Targeted Delivery to Tissues and Lesions Using Nanoparticles

Targeted Delivery to Tissues and Lesions Using Nanoparticles

Introduction to Nanoparticle-Based Targeted Delivery

Overview of Nanoparticle Drug Delivery Systems

Nanoparticle-based drug delivery systems are typically composed of three essential elements: the carrier matrix, the therapeutic payload, and the surface functional layer. The carrier materials can include lipids, polymers, inorganic scaffolds, or hybrid composites. The payload encompasses small molecules, nucleic acids, peptides, or proteins, while the surface layer is often engineered using polyethylene glycol (PEG), targeting ligands, or environmentally responsive moieties for dynamic performance modulation. The particle size is generally controlled within the range of 10-200 nm, which enables nanoparticles to avoid renal filtration while exploiting interstitial permeability for enhanced tissue penetration. The surface charge is usually maintained at slightly negative or near-neutral levels to minimize plasma protein adsorption and extend circulation half-life. Drug encapsulation strategies include physical entrapment, electrostatic adsorption, covalent conjugation, and hydrophobic insertion. Each method directly influences the drug loading capacity, release kinetics, and formulation stability.

Advantages of Nanoparticles in Targeted Therapeutics

Nanoparticle systems can increase the local drug concentration at the target site by one to two orders of magnitude while simultaneously reducing systemic exposure to less than one-tenth of that of conventional formulations. This results in a markedly expanded therapeutic window and improved efficacy-to-toxicity ratio. The carrier matrix can be engineered to respond to light, heat, enzymatic activity, or redox gradients, ensuring synchronized drug release in accordance with the lesion's microenvironment and thereby minimizing off-target effects. A high-density PEG layer on the particle surface forms a hydrated barrier that reduces recognition by the reticuloendothelial system, extending plasma half-life from mere minutes to several hours or even days. Porous and core-shell architectures allow the co-loading of multiple therapeutic agents, enabling synergistic or sequential release strategies. Moreover, inorganic frameworks containing high atomic number elements can provide imaging or sensitization capabilities, achieving integrated delivery and monitoring functions within a single platform.

Common Barriers in Conventional Drug Delivery

Traditional small-molecule formulations tend to distribute rapidly throughout the body after administration, with less than 1% of the administered dose reaching the intended target tissue. To maintain effective concentrations, substantially higher doses are often required, which increases systemic burden and undesired exposure to non-target tissues. Poorly soluble drugs frequently rely on organic solubilizers that can lead to precipitation or local irritation. Many compounds undergo rapid enzymatic degradation or protein binding in plasma, drastically reducing the free drug fraction available for activity. Physical barriers such as the blood-brain barrier, dense extracellular matrices, and elevated interstitial pressures collectively hinder deep tissue penetration, leaving core regions underexposed. Rapid systemic clearance further shortens the half-life, necessitating frequent dosing and causing concentration fluctuations that heighten the risk of cumulative toxicity.

Fig.1 Representation of nanoparticle-based drug delivery to tissues and lesions. Fig.1 Schematic of nanoparticle-mediated targeted drug delivery in vivo1,2.

Mechanisms of Tissue and Lesion Targeting

Passive Targeting via Enhanced Permeability and Retention (EPR) Effect

In pathological tissues such as tumors, infections, or inflammatory sites, the vascular endothelium exhibits enlarged fenestrations ranging from 100 to 800 nm. Nanoparticles can traverse these gaps and accumulate within the interstitial space through the Enhanced Permeability and Retention (EPR) effect. A slightly negative surface charge reduces electrostatic interactions with positively charged basement membranes, enhancing retention duration. Studies on long-circulating liposomes have demonstrated that, 24 hours post-administration, intratumoral drug concentrations can reach up to 15 times higher than those of free drugs, while cardiac exposure remains at one-third of the control. However, elevated interstitial pressure can impede deep penetration; to overcome this, strategies involving size-adaptive or enzyme-triggered expansion have been developed. These mechanisms enable nanoparticles to swell or aggregate within the lesion, resulting in secondary retention and improved spatial localization.

Active Targeting Through Ligand–Receptor Interactions

Building upon passive accumulation, nanoparticles can be modified with antibodies, peptides, carbohydrates, or nucleic acid aptamers that recognize overexpressed receptors on diseased cells or tissues. For example, folate receptors are expressed at levels 20-100 times higher in many epithelial-derived pathologies compared with normal tissues, and folate-functionalized nanoparticles exhibit nanomolar binding affinities. Antibody fragments such as single-chain variable fragments (scFv) retain high recognition specificity while reducing immunogenicity. Polymer-based nanoparticles functionalized with scFv have demonstrated fourfold deeper penetration in three-dimensional spheroid models. Dual-ligand designs exploit co-expression of multiple receptors. One high-affinity ligand anchors nanoparticles to vascular endothelium, while a secondary low-affinity ligand mediates transcellular transport, enabling cascade delivery from circulation to deep tissue layers.

Stimuli-Responsive Targeting (pH, Redox, Enzyme, or Temperature)

Many diseased microenvironments exhibit distinct physicochemical features that can be exploited for controlled release. In mildly acidic environments, pH-sensitive polymers become protonated below pH 6.5, transforming hydrophobic segments into hydrophilic ones, leading to carrier disassembly and rapid drug release. Redox-sensitive nanoparticles containing disulfide crosslinks respond to the intracellular glutathione concentration, up to 1000 times higher than in plasma, by cleaving their linkages upon cellular entry to liberate the payload. Enzyme-responsive systems leverage overexpressed proteases such as matrix metalloproteinases, which cleave peptide substrates to expose hidden ligands and induce surface charge reversal, thereby facilitating deeper tissue penetration. Thermosensitive liposomes undergo phase transitions near 42 ℃, markedly enhancing membrane permeability by two orders of magnitude; when combined with localized thermal fields, these systems achieve precise spatiotemporal control of drug release.

Biodistribution and Cellular Uptake Pathways

Following intravenous administration, nanoparticles rapidly interact with plasma proteins to form a dynamic "protein corona," which determines their initial biodistribution and capture efficiency in the liver, spleen, or lungs. Nanoparticles smaller than 50 nm can partially enter lymphatic circulation and accumulate in lymph nodes, where they are internalized by dendritic cells. Within tissue interstitia, particles migrate via diffusion, convection, or paracellular transport until they encounter target cells. Cellular internalization occurs through clathrin-mediated endocytosis, caveolae invagination, or macropinocytosis pathways. When surface ligand density exceeds 10 mol%, receptor-mediated uptake increases significantly; however, excessively dense functionalization may lead to receptor saturation and lysosomal degradation. The nuclear pore complex permits the entry of nanoparticles smaller than approximately 39 nm. For carriers delivering siRNA or gene-editing proteins, maintaining structural integrity is crucial to successfully traverse this barrier and achieve intracellular functional delivery.

Interested in Advanced Nanoparticle Delivery Strategies?

BOC Sciences provides versatile nanoparticles with diverse compositions and functional modifications, customized solutions for your delivery needs.

Design Considerations for Efficient Tissue-Specific Targeting

Neutral, hydrophilic PEG remains the most widely applied "stealth" coating; a surface density of ≥0.2 chains nm-2 can reduce plasma protein adsorption by up to 80%. Terminal functional groups such as active esters, maleimides, or azides enable covalent coupling of antibodies, peptides, aptamers, or glycans for specific recognition. For instance, PLGA nanoparticles functionalized with cyclic RGD peptides exhibit a 25-fold higher affinity toward αvβ3 integrin-positive endothelial cells and triple the accumulation in angiogenic vasculature. For central nervous system delivery, conjugation with lactoferrin or transferrin facilitates receptor-mediated transcytosis across the blood–brain barrier, resulting in 5–8 times higher brain accumulation. To minimize hepatic uptake, surface incorporation of anionic moieties such as sialic acid or phosphorylcholine can reduce Kupffer cell recognition and prolong systemic circulation.

The optimal size range for balancing vascular permeability and retention typically lies between 60 and 120 nm; particles exceeding this range struggle to penetrate dense stroma, while those smaller than 30 nm are rapidly filtered by the kidneys. Near-neutral or slightly negative zeta potentials maximize plasma half-life, though a mild positive charge (+5 to +10 mV) can promote cellular adhesion. "Charge-switching" polymers that transition from negative to positive under specific stimuli enable nanoparticles to circulate stealthily yet penetrate effectively at the target site. Particle shape also affects hemodynamic behavior: short rods with aspect ratios of 3-5 exhibit twice the wall-contact frequency of spherical counterparts, enhancing vascular adhesion. However, excessively elongated rods (>500 nm) risk mechanical entrapment. Techniques such as membrane stretching or microfluidic molding allow uniform rod fabrication with size variations below 6%.

Release profiles should align with the intended pharmacodynamic timeline. Rapid-release pulses (e.g., 80% release within 24 h) suit immunostimulatory or nucleic acid payloads, whereas zero-order sustained release over 7–14 days supports local microenvironmental modulation. Polymer molecular weight, crystallinity, and copolymer composition can tune the release half-life from hours to weeks. Liposomal formulations combining saturated phospholipids and cholesterol (molar ratio 60:40) maintain particle size growth below 10 nm after six months at 4 ℃. For PLGA nanoparticles, blending with 5% PEG-PLA mitigates autocatalytic hydrolysis, reducing encapsulation loss from 20% to less than 5%.

Pulmonary delivery requires evasion of mucociliary clearance. Nanoparticles sized at 200-300 nm and coated with nonadhesive polymers such as Pluronic F68 exhibit a tenfold improvement in mucus diffusivity. In joint environments rich in hyaluronic acid, co-delivery of hyaluronidase or decoration with hyaluronic acid–binding proteins facilitates deep cartilage penetration, increasing intratissue diffusion distance by up to fourfold. In fibrotic tumor tissues characterized by elevated matrix metalloproteinase expression, embedding the peptide sequence GPLGVRGC within lipid membranes enables enzyme-triggered surface charge reversal, promoting cellular uptake and feedback suppression of fibroblast activation. Physical adjuncts such as magnetic fields or focused ultrasound can further open intercellular junctions, enhancing three-dimensional tissue distribution.

Table 1. Correlation Between Key Design Parameters and In Vivo Performance of Nanocarriers.

Design Parameter Optimal Range Functional Role Risks if Deviated
Particle size60–120 nmBalances circulation and penetration<30 nm:="" renal="">200 nm: entrapment
Zeta potential−10 to +10 mVMinimizes protein corona formation>+20 mV: hemolysis; <−30 mV: aggregation
PEG density0.2–0.5 chains nm-2Balances stealth and targeting>0.8 chains nm-2: impedes ligand binding

Types of Nanoparticles for Targeted Delivery

Lipid-Based Nanoparticles

Liposomes consist of one or multiple concentric phospholipid bilayers surrounding an aqueous core, allowing simultaneous encapsulation of hydrophilic and lipophilic compounds. Upon PEGylation, their circulation half-life can extend from several minutes to more than ten hours, while passive accumulation in inflamed or neoplastic tissues increases by three- to fivefold. Solid lipid nanoparticles (SLNs) replace fluid phospholipids with biocompatible solid triglycerides or waxes, embedding drugs within a solid matrix rather than a liquid core. This architecture prevents leakage and maintains drug stability, retaining over 90% of lipophilic compounds such as retinoic acid after six months of storage. Both liposomes and SLNs can be fabricated using microfluidic rapid mixing, achieving particle sizes between 60 and 120 nm with batch-to-batch variations below 5%.

Polymeric Nanoparticles and Micelles

Biodegradable polyesters such as PLA, PLGA, and amphiphilic block copolymers like PCL-PEG can self-assemble into core-shell nanostructures. The hydrophobic core accommodates poorly soluble molecules such as paclitaxel or curcumin, with loading efficiencies exceeding 15%, while the PEG shell provides steric stabilization and minimizes uptake by the reticuloendothelial system. By adjusting the lactide-to-glycolide ratio, the degradation half-life at physiological pH can be tuned from 2 days to up to 4 weeks, supporting sustained release for long-term therapeutic regimens. Cationic polymers such as chitosan or polyethyleneimine condense nucleic acids via electrostatic complexation to form 100-200 nm nanocomplexes. Within cells, the "proton sponge" effect facilitates endosomal escape, enhancing mRNA transfection efficiency by more than two orders of magnitude compared with naked nucleic acids.

Inorganic Nanoparticles (Gold, Silica, Iron Oxide)

Gold nanoparticles exhibit tunable surface plasmon resonance peaks dependent on size and geometry. For instance, 50 nm gold spheres show strong absorption around 530 nm, enabling near-infrared photothermal conversion that elevates local temperatures to approximately 43 ℃ within minutes, transiently increasing membrane permeability and promoting cytosolic delivery of co-loaded molecules. Mesoporous silica nanoparticles offer adjustable pore diameters (2-10 nm) and exceptionally high surface areas exceeding 1000 m2 g-1, capable of adsorbing 0.3-0.5 mg of protein or peptide per milligram of carrier. Their open-channel architecture facilitates surface gating and stimuli-responsive release through redox or enzymatic triggers. Superparamagnetic iron oxide nanoparticles exhibit a saturation magnetization of about 70 emu g-1 under a 1 T magnetic field. Following systemic administration, application of a localized pulsed magnetic field can increase particle accumulation fourfold at the target site while simultaneously allowing real-time MRI monitoring.

Hybrid and Stimuli-Responsive Nanocarriers

Hybrid systems combine the advantages of lipid and polymeric architectures to achieve both membrane fusion capability and controlled release. For example, lipid-PLGA hybrid nanoparticles leverage a phospholipid shell for cellular affinity and a polymeric core for sustained drug retention; in vitro, less than 20% of the payload is released within seven days, yet more than 80% is liberated within 48 hours after enzymatic activation. Stimuli-responsive nanocarriers incorporate hydrazone, disulfide, or thermosensitive linkages within the matrix, enabling multi-level responsiveness to pH, redox potential, light, heat, or enzymatic activity. Disulfide-crosslinked micelles remain stable in circulation but disassemble rapidly in intracellular environments rich in glutathione, releasing the payload within minutes at a burst intensity up to tenfold higher than in plasma, substantially reducing systemic exposure.

Table 2. Overview of Nanoparticle-Related Products at BOC Sciences.

Product Name Description Applications Inquiry
Gold Nanoparticles (AuNPs)Offered in spherical or rod shapes, functionalized with carboxyl or thiol groups for biomolecule conjugation; available in sizes from 10-150 nm.Photothermal conversion, molecular imaging, SERS detection, hybrid nanocomposite systems.Inquiry
PLGA Nanoparticles Pre-fabricated PLGA nanoparticles (100-200 nm) with excellent batch-to-batch consistency; available with carboxyl, amino, or PEG-modified surfaces.Controlled-release systems, drug carrier evaluation, formulation and degradation studies.Inquiry
Lipid-Polymer Hybrid Nanocarriers Combine a lipid shell with a polymeric core to achieve both structural stability and membrane affinity; customizable particle size and composition.Co-delivery systems, smart and stimuli-responsive delivery platforms.Inquiry
Mesoporous Silica Nanoparticles (MSNs) Tunable pore sizes from 2-10 nm with surface areas up to 1000 m2/g; suitable for loading proteins or polymers and gatekeeper functionalization.Controlled release systems, enzyme-triggered delivery, co-loading of multiple drugs.Inquiry
Superparamagnetic Iron Oxide Nanoparticles (SPIONs) High magnetic saturation, 30-100 nm particle size; available with PEG or chitosan coatings for improved stability and dispersion.Magnetically guided delivery, cell labeling, magnetic-responsive release, MRI signal enhancement.Inquiry
Stimuli-Responsive Polymers Contain acylhydrazone, disulfide, or thermosensitive linkages that enable environment-triggered release behavior.Smart controlled-release design, site-specific delivery system development.Inquiry
Bioactive Inorganic Nanopowders Include hydroxyapatite-, silica-, and zinc oxide–based powders with enhanced bioactivity and mechanical strength.Scaffold reinforcement, regenerative medicine composites, bone tissue engineering.Inquiry

Applications of Nanoparticle-Mediated Targeted Delivery

Nanoparticles have emerged as highly versatile platforms for tumor-targeted delivery due to their tunable physicochemical properties and capacity for surface functionalization. The irregular vascular architecture and enhanced permeability of tumor tissues facilitate passive accumulation of nanoparticles via the enhanced permeability and retention (EPR) effect. Active targeting can be further achieved through the conjugation of ligands, antibody fragments, or peptides that recognize tumor-specific receptors. For instance, polymeric nanoparticles functionalized with cyclic RGD peptides can selectively bind to integrin receptors overexpressed in tumor neovasculature, significantly improving local drug retention. Inorganic nanoparticles such as gold nanoshells and iron oxide nanocrystals provide additional optical and magnetic responsiveness, enabling integration of therapeutic delivery with real-time imaging modalities such as optical or magnetic resonance imaging. This theranostic capability supports simultaneous treatment and visualization of pathological regions. Furthermore, co-loaded or sequential-release systems incorporating multiple agents allow dynamic modulation of tumor pathways, expanding the therapeutic window and improving precision at the cellular level.

Inflammatory lesions are characterized by vascular dilation, increased permeability, and the recruitment of immune cells, all of which promote the passive localization of nanoscale carriers. By tailoring surface chemistry, targeting specificity can be enhanced, for example, hyaluronic acid-modified nanoparticles can selectively interact with CD44 receptors enriched at inflamed sites. PEGylated liposomes exhibit extended circulation stability and enhanced penetration, resulting in substantially higher drug accumulation at target tissues. For environments dominated by oxidative stress, nanocarriers incorporating antioxidant motifs can undergo selective release in the presence of reactive oxygen species, minimizing off-target exposure. Stimuli-responsive mechanisms, such as infrared-triggered or ultrasound-triggered release, allow precise spatiotemporal control of drug liberation, enabling localized activation and improved therapeutic efficiency within inflammatory microenvironments.

The blood-brain barrier (BBB) represents a formidable obstacle for most therapeutic molecules due to its tight junctions and selective permeability. Rationally designed nanoparticles offer effective strategies for overcoming this biological barrier. Ligand conjugation with transferrin, lactoferrin, or glucose analogs can facilitate receptor-mediated transcytosis across endothelial cells. Polymeric micelles and lipid-based nanoparticles demonstrate enhanced uptake in brain capillary endothelial cell models, resulting in greater accumulation within neural tissues. Charge-reversible systems further improve delivery efficiency by maintaining a neutral surface charge during circulation to prolong stability, followed by a transition to a mildly positive charge upon reaching the BBB to promote membrane interaction and endocytosis. External field-assisted strategies, including magnetic targeting and focused ultrasound, have been employed to transiently modulate barrier permeability, thereby enhancing nanoparticle penetration into brain tissues while maintaining controlled delivery dynamics.

In regenerative medicine, nanoparticles function not only as carriers for bioactive molecules but also as integral components of engineered microenvironments. Biodegradable polymeric nanoparticles can provide sustained release of signaling molecules that guide stem cell differentiation or stimulate angiogenesis. Inorganic nanoparticles such as hydroxyapatite or silica variants can be integrated into scaffolds to improve mechanical strength and enhance bioactivity. Nanocarriers encapsulating peptides or nucleic acids allow controlled temporal release during tissue repair, synchronizing cellular migration, proliferation, and extracellular matrix remodeling. Stimuli-responsive carriers that react to local temperature or enzymatic activity enable adaptive release control, ensuring effective concentrations of growth factors throughout the regeneration process. Moreover, magnetic nanoparticles embedded in three-dimensional scaffolds facilitate external field-driven modulation of cell distribution and orientation, advancing the development of smart, responsive tissue engineering systems.

Analytical and Characterization Approaches

Particle Size, Zeta Potential, and Morphology Analysis

Particle size and surface charge are fundamental parameters determining the biological performance of nanoparticle systems, influencing distribution, colloidal stability, and cellular interactions. Dynamic light scattering (DLS) provides measurements of hydrodynamic diameter and polydispersity index, reflecting particle uniformity and aggregation behavior. Zeta potential analysis assesses surface charge, where near-neutral or slightly negative values typically correlate with extended systemic circulation and reduced nonspecific interactions. Transmission electron microscopy (TEM) and atomic force microscopy (AFM) reveal detailed morphological and structural features, including shell thickness, porosity, and core–shell architecture. For complex hierarchical systems, cryo-electron microscopy enables near-native visualization, supporting process optimization and reproducibility in nanoparticle synthesis.

Drug Loading Efficiency and Release Profiling

Drug encapsulation efficiency and loading capacity are critical determinants of nanocarrier performance. Quantification is typically performed using UV–visible spectroscopy, fluorescence spectrometry, or high-performance liquid chromatography, following the separation of unencapsulated drugs via centrifugation or dialysis. Drug release studies are conducted in simulated physiological media under controlled conditions, generating release profiles that describe diffusion and degradation behavior. Depending on the formulation, nanoparticles may exhibit burst, sustained, or multi-phase release kinetics. Mathematical modeling using Higuchi, Korsmeyer–Peppas, or zero-order equations provides insights into the underlying release mechanisms. For stimuli-responsive systems, comparative studies under varied pH, redox, or thermal environments confirm the precision and controllability of release behavior, ensuring system responsiveness to relevant biological conditions.

In Vitro and In Vivo Evaluation Methods

In vitro assays are essential for preliminary performance screening. Cellular uptake studies, often employing fluorescently labeled nanoparticles and confocal microscopy, elucidate endocytic pathways and intracellular distribution. Cytocompatibility is commonly evaluated through metabolic or membrane integrity assays such as MTT and LDH release tests. Barrier models like Transwell systems allow quantitative assessment of nanoparticle translocation across epithelial or endothelial layers. In vivo investigations provide complementary information on biodistribution and pharmacokinetic behavior. Fluorescence imaging, magnetic resonance tracking, and radiolabeling techniques enable visualization of nanoparticle fate within biological systems. Integration of these analytical methods facilitates a comprehensive understanding of the correlation between physicochemical properties and biological performance, guiding rational design and optimization of nanoparticle-mediated delivery systems.

BOC Sciences' Nanoparticle Development Solutions

BOC Sciences provides end-to-end nanoparticle development solutions spanning design, synthesis, characterization, and scale-up, offering both ready-to-use and fully customizable platforms for diverse applications, including tumor targeting, inflammation, central nervous system delivery, and regenerative medicine. The liposome production platform utilizes microfluidic continuous fabrication technology, ensuring high particle uniformity and enabling smooth transitions from early-stage experiments to larger-scale applications.

The polymeric nanoparticle platform covers a wide range of polyester systems, including PLGA, PCL, and PLA-PEG, with customizable ester bond ratios to fine-tune particle properties. Particles of various sizes can be prepared in a single step via solvent evaporation or nanoprecipitation, and hydrophilic and hydrophobic payloads can be compartmentalized within a single carrier to achieve coordinated co-delivery.

In the inorganic nanoparticle segment, BOC Sciences offers gold, silica, and iron oxide nanoparticles, with surfaces functionalizable through PEGylation, thiol, carboxyl, azide, and other active groups to facilitate conjugation with antibodies, peptides, or nucleic acids. The platform is complemented by a comprehensive characterization suite, including ICP-MS, UV-vis spectroscopy, DLS, and cryo-TEM, capable of providing full datasets on particle size, surface charge, drug loading, release profiles, and protein corona composition, supporting rapid carrier screening and mechanistic studies.

For stimuli-responsive nanoparticle systems, BOC Sciences maintains libraries of pH-, redox-, photothermal-, and enzyme-responsive linkers, enabling small-scale synthesis and in vitro trigger validation, which can be smoothly scaled up to gram-level quantities. By integrating materials design, surface functionalization, and multi-dimensional characterization capabilities, BOC Sciences delivers highly controllable and customizable nanoparticle platforms, supporting continuous development from fundamental research to functional applications.

Table 3. Overview of Nanoparticle-Related Services at BOC Sciences.

Service Scope of Service Supported Applications Inquiry
Custom Nanoparticle SynthesisProvides customized synthesis of lipid-based, polymeric, inorganic, metallic, or hybrid nanoparticles; options for particle size, surface modification, and loading schemes.Targeted delivery system development, drug screening, material validation studies.Inquiry
Particle Size and Zeta Potential Analysis Uses dynamic light scattering (DLS) and zeta potential measurement to report size distribution, polydispersity index, and surface charge.Stability testing, formulation optimization, quality control.Inquiry
Morphology and Structural Characterization High-resolution imaging via TEM, SEM, AFM, and cryo-EM; includes optional EDS elemental analysis.Nanostructure optimization, verification of core-shell or layered architectures.Inquiry
Drug Loading and Release Profiling Quantifies encapsulation efficiency and loading capacity using UV, fluorescence, or HPLC; provides release kinetics under simulated conditions.Performance evaluation, formulation refinement, mechanism studies.Inquiry
Surface Functionalization and Conjugation Offers PEGylation, antibody/ligand attachment, fluorescent or magnetic labeling, and reversible charge modification.Targeted delivery studies, imaging and tracking, barrier penetration analysis.Inquiry
In Vitro Evaluation and Cellular Modeling Includes cellular uptake assays, confocal imaging, cytocompatibility analysis, and Transwell permeability testing.Nanocarrier–cell interaction studies, transport mechanism research.Inquiry
In Vivo Distribution and Imaging Analysis Employs fluorescence, MRI, or radiotracer techniques to monitor nanoparticle biodistribution, accumulation, and metabolic profiles.Pharmacokinetic modeling, validation of targeting strategies.Inquiry
Stimuli-Responsiveness Testing Simulates pH, redox, or temperature variations to assess nanoparticle response and release sensitivity.Smart nanocarrier design, responsive delivery system optimization.Inquiry

Summary

Nanoparticle-mediated targeted delivery combines precise carrier design, surface functionalization, and responsive mechanisms to achieve accurate transport of drugs and bioactive molecules. Across tumors, inflammatory sites, the CNS, and regenerative tissues, these systems enhance local concentration while reducing off-target exposure, optimizing tissue distribution and biological response. Particle composition, size, charge, morphology, and release kinetics can be tuned to overcome biological barriers. Integrated characterization enables rapid assessment of physicochemical and biological performance, guiding formulation refinement. BOC Sciences' end-to-end platform offers customizable design, synthesis, characterization, and scale-up, supporting continuous development from research to functional applications. This technology establishes a controllable, adaptable framework for efficient and precise tissue-targeted delivery.

References

  1. Image retrieved from Figure 2 "Schematic illustration for the in vivo process of controlled and targeted drug delivery system based on MCNs," Zhao, Qinfu, et al., 2017, used under [CC BY 4.0] (https://creativecommons.org/licenses/by/4.0/). The title was changed to "Schematic of nanoparticle-mediated targeted drug delivery in vivo".
  2. Zhao, Qinfu, et al., Mesoporous carbon nanomaterials in drug delivery and biomedical application., Drug Delivery 24.2 (2017): 94-107. https://doi.org/10.1080/10717544.2017.1399300
  3. Yetisgin, Abuzer Alp, et al., Therapeutic nanoparticles and their targeted delivery applications., Molecules 25.9 (2020): 2193. https://doi.org/10.3390/molecules25092193
* Please kindly note that our services can only be used to support research purposes (Not for clinical use).
Online Inquiry
Verification code