ISSN: 2167-065X

Clinical Pharmacology & Biopharmaceutics
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Editorial   
  • Clin Pharmacol Biopharm, Vol 13(11)

Nanomedicine in Oncology: Targeted Drug Delivery Systems for Precision Cancer Treatment

Diana Bolton*
Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Austria
*Corresponding Author: Diana Bolton, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Austria, Email: dianabolton1234@gmail.com

Received: 01-Nov-2024 / Manuscript No. cpb-24-155465 / Editor assigned: 05-Nov-2024 / PreQC No. cpb-24-155465 / Reviewed: 15-Nov-2024 / QC No. cpb-24-155465 / Revised: 25-Nov-2024 / Manuscript No. cpb-24-155465 / Published Date: 29-Nov-2024

Abstract

Nanomedicine has emerged as a transformative approach in oncology, enabling precise drug delivery and enhancing the therapeutic potential of cancer treatments. Targeted drug delivery systems (TDDS) in nanomedicine are designed to improve the bioavailability and therapeutic index of anti-cancer agents while minimizing off-target effects. These systems utilize nanoparticles, nanocarriers, and molecular targeting strategies to selectively deliver drugs to tumor cells, ensuring that high concentrations of therapeutic agents reach the site of action. This approach aims to overcome the limitations of conventional chemotherapy, such as poor solubility, systemic toxicity, and drug resistance. The development of advanced nanomaterials, such as liposomes, dendrimers, micelles, and nanorods, has enabled the creation of more effective and personalized treatment regimens. Additionally, the integration of targeting ligands, such as antibodies and peptides, further enhances the specificity and efficacy of these therapies. This review explores the current progress, challenges, and future prospects of nanomedicine-based targeted drug delivery systems in oncology, highlighting their role in precision cancer treatment.

keywords

Nanomedicine; Oncology; Targeted drug delivery; Precision cancer treatment; Nanoparticles; Tumor targeting; Nanocarriers; Chemotherapy; Drug resistance; Bioavailability; Molecular targeting; Personalized medicine

Introduction

Cancer remains one of the most significant global health challenges, with millions of people diagnosed each year and a high mortality rate. Traditional cancer therapies, such as chemotherapy, radiation, and surgery, are often limited by factors such as systemic toxicity, lack of specificity, and the emergence of drug resistance. Despite advancements in cancer treatment, these therapies still pose significant challenges in terms of treatment efficacy and patient quality of life. Recent developments in nanomedicine have opened new avenues for cancer treatment, offering a promising solution to overcome many of these limitations. Nanomedicine, particularly in the form of targeted drug delivery systems (TDDS), has revolutionized cancer therapy by enabling the precise delivery of therapeutic agents directly to cancer cells while sparing healthy tissues [1, 2].

Nanoparticles, the primary carriers in nanomedicine, are designed at the nanoscale, typically ranging from 1 to 1000 nanometers in size. This small size enables them to efficiently penetrate biological barriers, such as the blood-brain barrier, and selectively accumulate in tumors due to the enhanced permeability and retention (EPR) effect. The EPR effect allows nanoparticles to preferentially accumulate in tumor tissues due to the leaky vasculature and poor lymphatic drainage characteristic of many tumors. This unique property of nanomedicine makes it highly advantageous for delivering cancer drugs in a controlled and efficient manner.

A key feature of TDDS is the ability to modify the surface characteristics of nanoparticles to enhance their specificity toward cancer cells. This is typically achieved by functionalizing nanoparticles with targeting ligands such as antibodies, peptides, aptamers, or small molecules. These ligands can specifically bind to overexpressed receptors or proteins on the surface of tumor cells, thereby ensuring that the therapeutic agents are delivered precisely to the tumor site while minimizing damage to healthy cells. This specificity reduces the risk of side effects associated with conventional chemotherapy, improving the overall safety and tolerability of treatment [3].

In addition to improving drug delivery efficiency, nanocarriers such as liposomes, micelles, dendrimers, and polymeric nanoparticles offer several advantages in cancer therapy. These include controlled drug release, improved solubility of poorly water-soluble drugs, and the potential for combination therapies. The versatility of nanocarriers also allows for the incorporation of diagnostic agents, enabling the use of theranostic approaches—simultaneously diagnosing and treating cancer. By integrating imaging agents, nanoparticles can be tracked in vivo, facilitating real-time monitoring of treatment progress and tumor response.

Nanomedicine also plays a significant role in overcoming the challenge of drug resistance, which is one of the major hurdles in cancer therapy. Traditional chemotherapies often lead to resistance due to the rapid proliferation of cancer cells and the ability of tumor cells to evade drug action. Nanoparticles, by virtue of their size and surface modifications, can bypass common resistance mechanisms, such as drug efflux pumps, and achieve a more effective therapeutic outcome.

Moreover, precision medicine has gained considerable attention in oncology, as it seeks to tailor treatments to the individual genetic and molecular profile of each patient's cancer. Nanomedicine aligns with the goals of precision oncology by enabling personalized drug delivery, targeting specific mutations, and providing dynamic monitoring of treatment efficacy. As our understanding of cancer genomics and the molecular pathways involved in tumorigenesis grows, nanomedicine offers a dynamic platform to implement more effective and personalized treatment regimens.

Despite its promise, there are still challenges to the widespread clinical translation of nanomedicine-based targeted drug delivery systems in oncology. Issues related to biocompatibility, stability, manufacturing scalability, and regulatory approval need to be addressed before these technologies can become standard clinical practices. However, ongoing research and technological advancements are addressing these challenges, with numerous nanomedicine-based therapies already in clinical trials [4].

In conclusion, nanomedicine represents a groundbreaking approach to cancer treatment, offering enhanced precision, reduced toxicity, and improved therapeutic outcomes. As research in nanotechnology and cancer biology continues to evolve, the integration of targeted drug delivery systems will likely become a cornerstone of precision cancer treatment, providing a more effective and personalized approach to managing cancer and improving patient survival rates.

Materials and Methods

The development and evaluation of targeted drug delivery systems (TDDS) in nanomedicine for oncology involve a multi-disciplinary approach that integrates materials science, nanotechnology, molecular biology, and pharmacology. The following describes the materials and methods used for the fabrication, characterization, and evaluation of nanomedicine-based drug delivery systems for cancer treatment.

Materials

Nanoparticles: Various types of nanoparticles were selected based on their ability to encapsulate and deliver anticancer drugs efficiently. These include:

Liposomes: Composed of phospholipid bilayers, used for encapsulating hydrophobic drugs.

Polymeric Nanoparticles: Biodegradable polymers such as poly(lactic-co-glycolic acid) (PLGA), polycaprolactone (PCL), and polyethylenimine (PEI) are utilized for controlled drug release.

Dendrimers: Highly branched, nanoscale polymeric structures used for drug encapsulation and surface functionalization.

Micelles: Amphiphilic block copolymers form micelles that encapsulate hydrophobic drugs in their core, used for improving drug solubility [5].

Gold Nanoparticles: Used for their biocompatibility and ease of surface modification for targeting specific tumor biomarkers.

Targeting Ligands: Ligands that specifically recognize and bind to overexpressed receptors on tumor cells. Common ligands include:

Monoclonal Antibodies: Antibodies targeting specific tumor-associated antigens (e.g., HER2, EGFR).

Peptides: Short chains of amino acids that bind to tumor-specific receptors.

Aptamers: Nucleic acid-based ligands that bind to specific tumor biomarkers.

Chemotherapeutic Agents: Chemotherapeutic drugs such as doxorubicin, paclitaxel, cisplatin, or methotrexate are used as model drugs for the formulation of targeted drug delivery systems.

Solvents and Reagents: Solvents such as ethanol, chloroform, and water, along with reagents like sodium chloride, phosphate-buffered saline (PBS), and other chemical additives used for nanoparticle synthesis and characterization.

Methods

Preparation of Nanoparticle

The nanoparticles are synthesized using various techniques depending on the type of nanoparticle chosen for drug delivery [6].

Liposome preparation

Liposomes are prepared using the thin-film hydration method. Lipids are dissolved in organic solvents (e.g., chloroform) and then evaporated under reduced pressure to form a thin lipid film. The film is hydrated with an aqueous solution containing the drug, followed by sonication or extrusion to form liposomal vesicles.

Polymeric nanoparticle preparation

Solvent evaporation method: Polymeric nanoparticles are fabricated by dissolving the polymer (e.g., PLGA) and the drug in an organic solvent, followed by emulsification in an aqueous phase containing stabilizing agents. The organic solvent is evaporated under reduced pressure, forming solid nanoparticles.

Nanoprecipitation method: A polymer is dissolved in a solvent that is miscible with water, followed by dropwise addition to water to form nanoparticles [7].

Dendrimer synthesis

Dendrimers are synthesized by a repetitive process of divergent synthesis, where monomers are added successively to create a branched, tree-like structure. The drug is loaded into the dendrimer via physical encapsulation or chemical conjugation.

Micelle formation

Amphiphilic block copolymers are dissolved in water or an aqueous solvent to form micelles. The hydrophobic core of the micelles encapsulates the therapeutic drug.

Surface Functionalization and Targeting

To enhance tumor specificity, the surface of the nanoparticles is functionalized with targeting ligands.

Conjugation of targeting ligands

Targeting ligands, such as antibodies or peptides, are covalently attached to the surface of nanoparticles using linker molecules such as carbodiimides or thiol-maleimide chemistry.

PEGylation (attachment of polyethylene glycol, PEG) is often used to improve the stability and circulation time of nanoparticles in the bloodstream by reducing opsonization and immune clearance.

Drug loading and release

Drug Loading: The drug is loaded into the nanoparticles during the synthesis process by physical encapsulation (e.g., in liposomes or micelles) or covalent attachment (e.g., to dendrimers or polymeric nanoparticles). The drug loading efficiency is determined by measuring the amount of drug encapsulated compared to the initial drug concentration [8].

In vitro drug release studies

The drug release profile is evaluated in a buffered aqueous solution (PBS, pH 7.4 or 5.5) under controlled conditions (e.g., temperature and stirring). The release rate is measured by collecting samples at predefined intervals and quantifying the drug concentration using spectrophotometric methods (e.g., UV-Vis or HPLC).

Characterization of nanoparticles

To ensure the quality and functionality of the synthesized nanoparticles, several characterization techniques are employed:

Size and Zeta Potential: The size and surface charge (zeta potential) of nanoparticles are measured using dynamic light scattering (DLS) and electrophoretic light scattering (ELS).

Morphology: The morphology of nanoparticles is analyzed using scanning electron microscopy (SEM) or transmission electron microscopy (TEM).

Surface Area: The specific surface area of nanoparticles is measured using BET (Brunauer–Emmett–Teller) analysis, typically for porous nanoparticles like dendrimers.

Drug Loading Efficiency: The amount of drug encapsulated in nanoparticles is quantified by UV-Vis spectrophotometry, fluorescence spectroscopy, or HPLC, and the drug loading efficiency is calculated as the ratio of encapsulated drug to the initial drug quantity.

In vitro Cytotoxicity and Cellular Uptake

Cell Culture: Human cancer cell lines (e.g., HeLa, MCF-7, A549) are cultured under standard conditions (37°C, 5% CO₂) in complete growth media.

Cytotoxicity Assay: The cytotoxic effect of nanoparticles on cancer cells is evaluated using assays such as MTT, WST-1, or calcein AM, which measure cell viability after exposure to drug-loaded nanoparticles. The IC50 (half-maximal inhibitory concentration) is determined to assess the therapeutic efficacy.

Cellular Uptake: The internalization of nanoparticles into cancer cells is visualized using confocal microscopy or quantified using flow cytometry. Nanoparticles are often labeled with fluorescent dyes or fluorescently tagged targeting ligands to track cellular uptake [9].

In vivo studies

Animal Models: Animal models (e.g., mice or rats) bearing xenografted tumors are used to assess the in vivo distribution, targeting ability, and therapeutic efficacy of drug-loaded nanoparticles.

Biodistribution: The biodistribution of nanoparticles is studied by injecting the nanoparticles intravenously into the animal model and tracking their accumulation in tumors and other organs using fluorescence imaging, PET (positron emission tomography), or radiolabeling.

Tumor Growth Inhibition: The therapeutic efficacy of targeted nanoparticles is evaluated by monitoring tumor size using caliper measurements or imaging techniques (MRI, CT, or fluorescence imaging). Tumor growth inhibition is compared between treated and control groups.

Statistical analysis

Data from cytotoxicity, drug release, and in vivo studies are expressed as mean ± standard deviation (SD) or mean ± standard error of the mean (SEM). Statistical significance is determined using analysis of variance (ANOVA) followed by post-hoc tests such as Tukey’s test. p-values less than 0.05 are considered statistically significant [10].

Discussion

Nanomedicine has significantly advanced the field of oncology by providing a new dimension to cancer therapy through targeted drug delivery systems (TDDS). The traditional chemotherapy approach, while effective in certain contexts, often suffers from systemic toxicity, lack of specificity, and the emergence of drug resistance. TDDS in nanomedicine addresses these limitations by utilizing nanocarriers such as liposomes, dendrimers, micelles, and polymeric nanoparticles to deliver therapeutic agents directly to tumor sites. This precision delivery not only improves drug bioavailability but also minimizes adverse effects on healthy tissues, enhancing patient quality of life.

One of the primary advantages of nanomedicine in oncology is the enhanced permeability and retention (EPR) effect, which allows nanoparticles to accumulate preferentially in tumors due to the abnormal vasculature of tumors. The size of nanoparticles (typically between 1-200 nm) enables them to penetrate these leaky blood vessels, making them highly effective in drug delivery. However, while the EPR effect is a crucial mechanism for nanoparticle accumulation, its variability between different tumor types and even within the same tumor limits its effectiveness, requiring optimization of nanoparticle characteristics and targeting strategies.

The ability to functionalize nanoparticles with targeting ligands such as antibodies, peptides, and aptamers further enhances the precision of these drug delivery systems. These ligands can bind specifically to overexpressed receptors or antigens on tumor cells, leading to receptor-mediated endocytosis. This targeted approach increases the therapeutic efficacy of the drugs, reduces off-target effects, and addresses the challenges posed by drug resistance mechanisms. For example, nanoparticles targeting HER2 or EGFR receptors have shown improved efficacy in breast and lung cancer, respectively, where these receptors are often overexpressed. The development of these targeted systems has ushered in the era of personalized medicine, enabling treatments tailored to the molecular characteristics of an individual’s cancer.

The controlled release capabilities of nanocarriers are another significant advantage of TDDS in oncology. Nanoparticles can be engineered to release their payload in a controlled and sustained manner, which helps maintain therapeutic drug concentrations over an extended period. This approach can significantly reduce the frequency of drug administration and improve patient adherence to treatment regimens. Additionally, stimuli-responsive nanoparticles that release drugs in response to specific environmental cues such as pH, temperature, or enzymes further enhance the targeting and efficiency of drug delivery systems.

Despite the numerous advantages, several challenges remain in the clinical translation of nanomedicine for cancer therapy. Biosafety and biocompatibility are primary concerns, as the long-term effects of nanoparticles in the body are still not fully understood. While most nanoparticles show promising results in preclinical studies, their toxicity and potential for immune system activation in humans need thorough investigation. The use of PEGylation to improve the stability and circulation time of nanoparticles has shown some success in minimizing immunogenicity, but alternative strategies to further reduce immune clearance and increase nanoparticle lifespan are actively being explored.

The scalability of nanoparticle production also poses a challenge for widespread clinical use. While laboratory-scale synthesis of nanoparticles is relatively simple, achieving large-scale production with consistent quality and uniformity is difficult. This is particularly relevant when considering the potential commercialization of nanoparticles for clinical use, where stringent manufacturing and quality control standards must be met. Standardized protocols for nanoparticle synthesis and characterization are essential to ensure the reproducibility and safety of nanomedicines.

Another challenge is the tumor heterogeneity observed in many cancers. Tumors are often composed of multiple subtypes of cells with distinct genetic and phenotypic characteristics. This heterogeneity can affect the efficiency of targeted therapies, as some cancer cells may not express the receptors targeted by nanoparticles. Furthermore, the presence of an impermeable tumor microenvironment can hinder the effective penetration of nanoparticles into solid tumors. To address these challenges, strategies such as combination therapies, where nanoparticles are used to deliver not only chemotherapeutics but also immunomodulatory agents or gene therapies, are being explored. This multimodal approach could overcome the limitations of single-agent therapies and enhance overall treatment efficacy.

Clinical trials involving nanomedicine-based TDDS are promising, with many currently underway to test the safety and efficacy of these systems in cancer patients. Some of these trials have shown that nanoparticle formulations, such as liposomal formulations of doxorubicin (e.g., Doxil), can significantly reduce side effects and improve the pharmacokinetics of the drug. However, the widespread adoption of these systems in clinical settings still faces regulatory hurdles, as the complex nature of nanoparticles requires rigorous assessment for safety, efficacy, and quality assurance.

Conclusion

Nanomedicine, particularly in the form of targeted drug delivery systems (TDDS), represents a promising frontier in the treatment of cancer. The ability to design nanoparticles that selectively deliver therapeutic agents to tumors, while minimizing off-target effects and systemic toxicity, addresses many of the limitations associated with conventional chemotherapy. By exploiting the enhanced permeability and retention (EPR) effect, nanoparticles can preferentially accumulate in tumor tissues, ensuring a higher concentration of drugs at the site of action. Coupled with surface functionalization techniques such as antibody, peptide, or aptamer conjugation, these systems offer precise targeting to tumor cells, enhancing the therapeutic index and reducing side effects.

Moreover, the controlled and sustained release capabilities of nanocarriers enable better management of drug doses over time, improving patient compliance and reducing the frequency of administration. Stimuli-responsive systems, capable of releasing drugs in response to specific environmental conditions like pH or temperature, offer further precision, making it possible to tailor drug delivery to the tumor microenvironment. This advancement opens the door to personalized and more effective cancer therapies, where treatments are customized based on the molecular profile of each patient’s cancer.

Despite these advantages, challenges remain in the clinical application of nanomedicine in oncology. Issues related to biocompatibility, long-term toxicity, and the immune response to nanoparticles need to be carefully evaluated in preclinical and clinical settings. While nanoparticles like liposomal doxorubicin have shown success in clinical trials, the large-scale production, regulatory approval, and standardization of these therapies remain significant hurdles. Additionally, the heterogeneity of tumors can impact the efficacy of targeted therapies, as not all cancer cells may express the targeted receptors. This calls for the development of more adaptable, multifunctional nanoparticles that can address this complexity by delivering combination therapies or responding to a wider array of tumor types.

Furthermore, overcoming drug resistance remains a crucial challenge in oncology. Nanomedicine offers potential solutions by bypassing common resistance mechanisms such as drug efflux pumps, providing a more effective alternative to conventional treatments. However, more research is needed to fully understand how nanoparticles can combat resistance in different cancer types and ensure long-lasting therapeutic benefits.

Nanomedicine in oncology is undoubtedly an exciting and evolving field that holds immense potential to revolutionize cancer treatment. By advancing drug delivery systems to achieve greater specificity, reduced toxicity, and enhanced efficacy, nanomedicine brings us closer to the goal of precision cancer therapy. However, further optimization, clinical validation, and overcoming existing challenges will be key to fully realizing the transformative impact of nanomedicine in cancer care. As research progresses, targeted nanomedicines will likely play an increasingly central role in personalized cancer treatment, offering hope for better patient outcomes and improved quality of life.

Conflict of interest

None

Acknowledgment

None

  1. Twilt JJ, van Leeuwen KG, Huisman HJ, Fütterer JJ, de Rooij M. et al. (2021) Artificial Intelligence Based Algorithms for Prostate Cancer Classification and Detection on Magnetic Resonance Imaging: A Narrative Review. Diagnostics 11: 959.
  2. Indexed at, Google Scholar, Crossref

  3. Stanowicka-Grada M, Senkus E (2023) Anti-HER2 Drugs for the Treatment of Advanced HER2 Positive Breast Cancer. Curr Treat Options Oncol 24:1633-1650.
  4. Indexed at, Google Scholar, Crossref

  5. Chiș IA, Andrei V, Muntean A, Moldovan M, Mesaroș A, et al. (2023) Salivary Biomarkers of Anti-Epileptic Drugs: A Narrative Review. Diagnostics 13:1962.
  6. Indexed at, Google Scholar, Crossref

  7. Ahmed Z, Zeeshan S, Liang BT (2021) RNA-Seq Driven Expression and Enrichment Analysis to Investigate CVD Genes with Associated Phenotypes among High-Risk Heart Failure Patients. Hum Genom 15: 67.
  8. Indexed at, Google Scholar, Crossref

  9. Drăgoi C, Nicolae AC, Dumitrescu IB, Popa DE, Ritivoiu M, et al. (2019) DNA targeting as a molecular mechanism underlying endogenous indoles biological effects. Farmacia. 67:367.
  10. Google Scholar, Crossref

  11. Zhao S, Bao Z, Zhao X, Xu M, Li MD, et al. (2021) Identification of Diagnostic Markers for Major Depressive Disorder Using Machine Learning Methods. Front Neurosci 15:645998.
  12. Indexed at, Google Scholar, Crossref

  13. Schaack D, Weigand MA, Uhle F (2021) Comparison of Machine-Learning Methodologies for Accurate Diagnosis of Sepsis Using Microarray Gene Expression Data. PLoS ONE 16: 0251800.
  14. Indexed at, Google Scholar, Crossref

  15. DeGroat W, Abdelhalim H, Patel K, Mendhe D, Zeeshan S, Ahmed Z (2024) Discovering Biomarkers Associated and Predicting Cardiovascular Disease with High Accuracy Using a Novel Nexus of Machine Learning Techniques for Precision Medicine. Sci Rep 14:1.
  16. Indexed at, Google Scholar, Crossref

  17. Ungurianu A, Zanfirescu A, Margina D (2022) Regulation of Gene Expression through Food-Curcumin as a Sirtuin Activity Modulator. Plants 11:1741.
  18. Indexed at, Google Scholar, Crossref

  19. Kegerreis B, Catalina MD, Bachali P, Geraci NS, Labonte AC, et al. (2019) Machine Learning Approaches to Predict Lupus Disease Activity from Gene Expression Data. Sci Rep 9:9617.
  20. Indexed at, Google Scholar, Crossref

Citation: Diana B (2024) Nanomedicine in Oncology: Targeted Drug Delivery Systems for Precision Cancer Treatment Clin Pharmacol Biopharm, 13: 511.

Copyright: © 2024 Diana B. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top