Abstract
Cancer remains a leading cause of morbidity and mortality worldwide despite advances in conventional therapies. Novel drug delivery systems (NDDS) offer promising strategies to enhance therapeutic efficacy and reduce systemic toxicity by targeted delivery of anticancer agents. However, the lack of standardized evaluation methods for these complex systems hampers reproducibility, regulatory approval, and clinical translation. This review focuses on current evaluation methodologies for NDDS in cancer therapy, highlights existing challenges, and proposes the development of standardized protocols to streamline preclinical and clinical assessments. Establishing universally accepted evaluation frameworks will accelerate the translation of innovative nanomedicines from bench to bedside.
Keywords
Novel drug delivery systems, cancer therapy, standardized evaluation, nanomedicine, pharmacokinetics, toxicity, clinical translation
Introduction
Cancer poses a significant global health burden, accounting for millions of deaths annually [1]. Conventional chemotherapy is often limited by poor drug solubility, nonspecific distribution, systemic toxicity, and multidrug resistance. Recent advances in nanotechnology and pharmaceutics have led to the development of novel drug delivery systems (NDDS), including liposomes, polymeric nanoparticles, dendrimers, and extracellular vesicles, which enable targeted and controlled drug release at tumour sites [2,3]. Despite promising preclinical results, many NDDS fail to translate into clinical success, largely due to inconsistencies in their evaluation. Standardization of in vitro, in vivo, and clinical evaluation methods is critical to ensure reliable data, facilitate regulatory approval, and optimize therapeutic outcomes [4,5]. This review aims to comprehensively analyze current evaluation approaches for NDDS in cancer therapy and discuss the need for standardized protocols to promote effective translation.
Overview of Novel Drug Delivery Systems in Cancer Therapy
NDDS are engineered platforms designed to improve the delivery of anticancer drugs by enhancing bioavailability, targeting tumor cells, and minimizing off-target effects [6]. These systems include:
- Liposomes: Phospholipid vesicles capable of encapsulating hydrophilic and hydrophobic drugs, improving solubility and circulation time [2].
- Polymeric nanoparticles: Biodegradable polymers forming nano-sized particles that provide controlled drug release and surface modification for targeting [7].
- Dendrimers: Branched, tree-like macromolecules with multiple functional groups allowing drug conjugation and targeting [8].
- Extracellular vesicles: Natural nanosized vesicles secreted by cells, used as biocompatible drug carriers with inherent targeting properties [9].
These platforms show potential in overcoming biological barriers, enhancing drug accumulation at tumor sites, and reducing systemic toxicity. However, their complexity demands rigorous evaluation to validate efficacy and safety.
Current Evaluation Methods for Novel Drug Delivery Systems (NDDS)
- In Vitro Evaluation Methods
In vitro methods serve as the preliminary step for assessing physicochemical properties, drug release, cellular uptake, and cytotoxicity of NDDS. Reliable in vitro assays are essential for screening potential formulations before animal studies [10].
-
- Physicochemical Characterization: Parameters such as particle size, surface charge (zeta potential), morphology, and stability are routinely measured using dynamic light scattering (DLS), transmission electron microscopy (TEM), and zeta potential analyzers [11]. These characteristics affect biodistribution and cellular interaction, but lack of standardized protocols leads to variability between labs.
- Drug Loading and Encapsulation Efficiency: Drug loading capacity and encapsulation efficiency are typically quantified using UV-Vis spectrophotometry or high- performance liquid chromatography (HPLC) after separation of free drug from loaded NDDS [12]. Discrepancies in sample preparation and assay conditions can influence results.
- Drug Release Kinetics: In vitro drug release profiles simulate how the drug diffuses or is released from the NDDS under physiological or pathological conditions. Techniques include dialysis bag diffusion, Franz diffusion cells, and agitation methods [13]. However, these models often fail to mimic the complex tumor microenvironment, such as pH variations and enzymatic activity.
- Cellular Uptake and Cytotoxicity Assays: Cell culture models employing cancer cell lines are used to evaluate cellular uptake by fluorescence microscopy or flow cytometry and cytotoxicity by MTT, WST-1, or LDH release assays \[14,15]. Inconsistency in cell lines, culture conditions, and assay protocols hampers reproducibility.
- Mechanistic Studies: Advanced studies investigate the pathways of endocytosis, intracellular trafficking, and drug release using confocal microscopy and molecular inhibitors 16]. These provide insight into NDDS behaviour but require standardization to compare results meaningfully.
- In Vivo Evaluation Methods
In vivo studies provide comprehensive data on pharmacokinetics, biodistribution, toxicity, and therapeutic efficacy in animal models, crucial for translational research [17].
-
- Pharmacokinetics and Biodistribution: Drug concentration in plasma and tissues over time is measured by techniques such as liquid chromatography-mass spectrometry (LC- MS) and radiolabelling [18]. Biodistribution studies focus on accumulation in tumors versus healthy organs, which correlates with therapeutic efficacy and side effects.
- Toxicity Assessment: Safety profiles are evaluated through acute and chronic toxicity studies assessing haematology, serum biochemistry, and histopathology [19]. Variations in animal species, doses, and assessment endpoints pose challenges in standardizing toxicity data.
- Therapeutic Efficacy: Efficacy is assessed via tumour volume measurement, survival analysis, and histological examination of tumor tissues [20]. Use of orthotopic and patient-derived xenograft (PDX) models provides more clinically relevant results but is not uniformly practiced.
- Clinical Evaluation and Regulatory Considerations
Few NDDS have progressed to clinical trials, hindered by manufacturing complexity and regulatory uncertainties [21]. Clinical evaluation encompasses safety, pharmacodynamics, and efficacy in humans. Regulatory agencies lack clear, unified guidelines specifically for NDDS, delaying approvals [22]. Collaborative efforts are underway to establish standards for clinical development.
- Challenges in Current Evaluation Methods
- Lack of universally accepted protocols for in vitro and in vivo assays leads to data variability and hinders comparability [10,14].
- In vitro models often fail to replicate the tumor microenvironment, limiting predictive accuracy [13].
- Animal models vary widely in species, tumor types, and evaluation endpoints, complicating extrapolation to humans [17,20].
- Regulatory pathways are not fully defined for complex NDDS, slowing clinical translation [21,22].
DISCUSSION
The advancement of novel drug delivery systems (NDDS) has revolutionized cancer therapy by improving the selective targeting of anticancer agents and minimizing systemic toxicity. This review has underscored the significance of robust and standardized evaluation methods to assess NDDS effectively across physicochemical properties, biological performance, and clinical relevance. The current lack of universally accepted protocols creates significant barriers to reproducibility and comparison of results, delaying the regulatory approval process and subsequent clinical translation [5,10]. This variability in evaluation methodologies hinders cross-study validation and impedes the integration of promising NDDS into mainstream oncology practice. Moreover, traditional in vitro and in vivo models fail to fully replicate the complexity of the tumor microenvironment, including factors such as heterogeneous cellular populations, extracellular matrix components, and dynamic biochemical gradients [13,20]. This discrepancy often leads to overestimation of therapeutic efficacy in preclinical studies, contributing to high attrition rates in clinical trials. The regulatory landscape for NDDS is evolving but remains fragmented, with differing requirements across regions and a lack of specific guidelines tailored to complex nanoscale systems [21,22]. Harmonization of regulatory frameworks is imperative to facilitate smoother approval pathways and foster innovation. Future directions emphasize the need for multi-disciplinary collaboration to develop consensus-driven standardized protocols. Integration of advanced 3D tumour models, microfluidic platforms, and computational simulations could improve predictive accuracy of preclinical assessments [13,16]. Additionally, scalable manufacturing processes and stringent quality control measures must be prioritized to ensure consistency and safety of clinical-grade NDDS. Personalized medicine approaches, incorporating patient-derived models and biomarker-driven targeting, offer promising avenues to tailor NDDS to individual tumor characteristics, potentially enhancing therapeutic outcomes [17]. In conclusion, overcoming the challenges in evaluation standardization is essential to unlock the full potential of NDDS in cancer therapy. Concerted efforts across academia, industry, and regulatory bodies will be pivotal in translating nanomedicine innovations from bench to bedside.
Challenges and Future Directions
- Challenges in Standardizing Evaluation Methods
Despite the advances in NDDS for cancer therapy, several challenges persist in establishing standardized evaluation protocols:
-
- Lack of Universal Guidelines: Currently, there is no consensus on standardized methods for physicochemical characterization, drug release, and biological evaluation, leading to inter-laboratory variability [10,14].
- Complexity of NDDS: The diverse compositions and functionalities of NDDS require tailored evaluation approaches, complicating the development of one-size-fits-all standards [23].
- Inadequate Tumour Models: Traditional 2D cell cultures and animal models often fail to mimic the tumor microenvironment accurately, limiting predictive value [13,20]. 3D spheroids and patient-derived xenografts are promising but not yet widely standardized.
- Regulatory Hurdles: Regulatory frameworks lag behind scientific advances in NDDS, with unclear pathways for clinical approval and quality control [21,22].
- Reproducibility Issues: Variability in materials, preparation methods, and assay conditions affects reproducibility of evaluation data, impeding clinical translation [5,10].
FUTURE DIRECTIONS
To overcome these challenges and accelerate clinical translation, the following strategies are recommended:
-
- Development of Consensus Protocols: Collaborative efforts by researchers, industry, and regulatory bodies to establish standardized, validated evaluation protocols for physicochemical and biological assays [5,10].
- Advanced In Vitro Models: Adoption of 3D cell culture systems, organoids, and microfluidic tumor-on-a-chip platforms to better simulate tumor physiology and predict in vivo behavior [13].
- Integrated Multi-Modal Evaluation: Combining physicochemical, biological, and computational modeling approaches to provide comprehensive assessment of NDDS [16].
- Regulatory Harmonization: Clear guidelines and frameworks specific to NDDS, developed through international cooperation, to streamline approval processes [21].
- Focus on Scalability and Quality Control: Emphasis on manufacturing consistency, reproducibility, and robust quality control methods for clinical-grade NDDS [18].
- Personalized Medicine Approaches: Leveraging biomarker-guided targeting and patient-specific tumor models to optimize NDDS design and evaluation [17].
CONCLUSION
Novel drug delivery systems represent a transformative approach to cancer therapy by enhancing targeted drug delivery and minimizing systemic toxicity. However, the lack of standardized, reproducible evaluation methods remains a significant barrier to clinical translation. This review highlights the need for development and adoption of universally accepted evaluation protocols encompassing physicochemical characterization, in vitro and in vivo assessments, and regulatory considerations. Future research should focus on innovative in vitro models, regulatory harmonization, and scalable manufacturing to realize the full potential of NDDS in cancer treatment.
REFERENCES
-
-
-
- Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7- 30.
- Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine. 2015; 10:975-999.
- Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751-760.
- Parveen S, Misra R, Sahoo SK. Nanoparticles: a boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine. 2012;8(2):147-166.
- Nobs L, Buchegger F, Gurny R, Allémann E. Current methods for attaching targeting ligands to liposomes and nanoparticles. J Pharm Sci. 2004;93(8):1980-1992.
- Wagner V, Dullaart A, Bock A-K, Zweck A. The emerging nanomedicine landscapes. Nat Biotechnol. 2006;24(10):1211-1217.
- Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric nanoparticles-based drug delivery systems. Colloids Surf B Biointerfaces. 2010;75(1):1-18.
- Caminade AM. Dendrimers as drug delivery agents: pharmacological and toxicological aspects. Drug Discov Today. 2015;20(5):536-541.
- Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm Sin B. 2016;6(4):287-296.
- Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17(1):20-37.
- Bhattacharjee S. DLS and zeta potential – What they are and what they are not? J Control Release. 2016; 235:337-351.
- Li M, Al-Jamal WT, Kostarelos K, Reineke J. Physiologically based pharmacokinetic modeling of nanoparticles. ACS Nano. 2010;4(11):6303-6317.
- Friedrich J, Seidel C, Ebner R, Kunz-Schughart LA. Spheroid-based drug screen: considerations and practical approach. Nat Protoc. 2009;4(3):309-324.
- Liu Y, Miyoshi H, Nakamura M. Nanomedicine for drug delivery and imaging: a promising avenue for cancer therapy and diagnosis using targeted functional nanoparticles. Int J Cancer. 2007;120(12):2527-2537.
- Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1-2):55-63.
- Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33(9):941-951.
- Alexis F, Pridgen E, Molnar LK, Farokhzad OC. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm. 2008;5(4):505-515.
- Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem Rev. 2016;116(4):2602-2663.
- Mitragotri S, Anderson DG, Chen X, et al. Accelerating the translation of nanomaterials in biomedicine. ACS Nano. 2015;9(7):6644-6654.
- Xu X, Ho W, Zhang X, Bertrand N, Farokhzad O. Cancer nanomedicine: from targeted delivery to combination therapy. Trends Mol Med. 2015;21(4):223-232.
- Etheridge ML, Campbell SA, Erdman AG, et al. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomedicine. 2013;9(1):1-14.
- European Medicines Agency. Reflection paper on nanotechnology-based medicinal products for human use. EMA/CHMP/806058/2011. 2011.
- Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm Sin B. 2016;6(4):287-296.