Crossing Biological Barriers: Nanoscale Strategies For Enhanced Drug Delivery in Brain Tumors (WWW - Kiu.ac - Ug)
Crossing Biological Barriers: Nanoscale Strategies For Enhanced Drug Delivery in Brain Tumors (WWW - Kiu.ac - Ug)
Page | 78
ABSTRACT
Brain tumors remain among the most challenging malignancies to treat due to the presence of complex
biological barriers, particularly the blood-brain barrier (BBB) and blood-tumor barrier (BTB), which restrict the
entry of therapeutic agents. Traditional chemotherapies often fail to achieve adequate drug concentrations at
the tumor site, resulting in suboptimal therapeutic outcomes and systemic toxicity. Nanoscale drug delivery
systems have emerged as promising tools to overcome these limitations by enhancing drug solubility, stability,
and site-specific delivery. This review explores the physiological and structural challenges posed by the BBB
and BTB and highlights the latest advancements in nanotechnology-based strategies designed to surmount
these barriers. Key nanocarrier platforms, including liposomes, polymeric nanoparticles, dendrimers, and
exosomes, are critically examined for their potential in targeting brain tumors. Furthermore, this review
discusses active and passive targeting mechanisms, stimuli-responsive delivery systems, and the integration of
theranostics for simultaneous imaging and therapy. Finally, the review considers clinical translation challenges
and future perspectives for nanoscale strategies in neuro-oncology.
Keywords: Blood-brain barrier, Brain tumors, Nanocarriers, Drug delivery, Targeted therapy
INTRODUCTION
Brain tumors, whether primary (originating in the brain) or metastatic (originating elsewhere and spreading to
the brain), remain among the most devastating forms of cancer[1]. Despite progress in oncology, these tumors
are associated with high morbidity and mortality due to the unique challenges presented by the central nervous
system (CNS) environment. A primary barrier to effective therapy is the presence of tightly regulated
physiological barriers, particularly the blood-brain barrier (BBB), which acts as a selective shield between
systemic circulation and the brain[2, 3]. This complex structure comprises endothelial cells with tight junctions,
supported by pericytes, astrocytic end-feet, and a basement membrane. While essential for protecting the brain
from toxins and pathogens, the BBB also inadvertently hinders the entry of many therapeutic agents, including
up to 98% of small-molecule drugs and nearly all macromolecules, thus complicating drug delivery strategies
for brain tumors[3–5].
The blood-tumor barrier (BTB), which forms in tumor-affected regions, is typically more permeable than the
intact BBB due to abnormal angiogenesis and leaky vasculature. However, this permeability is often inconsistent
and heterogeneous, varying with tumor subtype, grade, and anatomical location[6, 7]. This irregularity leads
to uneven drug distribution, making it difficult to achieve therapeutic concentrations across the entire tumor
mass and contributing to therapeutic failure and tumor relapse. As a result, conventional treatment strategies
such as surgical resection, radiation therapy, and systemic chemotherapy have shown limited efficacy, especially
in infiltrative or inoperable tumors such as glioblastoma multiforme[8, 9].
In response to these challenges, nanotechnology has emerged as a promising platform for improving drug
delivery across the BBB and BTB. Nanocarriers—engineered particles typically ranging from 1 to 200
nanometers—are being developed to exploit both passive and active targeting mechanisms to enhance
therapeutic delivery[10]. Passive targeting takes advantage of the enhanced permeability and retention (EPR)
effect in tumor vasculature, while active targeting employs ligands or antibodies that recognize tumor-specific
markers to enhance binding and uptake by tumor cells[11, 12]. These strategies can improve the
pharmacokinetic and pharmacodynamic profiles of anticancer drugs, increase their accumulation within the
tumor microenvironment, and reduce off-target toxicity in healthy tissues[13].
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
Furthermore, functionalized nanocarriers can be designed to respond to specific stimuli—such as pH, enzymes,
or temperature—within the tumor milieu, triggering controlled and localized drug release[14, 15]. Innovations
in nanomedicine also include biomimetic systems that cloak nanoparticles in cell membranes to evade immune
detection and increase circulation time. Collectively, these advances are shifting the paradigm of brain tumor
therapy from systemic and non-specific approaches to precision-guided, targeted interventions[14, 16]. This
review looks into the multifactorial barriers that hinder therapeutic delivery in brain tumors and critically
assesses the array of nanoscale strategies being developed to circumvent these barriers. By highlighting recent
breakthroughs and evaluating current limitations, we aim to provide an integrative overview of how Page | 79
nanomedicine can be leveraged for precision neuro-oncology, paving the way for improved treatment outcomes
and personalized therapeutic regimens.
2. The Challenge of Biological Barriers in Brain Tumor Therapy
2.1 The Blood-Brain Barrier (BBB): The blood-brain barrier (BBB) represents a highly specialized and tightly
regulated vascular interface that separates the systemic circulation from the central nervous system (CNS)[17].
This barrier is composed primarily of non-fenestrated endothelial cells that are interconnected by tight
junctions, creating a physical blockade that strictly limits paracellular transport. Supporting this structural
integrity are pericytes embedded in the basement membrane, astrocytic end-feet enveloping the capillaries, and
various transporters and enzymes that regulate molecular trafficking[18]. The primary physiological role of
the BBB is to maintain CNS homeostasis by selectively permitting the passage of essential nutrients while
preventing the entry of harmful substances, including pathogens, toxins, and xenobiotics[19].
However, in the context of brain tumor therapy, the very features that make the BBB protective also make it a
formidable obstacle[20]. The barrier restricts nearly all large therapeutic molecules and up to 98% of small-
molecule drugs from entering the brain parenchyma. Even promising chemotherapeutic agents with
demonstrated efficacy against tumor cells in vitro often fail in vivo due to inadequate penetration across the
BBB[21]. Moreover, active efflux transporters such as P-glycoprotein and multidrug resistance proteins further
reduce drug accumulation by actively pumping therapeutic agents out of the brain endothelial cells back into
the bloodstream[22].
In the setting of malignant brain tumors, the integrity of the BBB can be partially disrupted due to tumor-
induced angiogenesis and inflammatory responses. Nevertheless, this disruption is often limited and
inconsistent, failing to provide sufficient permeability across the entire tumor mass[23]. Additionally, the
surrounding normal brain tissue maintains an intact BBB, protecting areas where infiltrative tumor cells may
reside, thus shielding them from systemic therapies[23]. As a result, the BBB remains a critical bottleneck in
the successful treatment of brain tumors, necessitating the development of innovative strategies to enhance drug
penetration without compromising overall CNS function[24, 25].
2.2 The Blood-Tumor Barrier (BTB)
While the BBB presents a major hurdle in healthy brain tissue, brain tumors themselves often induce the
formation of a blood-tumor barrier (BTB), which arises due to pathological neovascularization and vascular
remodeling[26]. Unlike the tight and continuous structure of the BBB, the BTB is characterized by leaky,
irregular vasculature with disrupted tight junctions and increased fenestrations[27]. This leaky nature has led
to the idea that the BTB might be exploited for enhanced drug delivery through passive diffusion or the
enhanced permeability and retention (EPR) effect. However, this apparent advantage is often negated by the
heterogeneous nature of BTB permeability. Studies have shown that drug permeability within tumors varies
significantly depending on tumor type, grade, and intratumoral location[28, 29]. For instance, the core of a
glioblastoma may demonstrate relatively high permeability, while the infiltrative margins exhibit much lower
permeability due to retained BBB features. This spatial inconsistency results in uneven drug distribution,
creating sanctuaries where tumor cells can evade therapy and eventually lead to recurrence.
Furthermore, the abnormal architecture of tumor blood vessels within the BTB contributes to high interstitial
fluid pressure, sluggish blood flow, and compromised lymphatic drainage, all of which impair effective drug
transport and retention[30]. These physiological aberrations also affect nanoparticle distribution, potentially
limiting the therapeutic window. Additionally, the BTB is influenced by dynamic factors such as hypoxia,
inflammation, and immune cell infiltration, further complicating drug delivery[30].
The variability of the BTB underscores the necessity for drug delivery systems that can adapt to these spatial
and temporal inconsistencies. Advanced nanocarrier platforms that incorporate tumor-specific ligands,
responsiveness to tumor microenvironmental stimuli (e.g., acidic pH, proteolytic enzymes), and the ability to
penetrate deep into tumor tissue are currently under investigation[31]. A detailed understanding of the BTB’s
biophysical and biochemical properties is essential for the rational design of such systems[31]. Therefore,
overcoming the challenges posed by both the BBB and the BTB requires a multifaceted approach integrating
nanotechnology, molecular targeting, and systems biology for improved therapeutic success in brain tumor
management.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
3. Nanoscale Drug Delivery Platforms
3.1 Liposomes
Liposomes are spherical vesicles composed of one or more phospholipid bilayers surrounding an aqueous
core[32–34]. Their structure allows for the simultaneous encapsulation of both hydrophilic drugs (within the
aqueous interior) and lipophilic drugs (within the lipid bilayer). Due to their biocompatibility and ability to
reduce systemic toxicity, liposomes are widely used in nanomedicine[35–37]. The surface of liposomes can be
modified through PEGylation, adding polyethylene glycol chains to improve circulation time by reducing
opsonization and uptake by the mononuclear phagocyte system (MPS)[38]. Furthermore, liposomes can be Page | 80
functionalized with targeting moieties such as transferrin, folic acid, or antibodies to facilitate receptor-mediated
transport across the blood-brain barrier (BBB). These targeted liposomes can enhance drug accumulation in
brain tumor tissues through active transport mechanisms. Clinically approved formulations like Doxil have
paved the way for newer liposomal systems undergoing evaluation for glioblastoma, offering promise for more
effective and less toxic brain cancer therapies.
3.2 Polymeric Nanoparticles
Polymeric nanoparticles (PNPs) are solid colloidal particles typically ranging from 10 to 1000 nanometers in
size and are fabricated from biodegradable polymers such as poly(lactic-co-glycolic acid) (PLGA), chitosan, and
polyethylene glycol (PEG)-based copolymers[39–42]. These nanoparticles offer several advantages, including
the ability to fine-tune drug release profiles and surface properties. PNPs can encapsulate a variety of therapeutic
agents and release them in a controlled manner, either through passive diffusion or environmental stimuli like
pH changes or enzymatic degradation. In the context of brain tumor therapy, their size and surface chemistry
can be optimized to enhance penetration across the BBB. Moreover, their surfaces can be functionalized with
ligands to actively target tumor-associated receptors, improving specificity and reducing off-target effects[40].
Their excellent biocompatibility, low toxicity, and ease of large-scale production make polymeric nanoparticles
promising candidates for clinical translation in the treatment of gliomas and other central nervous system
malignancies.
3.3 Dendrimers
Dendrimers are highly branched, monodisperse macromolecules that exhibit a tree-like architecture with
multiple generations extending from a central core[43]. This unique structural design offers abundant terminal
functional groups for drug conjugation, surface modification, or targeting ligand attachment. Dendrimers such
as poly(amidoamine) (PAMAM) are widely explored for drug delivery because they can encapsulate therapeutic
molecules within their interior cavities or chemically attach them to surface groups[44]. Their nanometric size
and uniformity make them suitable for crossing the BBB, especially when further functionalized with
polyethylene glycol (PEG), antibodies, or peptides[45]. These modifications improve circulation time and
targeting specificity, minimizing off-target accumulation. Dendrimers are also capable of theranostic
applications, combining therapy and diagnostics by co-delivering imaging agents and drugs in a single
platform[45]. Their highly controlled architecture allows for precise engineering, offering versatility in
developing multifunctional nanocarriers tailored for personalized treatment of brain tumors, including
glioblastoma and metastatic brain lesions.
3.4 Inorganic and Hybrid Nanoparticles: Inorganic and hybrid nanoparticles integrate metal or mineral
components with organic materials to exploit their unique physical and chemical properties for drug delivery
and imaging[46, 47]. Examples include gold nanoparticles (AuNPs), which exhibit strong surface plasmon
resonance useful in photothermal therapy; iron oxide nanoparticles, which provide contrast in magnetic
resonance imaging (MRI) and magnetic targeting; and mesoporous silica nanoparticles (MSNs), which offer
high surface area and tunable pore sizes for drug loading[46, 48, 49]. These nanoparticles can be further
hybridized with polymers or lipids to improve biocompatibility and functionalization potential. In brain tumor
therapy, these platforms are explored for targeted delivery and real-time imaging to guide surgical resection or
monitor therapeutic efficacy[50]. However, while their multifunctionality is attractive, concerns remain about
their long-term safety, biodegradability, and accumulation in tissues. Thorough investigations into their
pharmacokinetics, clearance, and toxicity profiles are essential before they can be safely integrated into routine
clinical applications for central nervous system cancers.
3.5 Extracellular Vesicles and Exosomes: Extracellular vesicles (EVs), particularly exosomes, are nanosized
(30–150 nm) lipid bilayer-enclosed particles naturally secreted by cells for intercellular communication[51–53].
They transport proteins, lipids, and nucleic acids, and possess innate capabilities to traverse biological barriers
such as the BBB. Their endogenous origin provides superior biocompatibility, low immunogenicity, and
extended circulation, making them attractive vehicles for drug delivery. Exosomes can be isolated from various
biological fluids and engineered to carry therapeutic agents, including small molecules, siRNAs, and CRISPR
components. Surface modifications, such as ligand conjugation or genetic engineering of parental cells, can
enhance their targeting to tumor tissues. In brain tumor applications, engineered exosomes are being explored
for delivering chemotherapeutics or genetic payloads to glioblastoma cells.[54–56] Their capacity to mimic
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
natural cellular communication mechanisms offers a promising, minimally invasive approach to personalized
nanomedicine. However, challenges such as scalable production, efficient drug loading, and standardization of
isolation techniques need to be addressed for clinical translation.
4. Mechanisms for Crossing the BBB
4.1 Passive Targeting via EPR Effect: The Enhanced Permeability and Retention (EPR) effect refers to the
tendency of nanoparticles to accumulate in tumor tissues due to the leaky vasculature and impaired lymphatic
drainage typical of many tumors[57]. This passive targeting mechanism has been widely used in solid tumor
therapy to enhance drug concentration at the tumor site. However, the EPR effect in brain tumors is relatively Page | 81
limited compared to peripheral tumors[57, 58]. The presence of the highly selective BBB restricts passive
diffusion of most macromolecules and nanocarriers. Nonetheless, in glioblastoma and other high-grade brain
tumors, regions of the BBB may become partially disrupted, permitting some degree of nanoparticle
accumulation via the EPR effect[58]. Despite this, the heterogeneous and incomplete nature of BBB disruption
poses significant limitations for relying solely on passive targeting. As a result, passive accumulation through
the EPR effect often needs to be supplemented with active targeting strategies or stimuli-responsive systems to
achieve therapeutic concentrations in brain tumor tissues.
4.2 Active Targeting Strategies: Active targeting enhances nanoparticle delivery across the BBB and into
tumor cells by functionalizing the nanocarrier surface with ligands that bind specific receptors on endothelial or
tumor cells[59]. These ligands include transferrin, lactoferrin, folic acid, peptides (e.g., RGD), and antibodies
against overexpressed surface markers. Upon binding to their receptors, the nanoparticle-ligand complex is
internalized via receptor-mediated endocytosis or transcytosis, allowing the therapeutic payload to cross the
BBB efficiently. For example, transferrin receptor-targeting nanoparticles have shown promise in preclinical
models of glioma by improving drug accumulation in the brain. Similarly, insulin or integrin-targeted systems
exploit naturally occurring transport mechanisms to ferry drugs across endothelial barriers[60]. This strategy
not only increases specificity and uptake but also minimizes off-target side effects and systemic toxicity. Active
targeting remains a cornerstone of precision nanomedicine, particularly for brain tumors where traditional drug
delivery methods are hindered by the restrictive and protective nature of the BBB.
4.3 Stimuli-Responsive Delivery Systems: Stimuli-responsive delivery systems are intelligent nanocarriers
designed to release their therapeutic cargo in response to specific internal or external triggers[14, 61, 62]. In
the context of brain tumors, internal stimuli such as acidic pH, elevated redox potential, overexpressed enzymes,
or hypoxia within the tumor microenvironment can be exploited for controlled drug release. For example, pH-
sensitive nanoparticles can remain stable in the bloodstream (pH ~7.4) but undergo structural changes in the
acidic tumor milieu (pH ~6.5–6.8), triggering drug release specifically at the tumor site. Similarly, redox-
sensitive systems can respond to high intracellular glutathione concentrations in cancer cells to release
chemotherapeutics[63]. External stimuli such as heat, light, or magnetic fields can also be applied to trigger
release from thermosensitive, photosensitive, or magnetically-responsive carriers. These smart delivery systems
enhance therapeutic efficacy while reducing off-target toxicity. Their integration into brain tumor treatment
strategies holds great promise for overcoming the challenges of delivering potent agents across the BBB with
spatial and temporal precision.
5. Theranostic Nanomedicine and Imaging-Guided Therapy
Theranostic nanoparticles represent a cutting-edge advancement in nanomedicine by integrating diagnostic and
therapeutic functionalities into a single platform[64, 65]. This dual capability allows for simultaneous disease
imaging and targeted drug delivery, revolutionizing the way clinicians monitor and treat diseases, particularly
cancer. By enabling real-time tracking of the biodistribution of therapeutic agents and assessment of treatment
efficacy, theranostic nanoparticles can significantly enhance treatment personalization and responsiveness.
Several classes of nanoparticles have been explored for theranostic applications. Magnetic nanoparticles, such
as superparamagnetic iron oxide nanoparticles (SPIONs), serve as contrast agents in magnetic resonance
imaging (MRI) while also being capable of delivering drugs or mediating hyperthermia therapy[66, 67].
Quantum dots, owing to their unique optical properties, are used in fluorescence imaging and can be conjugated
with therapeutic molecules for targeted delivery. Gold nanoshells and nanorods, known for their tunable surface
plasmon resonance, can absorb near-infrared light and convert it into heat, enabling both photothermal therapy
and imaging.
The integration of these imaging modalities with targeted therapy helps improve treatment precision by
ensuring drugs are delivered specifically to diseased tissues while minimizing off-target effects. This reduces
systemic toxicity and enhances therapeutic outcomes. Furthermore, real-time imaging allows clinicians to assess
whether the therapeutic agent has reached its intended target and how the disease is responding to
treatment[68]. If necessary, treatment regimens can be rapidly adjusted, providing a level of adaptability not
possible with traditional methods.
In addition to oncology, theranostic nanoparticles are being studied for use in cardiovascular diseases,
neurodegenerative disorders, and infectious diseases[69]. As research progresses, improvements in
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
biocompatibility, targeting specificity, and imaging sensitivity will continue to drive the clinical translation of
theranostic nanotechnologies, making them a vital component of future precision medicine strategies.
6. Clinical Translation and Challenges
Despite promising preclinical results, clinical translation of nanomedicine for brain tumors faces significant
challenges. These include manufacturing scale-up, regulatory hurdles, reproducibility, immune response, and
long-term safety. Furthermore, inter-patient variability in BBB integrity and tumor biology complicates
treatment standardization. Clinical trials such as those involving nanoliposomal irinotecan or transferrin-
modified nanoparticles have shown potential but require further validation. Page | 82
7. Future Perspectives
The future of nanoscale brain tumor therapy lies in multi-functional, smart nanoparticles that integrate
targeting, stimuli-responsiveness, and real-time feedback. Advances in artificial intelligence, microfluidic
screening models, and precision medicine will accelerate the optimization of nanoparticle formulations.
Personalized nanomedicine approaches, leveraging patient-specific tumor profiles, hold the potential to
revolutionize neuro-oncology and overcome the current limitations of conventional treatments.
CONCLUSION
Crossing the BBB remains one of the greatest hurdles in brain tumor therapy. Nanoscale drug delivery systems
offer a promising avenue for circumventing this challenge by enhancing the delivery, specificity, and safety of
therapeutic agents. Continued interdisciplinary collaboration between nanotechnology, neuroscience, oncology,
and pharmacology will be critical for translating these innovative platforms into effective clinical therapies for
brain tumor patients.
REFERENCES
1. Koekkoek, J.A.F., Chang, S., Taphoorn, M.J.B.: Chapter 19 - Palliative care at the end-of-life in glioma
patients. In: Berger, M.S. and Weller, M. (eds.) Handbook of Clinical Neurology. pp. 315–326. Elsevier
(2016)
2. Mitusova, K., Peltek, O.O., Karpov, T.E., Muslimov, A.R., Zyuzin, M.V., Timin, A.S.: Overcoming the
blood–brain barrier for the therapy of malignant brain tumor: current status and prospects of drug
delivery approaches. J. Nanobiotechnology. 20, 412 (2022). https://doi.org/10.1186/s12951-022-01610-
7
3. Rechberger, J.S., Toll, S.A., Biswas, S., You, H.B., Chow, W.D., Kendall, N., Navalkele, P., Khatua, S.:
Advances in the Repurposing and Blood–Brain Barrier Penetrance of Drugs in Pediatric Brain Tumors.
Cancers. 17, 439 (2025). https://doi.org/10.3390/cancers17030439
4. Peters, J.J., Teng, C., Peng, K., Li, X.: Deciphering the Blood–Brain Barrier Paradox in Brain Metastasis
Development and Therapy. Cancers. 17, 298 (2025). https://doi.org/10.3390/cancers17020298
5. Zha, S., Liu, H., Li, H., Li, H., Wong, K.-L., All, A.H.: Functionalized Nanomaterials Capable of Crossing
the Blood–Brain Barrier. ACS Nano. 18, 1820–1845 (2024). https://doi.org/10.1021/acsnano.3c10674
6. Arvanitis, C.D., Ferraro, G.B., Jain, R.K.: The blood–brain barrier and blood–tumour barrier in brain
tumours and metastases. Nat. Rev. Cancer. 20, 26–41 (2020). https://doi.org/10.1038/s41568-019-0205-
x
7. Blethen, K.E., Arsiwala, T.A., Fladeland, R.A., Sprowls, S.A., Panchal, D.M., Adkins, C.E., Kielkowski,
B.N., Earp, L.E., Glass, M.J., Pritt, T.A., Cabuyao, Y.M., Aulakh, S., Lockman, P.R.: Modulation of the
blood-tumor barrier to enhance drug delivery and efficacy for brain metastases. Neuro-Oncol. Adv. 3,
v133–v143 (2021). https://doi.org/10.1093/noajnl/vdab123
8. Linville, R.M., Maressa, J., Guo, Z., Chung, T.D., Farrell, A., Jha, R., Searson, P.C.: A tissue-engineered
model of the blood-tumor barrier during metastatic breast cancer. Fluids Barriers CNS. 20, 80 (2023).
https://doi.org/10.1186/s12987-023-00482-9
9. Garg, P., Malhotra, J., Kulkarni, P., Horne, D., Salgia, R., Singhal, S.S.: Emerging Therapeutic Strategies
to Overcome Drug Resistance in Cancer Cells. Cancers. 16, 2478 (2024).
https://doi.org/10.3390/cancers16132478
10. Alum, E.U., Nwuruku, O.A., Ugwu, O.P.-C., Uti, D.E., Alum, B.N., Edwin, N.: Harnessing nature: plant-
derived nanocarriers for targeted drug delivery in cancer therapy. Phytomedicine Plus. 5, 100828 (2025).
https://doi.org/10.1016/j.phyplu.2025.100828
11. Kulkarni, M., Patel, K., Patel, A., Patel, S., Desai, J., Patel, M., Shah, U., Patel, A., Solanki, N.:
Nanomaterials as drug delivery agents for overcoming the blood-brain barrier: A comprehensive review.
ADMET DMPK. 12, 63–105 (2023). https://doi.org/10.5599/admet.2043
12. Yuan, S., Hu, D., Gao, D., Butch, C.J., Wang, Y., Zheng, H., Sheng, Z.: Recent advances of engineering
cell membranes for nanomedicine delivery across the blood–brain barrier. J. Nanobiotechnology. 23, 493
(2025). https://doi.org/10.1186/s12951-025-03572-y
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
13. Wang, K., Yang, R., Li, J., Wang, H., Wan, L., He, J.: Nanocarrier-based targeted drug delivery for
Alzheimer’s disease: addressing neuroinflammation and enhancing clinical translation. Front. Pharmacol.
16, (2025). https://doi.org/10.3389/fphar.2025.1591438
14. Mi, P.: Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics.
Theranostics. 10, 4557–4588 (2020). https://doi.org/10.7150/thno.38069
15. Alum, E.U.: AI-driven biomarker discovery: enhancing precision in cancer diagnosis and prognosis.
Discov. Oncol. 16, 313 (2025). https://doi.org/10.1007/s12672-025-02064-7
16. Sun, L., Liu, H., Ye, Y., Lei, Y., Islam, R., Tan, S., Tong, R., Miao, Y.-B., Cai, L.: Smart nanoparticles for Page | 83
cancer therapy. Signal Transduct. Target. Ther. 8, 418 (2023). https://doi.org/10.1038/s41392-023-
01642-x
17. Dotiwala, A.K., McCausland, C., Samra, N.S.: Anatomy, Head and Neck: Blood Brain Barrier. In:
StatPearls. StatPearls Publishing, Treasure Island (FL) (2025)
18. Kadry, H., Noorani, B., Cucullo, L.: A blood–brain barrier overview on structure, function, impairment,
and biomarkers of integrity. Fluids Barriers CNS. 17, 69 (2020). https://doi.org/10.1186/s12987-020-
00230-3
19. Scalise, A.A., Kakogiannos, N., Zanardi, F., Iannelli, F., Giannotta, M.: The blood–brain and gut–vascular
barriers: from the perspective of claudins. Tissue Barriers. 9, 1926190 (2021).
https://doi.org/10.1080/21688370.2021.1926190
20. Niazi, S.K.: Non-Invasive Drug Delivery across the Blood–Brain Barrier: A Prospective Analysis.
Pharmaceutics. 15, 2599 (2023). https://doi.org/10.3390/pharmaceutics15112599
21. Li, J., Zheng, M., Shimoni, O., Banks, W.A., Bush, A.I., Gamble, J.R., Shi, B.: Development of Novel
Therapeutics Targeting the Blood–Brain Barrier: From Barrier to Carrier. Adv. Sci. 8, 2101090 (2021).
https://doi.org/10.1002/advs.202101090
22. Chaves, J.C.S., Dando, S.J., White, A.R., Oikari, L.E.: Blood-brain barrier transporters: An overview of
function, dysfunction in Alzheimer’s disease and strategies for treatment. Biochim. Biophys. Acta BBA -
Mol. Basis Dis. 1870, 166967 (2024). https://doi.org/10.1016/j.bbadis.2023.166967
23. Sarkaria, J.N., Hu, L.S., Parney, I.F., Pafundi, D.H., Brinkmann, D.H., Laack, N.N., Giannini, C., Burns,
T.C., Kizilbash, S.H., Laramy, J.K., Swanson, K.R., Kaufmann, T.J., Brown, P.D., Agar, N.Y.R., Galanis,
E., Buckner, J.C., Elmquist, W.F.: Is the blood–brain barrier really disrupted in all glioblastomas? A
critical assessment of existing clinical data. Neuro-Oncol. 20, 184–191 (2018).
https://doi.org/10.1093/neuonc/nox175
24. Belykh, E., Shaffer, K.V., Lin, C., Byvaltsev, V.A., Preul, M.C., Chen, L.: Blood-Brain Barrier, Blood-Brain
Tumor Barrier, and Fluorescence-Guided Neurosurgical Oncology: Delivering Optical Labels to Brain
Tumors. Front. Oncol. 10, (2020). https://doi.org/10.3389/fonc.2020.00739
25. Dudley, A.C., Griffioen, A.W.: Pathological angiogenesis: mechanisms and therapeutic strategies.
Angiogenesis. 26, 313–347 (2023). https://doi.org/10.1007/s10456-023-09876-7
26. Zhang, L., Dimberg, A., Rasouli, J.: Editorial: The blood-brain barrier in brain tumors: molecular
mechanisms and therapeutic strategies. Front. Neurol. 14, (2023).
https://doi.org/10.3389/fneur.2023.1225594
27. Mo, F., Pellerino, A., Soffietti, R., Rudà, R.: Blood–Brain Barrier in Brain Tumors: Biology and Clinical
Relevance. Int. J. Mol. Sci. 22, 12654 (2021). https://doi.org/10.3390/ijms222312654
28. Pinkiewicz, M., Pinkiewicz, M., Walecki, J., Zaczyński, A., Zawadzki, M.: Breaking Barriers in Neuro-
Oncology: A Scoping Literature Review on Invasive and Non-Invasive Techniques for Blood–Brain
Barrier Disruption. Cancers. 16, 236 (2024). https://doi.org/10.3390/cancers16010236
29. Liu, D., Dai, X., Tao, Z., Zhou, H., Hong, W., Qian, H., Cheng, H., Wang, X.: Advances in blood–brain
barrier-crossing nanomedicine for anti-glioma. Cancer Nanotechnol. 14, 58 (2023).
https://doi.org/10.1186/s12645-023-00211-9
30. Stylianopoulos, T., Munn, L.L., Jain, R.K.: Reengineering the Tumor Vasculature: Improving Drug
Delivery and Efficacy. Trends Cancer. 4, 258–259 (2018). https://doi.org/10.1016/j.trecan.2018.02.010
31. Chen, B., Dai, W., He, B., Zhang, H., Wang, X., Wang, Y., Zhang, Q.: Current Multistage Drug Delivery
Systems Based on the Tumor Microenvironment. Theranostics. 7, 538–558 (2017).
https://doi.org/10.7150/thno.16684
32. Abbasi, H., Kouchak, M., Mirveis, Z., Hajipour, F., Khodarahmi, M., Rahbar, N., Handali, S.: What We
Need to Know about Liposomes as Drug Nanocarriers: An Updated Review. Adv. Pharm. Bull. 13, 7–23
(2023). https://doi.org/10.34172/apb.2023.009
33. Lombardo, D., Kiselev, M.A.: Methods of Liposomes Preparation: Formation and Control Factors of
Versatile Nanocarriers for Biomedical and Nanomedicine Application. Pharmaceutics. 14, 543 (2022).
https://doi.org/10.3390/pharmaceutics14030543
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
34. Rao, L., Zhu, P., Guo, M., Hu, M., Guo, X., Du, Y., Xu, G.: Nebulized inhalation of nintedanib-loaded
biomimetic nano-liposomes attenuated bleomycin-induced interstitial lung fibrosis in mice. Nano Today.
56, 102298 (2024). https://doi.org/10.1016/j.nantod.2024.102298
35. Alum, E.U.: Role of phytochemicals in cardiovascular disease management: Insights into mechanisms,
efficacy, and clinical application. Phytomedicine Plus. 5, 100695 (2025).
https://doi.org/10.1016/j.phyplu.2024.100695
36. Enzyme-responsive liposomes for controlled drug release. | Semantic Scholar,
https://www.semanticscholar.org/paper/Enzyme-responsive-liposomes-for-controlled-drug-Wei- Page | 84
Lv/b1d010ba3c8440075ad2d08a613991904f4bdf7a
37. Uti, D.E., Atangwho, I.J., Alum, E.U., Ntaobeten, E., Obeten, U.N., Bawa, I., Agada, S.A., Ukam, C.I.-O.,
Egbung, G.E.: Antioxidants in cancer therapy mitigating lipid peroxidation without compromising
treatment through nanotechnology. Discov. Nano. 20, 70 (2025). https://doi.org/10.1186/s11671-025-
04248-0
38. Makharadze, D., del Valle, L.J., Katsarava, R., Puiggalí, J.: The Art of PEGylation: From Simple Polymer
to Sophisticated Drug Delivery System. Int. J. Mol. Sci. 26, 3102 (2025).
https://doi.org/10.3390/ijms26073102
39. Begines, B., Ortiz, T., Pérez-Aranda, M., Martínez, G., Merinero, M., Argüelles-Arias, F., Alcudia, A.:
Polymeric Nanoparticles for Drug Delivery: Recent Developments and Future Prospects. Nanomaterials.
10, 1403 (2020). https://doi.org/10.3390/nano10071403
40. kapoor, D. u, Garg, R., Gaur, M., Prajapati, B.G., Agrawal, G., Bhattacharya, S., Elossaily, G.M.:
Polymeric nanoparticles approach and identification and characterization of novel biomarkers for colon
cancer. Results Chem. 6, 101167 (2023). https://doi.org/10.1016/j.rechem.2023.101167
41. Wang, X.-Q., Zhang, Q.: pH-sensitive polymeric nanoparticles to improve oral bioavailability of
peptide/protein drugs and poorly water-soluble drugs. Eur. J. Pharm. Biopharm. 82, 219–229 (2012).
https://doi.org/10.1016/j.ejpb.2012.07.014
42. Zhang, W., Mehta, A., Tong, Z., Esser, L., Voelcker, N.H.: Development of Polymeric Nanoparticles for
Blood–Brain Barrier Transfer—Strategies and Challenges. Adv. Sci. 8, 2003937 (2021).
https://doi.org/10.1002/advs.202003937
43. Pérez-Ferreiro, M., M. Abelairas, A., Criado, A., Gómez, I.J., Mosquera, J.: Dendrimers: Exploring Their
Wide Structural Variety and Applications. Polymers. 15, 4369 (2023).
https://doi.org/10.3390/polym15224369
44. Abedi-Gaballu, F., Dehghan, G., Ghaffari, M., Yekta, R., Abbaspour-Ravasjani, S., Baradaran, B.,
Dolatabadi, J.E.N., Hamblin, M.R.: PAMAM dendrimers as efficient drug and gene delivery nanosystems
for cancer therapy. Appl. Mater. Today. 12, 177–190 (2018). https://doi.org/10.1016/j.apmt.2018.05.002
45. Omidian, H., Cubeddu, L.X., Wilson, R.L.: Peptide-Functionalized Nanomedicine: Advancements in Drug
Delivery, Diagnostics, and Biomedical Applications. Molecules. 30, 1572 (2025).
https://doi.org/10.3390/molecules30071572
46. Magadani, R., Ndinteh, D.T., Roux, S., Nangah, L.P., Atangwho, I.J., Uti, D.E., Alum, E.U., Egba, S.I.:
Cytotoxic Effects of Lecaniodiscus Cupanioides (Planch.) Extract and Triterpenoids-derived Gold
Nanoparticles On MCF-7 Breast Cancer Cell Lines. Anti-Cancer Agents Med. Chem. Former. Curr. Med.
Chem. - Anti-Cancer Agents. 25, 841–850 (2025).
https://doi.org/10.2174/0118715206325529241004064307
47. Yanar, F., Carugo, D., Zhang, X.: Hybrid Nanoplatforms Comprising Organic Nanocompartments
Encapsulating Inorganic Nanoparticles for Enhanced Drug Delivery and Bioimaging Applications.
Molecules. 28, 5694 (2023). https://doi.org/10.3390/molecules28155694
48. Demishkevich, E., Zyubin, A., Seteikin, A., Samusev, I., Park, I., Hwangbo, C.K., Choi, E.H., Lee, G.J.:
Synthesis Methods and Optical Sensing Applications of Plasmonic Metal Nanoparticles Made from
Rhodium, Platinum, Gold, or Silver. Materials. 16, 3342 (2023). https://doi.org/10.3390/ma16093342
49. Uti, D.E., Alum, E.U., Atangwho, I.J., Ugwu, O.P.-C., Egbung, G.E., Aja, P.M.: Lipid-based nano-carriers
for the delivery of anti-obesity natural compounds: advances in targeted delivery and precision
therapeutics. J. Nanobiotechnology. 23, 336 (2025). https://doi.org/10.1186/s12951-025-03412-z
50. Wu, J., Ko ,Sungeun, Lee ,Eunseo, Son ,Euijin, Kang ,Gyeonghui, Hur ,Seoyoung, Lee ,Jung-Hoon, Oh
,Jeong-Wook, and Kim, Y.: Gold nanoparticles in imaging: advances, applications, and future perspectives.
Appl. Spectrosc. Rev. 0, 1–40. https://doi.org/10.1080/05704928.2025.2495022
51. Brezgin, S., Danilik, O., Yudaeva, A., Kachanov, A., Kostyusheva, A., Karandashov, I., Ponomareva, N.,
Zamyatnin, A.A., Parodi, A., Chulanov, V., Kostyushev, D.: Basic Guide for Approaching Drug Delivery
with Extracellular Vesicles. Int. J. Mol. Sci. 25, 10401 (2024). https://doi.org/10.3390/ijms251910401
52. Kou, M., Huang, L., Yang, J., Chiang, Z., Chen, S., Liu, J., Guo, L., Zhang, X., Zhou, X., Xu, X., Yan, X.,
Wang, Y., Zhang, J., Xu, A., Tse, H., Lian, Q.: Mesenchymal stem cell-derived extracellular vesicles for
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited
https://www.eejournals.org Open Access
immunomodulation and regeneration: a next generation therapeutic tool? Cell Death Dis. 13, 1–16 (2022).
https://doi.org/10.1038/s41419-022-05034-x
53. Li, S.-J., Cheng, R.-J., Wei, S.-X., Xia, Z.-J., Pu, Y.-Y., Liu, Y.: Advances in mesenchymal stem cell-derived
extracellular vesicles therapy for Sjogren’s syndrome-related dry eye disease. Exp. Eye Res. 237, 109716
(2023). https://doi.org/10.1016/j.exer.2023.109716
54. Shao, M., Rodrigues, J., Sousa-Oliveira, I., Moradialvand, M., Asadollahi, P., Veiga, F., Hameed, H., Jha,
N.K., Sillanpää, M., Sethi, G., Paiva-Santos, A.C., Makvandi, P.: Revolutionizing cancer treatment via
bioengineered extracellular vesicles: Exploring nanovesicles to fully synthetic solutions. Appl. Mater. Page | 85
Today. 40, 102395 (2024). https://doi.org/10.1016/j.apmt.2024.102395
55. Rozier, P., Maumus, M., Maria, A.T.J., Toupet, K., Lai-Kee-Him, J., Jorgensen, C., Guilpain, P., Noël, D.:
Mesenchymal stromal cells-derived extracellular vesicles alleviate systemic sclerosis via miR-29a-3p. J.
Autoimmun. 121, 102660 (2021). https://doi.org/10.1016/j.jaut.2021.102660
56. Wiklander, O.P.B., Mamand, D.R., Mohammad, D.K., Zheng, W., Jawad Wiklander, R., Sych, T., Zickler,
A.M., Liang, X., Sharma, H., Lavado, A., Bost, J., Roudi, S., Corso, G., Lennaárd, A.J., Abedi-Valugerdi,
M., Mäger, I., Alici, E., Sezgin, E., Nordin, J.Z., Gupta, D., Görgens, A., EL Andaloussi, S.: Antibody-
displaying extracellular vesicles for targeted cancer therapy. Nat. Biomed. Eng. 8, 1453–1468 (2024).
https://doi.org/10.1038/s41551-024-01214-6
57. Wu, J.: The Enhanced Permeability and Retention (EPR) Effect: The Significance of the Concept and
Methods to Enhance Its Application. J. Pers. Med. 11, 771 (2021). https://doi.org/10.3390/jpm11080771
58. Belyaev, I.B., Griaznova, O.Yu., Yaremenko, A.V., Deyev, S.M., Zelepukin, I.V.: Beyond the EPR effect:
Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv. Drug Deliv. Rev. 219, 115550
(2025). https://doi.org/10.1016/j.addr.2025.115550
59. Hong, L., Li, W., Li, Y., Yin, S.: Nanoparticle-based drug delivery systems targeting cancer cell surfaces.
RSC Adv. 13, 21365–21382. https://doi.org/10.1039/d3ra02969g
60. Tang, S., Han, E.L., Mitchell, M.J.: Peptide-functionalized nanoparticles for brain-targeted therapeutics.
Drug Deliv. Transl. Res. (2025). https://doi.org/10.1007/s13346-025-01840-w
61. Hou, J., Xue, Z., Chen, Y., Li, J., Yue, X., Zhang, Y., Gao, J., Hao, Y., Shen, J.: Development of Stimuli-
Responsive Polymeric Nanomedicines in Hypoxic Tumors and Their Therapeutic Promise in Oral
Cancer. Polymers. 17, 1010 (2025). https://doi.org/10.3390/polym17081010
62. Mi, P.: Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics.
Theranostics. 10, 4557–4588 (2020). https://doi.org/10.7150/thno.38069
63. Sabit, H., Pawlik, T.M., Radwan, F., Abdel-Hakeem, M., Abdel-Ghany, S., Wadan, A.-H.S., Elzawahri, M.,
El-Hashash, A., Arneth, B.: Precision nanomedicine: navigating the tumor microenvironment for
enhanced cancer immunotherapy and targeted drug delivery. Mol. Cancer. 24, 160 (2025).
https://doi.org/10.1186/s12943-025-02357-z
64. AlQurashi, D.M., AlQurashi, T.F., Alam, R.I., Shaikh, S., Tarkistani, M.A.M.: Advanced Nanoparticles in
Combating Antibiotic Resistance: Current Innovations and Future Directions. J. Nanotheranostics. 6, 9
(2025). https://doi.org/10.3390/jnt6020009
65. Azimizonuzi, H., Ghayourvahdat, A., Ahmed, M.H., Kareem, R.A., Zrzor, A.J., Mansoor, A.S., Athab, Z.H.,
Kalavi, S.: A state-of-the-art review of the recent advances of theranostic liposome hybrid nanoparticles
in cancer treatment and diagnosis. Cancer Cell Int. 25, 26 (2025). https://doi.org/10.1186/s12935-024-
03610-z
66. Gannot, I.: A multimodal nanoparticles‐based theranostic method and system. Wiley Interdiscip. Rev.
Nanomed. Nanobiotechnol. 14, e1796 (2022). https://doi.org/10.1002/wnan.1796
67. Rahman, M.: Magnetic Resonance Imaging and Iron-oxide Nanoparticles in the era of Personalized
Medicine. Nanotheranostics. 7, 424–449 (2023). https://doi.org/10.7150/ntno.86467
68. Kandasamy, G., Maity, D.: Multifunctional theranostic nanoparticles for biomedical cancer treatments -
A comprehensive review. Mater. Sci. Eng. C. 127, 112199 (2021).
https://doi.org/10.1016/j.msec.2021.112199
69. Mangadlao, J.D., Wang, X., McCleese, C., Escamilla, M., Ramamurthy, G., Wang, Z., Govande, M.,
Basilion, J.P., Burda, C.: Prostate-Specific Membrane Antigen Targeted Gold Nanoparticles for
Theranostics of Prostate Cancer. ACS Nano. 12, 3714–3725 (2018).
https://doi.org/10.1021/acsnano.8b00940
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited