA00145A
https://doi.org/10.1039/D4NA00145A
64 Naik K, Chaudhary S, Ye L, Parmar AS. A strategic review on carbon Quantum dots for cancer-diagnostics and treatment. Front Bioeng Biotechnol [Internet]. 2022;10. Available from: https://www.frontiersin.org/articles/10.3389/fbioe.2022.882100/full
https://doi.org/10.3389/fbioe.2022.882100
65 Harishbhai Tilala M, Kumar Chenchala P, Choppadandi A, Kaur J, Naguri S, Saoji R, et al. Ethical considerations in the use of artificial intelligence and machine learning in health care: A comprehensive review. Cureus [Internet]. 2024. Available from: https://www.cureus.com/articles/259236-ethical-considerations-in-the-use-of-artificial-intelligence-and-machine-learning-in-health-care-a-comprehensive-review
https://doi.org/10.7759/cureus.62443
66 Williamson SM, Prybutok V. Balancing privacy and progress: A review of privacy challenges, systemic oversight, and patient perceptions in AI-driven healthcare. Appl Sci [Internet]. 2024;14(2):675. Available from: https://www.mdpi.com/2076- 3417/14/2/675
https://doi.org/10.3390/app14020675
67 Stroud AM, Minteer SA, Zhu X, Ridgeway JL, Miller JE, Barry BA. Patient information needs for transparent and trustworthy artificial intelligence in healthcare [Internet]. 2024. Available from: http://medrxiv.org/lookup/doi/10.1101/2024.07.02.24309850
https://doi.org/10.1101/2024.07.02.24309850
68 Ghassemi M, Oakden-Rayner L, Beam AL. The false hope of current approaches to explainable artificial intelligence in health care. Lancet Digit Heal [Internet]. 2021;3(11):e745-50. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2589750021002089
https://doi.org/10.1016/S2589-7500(21)00208-9
69 Tjoa E, Guan C. A survey on explainable artificial intelligence (XAI): Toward medical XAI. IEEE Trans Neural Netw Learn Syst [Internet]. 2021;32(11):4793-813. Available from: https://ieeexplore.ieee.org/document/9233366/
https://doi.org/10.1109/TNNLS.2020.3027314
70 McKinney SM, Sieniek M, Godbole V, Godwin J, Antropova N, Ashrafian H, et al. International evaluation of an AI system for breast cancer screening. Nature [Internet]. 2020;577(7788):89-94. Available from: https://www.nature.com/articles/s41586-019-1799-6
https://doi.org/10.1038/s41586-019-1799-6
71 Kaissis GA, Makowski MR, Rückert D, Braren RF. Secure, privacy-preserving and federated machine learning in medical imaging. Nat Mach Intell [Internet]. 2020;2(6):305-11. Available from: https://www.nature.com/articles/s42256-020-0186-1
https://doi.org/10.1038/s42256-020-0186-1
72 Liu X, Glocker B, McCradden MM, Ghassemi M, Denniston AK, Oakden-Rayner L. The medical algorithmic audit. Lancet Digit Heal [Internet]. 2022;4(5):e384-97. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2589750022000036
https://doi.org/10.1016/S2589-7500(22)00003-6
73 Morley J, Machado CCV, Burr C, Cowls J, Joshi I, Taddeo M, et al. The ethics of AI in health care: A mapping review. Soc Sci Med [Internet]. 2020;260:113172. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0277953620303919
https://doi.org/10.1016/j.socscimed.2020.113172
74 Stern AD, Goldfarb A, Minssen T, Price II WN. AI Insurance: How liability insurance can drive the responsible adoption of artificial intelligence in health care. NEJM Catal [Internet]. 2022;3(4). Available from: http://catalyst.nejm.org/doi/10.1056/CAT.21.0242
https://doi.org/10.1056/CAT.21.0242
75 Allen B, Dreyer K, Stibolt R, Agarwal S, Coombs L, Treml C, et al. Evaluation and real-world performance monitoring of artificial intelligence models in clinical practice: Try it, buy it, check it. J Am Coll Radiol [Internet]. 2021;18(11):1489-96. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1546144021007407
https://doi.org/10.1016/j.jacr.2021.08.022
76 Obermeyer Z, Powers B, Vogeli C, Mullainathan S. Dissecting racial bias in an algorithm used to manage the health of populations. Science (80- ) [Internet]. 2019;366(6464):447-53. Available from: https://www.science.org/doi/10.1126/science.aax2342
https://doi.org/10.1126/science.aax2342
77 Powles J, Hodson H. Google DeepMind and healthcare in an age of algorithms. Health Technol (Berl) [Internet]. 2017;7(4):351-67. Available from: http://link.springer.com/10.1007/s12553-017-0179-1
https://doi.org/10.1007/s12553-017-0179-1








