5.1. Oral Nanosuspension
Oral NS are designed to improve the solubility, dissolution rate, and bioavailability of poorly water-soluble drugs in BCS Class II and IV. By decreasing the particle size to the nanometer scale, NS significantly increase the surface area of the drug, leading to an increase in saturation solubility and faster dissolution. This causes a higher concentration gradient in the gastrointestinal tract, resulting in enhanced drug absorption and bioavailability. Furthermore, NS provide versatility in drug formulation and help to overcome the food effects that typically affect oral drug absorption. An investigation was undertaken to assess the in vivo kinetics of ritonavir NS in rats (fed and fasted conditions) and was compared to various formulations [
108]. Under fed conditions, the NS with an average particle size of 545 nm increased C
max by 8.9 times and AUC
0−t by 12.5 times that of the coarse powder, and by 1.9 and 2.1 times that of the commercial product (Norvir
®), respectively.
Oral suspensions are highly preferred for geriatric and pediatric populations due to their liquid form, which ensures chemical stability and ease of administration. A novel NS of benznidazole, a class II drug aimed at the pediatric population for Chagas disease, was produced using an organic solvent-free nanomilling approach [
109]. The prepared NS exhibited a particle size of <500 nm, an acceptable PDI (0.23), high ZP, and physical stability for at least 90 days. The formulation showed tenfold higher solubility and exhibited improved in vitro dissolution behavior, toxicity profiles, and efficacy against
Trypanosoma cruzi than pure drugs. NS also offer the benefit of masking the taste of drugs and prolonging the drug action. A successful preparation of a taste-masked phospholipid stabilized tacrolimus NS using the microfluidization method has been reported [
110]. A spectral analysis revealed interactions between tacrolimus and phospholipids, while a thermal analysis showed tacrolimus transformed into an amorphous state within the nanodispersion. The dissolution rate was significantly improved (35-fold after 0.5 h and 15-fold after 2 h). The nanodispersion significantly reduced gastric irritation, as evidenced by lower ulcerative indices (2.45) compared to raw tacrolimus (6.73).
Controlled surface modification can enhance the efficacy of NS under in vivo circumstances, with the surface composition playing a crucial role in organ targeting. TPGS and Tween 80 were applied on the curcumin NS’s surface by physical adsorption [
111]. Higher brain concentrations of curcumin were observed with Tween 80-coated NS compared to both the curcumin solution and TPGS-coated NS (
p < 0.05). The enhanced brain delivery of Tween 80 coated NPs was attributed to the absorption of ApoE and/or ApoB from the blood, facilitating transport into the brain via receptor-mediated endocytosis. Although TPGS-coated NS also exhibited higher brain targeting than the curcumin solution (
p < 0.05), TPGS-NS resulted in higher curcumin levels in the liver, spleen, and lungs.
Recent advances in NS technology include surface modifications to improve stability, targeted drug delivery for precision medicine, and innovations in preparation techniques like sonication and SCF methods. With their increasing application in clinical trials and the biopharmaceutical industry, NS represent an interesting approach for enhancing the therapeutic efficacy of many drugs.
Table 2 summarizes the formulation composition, preparation methods, and key features of oral NS.
5.1.1. Powders
Liquid NS face various physical and chemical stability problems, such as particle aggregation, sedimentation, and Ostwald Ripening along with microbial growth during storage. To address these problems, liquid NS were modified into solid forms through processes like lyophilization, spray-drying, or spray freeze-drying. Powders are mixtures of drugs and excipients that can be processed into more complex formulations such as tablets, capsules, and granules. NSs typically encounter stability challenges related to physical phenomena like Ostwald ripening and agglomeration and chemical reactions like hydrolysis. Therefore, pharmaceutical NS containing thermostable adjuvants can be transformed into dry powders using methods like spray-drying and fluidized bed drying, while techniques like freeze-drying and vacuum drying are more suitable for thermosensitive substances.
The drying process to create a compaction-ready powder or granule is crucial, as it can lead to undesirable solid characteristics and the potential growth of NPs. Indomethacin NS were created using wet media milling and dried by the fluidized bed technique using carriers including lactose, microcrystalline cellulose, and crospovidone, with varying binder contents while granulating [
118]. The pellets were compressed into tablets, which showed an improved in vitro dissolution performance, especially with a higher binder content (PVP) and spray rates that minimized NP growth compared to raw indomethacin tablets.
In a recent study, poorly aqueous-soluble and poorly bioavailable albendazole was formulated into an NS using the twin centrifugation with Kollidon
® VA64 and sodium lauryl sulfate as the stabilizers [
119]. The NPs, with a size of less than 300 nm, were coated with microcrystalline cellulose to form nanopellets and encapsulated in EUDRACAP
® with the objective of targeting the colon. Both the NS and granules demonstrated a substantial drug release (~60% and ~55%, respectively) at a colonic pH. The NS also showed strong anticancer activity, with IC
50 values of 1.18 µM in HCT 116 and 3.59 µM in HT-29 colorectal cancer cells.
A study demonstrated the conversion of drug NS into stable, redispersible, inhalable nano-agglomerates using in situ thermal gelation and spray-drying [
120]. Itraconazole NS were co-spray-dried with methylcellulose to provide superior thermal protection. Optimized spray-drying preserved the particle size with a near-unity Sf/Si ratio. Although various factors influenced the aerosol performance, no clear trends emerged. The nano-agglomerates exhibited an excellent in vitro aerosol performance, with fine particle fractions exceeding 50% and aerodynamic diameters ranging from 2 to 3 µm, making them well suited for deep lung delivery applications.
A key challenge in the process of modification into solid forms is ensuring that the NPs can redisperse effectively when mixed with water or gastric fluids. Redispersants such as sucrose, trehalose, lactose, maltodextrin, and mannitol play a crucial role in preserving the integrity of NPs when NS are dried into powders through diverse techniques [
121]. Their primary function is to ensure that the dried NPs redisperse back to their original size upon reconstitution in water or biological fluids, preventing aggregation during the drying process. These sugars act as bulking agents to enable rapid redispersion forming while also providing cryoprotection during the freeze-drying process. Selecting the optimal redispersant based on the drying technique and drug properties is crucial for maintaining the particle size and stability, as well as preserving the enhanced dissolution, bioavailability, and stability of the drug [
122]. Suitable lyophilization protectants/stabilizers include a variety of substances such as sugars (e.g., sucrose or trehalose), hydroxy alcohols (e.g., glycerol or sorbitol), amino acids (e.g., glycine or arginine), and mixtures of monosaccharides. Two main theories explain their protective mechanisms during lyophilization: the water substitution hypothesis and the glassy state hypothesis. The water substitution hypothesis suggests that these protectants form hydrogen bonds with proteins or the surfaces of NPs, replacing water molecules and stabilizing the structure during the drying process. During dehydration, sugars replace water, which stabilizes the distance between NPs and maintains the stability of the NS. In contrast, the glassy state hypothesis posits that as water is removed, the solution becomes highly concentrated and forms a noncrystalline, glassy state. This viscous environment surrounds proteins, preventing their movement and preserving their structure and properties while also protecting the NPs.
A recent study identified the optimal formulation for the lyophilization protectant consisting of oligomeric mannose (0.46%
w/v), maltose (0.44%
w/v), and sorbitol (0.05%
w/v). Under these conditions, the lyophilized astaxanthin NP powder had a resoluble particle size of 472 nm, which is 1.32 times larger than the particle size before lyophilization. When the resultant powder was stirred with water, it formed a pink, fluffy coating that vanished entirely in ten seconds [
123].
A study systematically assessed the impact of freeze-drying, processing conditions, and different cryoprotectants on cilostazol NS, with a primary focus on the particle size after redispersion [
124]. The physical characteristics of formulations with trehalose (10%), maltodextrin (5%), or PEG 1500 (10%) were further investigated. The freeze-dried NCs successfully retained their original size and polymorphic form A, while significantly enhancing their dissolution rates (over 90% in 5 min) compared to untreated cilostazol (less than 30% for 60 min). Research has demonstrated that sucrose laurate is an effective stabilizer, allowing dried NCs of neutral drugs like fenofibrate, danazol, and probucol (150–300 nm) to disperse fully into their original sizes with mild agitation after drying by various methods [
125]. The preliminary findings suggest sucrose laurate can also disperse acidic and basic drugs, indicating wide applicability. Similarly, lactose laurate, another stabilizer, performed well.
Hummer acoustic resonance technology (HART) is an advanced approach for NS preparation, enabling the efficient development and evaluation of multiple formulations with high throughput capabilities and excellent scalability for process optimization [
126]. In a study, the optimal andrographolide NS was produced with a size of 223.99 ± 3.16 nm, PDI of 0.095 ± 0.007, and a ZP of –33.20 ± 0.58 mV, using PVP K30 and SDS as stabilizers. Significantly improved dissolution rates for NS and its solidified forms in both pH 1.2 and pH 6.8 buffers were reported as compared to the controls. In summary, HART offers a promising method for converting NS into solid powders, with benefits including precise control over the particle size and the preservation of sensitive drugs. However, its high energy consumption and complex scaling requirements may present challenges for large-scale applications.
Electrospray is a versatile technique used to produce powders from NS and surface coatings by applying an electric field to break the liquid into fine droplets, which then undergo solvent evaporation to form solid NPs [
127]. This method allows for precise control over the particle size and morphology, which is essential for optimizing drug delivery. Electrospraying is particularly advantageous for converting liquid NS into solid forms, making them easier to handle and store, while also preserving the stability of heat-sensitive drugs due to its relatively low operating temperature. However, challenges include scaling up the process and optimizing parameters such as the solvent choice, voltage, and flow rate [
128].
The electrospinning technique is an advanced process used in the preparation of NS, where an electric field is employed to produce fine polymer fibers or particles from a liquid solution or melt. This method enables the encapsulation of poorly water-soluble drugs within nanostructures, enhancing their solubility and bioavailability. It is particularly advantageous for its simplicity, scalability, controlled particle size and morphology, and ability to incorporate multiple therapeutic agents or excipients into the NS. A recent study demonstrated that polydopamine-coated paclitaxel-PEG NCs embedded in electrospun nanofibers enhanced the antitumor efficacy in a murine cervicovaginal tumor model. The nanofiber implant also achieved prolonged vaginal residence, improved transmucosal penetration, and minimal mucosal irritation [
129].
5.1.2. Pellets
Pellets, as multiparticulate dosage forms, provide numerous benefits, including a lower risk of dose dumping, shorter gastric residence time, and reduced dependence on gastric emptying. They also minimize intersubject variability in gastrointestinal transit times, enhance distribution to reduce local irritation, improve bioavailability, lower systemic toxicity risks, and offer more consistent pharmacokinetic behavior compared to traditional formulations. Additionally, pelletized NS offer several advantages, including excellent stability, good flow behavior, ease of dosing, low hygroscopicity, a high bulk density, and a dense, uniform surface that enhances their overall performance in various applications. Several pelletization techniques, including extrusion spheronization, powder layering, spray granulation, melt extrusion, and cryopelletization, can be used to convert NS into solid pellets, thereby improving their physical and chemical stability.
The hot melt extrusion (HME) method involves applying pressure and heat to melt or soften materials, which are subsequently extruded through a die to create uniformly shaped products. The advantages of the HME technique include the elimination of solvents and water, fewer processing steps that reduce the drying time, simplicity, continuous efficiency, the uniform dispersion of fine particles, and good stability under varying pHs and moisture conditions. HME is used for taste masking, enhancing the solubility of poorly soluble drugs, and preparing modified-release solid oral dosage forms. It is commonly employed to create amorphous solid dispersions that increase the dissolution rates, though they may face stability issues such as recrystallization during storage. Other key limitations of the HME method include high energy requirements, limited applicability to heat-sensitive materials due to the elevated temperatures, and the potential melting or softening of low melting binders during the handling and storage of the agglomerate. Dried amorphous clotrimazole NS were prepared by combining antisolvent precipitation with HME technology [
130]. The NS and matrix forming microcrystalline cellulose were directly fed into the extruder using separate feeding devices, improving drug uniformity in the extrudate, which is crucial for low-dose formulations. Key process factors like the feed rate and input temperature were found to influence the product’s redispersibility and moisture content. A moderate inlet temperature effectively removed the residual moisture without affecting the redispersibility of the dried NS. The drug remained amorphous post extrusion, as confirmed by DSC and XRPD, while polarized light microscopy was ineffective due to the presence of the semicrystalline nature of microcrystalline cellulose. It is important to note that the coexistence of crystalline and amorphous forms can be assessed using suspended-state NMR and Raman spectroscopies. In vitro dissolution studies showed enhanced solubility and dissolution, linked to the amorphous nature of the nanosized drug particles. A comparative evaluation of the impact of the matrix surface area of solid dispersions with hydrophilic polymers via the nanoextrusion technique and amorphous solid dispersions in enhancing the dissolution rate of a potent poorly aqueous-soluble model drug, griseofulvin, has been investigated [
131]. While fine grinding was necessary for the extrudates to achieve an immediate release, coarse milling was sufficient for the nanocomposite to rapidly release low-dose drugs. Lastly, this work suggests that drug loading, drug release, and the specific surface area of the milled dispersions interact in a complex way.
The advantages of extrusion spheronization include a uniform pellet size, controlled release properties, and suitability for both low- and high-dose formulations. Limitations include the need for specialized equipment, a time-consuming process, and challenges in scaling up for large-scale manufacturing. Wet-milled and normal suspensions were added to solid carriers made from microcrystalline cellulose, isomalt, and crospovidone to create matrix pellets via the extrusion/spheronization technique [
132]. The NPs were successfully reconstituted from the solid dosage forms, resulting in a decrease in particle size and a significant enhancement in the thermodynamic solubility and the dissolution rate of cilostazol in gastric media in comparison to pure surfactant dispersions. Differential scanning calorimetry and XRD confirmed that cilostazol transitioned from Form A to an amorphous state as a result of the extrusion process.
Simvastatin NS were prepared using 7% HPMC (stabilizer), 0.03% butylated hydroxyanisole (antioxidant), and 0.2% citric acid (synergistic antioxidant action) via low-temperature grinding [
133]. Drug and SDS in a 1:5 ratio were uniformly dispersed and applied to sugar pellets using a fluid bed granulator. The NS had an average particle size of 0.74 µm, with 80.6% of the particles being smaller than 1 µm. The relative bioavailability of the drug from the NS’s layered pellets was higher (probably due to a greater dissolution rate compared to a commercial tablet).
5.1.3. Tablets
Pharmaceutical tablets are solid dosage forms manufactured through compression or molding, typically using granules combined with appropriate excipients. NS can be transformed into dry powders by various techniques as well as stabilizers, as described in
Section 5.1.1. For instance, naproxen granules prepared from a nanodispersion using spray-drying were compressed into tablets using mannitol, which acted as a bulking agent, stabilizer, and disintegrant. These tablets exhibited rapid dissolution, completely dissolving within one minute under both sink and nonsink conditions [
134]. Similarly, lovastatin NCs were developed through spray-drying using PVP K17 (20%) and sodium lauryl sulfate (5%) as the stabilizers. The resulting sustained release tablets were optimized using lactose as a diluent, Avicel PH101 as a compression aid, and Ac-Di-Sol as a superdisintegrant [
135].
Spray-drying is a versatile, single-step, continuous process that transforms liquid feeds into dried particles, making it an effective platform for particle and crystal engineering. While spray-drying offers a straightforward method for converting NS into powders, its limitations include the risk of degradation of heat-sensitive drugs, the potential for particle agglomeration, high energy consumption, and challenges in controlling particle morphology. These limitations need to be considered carefully when using spray-drying for the preparation of pharmaceutical NS. The effect of dispersants on the dissolution of poorly aqueous-soluble itraconazole nanocomposites prepared using spray-drying techniques was studied [
136]. Superdisintegrants like sodium starch glycolate, crospovidone, and croscarmellose sodium, co-milled with itraconazole stabilized by HPC, demonstrated faster dissolution compared to sugars like mannitol and sucrose used as matrix formers. A correlation between the dispersants’ efficacy and swelling capacity points to a mechanism of erosion or disintegration brought on by swelling. This approach enabled the formulation of nanocomposites with a high drug-loading capacity (>60%
w/
w) and immediate drug release. Another study demonstrated the superior dissolution rate of risperidone spray-dried NS formulated into orally disintegrating tablets compared to commercially available products [
137]. Additionally, research has explored converting PLGA polymeric NP suspensions into solid dosage forms using fluid-bed processing. It was noted that dried intermediates containing polymeric NPs could successfully redisperse into their original nanoparticulate form after dispersion [
138]. Key factors such as the NP concentration in the granulation suspension and the ratio of the spraying rate to the atomization air pressure were found to critically influence the redispersibility and physicochemical properties of the granules.
A successful spray-dried NS formulation depends on the drying temperature, matrix-former type and content, and particle size. NS of naproxen and itraconazole, stabilized with Kollidon
® VA 64 and sodium lauryl sulfate, were spray-dried using lactose, trehalose, or sucrose as the matrix formers [
139]. The results revealed that the outlet temperature and drug content significantly influenced the redispersibility of the NS. The maximum achievable drug content for a redispersible product depended on the outlet temperature, the matrix former’s glass transition temperature, its proportion, and the NS’s particle size. Agomelatine NS were prepared using wet media milling using HPC as the stabilizer and subsequently solidified through spray-drying. A polymorphic change from agomelatine form I to form II occurred during the milling process. Crystal lattice simulations indicate that form I crystals exhibit high mechanical anisotropy, which may contribute to their rapid particle size reduction before undergoing polymorphic transformation. The spray-dried NS was then processed into mini tablets, which demonstrated significantly faster dissolution compared to physical mixtures of agomelatine [
60]. In another study, celecoxib was formulated into NS via precipitation and HPH, followed by spray-drying or freeze-drying [
140]. The choice of solvent, stabilizer, and surfactant significantly impacted the NP crystallization, size, and solubility. Both drying methods preserved the drug’s chemical stability and enhanced the dissolution rates compared to the raw drug. However, freeze-dried NS showed a brief initial delay in dissolution, followed by a lag phase due to particle aggregation.
Freeze-drying is a valuable pharmaceutical batch process for removing solvents, particularly water, while preserving the product integrity. It enhances the stability of thermolabile drugs, particularly for long-term storage, produces highly porous products with a low moisture content, and enables the preparation of reconstitutable NS. While effective, it is time consuming, energy intensive, and costly, which limits its scalability. The freeze-drying process can destabilize NPs due to freezing and desiccation stresses, where phase separation during freezing results in ice and a cryo-concentrated suspension with a high concentration of NPs, causing particle agglomeration. Ice crystallization can also exert mechanical stress, further impacting NP stability. To prevent product collapse, primary drying should occur below the collapsed temperature, and cryoprotectants should be added before lyophilization to mitigate these stability challenges [
141]. A study was conducted to evaluate the impact of freeze-drying with different cryoprotectants, such as TPGS and folate-modified distearoylphosphatidyl ethanolamine-PEG, on the physicochemical properties of resveratrol and quercetin NS [
142]. NS formulated using the antisolvent precipitation method exhibited resveratrol and quercetin NPs measuring approximately 210 nm and 110 nm, respectively. The dissolution rate of the drug NPs increased by 6–8-fold and exhibited long-term stability. Silymarin, which has limited water solubility and oral bioavailability, was formulated into lyophilized NS tablets using a sonoprecipitation technique accompanied by freeze-drying [
143]. PVA served as a stabilizer and binder, while mannitol functioned as a cryoprotectant and disintegrant. The optimized formulation displayed a porous structure, rapid disintegration, friability below 1%, and enhanced solubility and dissolution rate. The active freezing technique involves closely monitoring and regulating the freezing process to achieve a specific freezing rate or temperature profile. The method is used to convert metastable NS into redispersible nanocrystalline powders that are ideal for oral drug delivery [
144].
5.1.4. Capsules
Hard gelatin capsules offer multiple benefits, including ease of formulation, as they can accommodate various drug types like powders, granules, and even NS. For example, NS-loaded capsules of Novartis compound A and itraconazole demonstrated enhanced bioavailability in animal studies. The gelatin shell dissolves rapidly, ensuring the quick release and absorption of the drug. Capsules also mask unpleasant tastes and odors, improving patient compliance, and come in various sizes, providing flexibility in dosing. Additionally, they offer tamper evidence for added security and ensure precision and uniformity in dosing. Many patients find capsules easier to swallow than tablets, making them a preferred choice for oral medications.
A study optimized
Kaempferia parviflora NS to enhance intestinal absorption using antisolvent precipitation [
145]. The optimal formulation used sodium lauryl sulfate (3%), achieving stable particle sizes (100–300 nm) with high stability over one month. The NS encapsulated in a hard gelatin capsule showed rapid dissolution (over 80% within 30 min) and increased intestinal absorption by about 10-fold, demonstrating its effectiveness for improved bioavailability.
5.1.5. Film
Buccal films adhere firmly to the oromucosal membrane to provide precise dosing and enhanced drug absorption, making them ideal for both local and systemic treatments [
146,
147,
148]. These films are particularly suited for buccal use due to their versatility, comfort, flexibility, lightweight nature, adaptability, and resilience under mechanical stress, as well as their customizable size [
147,
149]. Integrating NS into films offers several benefits including a minimized premature or excessive drug release, and decreased fluctuations in plasma levels and interpatient variability. NS, particularly NCs, can accommodate a variety of hydrophobic drugs for sustained release. A three-layer buccal film containing carvedilol NS was developed, featuring an outer mucoadhesive layer, an NS core, and an innermost backing membrane [
145]. The NS had a mean particle size of 495 nm, a negative ZP of −17.21 mV, and a PDI of 0.203. When added to an HPMC and carbopol hydrogel layer, this film significantly enhanced the bioavailability (916%) and achieved a 7.3-fold higher Cmax and Tmax of 4 h compared to tablets, mainly due to the nanosized particles. Another study developed and evaluated oral mucoadhesive films containing clotrimazole NS to treat oral candidiasis [
150]. The clotrimazole NS were formulated using a bottom-up method with a surfactant, benzyl succinyl chitosan, and incorporated into a film made from catechol-functionalized hyaluronic acid and PVA. The films released clotrimazole slowly, achieving complete release within 6 h, and were nontoxic to normal cells while demonstrating significant antifungal activity compared to a traditional clotrimazole suspension.
The primary aim of orodispersible film is to dissolve or disintegrate rapidly in the oral cavity, forming a solution or suspension that is subsequently swallowed for absorption [
146]. Researchers improved the bioavailability of olmesartan medoxomil by formulating it as an NS using an antisolvent precipitation ultrasonication method, resulting in NPs with a size of 120 nm and ZP of −45 mV [
151]. The optimized fast-dissolving film showed rapid disintegration (20.50 s) and achieved high drug dissolution rates (87.53% in 6 min and 95.99% in 10 min). Compared to standard tablets, the fast-dissolving tablets substantially increased the bioavailability (209.28%), Cmax (from 66.62 to 179.28 ng/mL), and AUC
0–72 (from 498.36 to 1083.67 ng h/mL).
The ginkgolide B NS lyophilized powder-based orodispersible film was developed using solvent casting, with its formulation optimized through single-factor and orthogonal tests [
152]. The NS, prepared via media milling, was lyophilized with mannitol as a lyoprotectant. The final film, composed of NS (35.6%), PVA (49.4%), PEG 400 (10.7%), and sodium carboxymethyl starch (4.3%), exhibited ideal properties, including a rapid disintegration time of ~30 s and consistent particle sizes (~210 nm in a reconstituted form). It maintained stability and uniformity for 30 days. An electron microscopy image showed a smooth surface with evenly distributed particles (~200 nm), XRD analysis indicated reduced crystallinity, and in vitro testing revealed complete dissolution within 10 min.
A fast-dissolving orodispersible film incorporating nitrendipine NPs has been reported [
153]. Drug NS were prepared using the antisolvent sonoprecipitation method and subsequently processed into films through solvent casting. The optimized NS achieved a particle size of ~500 nm and significantly improved solubility (~26 times than pure drug). The film exhibited desirable properties, including thickness (0.148 mm), tensile strength (8.25 kg/cm
2), and a rapid disintegration time (24.60 s). Dissolution studies showed a complete drug release (8 min from lyophilized NCs and 3.5 min from film) compared to 30 min for conventional tablets. Pharmacokinetic tests in rabbits demonstrated higher bioavailability from films than from tablets.
The improper drying of drug NPs can cause irreversible aggregation, decreasing the dissolution rate. Loading drug NS onto films offers a simple and efficient solution to this issue. An optimized fast-dissolving oral film with paroxetine NS was developed using the solvent casting method with the aid of a full factorial design [
154]. The optimized film demonstrated strong mechanical properties, a short disintegration time (17 s), and greater dissolution (96.02% in 10 min). Permeation studies using the chicken buccal model showed over threefold increased drug absorption compared to the pure drug. Clinical testing in healthy volunteers confirmed the improvement in the relative bioavailability (178.43%) in comparison to a marketed tablet, highlighting the effectiveness of such a formulation in enhancing drug bioavailability.
The 3D printing of pharmaceuticals enables the creation of customized drug delivery systems, such as tablets and films, with tailored shapes, sizes, and release profiles. Incorporating NS into these films enhances the solubility and bioavailability of poorly soluble drugs by increasing the surface area and dissolution rates [
155]. This approach allows precise control over drug loading, release, and personalized dosing, offering flexibility for combining multiple drugs or excipients. A rapidly dissolving oral film loaded with indomethacin NCs was developed using a semi-solid extrusion 3D printing method [
156]. HPMC served as the film-forming polymer, glycerol as the plasticizer, and Poloxamer F68 as the stabilizer, producing particles with a size of 230 nm and PDI values below 0.20. NCs maintained their size range (300–500 nm) in all formulations. Films prepared with optimal polymer concentrations (2.85% and 3.5%
w/v) were flexible, homogeneous, and disintegrated within 1–2.5 min, achieving complete drug release in 23 min. Additionally, a solvent-free, low-temperature 3D printing technique, termed melting solidification printing, was used to embed albendazole NCs in printlets [
157]. These printlets demonstrated improved dissolution rates compared to spray-dried NCs, with physical and chemical stability maintained for six months during storage. A comparison illustrating the conversion of NS to various formulation types, along with their advantages, limitations, preparation methods, and typical applications, is summarized in
Table 3.
5.2. Ophthalmic
Conventional ophthalmic products face low ocular bioavailability due to anatomical, physiological, and biochemical barriers, and the physicochemical properties of the drug. With their smaller particle size and improved mucoadhesion, NS stay longer in the eye’s cul-de-sac, providing sustained release and avoiding frequent dosing. The growing use of NS in ocular drug delivery offers new possibilities for overcoming the limitations of conventional ophthalmic formulations, potentially improving treatment outcomes for conditions like infections and inflammation. For instance, voriconazole NS prepared using the emulsion solvent evaporation technique with Eudragit
® RS 100 as a stabilizer and N-methyl-2-pyrrolidone as a permeation enhancer demonstrated excellent ocular bioavailability in treating fungal keratitis caused by
Candida albicans [
85]. These findings emphasize the potential of NS as an effective platform for delivering poorly water-soluble drugs, including immunosuppressants, for the treatment of various ocular conditions. Utilizing advanced technologies can address stability challenges in ocular NS, resulting in more effective and dependable treatments for eye diseases [
158].
Table 4 lists NS formulations developed for various ocular conditions along with their attributes.
A mucoadhesive gellan gum-based in situ gelling NS of posaconazole has been developed to enhance the contact time in ocular tissues [
165]. The NS was formulated using microfluidization and optimized utilizing QBD principles with gellan gum (0.4%
w/v) to provide adequate viscosity and mucoadhesiveness. The NS exhibited a larger zone of inhibition (~15 mm) compared to the marketed itraconazole NS (~11 mm). It was determined to be nonirritant, receiving a potential irritancy score of 0.85. Furthermore, the NS achieved a higher drug release rate (~35%) compared to a coarse suspension (~10%), and permeation studies in goat corneas showed that around 70% of the drug was retained in the membrane. NS of moxifloxacin and pamoic acid, designed for mucus penetration, showed superior efficacy against
Staphylococcus aureus in bacterial keratitis with once daily dosing, outperforming standard eye drops administered thrice daily [
166]. Pharmacokinetic studies indicated effective drug distribution in anterior ocular tissues.
Unlike conventional suspensions, which may contain larger particles that cause irritation, NS minimize discomfort, enhance patient comfort, and require the minimal use of potentially harmful solvents [
59]. They also avoid the high osmolarity associated with some ophthalmic solutions, reducing potential damage to ocular tissues. Additionally, NS increase the saturation solubility of poorly water-soluble drugs, making them more effective in lachrymal fluids where conventional suspensions fall short. NS offer formulation versatility as they can be combined with hydrogels, ointments, or mucoadhesive bases to achieve tailored release profiles and extend residence times. Moreover, certain formulations containing Eudragit polymers help to stabilize sensitive drugs, prolonging their shelf life and effectiveness. These advantages make ocular NS particularly beneficial for treating chronic eye conditions and infections, where a sustained release and minimal irritation are crucial for successful treatment outcomes. Eudragit RS 100 was used as a polymer to optimize and enhance the intraocular delivery of itraconazole NS prepared via solvent evaporation [
167]. The data demonstrated that the optimized NS possess moderate particle sizes (332.7 to 779.2 nm), ZP (+0.609 to 16.3 mV), and entrapment efficiency (61.32% to 76.34%). Additionally, higher corneal permeation and antifungal efficacy against
Candida albicans and
Aspergillus flavus were shown by NS when compared to a marketed formulation and an itraconazole eye drop formulated with sulfobutyl ether-β-cyclodextrin.
Carbon dots, spherical NPs under 10 nm in size, are particularly effective due to their strong fluorescence, high water solubility, small size, ease of synthesis and modification, low toxicity, and good biocompatibility [
168,
169]. A composite ocular drug delivery system was developed for the topical administration of diclofenac, utilizing carbon dots synthesized through a one-step hydrothermal method from hyaluronic acid and carboxymethyl chitosan [
170]. These carbon dots were incorporated into a thermosensitive in situ gel made of poloxamer 407 and 188, creating a gel with sustained release capabilities for 12 h. Ex vivo fluorescence studies demonstrated that the gel enhances bioimaging and tracing in ocular tissues, with a 3.45-fold increase in drug bioavailability in the aqueous humor (compared to eye drops), indicating prolonged corneal retention.
Advancements in drug-loaded contact lenses have transformed ocular drug delivery by enhancing both efficacy and convenience over traditional eye drops [
171]. These lenses are designed with embedded drug reservoirs or NPs that gradually release therapeutic agents over extended periods, improving treatment adherence and reducing side effects. Recent innovations include hydrogel-based lenses and silicone hydrogel materials that support longer wear and precise drug dosing, making them promising for chronic conditions like glaucoma, dry eyes, and infections. On the other hand, these lenses often provide only a short release duration, which may not be sufficient for long-term treatment needs [
171]. Additionally, they can face issues with storage stability, as some drugs may degrade over time when stored in the lenses. A novel contact lens with a pH-sensitive inner layer provides an extended, stable drug release for ocular therapy [
172]. Fabricated with ethyl cellulose, Eudragit S100, and polyhydroxyethylmethacrylate hydrogel, the lens achieves an optimized daily release of diclofenac and can be stored with minimal drug loss. In vivo studies indicate sustained drug delivery for over 24 h, making it a promising option for treating various eye conditions.
Intravitreal injections can effectively deliver small molecules (less than 500 Da) directly to the vitreous humor. However, the repeated use of this route for drug administration may lead to complications related to the retina and an increase in the intraocular pressure [
173]. Research was undertaken to develop a hybrid system combining NS and DMNs as a minimally invasive alternative for the trans-scleral delivery of triamcinolone acetonide NS with a particle size of 246.65 ± 8.55 nm [
174]. The NS-loaded DMNs were capable of piercing excised porcine sclera, achieving insertion depths of over 80% of the needle height and dissolving rapidly (less than 3 min), while plain-drug-loaded DMNs took more than 8 min to dissolve. Trans-scleral deposition studies indicated that the NS-loaded DMNs deposited 56.46 ± 7.76 μg/mm
2 of the drug in the sclera after 5 min, a 4.5-fold increase compared to the plain-drug-loaded DMNs. The drug NS containing the DMN array demonstrated biocompatibility with ocular tissues when evaluated using the hen’s egg chorioallantoic membrane assay and cytotoxicity. Overall, this hybrid system of NS and DMNs offers a promising, minimally invasive approach to treating retinal diseases effectively.
NS are prone to instability, often resulting in agglomeration or sedimentation that reduces their efficacy. While stabilizers and surface modifiers are typically used, they can introduce toxicity and complicate regulatory approval. Researchers are investigating the use of advanced materials like biodegradable polymers and lipid-based carriers to improve long-term stability while ensuring biocompatibility and safety [
175]. In this context, stimuli-sensitive drug delivery systems that respond to pH, temperature, and enzyme activity appear to be interesting solutions to ocular NS stability issues. Microfluidics and 3D printing are promising techniques for creating stable, finely designed NS. Microfluidics provide precise NP production, lowering the possibility of aggregation, whereas 3D printing enables customized drug delivery systems, improving the ocular dosage accuracy and facilitating patient-specific therapies. The primary distinctions between microfluidizers and conventional HPH lie in the type of pump utilized, the pressure levels they can achieve, and the presence or absence of a nozzle [
71]. A study focused on enhancing the stability of iodine NS (
127I) through nanoprecipitation using microfluidic devices and examining the influence of preparation parameters on their stability [
176]. Artificial neural networks were employed to optimize the process, analyzing the relationships between the input variables (solvent temperature, antisolvent flow rate, and solvent flow rate) and output parameters (sedimentation time and PDI) across the test samples. The optimization revealed that the microfluidic preparation parameters significantly impacted the stability of the NS.
5.3. Transdermal
Transdermal drug delivery encounters significant formulation challenges, particularly due to the existence of intercellular lipids within the stratum corneum (SC), which form a strong permeability barrier, as well as the physical barrier provided by keratinocytes [
177]. However, small lipophilic molecules and certain hydrophilic or polar compounds can penetrate through the living epidermis and finally into the dermis, where there are blood capillaries. The physicochemical properties of a drug, governed by factors such as the molecular size, concentration, and solubility, are critical for successful transdermal delivery [
178]. According to Lipinski’s Rule of Five, drugs with a molecular weight under 500 Da, low polarity, and high lipophilicity are more likely to reach the dermis. The Stokes–Einstein equation further highlights the importance of molecular size and diffusion, as smaller molecules tend to diffuse more easily through the compact structure of the SC [
179,
180]. In addition, the nonionic state of NS at the site of application (i.e., SC with pH 4.2 to 5.6) can improve drug penetration [
181,
182]. Additionally, factors such as the application area, method, duration, skin age, and the use of carriers that modify the skin’s barrier function play a role in the overall effectiveness of transdermal therapy [
179].
NS-based transdermal delivery received much attention after the commercialization of cosmetic products of rutin (Juvedical
®) [
183]. Two possible mechanisms for the transdermal delivery of NS are suggested: a) an increased transdermal concentration gradient and b) extended retention on the skin’s surface to sustain the concentration gradient [
183]. The transdermal drug delivery of NS can occur through multiple pathways, including intracellular and intercellular penetration, as well as delivery via hair follicles, sweat glands, or sebaceous glands [
184]. For intracellular penetration through SC cells, the drug must exhibit favorable physicochemical properties such as optimal solubility, a distribution coefficient, drug concentration, cell membrane permeability, and the chosen mode of administration. The particle size of the NS significantly influences the efficiency and pathway of the transdermal transport. For example, a study comparing curcumin NPs of 140 nm, 400 nm, and 730 nm demonstrated that smaller particle sizes can enhance the penetration efficiency, highlighting the critical role of nanoscale dimensions in optimizing transdermal delivery. The 400 nm NS had a higher steady-state flux, but their cumulative penetration was lower than that of the 140 nm and 730 nm formulations [
183]. It was noticed that the curcumin molecules dissolved in the SC were rapidly distributed throughout the skin and hair follicles, whereas intact NS did not penetrate the skin. The NS were found to more easily accumulate in hair follicles due to their large openings, ranging from 10 to 210 μm in diameter.
Microneedles bypass the skin’s barrier, allowing the efficient administration of various therapeutics, including vaccines and peptides. Once inserted into the skin, DMNs dissolve, releasing the embedded drug or therapeutic agent into the body. It was proposed that drug NPs accumulated as a depot using DMNs could quickly dissolve in the skin’s interstitial fluid and subsequently be absorbed into the rich dermal microcirculation to achieve the desired plasma drug concentration levels [
185]. The successful integration of drug-loaded NS into DMNs for transdermal delivery has been reported [
186]. In this study, cholecalciferol NS were developed via sonoprecipitation to improve transdermal delivery by incorporating them into mechanically strong DMNs using a centrifugation-assisted micromolding method. The drug NS, stabilized with PVA, achieved a particle size of ~300 nm and demonstrated good mechanical strength when coupled with PVA. The DMNs penetrated approximately 375 μm into the skin model (Parafilm M
®). A permeation study using porcine skin showed that drug-NS-loaded DMNs significantly enhanced skin permeation (498.19 ± 89.3 μg) compared to drug NS patches without microneedles (73.2 ± 26.5 μg) over 24 h. Promising results were reported with other drugs like itraconazole [
187], metronidazole [
188], baclofen [
189], and ibuprofen [
190], highlighting the effectiveness of NS and microneedle technology in improving cutaneous drug delivery. Selected examples of NS formulations for transdermal delivery, detailing the active ingredient, formulation technique, and key highlights, are tabulated (
Table 5).
5.4. Pulmonary
Pulmonary drug delivery systems administer medications directly into the lungs through inhalation, making them highly effective for treating respiratory diseases such as asthma and chronic obstructive pulmonary disease. Key advantages include targeted delivery, rapid absorption via the alveoli’s large surface area, and lower systemic exposure, which reduces the side effects. However, these systems can be device-dependent, with efficacy influenced by proper inhaler use and patient inhalation patterns. For instance, conventional aerosols face several challenges, and particle agglomeration and aggregation lead to deposition in the pharynx and upper respiratory tract [
198]. The passive or active targeted administration of a drug to a specific tissue, either an intracellular compartment or cell, by managing release kinetics and shielding the therapeutic agents improves its therapeutic index by enhancing its specificity [
199]. Furthermore, the location of aerosol accumulation in the respiratory system is controlled by the aerosol’s physical qualities, inhalation circumstances, and the structure of the respiratory airways [
200].
The human lung has several mechanisms such as mucociliary clearance, mechanical clearance, alveolar macrophages, and enzymatic degradation in various regions which limits the diffusion and deposition of aerosols in the lung [
201]. While these mechanisms safeguard the respiratory tract from harmful exposure to foreign substances, they also present significant challenges for administering drugs via inhalation [
202]. Recent research on oropharyngeal drug deposition has revealed additional factors influencing transport and deposition, such as the particle velocity, mouthpiece diameter, and electrostatic effects associated with the delivery device [
198]. Apart from impaction, sedimentation, and diffusion, the fundamental deposition mechanisms depend mainly on the size of the inhaled particles and their inhaling rate [
203]. The pulmonary delivery of medications via aerosol is mainly facilitated by different inhalation devices (nebulizers, metered dose inhalers, dry powder inhalers, and soft mist inhalers), categorized according to the physical state [
204].
Targeted NP delivery to the lungs is an emerging field, offering the advantages of a reduced dosage and minimized side effects by limiting systemic exposure. NS offer significant benefits in pulmonary drug delivery [
205]. They enhance the bioavailability of poorly soluble drugs, extend the retention time by adhering to the lung’s mucus layer, and allow for reduced dosages by concentrating the drug in the lungs. Recent advances in this field include NP-based inhalers with improved aerosol properties, dry powder NS for better stability, and functionalized NPs targeting specific lung cells. These advancements have enormous potential to enhance drug solubility, deliver drugs precisely, and reduce adverse effects in pulmonary therapy. The main physical parameters affecting the overall and regional deposition of inhaled chemicals in the respiratory tract are aerodynamic particle size distribution and particle velocity [
206]. It is common practice to use the mass median aerodynamic diameter to assess the mass distribution of aerosol particles. For the fine particle fraction, which represents the percentage of particles with an aerodynamic diameter of less than 5 μm, the optimal size for lung deposition is calculated by the ratio of the fine particle dose to the delivered dose [
207]. To achieve a respirable particle size and maximize the shelf life, excipients like mannitol, sucrose, sodium chloride, and trehalose are often incorporated [
208]. Several biopharmaceutical products, predominantly protein-based, are currently approved in the United States and Europe for treating various diseases [
209]. Given the large size of biopharmaceutical molecules, absorption enhancers are needed for their effective uptake at the alveolar level, although these enhancers may increase the particle size due to entrapment or encapsulation. It is crucial to control both the aerodynamic size and surface properties of the particles, as micronization can increase the particle charge and aggregation risk. Additionally, the rapid elimination of proteins from the lungs necessitates frequent dosing, which can affect patient compliance [
210].
The drug’s nanoparticulate form allows for a quicker onset of action since it diffuses and dissolves more quickly in alveolar fluids. Additionally, the nano-products can prolong the drug release because of their enhanced affinity for mucosal surfaces. The NPs’ unique physicochemical properties, including uniform and narrow size distribution, make uneven drug distribution and delivery to the lungs unlikely compared to aerosols. When comparing hydrophobic budesonide NS to coarse and micronized drug particles, the lung distribution rate of the former was substantially higher (872.9 ng/g) and distinct (
p < 0.05) [
211].
Spray-freeze-drying is an effective approach for converting NS into dry powders and is particularly suitable for producing low-density porous particles for inhalation drug delivery [
212]. In this process, active agents and excipients are co-sprayed into liquid nitrogen, quickly freezing the droplets, which are then freeze-dried into powders. Cefixime NS were solidified by spray-freeze-drying to produce inhalable microparticles [
213]. The fine particle fraction values varied from 18.96% to 79.28%, with the highest achieved using trehalose at a 1:1 NP/carrier ratio and 20% leucine. Particle sizes ranged between 5.24 and 10.17 μm, with mannitol formulations having the broadest distribution and trehalose the narrowest. Most particles were spherical with varying porosity and no needle-like structures. The release rates for the selected formulations were 89.33% and 93.54% within the first 10 min, respectively. Recent examples of NS used in pulmonary drug delivery and their highlights are illustrated in
Table 6.
5.5. Parenteral
The parenteral administration, especially via the intravenous route, is often preferable for quick action, bypassing the first-pass liver metabolism, and delivering drugs directly to the target site. However, developing injectable products for poorly aqueous-soluble drugs presents major challenges. Techniques currently used in increasing the drug solubility are by making salts or by adding co-solvents, surfactants, or cyclodextrin complexes, but they have limitations such as incomplete solubilization, hypersensitivity, toxicity, and restricted applicability to certain molecules. Additionally, the limited concentration of these agents as mandated by regulatory authorities presents a formidable challenge for formulation development scientists. On the other hand, the NS, along with other carriers, can be an option for drug targeting to various organs [
220,
221]. The targeting potential of nevirapine was improved by developing NS and surface-modifying them with serum albumin, polysaccharide, and PEG [
222]. NS, which contain NCs of poorly soluble drugs suspended in an aqueous medium with a restricted concentration of stabilizers, are emerging as a promising solution for this issue. Parenteral NS are a widely used formulation approach in drug development, particularly when evaluating the safety of a new chemical entity in experimental animals during dose escalation studies [
223].
Parenteral NS delivery utilizing NCs reduces the toxicity of nonaqueous formulations while enabling targeted drug delivery, making them valuable for precision therapies [
224]. Parenteral NS offer advantages such as the enhanced solubility of poorly aqueous-soluble drugs and improved bioavailability of BCS class IIa drugs with high doses and BCS class IIb drugs with limited solubility and high melting points [
28]. In general, intravenous NS are a promising option for high-dose, poorly soluble drugs [
21]. Particles are typically stabilized by agents such as polymers or surfactants, but selecting safe and effective stabilizers is crucial for parenteral use due to concerns over toxicity and microbial risks. Moreover, the concentration of stabilizers beyond the critical flocculation concentration and critical micellar concentration may result in micellar solubilization leading to Ostwald ripening and system destabilization [
225]. NS with particle sizes between 100 and 300 nm are ideal for utilizing the enhanced permeability and retention effect, beneficial particularly in solid tumors and hence for reduced toxicity [
226]. Nanocrystalline suspensions have become a promising approach for developing parenteral prolonged release systems for insoluble drugs [
227]. Crystalline transformation methods, including solid–solid transformations, solution-mediated transformations, transformations through raw material melting, and those from drug solutions, are often to produce stable crystalline pharmaceutical solids [
228]. A uniform-sized nanocrystalline suspension-based montelukast parenteral prolonged-release delivery system was developed using the bead milling technique, utilizing polysorbate 80 as a suspending agent [
4]. In rats, the NS with 200 nm and 500 nm particle sizes showed a longer pharmacokinetic profile for up to 4 weeks, with greater plasma drug concentrations than the injectable suspensions with 3 μm particle sizes. According to the histopathological investigation, the generated NS resulted in persistent granulomatous inflammation at the injection site, which disappeared after four weeks. Formulation challenges include the risk of aggregation, which can block intravenous lines and fine capillaries, which can potentially cause serious complications such as thrombus, embolism, or stroke. For example, particles larger than 7 μm and larger agglomerates can lead to a pulmonary embolism and pose a risk to patient safety. In recent years, parenteral NS use has been observed in toxicological and clinical formulations [
229].
Parenteral NS require the careful consideration of key parameters to ensure safety and efficacy. Isotonicity and pH are adjusted for physiological compatibility, while particle size and ZP affect stability and drug delivery. Lyophilized formulations need to be easily redispersed, and sterility is ensured through filtration or aseptic processing. Multi-dose formulations include antimicrobial preservatives, and viscosity is controlled for easy administration. Stability studies ensure the formulation remains effective over time, and drug release profiles determine how the drug is released in the body. These factors are critical in developing an optimal NS for parenteral use. In addition, parenteral NS must meet stringent regulatory requirements, including QBD and current good manufacturing practice standards to ensure safety, quality, sterility, and stability. The FDA’s guidance for drug products containing nanomaterials outlines considerations for managing risks associated with these materials, emphasizing that the same safety, efficacy, and quality standards apply to nanomaterial-based products as to other drugs. Nanomaterials can be sensitive to process conditions and may face stability issues, such as changes in size or aggregation. Despite these challenges, drug developers must adhere to current good manufacturing practices, thoroughly assess CQAs, and evaluate the safety, efficacy, and quality of these nanomaterial based-drug products.
Amino-acid-derived copolymers such as albumin, lysine, leucine, and transferrin and synthetic nonionic polymers such as poloxamer 188 (Pluronics F68) and poloxamer 407 (Pluronics F127) are widely regarded as safe for parenteral applications by FDA. A comparative study of stabilizing excipients revealed that polysorbate 80-based NS caused stronger inflammation and slower recovery in male rats, while TPGS-based formulations showed a moderate response, combining features of both the polysorbate and poloxamer [
230]. Soluplus, a polyvinyl caprolactam-polyvinyl acetate-PEG graft copolymer, serves as a solubilizer for poorly water-soluble drugs in parenteral formulations at moderate concentrations. It is compatible with various processes like spray-drying, high shear milling, solvent evaporation, and electrospinning. Its high flowability and controlled extrudability make it an effective stabilizer in pharmaceuticals [
231]. Additionally, below its critical micelle concentration, Soluplus inhibits drug precipitation by preventing nucleation and crystal growth, while providing steric stabilization in a supersaturated state [
232]. Similar to coarse suspensions, intravenous nanocrystalline suspensions can be categorized into two types: (a) pre-mixed and (b) dry powder NS for reconstitution.
Parenteral NS can be prepared through the bottom-up technique, which forms NPs by dissolving the drug in an organic solvent and using an antisolvent to precipitate the drug with a stabilizer. Common methods include SCF processes, sonocrystallization, spray-drying and controlled crystallization using freeze-drying [
56]. In the top-down wet media milling technique, highly cross-linked polystyrene beads are commonly used as the milling media to minimize cracks and abrasions. For example, the commercial technology NanoCrystal
® utilizes PolyMill media made from polystyrene beads to prepare NS meant for parenteral use [
28]. Aseptic processing, autoclaving, or gamma radiation (for dry powders) are the three methods used to sterilize injectables. Typically, the active pharmaceutical ingredient and excipients are separate, and then combined under aseptic conditions. Alternatively, the excipients can be sterilized in solution and mixed with the drug aseptically. The sterile filtration of aqueous NS is another option, as demonstrated by NanoCrystal™ for the X-ray contrast agent iodipamide, which used a 0.2 μm Supor
® filter to retain 100% of
Pseudomonas diminuta.
Nanocrystalline suspensions can be utilized for drug targeting owing to their surface potential and in vivo behavior. After an intravenous injection, drug NCs can passively target tumors via the enhanced permeability and retention effect or be absorbed by macrophages if dissolution is delayed [
233]. This leads to a prolonged release, reducing toxicity while maintaining drug efficacy, particularly beneficial for certain drugs like antineoplastics [
234]. Differing pharmacokinetics were observed between the itraconazole NS and marketed injections, with greater concentrations detected in the liver, spleen, and lungs, indicating potential for passive targeting to the mononuclear phagocyte systems [
235]. A comparative study evaluated the tissue distribution as well as the pharmacokinetics of two oridonin NCs with markedly different sizes (103.3 ± 1.5 nm and 897.2 ± 14.2 nm) after intravenous delivery in rabbits [
236]. It was noticed that the smaller NCs acted similar to a drug solution, while the larger NCs exhibited increased accumulation in the liver, lungs, and spleen. The studies demonstrate that modifying the particle size is crucial for effectively targeting NCs. Smaller crystals target tumor cells by an enhanced permeability and retention effect, while larger crystals are predominantly absorbed by the mononuclear phagocyte system, especially in the spleen and liver. Stabilizers and targeting ligands are non-covalently attached to the surface of NCs, where nonspecific interactions, such as adsorption, play a dominant role during surface modification [
28]. Hyaluronic-acid-anchored paclitaxel NCs significantly extended the systemic circulation of the drug, increasing the AUC by 8.4 times compared to the commercial formulation (Taxol™). Additionally, the NCs demonstrated reduced lung metastasis, enhanced antitumor efficacy, and lower toxicity in an LA-7 tumor-bearing rat model compared to Taxol™. In another study, paclitaxel NCs were modified with hyaluronic acid and apo-transferrin (bilobar protein) to evaluate their ability to inhibit cell growth. In MCF-7 cells, the surface-modified NCs achieved 60% cell growth inhibition, though this effect was less potent than that of normal NCs and the pure drug in normal cell lines [
237]. The evaluation of Herceptin-functionalized paclitaxel NCs on HER-2-positive breast cancer cells showed that the surface-modified NCs had a good binding affinity and cell-specific uptake in comparison to the drug NCs [
238]. Docetaxel NCs stabilized with a chondroitin sulfate A and PEG conjugate, demonstrating key benefits such as pegylation, stabilization, and CD44 receptor targeting [
239]. In studies with MDA-MB-231 cells, the drug chondroitin sulfate A NCs exhibited an increased cellular uptake, deeper tissue penetration, and enhanced cytotoxicity, primarily due to the enhanced permeability and retention effect and receptor-mediated endocytosis.
The excessive dilution of the formulation can cause the detachment of stabilizers or targeting ligands. As a result, this shedding poses a significant challenge when the goal is to achieve targeted delivery to a specific site [
240]. To reduce stabilizer shedding, strategies include chemically modifying stabilizers to increase the binding sites or using crosslinking techniques to physically entrap the stabilizer around the NCs. A study showed that the layer-by-layer assembly of polyelectrolyte-coated paclitaxel NCs dissolved more slowly compared to uncoated NCs and the commercial formulation (Abraxane
®) [
241]. However, they were rapidly removed from circulation, likely due to the shedding of the PEGylated coating from the surface of the NCs. Even though drug NCs have been rapidly developed for oral and other administration routes, the parenteral NS face a number of challenges including sterility, long-term stability, and translation to clinical use. Currently, only one intravenous NC product, Ryanodex
® (dantrolene sodium), is available on the market. Other products, such as meloxicam and ubidecarenone NCs, are in advanced clinical trials.
Clinical trials focus on evaluating safety, efficacy, pharmacokinetics, and optimal dosing across phases, with an emphasis on bioavailability and reduced adverse effects. Patents often cover novel formulation methods, such as advanced manufacturing techniques, surface modifications, and lyophilization processes that enhance stability and dispersibility. Recent advances include the use of SCF technology, targeted delivery systems, sustained release formulations, nanocrystal technology, and smart, stimuli-responsive NS, all of which improve drug solubility, bioavailability, and therapeutic outcomes, particularly in cancer and chronic disease treatments. In summary, NS significantly enhance drug delivery across various administration routes, including oral, pulmonary, ocular, and transdermal. The key features of the most vital NS, their preparation techniques, delivery routes, and applications are summarized in
Table 7.
NS offer unique advantages for enhancing the solubility and bioavailability of poorly soluble drugs, featuring simple production processes and broad applications. However, formualtions like liposomes, nanoemulsion, polymeric micelles, solid lipid NPs, and nanostructured lipid carriers excel in targeted delivery and biocompatibility, albeit at higher costs and with stability challenges. Contextualizing NS alongside these alternatives highlights their role as a versatile and cost-effective platform for drug delivery, particularly for hydrophobic drugs (
Table 8).