Science and Technology Development Journal

An official journal of Viet Nam National University Ho Chi Minh City, Viet Nam since 1997

Skip to main content Skip to main navigation menu Skip to site footer

 Section: HEALTH SCIENCES

HTML

0

Total

0

Share

Niosomes: recent advances and applications in targeted drug delivery






 Open Access

Downloads

Download data is not yet available.

Abstract

In drug delivery, niosomes, novel potential biomaterials, were inspired by vesicular surfactant systems. The niosome particles form a bilayered structure containing nonionic surfactants and an aqueous region, allowing them to carry drug molecules with a wide solubility spectrum. Additionally, niosomes provide a cost-effective, biocompatible alternative, ensuring long-term, focused medication and targeted drug delivery. The advantages of niosomes over other drug carrier systems have attracted increasing attention from researchers, who recently published numerous studies on this material as a targeted drug delivery system. Niosomal drugs with high bioavailability can be delivered via various routes of administration, such as oral, dermal, ocular, and respiratory routes. This review presents the fundamentals of the characterization and preparation methods of niosomes. We also highlight recent advancements in the field of targeted drug delivery of niosomes and their applications for routes of administration, highlighting future research trends in niosomes.

Introduction

Currently, addressing drug development related to delivering drugs to their specific sites of action has become increasingly important 1 . In the early 20 th century, Paul Ehrlich introduced the concept of targeted drug delivery and promoted drug delivery system research. The basic principle of this terminology is the delivery of drugs with a sufficient concentration directly to the targeted region, reducing their distribution to the nontargeted area 2 . Consequently, it produces fewer undesirable effects at other sites and could reduce the amount of drug required to achieve therapeutic effects. In terms of therapeutic agents, a targeted drug delivery system aims to manage and control the pharmacokinetics, pharmacodynamics, specific toxicity, immunogenicity, and biorecognition of drugs 3 . Therefore, their most crucial application is treating tumor cells because of their better ability to penetrate macrophages, high concentration at the affected site, and fewer side effects 4 . Furthermore, targeted drug delivery systems have various applications because of their ability to deliver agents via many routes, including ocular, brain, oral, and transdermal routes with diverse carriers and bioactive compounds 2 , 5 .

Direct techniques can be utilized for targeted drug delivery, such as direct injection, catheters, and gene guns, but they are uncomfortable and costly for patients in many cases 1 . Therefore, the use of specific engineered vectors capable of incorporating drug molecules through encapsulation seems to be an excellent approach 6 . Numerous novel nanocarriers, including nanoparticles, micelles, nanogels, transferosomes, liposomes, and niosomes, have been developed and applied as drug delivery systems 2 , 7 , 8 , 9 .

Among these systems, niosomes have recently been acknowledged as promising targeted drugs inspired by lipid-based nanovesicles. Niosomes, with surfactants as their main components, exhibit greater chemical stability and lower production costs despite having a similar structure and utility as other drug delivery vehicles 10 . Owing to the amphiphilicity of the niosomal structure, hydrophilic molecules can be incorporated into the central cavity, whereas lipophilic molecules can be entrapped within the lipid bilayer. Previous studies have shown that the use of niosomes is a versatile strategy for dermal and transdermal 11 , nose-to-brain 12 , oral 13 , ocular 14 , and respiratory 15 drug delivery.

This review presents the fundamentals of the characterization and updated preparation methods of niosomes. We also provide an overview of the latest advances in the field of targeted niosomal drug carriers for various routes of administration.

Overview of niosomes

Structure

Niosomes are bilayered vesicles composed of polyglycerol alkyl/dialkyl ether nonionic surfactants, such as polysorbate 80 or Span 60, and cholesterol. Nonionic surfactants are amphiphilic molecules with hydrophobic (tails) and hydrophilic (heads) groups that can easily fit into any geometry. Moreover, compared with anionic, cationic, and amphoteric surfactants, nonionic surfactants are more stable and less toxic, enhancing their cellular permeability and solubility. Moreover, cholesterol improves the stability of vesicles by reducing bilayer acyl chain movement through hydrogen bonds between the hydroxyl groups of cholesterol and the alkyl chains of surfactants 16 . In combination with the type of surfactant, cholesterol significantly affects niosome size. As the cholesterol concentration increases, the hydrophobicity of bilayers increases, decreasing the surface free energy and thereby decreasing the size of niosomes 17 . The structure of a niosome is shown in Figure 1 , which also illustrates the entrapment areas of hydrophilic/lipophilic drugs. The hydrophobic tails remain in contact, whereas the hydrophilic head groups are oriented toward the aqueous interior and exterior of the vesicle. Therefore, with hydrophobic and hydrophilic groups, niosomes can accommodate drug molecules with diverse solubilities 8 .

Figure 1 . The bilayer structure of a niosome.

Types of niosomes

Most categorize niosomes into three basic types on the basis of size and the number of lamellae: small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs), and multilamellar vesicles (MLVs) ( Figure 2 ) 18 . Typically, SUVs range in size from 10–100 nm 18 , LUVs from 100–1000 nm (with the potential to reach up to 5000 nm), and MLVs from 500–1000 nm or without a defined upper limit 8 , 17 , 18 . SUVs and LUVs are unilamellar, differing from MLVs, which are multilamellar. The increase in size and number of layers is directly proportional to the number of hydrophilic head groups. In practical terms, this means that SUVs, with their lower aqueous and bilayer ratios than LUVs do, are more suitable for lipophilic drugs, whereas LUVs are better for hydrophilic drugs 18 , 19 . MLVs have bilayers adjacent to aqueous compartments, as shown in Figure 2 . Additionally, larger niosomes provide advantages for loading lipophilic drugs because of the presence of multiple bilayers 20 .

Figure 2 . Niosome classification into small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs), and multilamellar vesicles (MLVs)

Preparation of niosomes

Niosomes can be prepared via various methods to optimize the desired particle size and size distribution, number of bilayers, aqueous phase encapsulation efficiency, and vesicular membrane permeability to transport different drugs, hormones, and antigens 21 . In thin-film hydration, ether injection, reversed-phase evaporation, or transmembrane pH gradient drug uptake methods, the primary advantages of those methods are easy handling and fast production, which do not require sophisticated equipment. In addition, the thin-film hydration method is a straightforward and convenient technique for obtaining MLVs, and the reverse evaporation method is commonly used to produce LUVs 22 . The sonication method is the most common size reduction method, which increases drug solubility, enhances bioavailability, and creates niosomes at the nanoscale 22 . Other novel methods have recently been reported; for example, the microfluidization approach quickly and precisely controls the mixing process of aqueous and surfactant cholesterol solutions in microchannels 23 . Another form of niosome, the proniosome, is obtained as a dry powder to increase physical stability. The resulting proniosomes are stable throughout storage and transportation and are suitable for making unit dosages 24 , 25 .

Furthermore, the classification of niosome synthesis considers the active and passive trapping approaches by the niosome. The majority of niosome synthesis methods employ the passive trapping approach. This procedure aids in the encapsulation of the medication during niosome synthesis. In contrast, active trapping techniques make it more accessible for the medicine to be absorbed inside the niosome, as the niosome is produced on the basis of an ion gradient or pH gradient. The advantages of this method include cost-effectiveness, high drug–lipid ratios, reduced leakage, and applicability for thermosensitive compounds 20 . The preparation category, types of vesicle size of each method, and their benefits and drawbacks for a clearer understanding of all the approaches discussed above are displayed in Table 1 .

Table 1 Advantages and disadvantages of niosome preparation methods.

Although thin film methods, solvent injection, and heating methods are simple and universal, implementing these processes in clinical practice is constrained by several steps, leading to time-consuming and challenging conversion to an industrial scale 46 , 47 . Additionally, the above methods involve complex mechanical and chemical environments for control. The resulting vesicles are often large and polydisperse 48 ; thus, they need an extra step of size reduction by sonication, extrusion or freezing and thawing cycles. However, a novel preparation, called the microfluidization method, can overcome these restrictions and support niosome creation on an industrial scale 49 , 50 .

In recent years, microfluidization has been one of the most commonly employed methods to produce niosomes. By optimizing parameters, such as mixing conditions, the type and ratio of surfactants, and other materials, niosomes with desired particle sizes and size distributions can be produced with better reproducibility than other techniques 51 , which are based on the jet principle 52 . In the microfluidization method, the drug and surfactant are mixed and pumped under reservoir pressure into an ice-packed interaction chamber. The generated heat is removed during the passage of the solution through a cooling loop 21 , 23 , 53 . Compared with other methods, microfluidization is uniquely superior to conventional modification procedures in some cases, such as continuous rapid processing time 54 . Furthermore, vesicles of a specific size have been strictly controlled by precisely adjusting the flow rates of the aqueous and organic streams while minimizing the necessary liquid volumes and thereby being able to synthesize multiple types of niosomes simultaneously, reducing preparation time and development costs as well as meeting specific quality requirements 8 , 50 . Additionally, ethanol or isopropanol, which are typically used in this method's solvents, make it more ecologically friendly than other methods are. The microfluidizer is one of several commercially available homogenizers that may produce niosomes on a large scale, hence avoiding intermediary research. However, no studies have described this technique of niosome preparation 55 . Owing to these benefits, the microfluidization method potentially has a strong chance of being applied at an industrial scale.

Characterization of niosomes

Entrapment efficiency (EE%) and loading efficacy (LE%)

For drug delivery, the crucial parameters of niosomes are entrapment efficiency (EE%) and loading efficacy (LE%) 56 . The entrapment efficacy (EE%) is the percentage of the drug successfully encapsulated within lipid nanoparticles relative to the total amount of drug used in the formulation process and is aimed at assessing the appropriateness and minimizing the waste of drug usage. In contrast, loading efficacy (LE%) is the ratio of the encapsulated drug to the total weight of the nanoparticles, including both the drug and the lipid carrier, aimed at determining the average drug dosage in each nanoparticle, thereby optimizing distribution efficiency and therapeutic impact 57 .

The EE% of niosomes depends on the characteristics of drug molecules, surfactants, and additives and the preparation method. In particular, the EE% values of the niosomes obtained via the ether injection method are superior to those obtained via the thin-film hydration method. Adding cholesterol or diacetyl phosphate to nonionic surfactants alters the EE% 58 . Moreover, the size and number of bilayers affect the choice of loaded drugs and the EE%. In fact, SUVs are less thermodynamically stable, resulting in poor hydrophilic drug loading capacity and easy aggregation. Conversely, LUVs with large aqueous compartments are favorable for hydrophilic drugs. With multiple bilayer membranes, MUVs have a relatively high EE for lipophilic drugs 18 . The EE% and LE% are calculated via the following formulas:

Several methods have been applied to calculate the EE%, such as spectrophotometry 59 , 60 , 61 , 62 , 63 , 64 and reverse-phase high-performance liquid chromatography 65 . The purpose of these methods is to quantify the amount of unentrapped drug in the supernatant after centrifugation. When Span is used in formulations, hydrophobic drugs are entrapped more effectively than Tween because the lipophilic part of Span results in more significant space in the bilayer domain 65 . In addition, the structure of the bilayer, size, and surface charge of niosomes affect the EE% 66 . Damera’s study showed that a hydrophobic drug (coumarin-153) was entrapped effectively by Span 60-niosomes, whereas Span 80-niosomes had better EE% for a hydrophilic molecule, rhodamine 6G (R6G). Owing to the structural difference between Span 60 and Span 80, the latter contains an unsaturation at the 8 th carbon in the chain, resulting in a more disordered bilayer structure. The greater entrapment efficacy of R6G when Span 80 is used may be due to its hydrophilic nature; the R6G molecule can penetrate disordered bilayer areas, allowing it to reach the air‒water interface. This results in leakage and decreased entrapment efficiency in Span 80-niosomes for hydrophobic dyes 66 .

Size, zeta potential, and stability

During the preparation of niosomes, size and zeta potential (ζ-potential) are two factors that should be monitored carefully. The ζ-potential refers to the potential at the slipping or shear plane of a colloid particle moving under an electric field 67 , a parameter of particular interest owing to its profound impact on the pharmacokinetics and efficacy of nanomedicine 68 . Although niosomes can reach sizes of several micrometres, for successful drug encapsulation, good stability, and optimal release kinetics, the particle size should be smaller than 1000 nm 40 . The particle size can be influenced and adjusted by various factors, such as the synthetic materials and the synthesis process. When selecting components for niosome synthesis, the desired type of particle to be synthesized, the type of surfactant, and the ratio of surfactant to cholesterol are essential to consider. The selection of surfactant is crucial because particle size generally increases with increasing hydrophilic‒lipophilic balance (HLB) index 16 , which is defined as the ratio of the weight percentages of hydrophilic and lipophilic groups within a molecule 69 . Similarly, Hedayati demonstrated that the particle size of niosomes prepared via the thin film hydration method depends on the surfactant-to-cholesterol ratio 62 . Additionally, several methods can be used to reduce the particle size to the desired level. Fatemeh Nowroozi et al . utilized the thin film hydration method to compare the effectiveness of current size reduction techniques, including sonication, extrusion, and high-pressure homogenization. For niosomes prepared using tween 60 and span 60, the most effective methods for reducing particle size are extrusion and sonication 16 .

Another significant aspect, the ζ-potential, significantly impacts the behavior and properties of vesicles 70 . Because of the direct correlation with the repulsion of niosomes, niosomes with largely negative or positive ζ-potentials tend to be prevented from aggregating 65 . Jain et al. reported that an increase in the HLB value of a surfactant directly correlates with a more negative zeta potential due to higher surface energy and the resulting repulsion of like-charged particles, which inhibits niosomal particle aggregation 71 . These results can be attributed to the highly hydrophilic HLB, which envelops the niosome in the hydrophilic layer, leading to pronounced negative charges on the surface of the niosome.

Niosome stability is crucial for maintaining the structure of the niosome and preventing drug leakage from its carrier before administration or reaching the targeted site in the body. Niosome stability depends on many factors, such as the nature of the drug molecule, the surfactant properties, the concentration of additives, and the storage conditions. The stability is directly evaluated through the particle size and zeta potential. According to Moazeni et al. , niosomes ranging from 1–10 µm are more stable than those smaller than submicrons. Thermodynamically, smaller niosomes have higher surface free energy and tend to aggregate to minimize excess free energy 72 . Additionally, MLVs can be easily prepared without complex techniques and offer better stability than can SUVs and LUVs under normal storage conditions. However, it is crucial to consider the desired target effect to determine the appropriate size and to select a suitable surfactant, as mentioned previously. Moreover, particles in the LNP group should have a zeta potential greater than +30 mV or less than -30 mV to indicate electrical stability 73 . However, in some instances where nonionic surfactants are used, the zeta potential falls within the range of ±30 mV, which does not necessarily mean that the niosome lacks stability. A study by Junyaprasert et al . revealed that the highest stability is achieved when the zeta potential is less than -30 mV. Nonetheless, samples with zeta potentials lower than this range can still demonstrate stability. Spatial stability enhances the physical stability of niosomes with nonionic surfactants. These findings indicate that electrical and spatial stability are the most critical indicators of niosomal stability, as reflected by the average particle size and entrapment efficacy 74 . The HLB value is widely recognized as a crucial factor in niosome synthesis, as it is regarded as an indirect determinant influencing the stability of the particles. In addition to these properties, Akbarzadeh performed a stability study under two conditions, at room temperature (25±1 °C) and under refrigeration (4±1 °C) for three months, and the results revealed that the niosome stability was excellent under cold conditions 75 . A similar experiment was conducted by Suthinee Sangkana et al. to assess the stability of GME-loaded niosomes stored at 4 and 25 °C. The study revealed that niosomes exhibited better stability at 4 °C. Interestingly, GME-loaded niosomes become nearly neutral when stored at 25 °C for 4 weeks, likely due to surfactant degradation, which can disrupt their structure and surface charge. High temperatures can cause surfactants to lose their amphiphilic properties, leading to their degradation. Additionally, vesicle fusion may occur due to increased molecular motion at higher temperatures, causing niosomes to merge and dilute the surface charge 76 . Similarly, Hashim's findings revealed that the chosen niosomal formulation, which was stable in the refrigerator, had only 2% encapsulated drug leakage at the end of six months 61 .

In vitro and in vivo release

The drug concentration, hydration volume, and other factors can impact drug release from niosomal particles. The drug molecules released from the niosomes diffuse through a dialysis membrane at 37 °C in phosphate-buffered saline (pH 7.4 and 5.2) 58 , 59 , 63 , 75 , 77 . In the next step, high-performance liquid chromatography methods are utilized to calculate the amount of drug released 59 , 65 , 75 . The drug release process consists of two phases: a fast-release phase at the beginning and a prolonged-release phase 60 , 65 . While the drug released in the initial phase is from the niosome surface, the drug molecules from the core of the particles diffuse out in the second phase. These two phases depend on several factors, such as particle size, drug characteristics, and surfactants 60 , 65 .

The effectiveness of nanoparticle synthesis depends critically on the capacity to manage the features of the synthesized particles, such as the particle size, size distribution, and encapsulation efficiency, as these factors affect the in vivo performance 78 . An increase in encapsulation efficiency increases the size of the network, which is related to physical factors such as the vehicle loading volume capacity 79 . The half-life of drug carrier systems is significantly affected by their size, as more giant vesicles with short serum half-lives tend to accumulate in some organs, such as the lungs, liver, and spleen. Additionally, niosomes may aggregate due to improper zeta potential, which may have unwanted consequences, such as toxicity and reduced targeting effectiveness 38 . Size also influences the ability to pass through physiological barriers, such as the blood‒brain barrier 80 , the blood‒retinal barrier 81 , and the blood‒placental barrier 82 . The correlation between nanoparticle size and blood‒brain barrier crossing was shown to be nonmonotonic. Among the polystyrene spherical nanoparticles that range between 100 and 500 nm in size, 200 nm particles can penetrate more effectively than 100 nm or 500 nm particles can 83 . In addition to the particle size, the size distribution parameters also significantly affect the pharmacokinetics since particles with a broader size distribution have greater liver uptake and shorter blood clearance 84 .

Functionalization of niosomes enhances targeted delivery

By concentrating drugs at specific sites, surface ligands on niosomes have been developed to increase the efficiency of targeted drug delivery. These ligands facilitate the binding or absorption of niosomes precisely by target cells, thereby increasing the effectiveness of drug delivery to the intended cells. The functional biomolecules frequently employed in the surface modification of niosomes include aptamers, chitosan, folic acid, peptides, and transferrin, as outlined in Table 2 .

Table 2 Niosome formulations functionalized with various biomolecules

Routes of administration and applications

Oral administration

Overview

Oral administration is the most common route because of its advantages, including ease of use, patient preference, cost-effectiveness, suitability for repeated and prolonged use, and the lack of sterile precautions needed 99 , 100 . However, drugs are easily degradable through oral administration because of their first-pass metabolism, highly acidic gastric environment, and mucosal enzymes before they enter the systemic circulation 101 . Numerous drugs may not be absorbed because of inadequate solubility, lipophilicity, and high molecular weight 101 . Therefore, many scientists have investigated the preparation of orally active substances loaded with niosomes to overcome the limitations of classic oral formulations 102 .

Niosomes can cross the intestinal membrane, involving paracellular permeation, M-cell-mediated absorption, and endocytosis, as shown in Figure 3 . The endocytosis process involves four submechanisms: clathrin-mediated endocytosis, caveolae, micropinocytosis, and phagocytosis 103 .

Figure 3 . Mechanisms of niosome absorption, including A) paracellular permeation, B) M-cell-mediated absorption, and C) endocytosis

Applications

Several compounds have been formulated and innovated to overcome the first-pass metabolism effect and take advantage of oral administration. Coenzyme Q10 (CoQ10) is an endogenous substance with various pharmacological properties. CoQ10 is unstable under light, thermolabile, poorly water soluble, and has low bioavailability; therefore, CoQ10 is entrapped in niosomes coated with polyethylene glycol (PEG) and chitosan (CS) to increase its therapeutic efficacy and improve its release pharmacokinetics. The results revealed that PEG and CS modification significantly increased the hydroxyl radical scavenging capacity of CoQ10. The best PEG coating was 1:2 PEG/surfactant, which significantly increased the stability of the preloaded formulation against light, heat, and storage conditions. Furthermore, the PEG-coated niosomes had more sustained release effects than did the CoQ10 solution in the in vitro study in which the dialysis method was used for 24 hours. However, the CS modification caused some adverse effects, such as disturbing the bilayer structure, leading to a leak of CoQ10 104 .

In addition, the gastrointestinal residence time increased when chitosan was applied as a bioadhesive coating for niosomes 105 . Owing to the strong mucoadhesive and absorption-enhancing properties of chitosan, encapsulating poorly water-soluble drugs into chitosan-coated niosomes could improve their bioavailability. In a study by AbuElfadl et al ., candesartan cilexetil was loaded onto a modified-surface niosome, and an increase in bioavailability of 37% was observed 106 . Another active drug is famotidine, which is also loaded in chitosan-modified niosomal vesicles. Compared with the uncoated formulation, the results demonstrated that 0.1 mg/mL chitosan-coated niosome had greater mucoadhesive effectiveness and stability 107 .

Niosomes are also used as carriers of antitumor substances. Galangin is a flavonoid compound with antitumor activity that is a promising agent against liver cancer but has poor solubility 108 . Thus, galangin-loaded niosomes constitute a promising targeted system for enhancing antitumor activity against hepatocarcinoma. Niosomes have longer storage times than liposomes do, while the amount of galangin leaching in the in vitro drug release assay increases. Galangin-loaded niosomes effectively decreased the number of neoplastic hepatic lesions with a small number of hepatic adenomas and minichromosome maintenance in three immunostained hepatocytes 108 . In a study by Sabry et al ., BiloNiosome-core/chitosan-shell hybrid nanocarriers with cordycepin were studied for their ability to improve oral drug efficacy. The self-association of nonionic surfactants, lipids, and cholesterol produced vesicular nanoparticles to deliver cordycepin. The incorporation of bile salts into these nanocarriers prevents gastrointestinal degradation. Moreover, mucoadhesive chitosan on the surface of the nanoparticles improved their ability to target mucosal surfaces. The results revealed that hybrid nanocarriers were stable in gastric fluid, mucoadhesive, and permeable across intestinal epithelium models and had greater anticancer effects than did the standard cordycepin 109 . Q. Wang et al. produced ginsenoside Rb1-loaded niosomes via response surface methodology and a central composite design tool to increase its oral bioavailability. Notably, the resulting niosomes showed an increase in oral bioavailability of 181.93% (p < 0.05) compared with the free ginsenoside Rb1 110 .

In addition, proniosomes are dehydrated forms of niosomes and should be soaked before usage to produce an aqueous niosome dispersion 45 . In these dry forms, proniosomes have some benefits over niosomes in terms of aggregation, fusion, caking, transportation and distribution 111 . According to Ibrahim and Shehata, the application of tramadol HCl-loaded niosomes via proniosome technology could help prolong the pharmacological effects of niosomes compared with those of the solution in mice. The improved efficacy of tramadol HCl is due to an increase in the hydrophilic‒lipophilic balance of the surfactant. Drug release increased as the cholesterol content increased from 0% to 50%. At 10% cholesterol, drug release increased in a specific order: Span 80 > Span 40 > Tween 80 > Tween 40 112 .

Advantages and disadvantages

As a drug delivery vehicle via the oral route, the advantages of niosomal formulations include improved stability and bioavailability 113 . Both lipophilic and hydrophilic drugs can be formulated easily and combined in the same vesicles 114 . Coating vesicular systems protects delicate molecules, increasing their bioavailability through the intestine. Nonionic surfactants such as Span, Tween, Brij, and poloxamer are permeation enhancers that increase the ability of drugs to permeate through intestinal tissue 115 . Niosomes may significantly extend the half-life of drug molecules in the plasma, making it possible to minimize the frequency of drug administration 116 . A study by Kamboj et al. on niosomes loaded with tenofovir disoproxil fumarate, an anti-HIV drug, demonstrated a significant increase in the mean residence time and more than a twofold increase in oral bioavailability compared with the same dose of the drug in its standard form 116 . The release of niosomes can be controlled or sustained in transit and at the localization site 61 . The stability of nanocarriers under gastrointestinal conditions is a serious concern, influencing the therapeutic efficacy of the formulation 117 . In the future, niosomes can be engineered to transport drug molecules to desired locations by surface modification via the attachment of various ligands or magnetic guidance.

Transdermal delivery

Overview

The topical application of niosomes can improve drug penetration into the epidermis and dermis, as reported by many previous studies 118 , 119 , 120 , 121 , 122 , 123 . The following mechanisms explain this effect. First, when drug-containing niosomes are absorbed on the skin surface, a high degree of drug thermodynamic activity occurs at the vesicle and stratum corneum (SC) surfaces, which facilitates the penetration of drugs through the SC 124 , 125 , 126 , 127 , 128 . Second, niosomal formulations disrupt the densely packed lipids that fill the extracellular space of the SC, enhancing drug permeability through conformational alteration of the SC. Third, transdermal penetration is improved in the presence of nonionic surfactants as penetration enhancers, which alter the intercellular lipids of the SC and increase the overall membrane fluidity 24 , 124 , 125 , 127 , 129 , 130 , 131 , 132 , 133 , 134 , 135 . Finally, niosomes induce a hydrating effect, which loosens the tight cellular arrangement of the SC 124 , 127 . The dermal and transdermal routes of delivery of niosomes are briefly presented in Figure 4 .

Figure 4 . Skin penetration pathway of niosomes and proniosomes in dermal and transdermal drug delivery. (A) Niosomes release drug particles. (B) Niosomes disrupt the densely packed lipids that fill the extracellular space of the SC. (C) Niosomes disrupt their structure, and niosomal fragments and drug particles diffuse through the SC extracellular space. (D) Niosomal surfactants act as penetration enhancers and increase membrane fluidity. (E) Niosomes diffuse through hair follicles.

Applications

Since 1975, many dermatological products applying niosome technology, such as the antiaging LancĂ´me Niosome Plus, have been developed and put on the market 136 . Current efforts focus on the clinical application of niosomes as local drug delivery systems 137 .

Local anesthesia

Before performing skin procedures, dermatologists take advantage of the benefits of local anesthetics for pain relief. Ineffective local anesthetic formulations can lead to inadequate anesthesia, severe dermatitis, and systemic toxicity 138 . The importance of the delivery system and its surfactants was demonstrated by Shabery et al. semisolid niosomal encapsulated lidocaine and prilocaine were prepared via a mixture of Span 40 or Span 60, cholesterol, and a patented palm oil base. The results showed that among the formulations, formulation F1 containing Span 40 had the greatest EE%, as well as the highest permeability, of the two anesthetics. Furthermore, the formulation of the F1-C emulsion was optimized via a cold process, resulting in an even higher permeability rate. This led to a strong anesthetic effect at the administration site. Compared with the EMLA ® cream, the F1-C emulgel had a comparable local anesthetic effect on healthy subjects 139 .

Psoriasis

Psoriasis is a chronic systemic inflammation of the skin that causes discomfort, pain, itch and disfiguring skin lesions, which can trigger the psychosocial burden of the disease 140 . The effectiveness and safety of antipsoriatic treatments could be improved through the use of topical niosomal formulations. Methotrexate (MTX) is an antifolate used to treat psoriasis that causes many side effects, particularly hepatotoxicity, when it is systemically administered 141 . Dermal delivery of MTX could be a promising strategy to avoid systemic side effects 142 . Lakshmi et al. prepared MTX-loaded niosomes in a chitosan gel, and the clinical evaluation results revealed that this niosomal system was more effective than the placebo and marketed MTX gels 143 . In addition, niosomes encapsulating celastrol, a triterpenoid found in the genus Tripterygium, significantly improved its antipsoriatic activity in rats by increasing its water solubility and skin penetration 144 .

Vitiligo

Vitiligo is caused by locally damaged melanocytes, resulting in white, discolored areas of the skin. Although vitiligo is not a life-threatening problem, it harms quality of life 145 . The application of transdermal drug delivery systems could play an essential role in overcoming the drawbacks of conventional topical formulations in terms of efficacy and safety 146 . Defects in human tyrosinase, an enzyme involved in melanogenesis, lead to the loss or inactivity of melanocytes and have been demonstrated to be one of the causes of vitiligo or depigmented skin 147 , 148 . Elastic cationic niosomes, including Tween 61/cholesterol/dimethyl dioctadecyl ammonium bromide with a molar ratio of 1:1:0.5, were studied by Manosroi et al. (2010) for efficient transdermal delivery of pMEL34 (tyrosinase-encoding plasmid). The results showed that this system produced better permeability flux in the viable epidermis and dermis than inelastic niosomes did 149 . By applying elastic cationic niosomes loaded with pMEL34 in melanoma cell lines, tyrosinase gene expression was approximately four times higher than that of free plasmids and pMEL34 loaded in inelastic niosomes 149 . In a study by Manosroi et al . (2009), luciferase plasmids were enclosed in elastic and inelastic cationic niosomes, creating a novel method for the transdermal delivery of genetic material in gene therapy via electrophoresis 150 . In particular, elastic cationic niosomes are promising because they do not require additional equipment. This study revealed the superiority of niosomes in their ability to deliver luciferase plasmids through the skin 150 . Both studies indicated that elastic cationic niosomes can effectively transport genetic materials for local gene therapy.

Cutaneous inflammation

Celecoxib, a selective cyclooxygenase-2 inhibitor, was encapsulated in a niosomal gel for site-specific retention and delivery. Auda et al . prepared gels with Span 60 or Span 40 and cholesterol using different polymers as bases. As a result, the formula with a 1:1 ratio of Sponge 60:cholesterol resulted in the highest encapsulation and significantly increased the drug's in vitro release (to 80%). The anti-inflammatory activity of the gel formulations was investigated, and the niosomal poloxamer gel significantly reduced rat paw edema (75.4%), resulting in better permeation flux. The results revealed that niosomal gel formulations made with polymers are suitable carriers for skin accumulation and anti-inflammatory effects 151 . Nasr et al . employed thin film hydration to create multilamellar liposomes and niosomes with aceclofenac, a potent analgesic, antipyretic, and anti-inflammatory agent. Compared with commercial products, both systems were shown to prolong the pharmacological effect of the encapsulated drugs 151 . In particular, the efficacy of niosomes outperformed that of liposomes in animal models 152 . In the studies by Bhardwaj and Bhatia, Tweens and the natural mucilage of Lallemantia royaleana were combined to create niosome gel-loaded ibuprofen. With increasing concentrations of L. royaleana , the results demonstrated a reduction in drug permeation and drug flux values. These findings suggest that mucilage has good binding ability and thus regulates the release of ibuprofen from the niosomal gel system. Ex vivo and in vivo tests revealed that niosomal gel formulations combined with carbopol and L. royaleana had superior penetration and efficacy than carbopol formulations alone. This may be due to better skin retention and deposition, which may have increased drug partitioning into the rat paw and prolonged and boosted its anti-inflammatory effect 153 .

Topical corticosteroids are used for various dermatological disorders because corticosteroids with an appropriate carrier can prolong their effect, thereby reducing the frequency of doses and associated side effects. In a study by Sun Zhe, clobetasol propionate, a potent anti-inflammatory and antipruritic agent, was incorporated into a niosomal gel. The performance of the niosomal clobetasol gel was compared to commercially available gels, considering drug deposition in the stratum corneum (SC) layer and in vivo studies. The findings revealed that the niosomal gel formulation exhibited superior efficacy, reducing the psoriasis area severity index 154 .

Chlorpheniramine maleate is an antihistamine commonly used topically to treat sunburn, urticaria, angioedema, itching, and insect bites. The proniosomes composed of Span 40/lecithin/cholesterol prepared with ethanol exhibited ideal stability, encapsulation efficiency, particle size, and release kinetics, making them well suited for the transdermal delivery of chlorpheniramine maleate. The simple preparation and ease of use of proniosomes are the most significant advantages this dosage form offers 155 . Afreen et al . assessed a niosome-based gel containing chlorpheniramine for treating mild to moderate skin allergies. The optimal niosome dispersion with low cholesterol and span-80 and a high level of span-60 demonstrated a high entrapment efficiency value and maximum drug release. The entrapment efficiency is influenced by the amount of cholesterol and nonionic surfactants, which affects the hydrophobicity, rigidity, and stability of the niosomal layer 156 .

Alopecia

Hormonal alopecia (male pattern alopecia) is the most common cause of hair loss in men 157 . Pumpkin seed oil contains β-sitosterol, which has been shown to prevent hair loss by inhibiting 5α-reductase type 1 (a major enzyme responsible for increased dihydrotestosterone production in male patients with hormonal alopecia) 158 . Research by Teeranachaideekul et al . revealed that hair loss significantly decreased by 44.42% in a 60-second hair count experiment after eight weeks of pumpkin seed oil-loaded niosomes 159 . Finasteride, a 5α-reductase inhibitor, has several undesirable systemic side effects that can be alleviated if it acts locally in hair follicles 160 . Finasteride-loaded proniosomes (FLP) greatly increased the quantity and size of hair follicles in the dorsal areas of C57BL/6MLac mice. In addition, although the concentration of finasteride in FLP (1%) was lower than that in the minoxidil solution used (2%), the therapeutic effects of these two treatments were comparable. Accordingly, the results showed that FLP might be utilized to prevent hair loss by inhibiting type I 5α-reductase 161 . Minoxidil is the most commonly used drug to treat male hormonal alopecia 162 , 163 . Mali et al . reported that Span 60 created stable niosomes with the highest entrapment effectiveness (31.27±1.5%) and the smallest particle size. Compared with those of conventional minoxidil gels, the in vitro permeability and deposition of minoxidil in niosomal gels were significantly improved 162 .

Acne

Acne is a prevalent skin disorder during adolescence. Treatment options often involve topical analgesics, although their side effects might compromise efficacy and patient satisfaction. Niosomes with excellent potential for dermal administration may help improve the transdermal delivery of antacid agents by facilitating their local accumulation with reduced side effects 164 . Benzoyl peroxide, a first-line acne treatment with poor water solubility, the formation of larger clumps, and modest dermal penetration when applied topically, necessitates high amounts of the drug to achieve the desired effect, resulting in skin irritation. Gagan Goyal et al. developed a carbopol niosomal gel formulation containing benzoyl peroxide, which significantly improved the skin penetration of the active ingredient. Compared with conventional medicine and a simple niosomal formulation, the application of niosomal gel had a significant antibacterial effect after four days of treatment, reducing skin inflammation and irritation 165 . Manosroi et al. developed an anionic niosome formulation loaded with gallidermin formed from Tween 61, cholesterol, and diacetylphosphate. The results from the absorption through rat skin revealed that the accumulation in the epidermis and dermis of the rat skin was more than two times greater than that of an aqueous solution of gallidermin; furthermore, gallidermin loaded in niosomes at high temperatures exhibited better chemical stability 166 . This research suggests the potential of the anionic niosomes of gallidermin as effective topical antibacterial formulations without the risk of systemic effects 166 . The percutaneous delivery of tretinoin-loaded niosomal formulations across neonatal pig skin was investigated via Franz diffusion cells. The results from these assays revealed better skin retention than that of liposomal and commercial formulations 167 . Rosemarinic acid is a natural ester of caffeic acid with antibacterial and anti-inflammatory properties. Rosemarinic acid-loaded niosomes have been reported to deliver biological antibacterial agents into deeper layers of the skin 168 . A clinical study was also performed with niosomes loaded with retinoids, critical ingredients in topical acne treatment. The results showed that niosomal retinoids improved antiacne efficacy and were safe for long-term use 168 . Recently, the development of new antibiotics for treating acne has attracted considerable interest. For skin delivery, various solid forms of roxithromycin are packaged into vesicular systems (niosomes, proniosomes, ufosomes, and pro-ufosomes). In particular, niosomes are the most effective formulation for drug delivery to the dermis, where they need to be active against Cutibacterium acnes bacteria 169 .

Advantages and disadvantages

Niosomal formulations have been proven to be well-controlled and advantageous drug delivery systems for topical and transdermal administration. Topical niosomes provide stable dosage forms and cause minimal systemic side effects, as drug release is localized at the application site 170 , 171 . Transdermal drug formulations can deliver active ingredients to the systemic circulation. Remarkably, drug delivery via this route is noninvasive (no needles are needed), has relatively high bioavailability because first-pass metabolism and acidic/enzymatic breakdown in the gastrointestinal tract can be avoided, eliminating food‒drug interactions 17 . Vesicular carriers can also help overcome the physical and chemical instability of some drug molecules. Niosomes, which are more stable and permeable, appear to be a preferred drug delivery system over liposomes 137 .

In addition to the multiple benefits of transdermal routes, the absorption of drugs could be limited by the transdermal barrier, such as the stratum corneum, and its protective nature 172 , 173 . Ideal drug molecules for transdermal formulations often have small molecular weights (under 500 Da) and high hydrophobicity and preferably have low effective doses 166 . Consequently, only a limited number of drugs can satisfy these criteria. Furthermore, the liquid nature of the niosomal preparations is unfavorable for topical application 17 .

Ocular delivery

Overview

The eye is a crucial human sense organ; therefore, any ocular disorder can lead to profound impairment. Prevalent ocular conditions include glaucoma, keratitis, genetic disorders, and diabetic macular edema 174 , all of which have the potential to impact vision detrimentally and, if not promptly treated, even lead to blindness. Thus, ocular disease treatment is essential and should be prioritized. Nevertheless, the complex structure of the eye, which has numerous barriers and defense mechanisms, leads to several drug delivery challenges. However, antibiotic-loaded niosomes can effectively contact the corneal surface and then release drugs, as shown in Figure 5 . In this case, by removing the mucus layer, the surfactants enhance the penetration of antibiotics.

Figure 5 . Pathway of drug release from antibiotic-loaded niosomes at the site of action for ocular delivery.

Applications

The common forms used for treating ocular diseases include topical eye drops and ointments 175 , with the cornea serving as the primary pathway for drug administration to the anterior segment of the eye 14 . The drug delivery efficiency through this route mainly depends on the molecular weight, lipophilicity, and diffusion of the drugs. However, this drug delivery route still has several disadvantages, such as low bioavailability and challenges in permeability retention time 176 . Additionally, owing to insufficient time to settle in the conjunctiva, many drugs are washed away from the ocular surface into the nasolacrimal ducts or lacrimal glands. Intravenous and intravitreal administrations are the primary routes for delivery to the posterior part of the eye 177 . In the clinic, the time interval between injections is usually very long, making it difficult to maintain patient compliance and reducing the efficacy of treatment. Therefore, developing novel ocular drug delivery systems is essential for increasing drug residence time, permeability, and bioavailability.

Glaucoma

In recent years, numerous studies have highlighted the significant potential of niosomes in drug delivery for ocular disease treatment. Glaucoma, a chronic and progressive disorder, often requires prolonged treatment 178 . The most common antiglaucoma drugs are prostaglandin analogs, beta blockers, carbonic anhydrase inhibitors, adrenergic agonists, and miotics 178 . Hasan designed and developed a formulation of niosomes loaded with dorzolamide hydrochloride for glaucoma treatment. The results showed that this formulation has a high drug EE% with small niosomes and a longer drug release rate than conventional preparations do 179 . Jain et al . developed a formulation of niosomal gel containing pilocarpine hydrochloride to increase the retention time and improve the bioavailability of this antiglaucoma drug. The results revealed that this niosomal gel has a bioavailability 2.64 times greater than that of the commercial pilopine HS gel 71 . Fathalla et al . formulated niosomes containing latanoprost, a highly effective drug for glaucoma therapy 180 . The latanoprost niosomal gel showed a remarkable ability to maintain drug release. Additionally, the results from in vivo experiments indicated that the gel has no toxic or irritant effects and reduces intraocular pressure after treating rabbits' eyes for three days 180 . Recently, Abdelmonem et al . developed a niosomal formulation comprising acetazolamide and carvedilol to limit their side effects 181 . Within just one hour after administration, the sustained gel formula of the combination normalized the intraocular pressure, which continued to stabilize for four days. Moreover, histological studies of glaucomatous eyeballs from treated rabbits revealed improvements in retinal atrophy. In addition, other antiglaucoma drugs, such as dorzolamide hydrochloride or brimonidine tartrate, have also been incorporated into proniosome gels, which induce sustained drug action after reconstitution in water 182 , 183 .

Fungal keratitis

Fungal keratitis or keratomycosis is a severe corneal infection caused by corneal trauma, chronic ocular surface disease, topical corticosteroids, or contact lenses 184 . Fluconazole, voriconazole, natamycin, or ketoconazole preparations are antifungal drugs often used to treat keratomycoses; however, the bioavailability of these drugs is a significant challenge. El-Emam et al . successfully developed the voriconazole-loaded proniosomal formula, which has a high capture efficiency (87.4±2.55%) 185 . In an in vitro experiment against Candida albicans and Aspergillus nidulans , the formulation of proniosomes exhibited excellent antifungal effects and stable drug release. Other antifungal drugs for fungal keratitis treatment have also been studied for their ability to increase bioavailability. Soliman et al . reported that a niosomal gel containing fluconazole was twice as bioavailable as the corresponding microemulsion 186 . A study on fluconazole loaded in niosomes for ocular drug delivery was performed by Elmotasem et al . with the FL-HP-β-CD complex enclosed in eudragit nanoparticles. The results showed that niosomes coated with cationic chitosan improved adhesion, increased corneal permeability, and prolonged action 187 . As reported by Abdelbary et al ., ketoconazole-loaded proniosomal gels presented increased corneal exposure, penetration, and ocular retention times, resulting in a sustained effect and increased bioavailability by 20 times compared with that of the conventional suspension 188 .

Bacterial keratitis

The treatment of bacterial keratitis involves the topical use of antibiotics in combination with corticosteroids to improve patient compliance 189 . Niosomes containing ethanol and cholesterol have been studied for the intraocular delivery of prednisolone acetate and prednisolone sodium phosphate, and the results have shown promise in drug delivery 190 . Abdelbary et al. reported that the formulation of lomefloxacin HCl (LXN) in the niosomal form resulted in an increase of 166% in drug permeation across the cornea compared with that of an LXN solution 191 . Khalil et al. also studied niosomes with LXN on an optimized formula that remained stable under refrigerated conditions for up to three months, and there was a significantly greater percentage of inhibition of S. aureus than that of a commercial product 192 . Clinical observations and colony counts of infected eyes revealed considerable improvement in the treatment response. In infected rabbit eyes, the effectiveness of the niosomal formula was evaluated compared with that of a commercial drug. After eight days, the group treated with the LXN-vesicular formula presented no viable bacteria or any signs of inflammation, indicating an improvement in the treatment response. Thus, the niosomal dispersion of LXN could serve as a superior ocular delivery system that holds promise for treating bacterial conjunctivitis 192 . Further research by Khalil et al . was carried out to improve the drug bioavailability of LXN in proniosomes 193 . The optimized proniosomal gel formulation (P-LXN 7) appeared as spherical vesicles with high adhesion efficiency (> 80%) and exhibited controlled drug release for 12 h. P-LXN 7 was shown to be safe and suitable for ophthalmic use, having a greater antibacterial effect than commercially available LXN eye drops. Allam et al. developed a vancomycin-loaded niosomal formulation with high drug capture efficiency and increased eye residence time 194 . An in vivo study on MRSA-infected rabbits revealed that this formulation was 2.5 times more effective in terms of antibacterial activity than a vancomycin-free drug solution 194 . Gugleva et al . studied doxycycline-loaded niosomes, which demonstrated sustained release properties and satisfactory stability after two months of storage at 4 °C 195 . Further research on doxycycline niosomal topical heat-sensing gels also revealed enhanced antibacterial activity compared with that of a doxycycline solution 196 . To improve the corneal residence time of ciprofloxacin (an antibiotic commonly used to treat bacterial conjunctivitis), Ameeduzzafar et al. developed a formulation of chitosan-coated niosomes loaded with the drug. Compared with commercially available ciprofloxacin eye drops, the optimized formula resulted in a 1.79-fold increase in corneal permeability 197 . Chitosan-coated niosomes were found to be promising drug transporters for treating bacterial conjunctivitis, as they significantly inhibited bacterial growth and increased the corneal contact time. A niosomal formulation loaded with flurbiprofen to increase the ocular drug retention time and improve its anti-inflammatory activity was reported by El-Saved et al . Later, in vitro and in vivo studies revealed reduced drug crystallinity, higher interaction rates with other niosomal contents, and elevated ocular bioavailability than the corresponding flurbiprofen ophthalmic solutions 198 . Recently, curcumin has been shown to replace common active anti-inflammatory ingredients for the treatment of eye inflammation. The curcumin-loaded PG formulation exhibited a high capture efficiency (96.0 ± 0.1%). The resulting permeability was 3.22 times and 1.76 times greater than that of the dispersed and lyophilized forms, respectively 199 .

Other niosomal formulations have also been developed to deliver topical medications to the eye. Studies carried out by Puras et al . revealed that when protamine was incorporated into niosomal DNA vectors, the transformation efficiency, cell viability, and DNA concentration improved. This study highlighted the enhanced properties of protamine/DNA/niosome tertiary vectors for efficient and safe gene delivery to the mouse retina, which could become a new promising genetic material delivery system for the retina 200 . The azithromycin chitosan-coated niosomes constructed and optimized by Eid et al . presented a novel colloidal system that increased the residence time, ocular permeability, and bioavailability. The optimized niosomes markedly improved the apparent osmolarity, corneal permeability, and azithromycin concentration in rabbit eyes without significant irritant effects 201 .

Advantages and disadvantages

As reported in many studies, various forms of niosomes have shown outstanding abilities in ocular drug delivery. They can be considered promising drug carriers for treating different eye conditions. They were demonstrated to enhance the chemical stability, bioavailability, and permeability of drug molecules while reducing side effects. Additionally, nanosize niosomes have been reported to resist drainage due to reflex tearing and blinking; thus, they can be better retained and widely spread on the eye surface than other carriers 202 . However, more studies on niosomal formulations should be carried out before their commercialization as ophthalmic medicines. It has been reported that niosomes cannot maintain stability under ambient conditions, leading to drug leakage and decreased encapsulation efficiency 203 , 204 . Moreover, targeted drug delivery strategies to the site of action of the eye should be integrated into niosomal formulations to increase the therapeutic effect. In addition, investigating the cytotoxicity and other side effects is essential for the preclinical or clinical stages to ensure the safety of niosomal drugs.

Respiratory delivery

Overview

Recently, the delivery of drugs through the respiratory system has been shown to have both local and systemic effects. Additionally, this application site can offer an attractive, noninvasive route of administration 205 . Pulmonary drug delivery therapies are widely used to treat respiratory diseases such as inflammation, asthma, chronic obstructive pulmonary disease, cystic fibrosis, diabetes, and other systemic diseases 206 . The advantages of pulmonary drug delivery include a relatively high degree of convenience and excellent absorption (air-to-blood delivery) because of the large surface area of the lung (i.e., 100 m 2 ) and the thin epithelial layer (0.2--0.7 ÎĽm) 207 . On the other hand, local pulmonary drug delivery to treat lung diseases could reduce systemic distribution and adverse effects 208 .

However, this route of administration also has several limitations, such as the low effectiveness of inhalation systems, the small amount of drug delivered per puff, and the instability of some drugs in these pulmonary formulations 58 . To reach the lower respiratory tract, the particle size should be between 1 and 5 μm. Particles smaller than 1 μm cannot be retained in the lungs, whereas those larger than 1 μm are deposited mainly in the upper respiratory tract 209 . In particular, those ranging from 2–5 μm are deposited in the bronchioles by sedimentation, whereas particles less than 2 μm are deposited in the alveoli by Brownian diffusion, as illustrated in Figure 6 . Notably, the various transportation mechanisms involved in drug release and absorption across the epithelium depend on the position of the deposition.

Figure 6 . Pathway of drug release from niosomes in respiratory drug delivery.

Applications

Gemcitabine (Gem) and cisplatin (Cis) are common drugs used for lung cancer treatment. A study by Mohamad Saimi et al. developed an aerosolized niosome formulation containing Gem and Cis, aiming to reduce the number of dogs due to the targeted capability of the niosome. The authors determined a suitable optimized niosome formulation with an aerosol output of 96.22%. Cytotoxicity to normal lung MRC5 and cancerous A549 pulmonary cell lines was detected, and the results revealed moderate toxicity to A549 cells, with an IC 50 = 46.0 µg/mL, but weak toxicity to MCR5 cells (IC 50 = 280.0 µg/mL). Therefore, aerosolized niosome formulations are promising candidates for aerosolized delivery methods to treat lung cancer with fewer side effects on normal lung cells 210 . Nintedanib has been used to treat idiopathic pulmonary fibrosis and other lung diseases 211 . In the study of Shukla et al ., nintedanib was formulated as an inhalable agent for non-small cell lung cancer treatment. Cytotoxicity studies have indicated that niosomal formulations have greater cytotoxic effects on lung cancer cells (A549, H2122, H1299, H358, and H460) than plain nintedanib does 212 . Curcumin, a natural phenolic compound isolated from turmeric, shows promise as a chemopreventive and anticancer agent, with multiple studies suggesting its ability to inhibit the onset and improvement of non-small cell lung carcinoma 213 , 214 . Curcumin-loaded niosomes were shown to have anti-inflammatory effects on respiratory syncytial virus-induced respiratory disease in a BALB/c mouse model. The mice were treated daily with the niosome, and after five injections, the lung pathology was alleviated, which reduced immune cell infiltration and the synthesis of inflammatory mediators (MIP-1, TNF-α, and IFN-γ) in the lungs 215 .

Advantages and disadvantages

Pulmonary drug absorption benefits from an extensive contact area facilitated by the dense capillary network in the alveoli, enhancing the interaction between air and blood 216 . Niosomal drugs can help improve permeation via pulmonary administration via airway mucus, and these vesicles were also found to significantly increase drug uptake by human lung fibroblasts 217 . Moreover, they can provide a tangible strategy to resolve formulation-related problems associated with many poorly soluble therapeutic molecules to treat local respiratory and systemic diseases. The niosomal delivery of a drug molecule through the pulmonary route extends its residence time in the lung, preventing systemic side effects. However, limitations exist regarding drug irritation, toxicity, and in vivo stability; ensuring drug transport to the intended site of action is not guaranteed, and challenges may arise in drug retention and clearance owing to the efficient clearance mechanisms of the lungs 216 , 218 .

Discussion

During the past decade, with remarkable efficiency and safety as drug carriers, several lipid-based nanoparticles, such as AmBisome® (Amphotericin B), DaunoXome® (daunorubicin), and Doxyl® (doxorubicin), have been successfully commercialized 219 . Because lipid-based particles are similar in structure to the phospholipid bilayer in cell membranes 220 , the promise of lipid-based carriers is enormous, leading to the first appearance of liposomes. Since then, an increasing number of different types of lipid particles, such as ethosomes, sphingosomes, transfersomes, and niosomes, whose characteristics have been modified and improved to control drug release and suit different routes of administration. Niosomes offer notable benefits over other lipid nanoparticle varieties, including increased chemical stability, reduced production expenses, abundant nonionic surfactant options, biocompatibility, and biodegradability 221 , 222 .

In addition to some advantages, well-known and popular methods for the preparation of niosomes, such as thin-film hydration, ether injection, and reverse-phase evaporation, involve the use of organic solvents, thereby increasing the risk of their contamination in the final products 37 . Furthermore, the large particle size and heat sterilization significantly reduce vesicle stability because of their potential to disrupt lipid structures or surfactant-based formulations 17 . In addition, surfactants play an essential role in the formation of niosomes, determining some properties or limitations of niosomes, such as toxicity, drug release ability, and physicochemical stability 223 . Numerous papers have recently revealed information about the cytotoxicity of surfactants and niosome molecules 224 , 225 . However, long-term toxicity in animal models after administration has yet to be reported; thus, further studies on tolerability and clinical implications are needed.

Therefore, the current trend of niosome preparation is to increase the efficiency of controlled drug release, avoid the use of organic solvents, and reduce the particle size in just one step; multiple methods, such as the ball milling method 222 and bubble method 14 , have recently been studied. Another development goal of niosomes is to optimize their preparation technique, aiming for improved quality and scalability to produce a greater number of niosomes. Furthermore, the future goal of niosomes is not only to encapsulate the active ingredient into vesicles but also to load the targeted drug at the site of action, such as the central nervous system, to significantly increase permeability, cross the blood‒brain barrier and increase bioavailability via modification with ligands 226 . Niosomes decorated with ligands have been shown to prolong their presence in blood circulation 227 and enhance their ability to traverse the blood‒brain barrier 19 .

Although there are reports of niosomes loaded with antifungal, antineoplastic, and antidiabetic compounds for intravenous, oral, and transdermal administration, compared with liposomes, most studies are still in vitro and in vivo 228 . The main application of niosomes is in transdermal drug delivery systems, which are mainly topical products, such as LancĂ´me's 229 , 15 . Since then, no drug-based niosomes have been marketed orally or injected. The limitation of the transdermal route for the treatment of certain diseases, such as cancer or infection, is extending the duration of treatment and leading to poor patient compliance. Because niosomes provide a large area and many ligand binding sites, such as antibodies, peptide chains, and folic acid 230 , they can be modified to diversify drug delivery pathways and increase the application efficiency of niosomes. In addition to loading pharmaceutical compounds, the use of natural therapeutic molecules (e.g., curcumin 231 and morusin 232 ) is also a focus of research because of their ability to encapsulate both lipophilic and hydrophilic molecules owing to the presence of amphiphilic surfactants, which help absorb natural-origin molecules into the body, reduce the side effects of some therapeutic drugs and synergistically increase the therapeutic potential of drugs.

Conclusion

Niosomes have long been known as effective drug delivery vehicles. The characteristics of niosomes can be altered by adjusting their size and structure during the preparation process. Niosomes may offer additional benefits in terms of stability and cost compared with liposomes. Additionally, the application of niosomes via oral, dermal, ocular, and respiratory routes is viable for safe long-term treatment and focused medication delivery. Although topical niosomes appear to be a feasible and promising strategy for therapeutic and cosmetic applications, further investigation is needed to understand the mode of interaction between niosomal particles and stratum corneum lipids and their impact on drug penetration. More clinical investigations and postmarketing surveillance should be conducted to obtain more evidence for the safety and efficacy of niosomes in humans. Although controlled clinical studies are needed to acquire additional information about niosome safety for long-term treatment, stability, and bioavailability effects, niosomes are attractive candidates for treating ocular diseases. Vaccine and biotechnology product delivery are also potential areas of research that should attract more attention. The encouraging results from niosomal systems and the emerging discoveries in this field will likely lead to more viable commercial niosomal products.

List of abbreviations

CS Chitosan

EE% Entrapment efficiency

FLP Finasteride-loaded proniosomes

HLB Hydrophilic‒lipophilic balance

LUVs Large unilamellar vesicles

LXN Lomefloxacin HCl

MLVs Multilamellar vesicles

MTX Methotrexate

PDI Polydispersity index

PEG Polyethylene glycol

R6G Rhodamine 6G

SC Stratum corneum

SUVs Small unilamellar vesicles

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

This research is funded by Vietnam National University Ho Chi Minh City (VNU-HCM) under grant number DN2022-44-01.

Authors’ contributions

The concepts were proposed by Minh Hien Nguyen and Minh Tri Le. Minh Hien Nguyen, Thien Han Nguyen Le, Tran Phuoc Thuan Nguyen, Thi Ngoc Tam Le, Thi Yen Nhi Nguyen, Kim Anh Nguyen, and Thi Tan Pham undertook to collect, summarize, and evaluate researches, news. All authors contributed to drafting and revising the final manuscript.

References

  1. Devarajan PV, Jain S. Targeted Drug Delivery: Concepts and Design. first ed. New York: Springer Cham; 2015. . ;:. Google Scholar
  2. Tewabe A, Abate A, Tamrie M, Seyfu A, Siraj EA. Targeted Drug Delivery-From Magic Bullet to Nanomedicine: Principles, Challenges, and Future Perspectives. J Multidiscip Healthc. 2021;14:1711-1724. . ;:. Google Scholar
  3. Ali Y, Alqudah A, Ahmad S, Hamid SA, Farooq U. Macromolecules as targeted drugs delivery vehicles: an overview. Des Monomers Polym. 2019;22(1):91-97. . ;:. Google Scholar
  4. Kumar A, Nautiyal U, Kaur C, Goel V, Piarchand N. Targeted drug delivery system: current and novel approach. Int J Pharm Med Res. 2017;5(2):448-454. . ;:. Google Scholar
  5. Liyanage PY, Hettiarachchi SD, Zhou Y, Ouhtit A, Seven ES, Oztan CY, et al. Nanoparticle-mediated targeted drug delivery for breast cancer treatment. Biochim Biophys Acta Rev Cancer. 2019;1871(2):419-433. . ;:. Google Scholar
  6. Ertas YN, Abedi Dorcheh K, Akbari A, Jabbari E. Nanoparticles for Targeted Drug Delivery to Cancer Stem Cells: A Review of Recent Advances. Nanomaterials. 2021;11(7):1755. . ;:. Google Scholar
  7. Crommelin DJA, van Hoogevest P, Storm G. The role of liposomes in clinical nanomedicine development. What now? Now what? J Control Release. 2020;318:256-263. . ;:. Google Scholar
  8. Bhardwaj P, Tripathi P, Gupta R, Pandey S. Niosomes: A review on niosomal research in the last decade. J Drug Deliv Sci Technol. 2020;56:101581. . ;:. Google Scholar
  9. Opatha SAT, Titapiwatanakun V, Chutoprapat R. Transfersomes: A promising nanoencapsulation technique for transdermal drug delivery. Pharmaceutics. 2020;12(9):855. . ;:. Google Scholar
  10. Ghanbarzadeh S, Khorrami A, Arami S. Nonionic surfactant-based vesicular system for transdermal drug delivery. Drug Deliv. 2015;22(8):1071-1077. . ;:. Google Scholar
  11. Chacko IA, Ghate VM, Dsouza L, Lewis SA. Lipid vesicles: A versatile drug delivery platform for dermal and transdermal applications. Colloids Surf B. 2020;195:111262. . ;:. Google Scholar
  12. Sita VG, Jadhav D, Vavia P. Niosomes for nose-to-brain delivery of bromocriptine: Formulation development, efficacy evaluation and toxicity profiling. J Drug Deliv Sci Technol. 2020;58:101791. . ;:. Google Scholar
  13. Sadeghi-Ghadi Z, Ebrahimnejad P, Talebpour Amiri F, Nokhodchi A. Improved oral delivery of quercetin with hyaluronic acid containing niosomes as a promising formulation. J Drug Target. 2021;29(2):225-234. . ;:. Google Scholar
  14. Durak S, Esmaeili Rad M, Alp Yetisgin A, Eda Sutova H, Kutlu O, Cetinel S, et al. Niosomal Drug Delivery Systems for Ocular Disease—Recent Advances and Future Prospects. Nanomaterials. 2020;10(6):1191. . ;:. Google Scholar
  15. Madane VB, Aloorkar NH, Mokale VJ. Niosomes As an Ideal Drug Delivery System. J NanoSci Res Rep. 2021;3(3):1-9. . ;:. Google Scholar
  16. Nowroozi F, Almasi A, Javidi J, Haeri A, Dadashzadeh S. Effect of Surfactant Type, Cholesterol Content and Various Downsizing Methods on the Particle Size of Niosomes. Iran J Pharm Res. 2018;17(Suppl2):1-11. . ;:. Google Scholar
  17. Chen S, Hanning S, Falconer J, Locke M, Wen J. Recent advances in nonionic surfactant vesicles (niosomes): Fabrication, characterization, pharmaceutical and cosmetic applications. Eur J Pharm Biopharm. 2019;144:18-39. . ;:. Google Scholar
  18. Marianecci C, Di Marzio L, Rinaldi F, Celia C, Paolino D, Alhaique F, et al. Niosomes from 80 s to present: The state of the art. Adv Colloid Interface Sci. 2014;205:187-206. . ;:. Google Scholar
  19. Gharbavi M, Amani J, Kheiri-Manjili H, Danafar H, Sharafi A. Niosome: A Promising Nanocarrier for Natural Drug Delivery through Blood‒Brain Barrier. Adv Pharmacol Sci. 2018;2018:6847971. . ;:. Google Scholar
  20. Sankhyan A, Pawar P. Recent Trends in Niosome as Vesicular DrugDelivery System. J Appl Pharm Sci. 2012(Issue):20-32. . ;:. Google Scholar
  21. Kazi KM, Mandal AS, Biswas N, Guha A, Chatterjee S, Behera M, et al. Niosome: A future of targeted drug delivery systems. J Adv Pharm Technol Res. 2010;1(4):374-373 s380. . ;:. Google Scholar
  22. Homaei M. Preparation and characterization of giant niosomes [Master Thesis]. Gothenburg, Sweden Chalmers University of Technology; 2016. . ;:. Google Scholar
  23. Moghassemi S, Hadjizadeh A. Nanoniosomes as nanoscale drug delivery systems: an illustrated review. J Control Release. 2014;185:22-36. . ;:. Google Scholar
  24. Sambhakar S, Paliwal S, Sharma S, Singh B. Formulation of risperidone loaded proniosomes for effective transdermal delivery: An in vitro and in vivo study. Bull Fac Pharm Cairo Univ. 2017;55(2):239-247. . ;:. Google Scholar
  25. Mittal S, Chaudhary A, Chaudhary A, Kumar A. Proniosomes: the effective and efficient drug-carrier system. Ther Deliv. 2020;11(2). . ;:. Google Scholar
  26. Martin F. Pharmaceutical manufacturing of liposomes. Drug Pharm Sci. 1990;41:267-316. . ;:. Google Scholar
  27. Basiri L, Rajabzadeh G, Bostan A. α-Tocopherol-loaded niosome prepared by heating method and its release behavior. Food Chem. 2017;221:620-628. . ;:. Google Scholar
  28. Naderi R, Pardakhty A, Abbasi MF, Ranjbar M, Iranpour MJSR. Preparation and evaluation of crocin loaded in nanoniosomes and their effects on ischemia–reperfusion injuries in rat kidney. Sci Rep. 2021;11(1):23525. . ;:. Google Scholar
  29. Ge X, Wei M, He S, Yuan W‒E. Advances of Non-Ionic Surfactant Vesicles (Niosomes) and Their Application in Drug Delivery. Pharmaceutics. 2019;11(2):55. . ;:. Google Scholar
  30. Yuksel N, Bayindir ZS, Aksakal E, Ozcelikay AT. In situ niosome forming maltodextrin proniosomes of candesartan cilexetil: In vitro and in vivo evaluations. Int J Biol Macromol. 2016;82:453-463. . ;:. Google Scholar
  31. Hu C, Rhodes DG. Proniosomes: a novel drug carrier preparation. Int J Pharm. 1999;185(1):23-35. . ;:. Google Scholar
  32. Yeo PL, Lim CL, Chye SM, Kiong Ling AP, Koh RY. Niosomes: a review of their structure, properties, methods of preparation, and medical applications. Asian Biomed. 2017;11(4):301-314. . ;:. Google Scholar
  33. Bhaskaran S, Lakshmi P. Comparative evaluation of niosome formulations prepared by different techniques. Acta Pharm Sci. 2009;51(1). . ;:. Google Scholar
  34. Mayer LD, Bally MB, Hope MJ, Cullis PR. Uptake of antineoplastic agents into large unilamellar vesicles in response to a membrane potential. Biochim Biophys Acta Biomembr. 1985;816(2):294-302. . ;:. Google Scholar
  35. Kumavat S, Sharma PK, Koka SS, Sharma R, Gupta A, Darwhekar GN. A review on niosomes: potential vesicular drug delivery system. J Drug Deliv Ther. 2021;11(5):208-212. . ;:. Google Scholar
  36. Bagheri A, Chu BS, Yaakob H. Niosomal drug delivery systems: Formulation, preparation and applications. World Appl Sci J. 2014;32(8):1671-1685. . ;:. Google Scholar
  37. Khan DH, Bashir S, Figueiredo P, Santos HA, Khan MI, Peltonen L. Process optimization of ecological probe sonication technique for production of rifampicin loaded niosomes. J Drug Deliv Sci Technol. 2019;50:27-33. . ;:. Google Scholar
  38. Abdelkader H, Alani AWG, Alany RG. Recent advances in nonionic surfactant vesicles (niosomes): self-assembly, fabrication, characterization, drug delivery applications and limitations. Drug Deliv. 2014;21(2):87-100. . ;:. Google Scholar
  39. Abdelkader H, Ismail S, Kamal A, Alany RG. Design and evaluation of controlled-release niosomes and discomes for naltrexone hydrochloride ocular delivery. J Pharm Sci. 2011;100(5):1833-1846. . ;:. Google Scholar
  40. Amoabediny G, Haghiralsadat F, Naderinezhad S, Helder MN, Akhoundi Kharanaghi E, Mohammadnejad Arough J, et al. Overview of preparation methods of polymeric and lipid-based (niosome, solid lipid, liposome) nanoparticles: A comprehensive review. Int J Polym Mater Polym Biomater. 2018;67(6):383-400. . ;:. Google Scholar
  41. Andra V, Pammi S, Bhatraju L, Ruddaraju L. A comprehensive review on novel liposomal methodologies, commercial formulations, clinical trials and patents. BioNanoSci. 2022;12:274-291. . ;:. Google Scholar
  42. Samed N, Sharma V, Sundaramurthy A. Hydrogen bonded niosomes for encapsulation and release of hydrophilic and hydrophobic anti-diabetic drugs: an efficient system for oral anti-diabetic formulation. Appl Surf Sci. 2018;449:567-573. . ;:. Google Scholar
  43. Masjedi M, Montahaei T. An illustrated review on nonionic surfactant vesicles (niosomes) as an approach in modern drug delivery: Fabrication, characterization, pharmaceutical, and cosmetic applications. J Drug Deliv Sci Technol. 2021;61:102234. . ;:. Google Scholar
  44. Ag Seleci D, Seleci M, Walter J-G, Stahl F, Scheper T. Niosomes as nanoparticular drug carriers: fundamentals and recent applications. J Nanomater. 2016;2016:7372306. . ;:. Google Scholar
  45. Moammeri A, Chegeni MM, Sahrayi H, Ghafelehbashi R, Memarzadeh F, Mansouri A, et al. Current advances in niosomes applications for drug delivery and cancer treatment. Materials Today, Bio. 2023;23:100837. . ;:. Google Scholar
  46. Webb C, Forbes N, Roces CB, Anderluzzi G, Lou G, Abraham S, et al. Using microfluidics for scalable manufacturing of nanomedicines from bench to GMP: A case study using protein-loaded liposomes. Int J Pharm. 2020;582:119266. . ;:. Google Scholar
  47. Obeid MA, Al Qaraghuli M, Ruano M, Sangboonruang S, Alsaadi M, Tragoolpua Y, et al. Lipid-based nanomaterials in cancer treatment and diagnosis. In: Aljabali AA, Pal K, editors. Bionanotechnology: Next-Generation Therapeutic Tools. Sharjah: Bentham Science Publisher; 2022. p. 49-83. . ;:. Google Scholar
  48. Obeid MA, Khadra I, Mullen AB, Tate RJ, Ferro VA. The effects of hydration media on the characteristics of nonionic surfactant vesicles (NISV) prepared by microfluidics. Int J Pharm. 2017;516(1-2):52-60. . ;:. Google Scholar
  49. Dittrich PS, Manz A. Lab-on-a-chip: microfluidics in drug discovery. Nat Rev Drug Discov. 2006;5(3):210-218. . ;:. Google Scholar
  50. Seleci DA, Maurer V, Stahl F, Scheper T, Garnweitner G. Rapid Microfluidic Preparation of Niosomes for Targeted Drug Delivery. Int J Mol Sci. 2019;20(19):4696. . ;:. Google Scholar
  51. Verma S, Singh SK, Syan N, Mathur P, Valecha V. Nanoparticle vesicular systems: a versatile tool for drug delivery. J Chem Pharm Res. 2010;2(2):496-509. . ;:. Google Scholar
  52. Kazi KM, Mandal AS, Biswas N, Guha A, Chatterjee S, Behera M, et al. Niosome: a future of targeted drug delivery systems. J Adv Pharm Technol Res. 2010;1(4):374. . ;:. Google Scholar
  53. Kumar GP, Rajeshwarrao P. Nonionic surfactant vesicular systems for effective drug delivery-an overview. Acta Pharm Sin B. 2011;1(4):208-219. . ;:. Google Scholar
  54. He X, Chen J, He X, Feng Z, Li C, Liu W, et al. Industry-scale microfluidization as a potential technique to improve solubility and modify structure of pea protein. Innov Food Sci Emerg Technol. 2021;67:102582. . ;:. Google Scholar
  55. Zidan AS, Rahman Z, Khan MA. Product and process understanding of a novel pediatric anti-HIV tenofovir niosomes with a high-pressure homogenizer. Eur J Pharm Sci. 2011;44(1-2):93-102. . ;:. Google Scholar
  56. Mehta M, Bui TA, Yang X, Aksoy Y, Goldys EM, Deng W. Lipid-Based Nanoparticles for Drug/Gene Delivery: An Overview of the Production Techniques and Difficulties Encountered in Their Industrial Development. ACS Materials Au. 2023;3(6):600-619. . ;:. Google Scholar
  57. Puri A, Loomis K, Smith B, Lee JH, Yavlovich A, Heldman E, et al. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to clinic. Crit Rev Ther Drug Carrier Syst. 2009;26(6):523-580. . ;:. Google Scholar
  58. Khoee S, Yaghoobian M. Chapter 6 - Niosomes: a novel approach in modern drug delivery systems. In: Andronescu E, Grumezescu AM, editors. Nanostructures for Drug Delivery. Amsterdam: Elsevier; 2017. p. 207-237. . ;:. Google Scholar
  59. Amiri B, Ahmadvand H, Farhadi A, Najmafshar A, Chiani M, Norouzian D. Delivery of vinblastine-containing niosomes results in potent in vitro/in vivo cytotoxicity on tumor cells. Drug Dev Ind Pharm. 2018;44(8):1371-1376. . ;:. Google Scholar
  60. El-Ridy MS, Abdelbary A, Essam T, El-Salam RM, Kassem AA. Niosomes as a potential drug delivery system for increasing the efficacy and safety of nystatin. Drug Dev Ind Pharm. 2011;37(12):1491-1508. . ;:. Google Scholar
  61. Hashim F, El-Ridy M, Nasr M, Abdallah Y. Preparation and characterization of niosomes containing ribavirin for liver targeting. Drug Deliv. 2010;17(5):282-287. . ;:. Google Scholar
  62. Hedayati Ch M, Abolhassani Targhi A, Shamsi F, Heidari F, Salehi Moghadam Z, Mirzaie A, et al. Niosome-encapsulated tobramycin reduced antibiotic resistance and enhanced antibacterial activity against multidrug-resistant clinical strains of Pseudomonas aeruginosa. J Biomed Mater Res A. 2021;109(6):966-980. . ;:. Google Scholar
  63. Jin-Ying Wong, Zhao Yin Ng, Meenu Mehta, Shakti D Shukla, Jithendra Panneerselvam, Thiagarajan Madheswaran, et al. Curcumin-loaded niosomes downregulate mRNA expression of pro-inflammatory markers involved in asthma: an in vitro study. Nanomedicine. 2020;15(30):2955–2970. . ;:. Google Scholar
  64. Khan S, Akhtar MU, Khan S, Javed F, Khan AA. Nanoniosome-encapsulated levoflaxicin as an antibacterial agent against Brucella. J Basic Microbiol. 2020;60(3):281-290. . ;:. Google Scholar
  65. Gilani SJ, Imam SS, Ahmed A, Chauhan S, Mirza MA, Taleuzzaman M. Formulation and evaluation of thymoquinone niosomes: application of developed and validated RP-HPLC method in delivery system. Drug Dev Ind Pharm. 2019;45(11):1799-1806. . ;:. Google Scholar
  66. Damera DP, Venuganti VVK, Nag A. Deciphering the Role of Bilayer of a Niosome toward Controlling the Entrapment and Release of Dyes. ChemistrySelect. 2018;3(14):3930-3938. . ;:. Google Scholar
  67. Kaszuba M, Corbett J, Watson FM, Jones A. High-concentration zeta potential measurements using light-scattering techniques. Philosophical transactions of the royal society a: mathematical, physical and engineering sciences. 2010;368(1927):4439-4451. . ;:. Google Scholar
  68. Honary S, Zahir F. Effect of zeta potential on the properties of nanodrug delivery systems-a review (Part 1). Trop J Pharm Res. 2013;12(2):255-264. . ;:. Google Scholar
  69. Griffin WC. Classification of surface-active agents by" HLB". J Soc Cosmet Chem. 1949;1:311-325. . ;:. Google Scholar
  70. Agrati C, Marianecci C, Sennato S, Carafa M, Bordoni V, Cimini E, et al. Multicompartment vectors as novel drug delivery systems: selective activation of Tγδ lymphocytes after zoledronic acid delivery. Nanomedicine. 2011;7(2):153-161. . ;:. Google Scholar
  71. Jain N, Verma A, Jain N. Formulation and investigation of pilocarpine hydrochloride niosomal gels for the treatment of glaucoma: intraocular pressure measurement in white albino rabbits. Drug Deliv. 2020;27(1):888-899. . ;:. Google Scholar
  72. Moazeni E, Gilani K, Sotoudegan F, Pardakhty A, Najafabadi AR, Ghalandari R, et al. Formulation and in vitro evaluation of ciprofloxacin containing niosomes for pulmonary delivery. J Microencapsul. 2010;27(7):618-627. . ;:. Google Scholar
  73. Mishra V, Bansal KK, Verma A, Yadav N, Thakur S, Sudhakar K, et al. Solid Lipid Nanoparticles: Emerging Colloidal Nano Drug Delivery Systems. Pharmaceutics [Internet]. 2018; 10(4). . ;:. Google Scholar
  74. Junyaprasert VB, Teeranachaideekul V, Supaperm T. Effect of charged and nonionic membrane additives on physicochemical properties and stability of niosomes. AAPS PharmSciTech. 2008;9(3):851-859. . ;:. Google Scholar
  75. Akbarzadeh I, Saremi Poor A, Yaghmaei S, Norouzian D, Noorbazargan H, Saffar S, et al. Niosomal delivery of simvastatin to MDA-MB-231 cancer cells. Drug Dev Ind Pharm. 2020;46(9):1535-1549. . ;:. Google Scholar
  76. Sangkana S, Eawsakul K, Ongtanasup T, Boonhok R, Mitsuwan W, Chimplee S, et al. Preparation and evaluation of a niosomal delivery system containing G. mangostana extract and study of its anti-Acanthamoeba activity. Nanoscale Adv. 2024;6(5):1467-1479. . ;:. Google Scholar
  77. Shah A, Boldhane S, Pawar A, Bothiraja C. Advanced development of a nonionic surfactant and cholesterol material based niosomal gel formulation for the topical delivery of anti-acne drugs. Adv Mater. 2020;1(6):1763-1774. . ;:. Google Scholar
  78. Lo CT, Jahn A, Locascio LE, Vreeland WN. Controlled self-assembly of monodisperse niosomes by microfluidic hydrodynamic focusing. Langmuir. 2010;26(11):8559-8566. . ;:. Google Scholar
  79. García-Manrique P, Machado ND, Fernández MA, Blanco-López MC, Matos M, Gutiérrez G. Effect of drug molecular weight on niosomes size and encapsulation efficiency. Colloids Surf B: Biointerfaces. 2020;186:110711. . ;:. Google Scholar
  80. Luo Y, Yang H, Zhou Y-F, Hu B. Dual and multitargeted nanoparticles for site-specific brain drug delivery. J Control Release. 2020;317:195-215. . ;:. Google Scholar
  81. Díaz-Coránguez M, Ramos C, Antonetti DA. The inner blood‒retinal barrier: Cellular basis and development. Vision Res. 2017;139:123-137. . ;:. Google Scholar
  82. Tetro N, Moushaev S, Rubinchik-Stern M, Eyal S. The placental barrier: the gate and the fate in drug distribution. Pharm Res. 2018;35:1-16. . ;:. Google Scholar
  83. Nowak M, Brown TD, Graham A, Helgeson ME, Mitragotri S. Size, shape, and flexibility influence nanoparticle transport across brain endothelium under flow. Bioeng Transl Med. 2020;5(2):e10153. . ;:. Google Scholar
  84. Rijcken CJ, Snel CJ, Schiffelers RM, van Nostrum CF, Hennink WE. Hydrolysable core-crosslinked thermosensitive polymeric micelles: synthesis, characterization and in vivo studies. Biomaterials. 2007;28(36):5581-5593. . ;:. Google Scholar
  85. Rezaei T, Rezaei M, Karimifard S, Mahmoudi Beram F, Dakkali MS, Heydari M, et al. Folic Acid-Decorated pH-Responsive Nanoniosomes With Enhanced Endocytosis for Breast Cancer Therapy: In Vitro Studies. Front Pharmacol. 2022;13. . ;:. Google Scholar
  86. Yadavar-Nikravesh M-S, Ahmadi S, Milani A, Akbarzadeh I, Khoobi M, Vahabpour R, et al. Construction and characterization of a novel Tenofovir-loaded PEGylated niosome conjugated with TAT peptide for evaluation of its cytotoxicity and anti-HIV effects. Adv Powder Technol. 2021;32(9):3161-3173. . ;:. Google Scholar
  87. d’Avanzo N, Sidorenko V, Simón-Gracia L, Rocchi A, Ottonelli I, Ruozi B, et al. C-end rule peptide-guided niosomes for prostate cancer cell targeting. J Drug Deliv Sci Technol. 2024;91:105162. . ;:. Google Scholar
  88. Karim H, Rehman K, Khan SN, Shuja AA, Kanwal T, Simjee SU, et al. Synthesis and Characterization of Novel Amphiphilic Peptide and its Application in the Development of Niosomal Formulation as a Drug Delivery Carrier for Curcumin. J Clust Sci. 2024;35(5):1587-1599. . ;:. Google Scholar
  89. Hong M, Zhu S, Jiang Y, Tang G, Pei Y. Efficient tumor targeting of hydroxycamptothecin loaded PEGylated niosomes modified with transferrin. J Control Release. 2009;133(2):96-102. . ;:. Google Scholar
  90. Tavano L, Muzzalupo R, Mauro L, Pellegrino M, Andò S, Picci N. Transferrin-Conjugated Pluronic Niosomes as a New Drug Delivery System for Anticancer Therapy. Langmuir. 2013;29(41):12638-12646. . ;:. Google Scholar
  91. Barlas FB, Olceroglu B, Ag Seleci D, Gumus ZP, Siyah P, Dabbek M, et al. Enhancing chemotherapeutic efficacy: Niosomeâ€encapsulated Doxâ€Cis with MUCâ€1 aptamer. Cancer Med. 2024;13(15):e70079. . ;:. Google Scholar
  92. Sangboonruang S, Semakul N, Suriyaprom S, Kitidee K, Khantipongse J, Intorasoot S, et al. Nano-Delivery System of Ethanolic Extract of Propolis Targeting Mycobacterium tuberculosis via Aptamer-Modified-Niosomes. Nanomaterials [Internet]. 2023; 13(2). . ;:. Google Scholar
  93. Seleci DA, Seleci M, Jochums A, Walter J-G, Stahl F, Scheper T. Aptamer mediated niosomal drug delivery. RSC Adv. 2016;6(91):87910-87918. . ;:. Google Scholar
  94. Miatmoko A, Hariawan BS, Cahyani DM, Sari R, Dinaryanti A, Hendrianto E. The effect of chitosan addition on cellular uptake and cytotoxicity of ursolic acid niosomes. An Acad Bras Cienc. 2021;93(3):e20201850. . ;:. Google Scholar
  95. You L, Liu X, Fang Z, Xu Q, Zhang Q. Synthesis of multifunctional Fe3O4@PLGA-PEG nanoniosomes as a targeting carrier for treatment of cervical cancer. Mater Sci Eng C. 2019;94:291-302. . ;:. Google Scholar
  96. Bragagni M, Mennini N, Furlanetto S, Orlandini S, Ghelardini C, Mura P. Development and characterization of functionalized niosomes for brain targeting of dynorphin-B. Eur J Pharm Biopharm. 2014;87(1):73-79. . ;:. Google Scholar
  97. Akbarzadeh I, Tavakkoli Yaraki M, Ahmadi S, Chiani M, Nourouzian D. Folic acid-functionalized niosomal nanoparticles for selective dual-drug delivery into breast cancer cells: An in vitro investigation. Adv Powder Technol. 2020;31(9):4064-4071. . ;:. Google Scholar
  98. Safari Sharafshadeh M, Tafvizi F, Khodarahmi P, Ehtesham S. Folic acid-functionalized PEGylated niosomes coencapsulated cisplatin and doxoribicin exhibit enhanced anticancer efficacy. Cancer Nanotechnology. 2024;15(1):14. . ;:. Google Scholar
  99. Talevi A, Quiroga PA. ADME Processes in Pharmaceutical Sciences. Switzerland: Springer; 2018. . ;:. Google Scholar
  100. Raj GM, Raveendran R. Introduction to Basics of Pharmacology and Toxicology. first ed. Singapore: Springer; 2019. . ;:. Google Scholar
  101. Gavhane YN, Yadav AV. Loss of orally administered drugs in GI tract. Saudi Pharm J. 2012;20(4):331-344. . ;:. Google Scholar
  102. Arzani G, Haeri A, Daeihamed M, Bakhtiari-Kaboutaraki H, Dadashzadeh S. Niosomal carriers enhance oral bioavailability of carvedilol: effects of bile salt-enriched vesicles and carrier surface charge. Int J Nanomedicine. 2015;10:4797-4813. . ;:. Google Scholar
  103. Lundquist P, Artursson P. Oral absorption of peptides and nanoparticles across the human intestine: Opportunities, limitations and studies in human tissues. Adv Drug Deliv Rev. 2016;106:256-276. . ;:. Google Scholar
  104. Cheng R, Xu T, Wang C, Gan C. The stabilization and antioxidant performances of coenzyme Q10-loaded niosomes coated by PEG and chitosan. J Mol Liq. 2021;325:115194. . ;:. Google Scholar
  105. Zubairu Y, Negi LM, Iqbal Z, Talegaonkar S. Design and development of novel bioadhesive niosomal formulation for the transcorneal delivery of anti-infective agent: In vitro and ex-vivo investigations. Asian J Pharm Sci. 2015;10(4):322-330. . ;:. Google Scholar
  106. AbuElfadl A, Boughdady M, Meshali M. New Peceol™/Span™ 60 Niosomes Coated with Chitosan for Candesartan Cilexetil: Perspective Increase in Absolute Bioavailability in Rats. Int J Nanomedicine. 2021;16:5581-5601. . ;:. Google Scholar
  107. Moghassemi S, Hadjizadeh A, Omidfar K. Formulation and Characterization of Bovine Serum Albumin-Loaded Niosome. AAPS PharmSciTech. 2017;18(1):27-33. . ;:. Google Scholar
  108. Sabry S, El hakim Ramadan A, Abd elghany M, Okda T, Hasan A. Formulation, characterization, and evaluation of the anti-tumor activity of nanosized galangin loaded niosomes on chemically induced hepatocellular carcinoma in rats. J Drug Deliv Sci Technol. 2021;61:102163. . ;:. Google Scholar
  109. Kengkittipat W, Kaewmalun S, Khongkow M, Iempridee T, Jantimaporn A, Bunwatcharaphansakun P, et al. Improvement of the multiperformance biocharacteristics of cordycepin using BiloNiosome-core/chitosan-shell hybrid nanocarriers. Colloids Surf B. 2021;197:111369. . ;:. Google Scholar
  110. Wang Q, Wang Y, Xie Y, Adu-Frimpong M, Wei C, Yang X, et al. Nonionic surfactant vesicles as a novel drug delivery system for increasing the oral bioavailability of Ginsenoside Rb1. Food Bioscience. 2021;42:101064. . ;:. Google Scholar
  111. Song S, Tian B, Chen F, Zhang W, Pan Y, Zhang Q, et al. Potentials of proniosomes for improving the oral bioavailability of poorly water-soluble drugs. Drug Dev Ind Pharm. 2015;41(1):51-62. . ;:. Google Scholar
  112. Ibrahim MM, Shehata TM. Tramadol HCl encapsulated niosomes for extended analgesic effect following oral administration. J Drug Deliv Sci Technol. 2018;46:14-18. . ;:. Google Scholar
  113. Shilakari Asthana G, Sharma PK, Asthana A. In Vitro and In Vivo Evaluation of Niosomal Formulation for Controlled Delivery of Clarithromycin. Scientifica (Cairo). 2016;2016:6492953-6492953. . ;:. Google Scholar
  114. Karandikar S, Mirani A, Waybhase V, Patravale VB, Patankar S. Chapter 10 - Nanovaccines for oral delivery-formulation strategies and challenges. In: Andronescu E, Grumezescu AM, editors. Nanostructures for Oral Medicine. Amsterdam: Elsevier; 2017. p. 263-293. . ;:. Google Scholar
  115. Khalifa A-ZM, Rasool BKA. Optimized mucoadhesive coated niosomes as a sustained oral delivery system of famotidine. AAPS PharmSciTech. 2017;18(8):3064-3075. . ;:. Google Scholar
  116. Kamboj S, Saini V, Bala S. Formulation and characterization of drug loaded nonionic surfactant vesicles (niosomes) for oral bioavailability enhancement. Sci World J. 2014;2014:959741. . ;:. Google Scholar
  117. Yaghoobian M, Haeri A, Bolourchian N, Shahhosseni S, Dadashzadeh S. The Impact of Surfactant Composition and Surface Charge of Niosomes on the Oral Absorption of Repaglinide as a BCS II Model Drug. Int J Nanomedicine. 2020;15:8767-8781. . ;:. Google Scholar
  118. El-Menshawe S. A novel approach to topical acetazolamide/PEG 400 ocular niosomes. J Drug Deliv Sci Technol. 2012;22(4):295-299. . ;:. Google Scholar
  119. Kong M, Park H, Feng C, Hou L, Cheng X, Chen X. Construction of hyaluronic acid nisome as functional transdermal nanocarrier for tumor therapy. Carbohydr Polym. 2013;94(1):634-641. . ;:. Google Scholar
  120. Manconi M, Sinico C, Valenti D, Lai F, Fadda AM. Niosomes as carriers for tretinoin. III. A study into the in vitro cutaneous delivery of vesicle-incorporated tretinoin. Int J Pharm. 2006;311(1-2):11-19. . ;:. Google Scholar
  121. Manosroi A, Chankhampan C, Manosroi W, Manosroi J. Transdermal absorption enhancement of papain loaded in elastic niosomes incorporated in gel for scar treatment. Eur J Pharm Sci. 2013;48(3):474-483. . ;:. Google Scholar
  122. Manosroi J, Lohcharoenkal W, Gotz F, Werner RG, Manosroi W, Manosroi A. Transdermal absorption enhancement of N-terminal Tat-GFP fusion protein (TG) loaded in novel low-toxic elastic anionic niosomes. J Pharm Sci. 2011;100(4):1525-1534. . ;:. Google Scholar
  123. Shah J, Nair AB, Shah H, Jacob S, Shehata TM, Morsy MA. Enhancement in antinociceptive and anti-inflammatory effects of tramadol by transdermal proniosome gel. Asian J Pharm Sci. 2020;15(6):786-796. . ;:. Google Scholar
  124. Kassem AA, Abd El-Alim SH, Asfour MH. Enhancement of 8-methoxypsoralen topical delivery via nanosized niosomal vesicles: Formulation development, in vitro and in vivo evaluation of skin deposition. Int J Pharm. 2017;517(1-2):256-268. . ;:. Google Scholar
  125. Fang JY, Yu SY, Wu PC, Huang YB, Tsai YH. In vitro skin permeation of estradiol from various proniosome formulations. Int J Pharm. 2001;215(1-2):91-99. . ;:. Google Scholar
  126. El Maghraby GM, Williams AC, Barry BW. Can drug-bearing liposomes penetrate intact skin? J Pharm Pharmacol. 2006;58(4):415-429. . ;:. Google Scholar
  127. Zhang Y, Zhang K, Wu Z, Guo T, Ye B, Lu M, et al. Evaluation of transdermal salidroside delivery using niosomes via in vitro cellular uptake. Int J Pharm. 2015;478(1):138-146. . ;:. Google Scholar
  128. Singh S, Parashar P, Kanoujia J, Singh I, Saha S, Saraf SAJJodds, et al. Transdermal potential and anti-gout efficacy of Febuxostat from niosomal gel. J Drug Deliv Sci Technol. 2017;39:348-361. . ;:. Google Scholar
  129. Jamal M, Imam SS, Aqil M, Amir M, Mir SR, Mujeeb M. Transdermal potential and anti-arthritic efficacy of ursolic acid from niosomal gel systems. Int Immunopharmacol. 2015;29(2):361-369. . ;:. Google Scholar
  130. Tavano L, Muzzalupo R, Picci N, de Cindio B. Coencapsulation of lipophilic antioxidants into niosomal carriers: percutaneous permeation studies for cosmeceutical applications. Colloids Surf B: Biointerfaces. 2014;114:144-149. . ;:. Google Scholar
  131. Gupta PN, Mishra V, Rawat A, Dubey P, Mahor S, Jain S, et al. Noninvasive vaccine delivery in transfersomes, niosomes and liposomes: a comparative study. Int J Pharm. 2005;293(1-2):73-82. . ;:. Google Scholar
  132. Tavano L, Gentile L, Oliviero Rossi C, Muzzalupo R. Novel gel-niosomes formulations as multicomponent systems for transdermal drug delivery. Colloids Surf B: Biointerfaces. 2013;110:281-288. . ;:. Google Scholar
  133. Junyaprasert VB, Singhsa P, Jintapattanakit A. Influence of chemical penetration enhancers on skin permeability of ellagic acid-loaded niosomes. Asian J Pharm Sci. 2013;8(2):110-117. . ;:. Google Scholar
  134. Maheshwari C, Pandey RS, Chaurasiya A, Kumar A, Selvam DT, Prasad GB, et al. Nonionic surfactant vesicles mediated transcutaneous immunization against hepatitis B. Int Immunopharmacol. 2011;11(10):1516-1522. . ;:. Google Scholar
  135. Ammar HO, Ghorab M, El-Nahhas SA, Higazy IM. Proniosomes as a carrier system for transdermal delivery of tenoxicam. Int J Pharm. 2011;405(1-2):142-152. . ;:. Google Scholar
  136. Azeem A, Anwer MK, Talegaonkar S. Niosomes in sustained and targeted drug delivery: some recent advances. J Drug Target. 2009;17(9):671-689. . ;:. Google Scholar
  137. Sezer AD. Recent Advances in Novel Drug Carrier Systems. first ed. Sezer AD, editor. United Kingdom: IntechOpen; 2012. 165-200 p. . ;:. Google Scholar
  138. El-Boghdadly K, Pawa A, Chin KJ. Local anesthetic systemic toxicity: current perspectives. Local and regional anesthesia. 2018;11:35-44. . ;:. Google Scholar
  139. Shabery AM, Widodo RT, Chik Z. Formulation and In Vivo Pain Assessment of a Novel Niosomal Lidocaine and Prilocaine in an Emulsion Gel (Emulgel) of Semisolid Palm Oil Base for Topical Drug Delivery. Gels. 2023;9(2):96. . ;:. Google Scholar
  140. Wojtyna E, Ĺakuta P, Marcinkiewicz K, Bergler-Czop B, BrzeziĹ„ska-WcisĹ‚o L. Gender, Body Image and Social Support: Biopsychosocial Determinants of Depression Among Patients with Psoriasis. Acta Derm Venereol. 2016;97(1):91-97. . ;:. Google Scholar
  141. Ezhilarasan D. Hepatotoxic potentials of methotrexate: Understanding the possible toxicological molecular mechanisms. Toxicology. 2021;458:152840. . ;:. Google Scholar
  142. Javadzadeh Y, Hamishehkar H. Enhancing percutaneous delivery of methotrexate using different types of surfactants. Colloids Sur B. 2011;82(2):422-426. . ;:. Google Scholar
  143. Lakshmi PK, Devi GS, Bhaskaran S, Sacchidanand S. Niosomal methotrexate gel in the treatment of localized psoriasis: phase I and phase II studies. Indian journal of dermatology, venereology and leprology. 2007;73(3):157-161. . ;:. Google Scholar
  144. Meng S, Sun L, Wang L, Lin Z, Liu Z, Xi L, et al. Loading of water-insoluble celastrol into niosome hydrogels for improved topical permeation and anti-psoriasis activity. Colloids Surf B: Biointerfaces. 2019;182:110352. . ;:. Google Scholar
  145. Dabas G, Vinay K, Parsad D, Kumar A, Kumaran MS. Psychological disturbances in patients with pigmentary disorders: a cross-sectional study. J Eur Acad Dermatol Venereol. 2020;34(2):392-399. . ;:. Google Scholar
  146. Garg BJ, Saraswat A, Bhatia A, Katare OP. Topical treatment in vitiligo and the potential uses of new drug delivery systems. Indian journal of dermatology, venereology and leprology. 2010;76(3):231-238. . ;:. Google Scholar
  147. Garcia‑Melendez ME, Salinas‑Santander M, Sanchez‑Dominguez C, Gonzalez‑Cardenas H, Cerda‑Flores RM, Ocampo‑Candiani J, et al. Protein tyrosine phosphatase PTPN22 +1858C/T polymorphism is associated with active vitiligo. Exp Ther Med. 2014;8(5):1433-1437. . ;:. Google Scholar
  148. Pillaiyar T, Manickam M, Namasivayam V. Skin whitening agents: medicinal chemistry perspective of tyrosinase inhibitors. J Enzyme Inhib Med Chem. 2017;32(1):403-425. . ;:. Google Scholar
  149. Manosroi J, Khositsuntiwong N, Manosroi W, Gotz F, Werner RG, Manosroi A. Enhancement of transdermal absorption, gene expression and stability of tyrosinase plasmid (pMEL34)-loaded elastic cationic niosomes: potential application in vitiligo treatment. J Pharm Sci. 2010;99(8):3533-3541. . ;:. Google Scholar
  150. Manosroi A, Khositsuntiwong N, Götz F, Werner RG, Manosroi J. Transdermal enhancement through rat skin of luciferase plasmid DNA loaded in elastic nanovesicles. J Liposome Res. 2009;19(2):91-98. . ;:. Google Scholar
  151. Auda SH, Fathalla D, Fetih G, El-Badry M, Shakeel F. Niosomes as transdermal drug delivery system for celecoxib: in vitro and in vivo studies. Polym Bull. 2016;73(5):1229-1245. . ;:. Google Scholar
  152. Nasr M, Mansour S, Mortada ND, Elshamy AA. Vesicular aceclofenac systems: a comparative study between liposomes and niosomes. J Microencapsul. 2008;25(7):499-512. . ;:. Google Scholar
  153. Bhardwaj S, Bhatia S. Development and Characterization of Niosomal Gel System using Lallementia royaleana Benth. mucilage for the treatment of Rheumatoid Arthritis. Iran J Pharm Res. 2020;19(3):465-482. . ;:. Google Scholar
  154. Sun Z. Optimization of clobetasol propionate loaded niosomal gel for the treatment of psoriasis: Ex vivo and efficacy study. J Dispers Sci Technol. 2022:1-11. . ;:. Google Scholar
  155. Varshosaz J, Pardakhty A, Baharanchi SM. Sorbitan monopalmitate-based proniosomes for transdermal delivery of chlorpheniramine maleate. Drug Deliv. 2005;12(2):75-82. . ;:. Google Scholar
  156. Afreen U, Fahelelbom KM, Shah SNH, Ashames A, Almas U, Khan SA, et al. Formulation and evaluation of niosomes-based chlorpheniramine gel for the treatment of mild to moderate skin allergy. J Exp Nanosci. 2022;17(1):467-495. . ;:. Google Scholar
  157. Kische H, Arnold A, Gross S, Wallaschofski H, Völzke H, Nauck M, et al. Sex Hormones and Hair Loss in Men From the General Population of Northeastern Germany. JAMA Dermatol. 2017;153(9):935-937. . ;:. Google Scholar
  158. Morakul B, Teeranachaideekul V, Buraphacheep Junyaprasert V. Niosomal delivery of pumpkin seed oil: development, characterization, and physical stability. J Microencapsul. 2019;36(2):120-129. . ;:. Google Scholar
  159. Teeranachaideekul V, Parichatikanond W, Junyaprasert VB, Morakul B. Pumpkin Seed Oil-Loaded Niosomes for Topical Application: 5α-Reductase Inhibitory, Anti-Inflammatory, and In Vivo Anti-Hair Loss Effects. Pharmaceuticals. 2022;15(8):930. . ;:. Google Scholar
  160. Javadzadeh Y, Shokri J, Hallaj-Nezhadi S, Hamishehkar H, Nokhodchi A. Enhancement of percutaneous absorption of finasteride by cosolvents, cosurfactant and surfactants. Pharm Dev Technol. 2010;15(6):619-625. . ;:. Google Scholar
  161. Rungseevijitprapa W, Wichayapreechar P, Sivamaruthi BS, Jinarat D, Chaiyasut C. Optimization and Transfollicular Delivery of Finasteride-Loaded Proniosomes for Hair Growth Stimulation in C57BL/6Mlac Mice. Pharmaceutics. 2021;13(12):2177. . ;:. Google Scholar
  162. Mali N, Darandale S, Vavia P. Niosomes as a vesicular carrier for topical administration of minoxidil: formulation and in vitro assessment. Drug Deliv Transl Res. 2013;3(6):587-592. . ;:. Google Scholar
  163. Tampucci S, Paganini V, Burgalassi S, Chetoni P, Monti D. Nanostructured Drug Delivery Systems for Targeting 5-α-Reductase Inhibitors to the Hair Follicle. Pharmaceutics. 2022;14(2):286. . ;:. Google Scholar
  164. Choi MJ, Maibach HI. Liposomes and niosomes as topical drug delivery systems. Skin Pharmacol Physiol. 2005;18(5):209-219. . ;:. Google Scholar
  165. Goyal G, Garg T, Malik B, Chauhan G, Rath G, Goyal AK. Development and characterization of niosomal gel for topical delivery of benzoyl peroxide. Drug Deliv. 2015;22(8):1027-1042. . ;:. Google Scholar
  166. Manosroi A, Khanrin P, Lohcharoenkal W, Werner RG, Gotz F, Manosroi W, et al. Transdermal absorption enhancement through rat skin of gallidermin loaded in niosomes. Int J Pharm. 2010;392(1-2):304-310. . ;:. Google Scholar
  167. Budhiraja A, Dhingra G. Development and characterization of a novel antiacne niosomal gel of rosmarinic acid. Drug Deliv Transl Res. 2015;22(6):723-730. . ;:. Google Scholar
  168. Kim J, Kim J, Lee YI, Suk J, Lee D, Lee JH. A pilot study evaluating the efficacy and safety of retinaldehyde-loaded niosomes against mild-to-moderate acne. J Cosmet Dermatol. 2021;20(11):3586-3592. . ;:. Google Scholar
  169. Csongradi C, du Plessis J, Aucamp ME, Gerber M. Topical delivery of roxithromycin solid-state forms entrapped in vesicles. Eur J Pharm Biopharm. 2017;114:96-107. . ;:. Google Scholar
  170. Alomrani AH, Al-Agamy MH, Badran MM. In vitro skin penetration and antimycotic activity of itraconazole loaded niosomes: Various nonionic surfactants. J Drug Deliv Sci Technol. 2015;28:37-45. . ;:. Google Scholar
  171. Muzzalupo R, Perez L, Pinazo A, Tavano L. Pharmaceutical versatility of cationic niosomes derived from amino acid-based surfactants: Skin penetration behavior and controlled drug release. Int J Pharm. 2017;529(1-2):245-252. . ;:. Google Scholar
  172. Pando D, Matos M, Gutierrez G, Pazos C. Formulation of resveratrol entrapped niosomes for topical use. Colloids Surf B. 2015;128:398-404. . ;:. Google Scholar
  173. Abdelbary AA, AbouGhaly MH. Design and optimization of topical methotrexate loaded niosomes for enhanced management of psoriasis: application of Box‒Behnken design, in vitro evaluation and in vivo skin deposition study. Int J Pharm. 2015;485(1-2):235-243. . ;:. Google Scholar
  174. Krishnaswami V, Kandasamy R, Alagarsamy S, Palanisamy R, Natesan S. Biological macromolecules for ophthalmic drug delivery to treat ocular diseases. Int J Biol Macromol. 2018;110:7-16. . ;:. Google Scholar
  175. Bertens CJ, Gijs M, van den Biggelaar FJ, Nuijts RM. Topical drug delivery devices: A review. Exp Eye Res. 2018;168:149-160. . ;:. Google Scholar
  176. Farkouh A, Frigo P, Czejka M. Systemic side effects of eye drops: a pharmacokinetic perspective. Clin Ophthalmol. 2016;10:2433. . ;:. Google Scholar
  177. Willoughby CE, Ponzin D, Ferrari S, Lobo A, Landau K, Omidi Y. Anatomy and physiology of the human eye: effects of mucopolysaccharidoses disease on structure and function–a review. Clin Experiment Ophthalmol. 2010;38:2-11. . ;:. Google Scholar
  178. Zhai Z, Cheng Y, Hong J. Nanomedicines for the treatment of glaucoma: current status and future perspectives. Acta Biomater. 2021;125:41-56. . ;:. Google Scholar
  179. Hasan AA. Design and in vitro characterization of small unilamellar niosomes as ophthalmic carrier of dorzolamide hydrochloride. Pharm Dev Technol. 2014;19(6):748-754. . ;:. Google Scholar
  180. Fathalla D, Fouad EA, Soliman GM. Latanoprost niosomes as a sustained release ocular delivery system for the management of glaucoma. Drug Dev Ind Pharm. 2020;46(5):806-813. . ;:. Google Scholar
  181. Abdelmonem R, Elhabal SF, Abdelmalak NS, El-Nabarawi MA, Teaima MH. Formulation and Characterization of Acetazolamide/Carvedilol Niosomal Gel for Glaucoma Treatment: In Vitro, and In Vivo Study. Pharmaceutics. 2021;13(2):221. . ;:. Google Scholar
  182. Emad Eldeeb A, Salah S, Ghorab M. Proniosomal gel-derived niosomes: an approach to sustain and improve the ocular delivery of brimonidine tartrate; formulation, in vitro characterization, and in vivo pharmacodynamic study. Drug Deliv. 2019;26(1):509-521. . ;:. Google Scholar
  183. Fouda NH, Abdelrehim RT, Hegazy DA, Habib BA. Sustained ocular delivery of Dorzolamide-HCl via proniosomal gel formulation: in vitro characterization, statistical optimization, and in vivo pharmacodynamic evaluation in rabbits. Drug Deliv. 2018;25(1):1340-1349. . ;:. Google Scholar
  184. Bourcier T, Sauer A, Dory A, Denis J, Sabou M. Fungal keratitis. J Fr Ophtalmol. 2017;40(9):e307-e313. . ;:. Google Scholar
  185. El-Emam GA, Girgis GN, El-Sokkary MMA, Soliman OAE-A, Abd El AEGH. Ocular Inserts of Voriconazole-Loaded Proniosomal Gels: Formulation, Evaluation and Microbiological Studies. Int J Nanomedicine. 2020;15:7825. . ;:. Google Scholar
  186. Soliman OAE-A, Mohamed EA, Khatera NAA. Enhanced ocular bioavailability of fluconazole from niosomal gels and microemulsions: Formulation, optimization, and in vitro–in vivo evaluation. Pharm Dev Technol. 2019;24(1):48-62. . ;:. Google Scholar
  187. Elmotasem H, Awad GE. A stepwise optimization strategy to formulate in situ gelling formulations comprising fluconazole-hydroxypropyl-beta-cyclodextrin complex loaded niosomal vesicles and Eudragit nanoparticles for enhanced antifungal activity and prolonged ocular delivery. Asian J Pharm. 2020;15(5):617-636. . ;:. Google Scholar
  188. Abdelbary GA, Amin MM, Zakaria MY. Ocular ketoconazole-loaded proniosomal gels: formulation, ex vivo corneal permeation and in vivo studies. Drug Deliv. 2017;24(1):309-319. . ;:. Google Scholar
  189. Palioura S, Henry CR, Amescua G, Alfonso EC. Role of steroids in the treatment of bacterial keratitis. Clin Ophthalmol. 2016;10:179-186. . ;:. Google Scholar
  190. Gaafar PM, Abdallah OY, Farid RM, Abdelkader H. Preparation, characterization and evaluation of novel elastic nanosized niosomes (ethoniosomes) for ocular delivery of prednisolone. J Liposome Res. 2014;24(3):204-215. . ;:. Google Scholar
  191. Abdelbary A, Salem HF, Khallaf RA, Ali AM. Mucoadhesive niosomal in situ gel for ocular tissue targeting: in vitro and in vivo evaluation of lomefloxacin hydrochloride. Pharm Dev Technol. 2017;22(3):409-417. . ;:. Google Scholar
  192. Khalil RM, Abdelbary GA, Basha M, Awad GE, El-Hashemy HA. Enhancement of lomefloxacin Hcl ocular efficacy via niosomal encapsulation: in vitro characterization and in vivo evaluation. J Liposome Res. 2017;27(4):312-323. . ;:. Google Scholar
  193. Khalil RM, Abdelbary GA, Basha M, Awad GE, El-Hashemy HA. Design and evaluation of proniosomes as a carrier for ocular delivery of lomefloxacin HCl. J Liposome Res. 2017;27(2):118-129. . ;:. Google Scholar
  194. Allam A, El-Mokhtar MA, Elsabahy M. Vancomycin-loaded niosomes integrated within pH-sensitive in situ forming gel for treatment of ocular infections while minimizing drug irritation. J Pharm Pharmacol. 2019;71(8):1209-1221. . ;:. Google Scholar
  195. Gugleva V, Titeva S, Rangelov S, Momekova D. Design and in vitro evaluation of doxycycline hyclate niosomes as a potential ocular delivery system. Int J Pharm. 2019;567:118431. . ;:. Google Scholar
  196. Gugleva V, Titeva S, Ermenlieva N, Tsibranska S, Tcholakova S, Rangelov S, et al. Development and evaluation of doxycycline niosomal thermoresponsive in situ gel for ophthalmic delivery. Int J Pharm. 2020;591:120010. . ;:. Google Scholar
  197. Alruwaili NK, Imam SS, Alotaibi NH, Alhakamy NA, Alharbi KS, Alshehri S, et al. Formulation of chitosan polymeric vesicles of ciprofloxacin for ocular delivery: box-behnken optimization, in vitro characterization, HET-CAM irritation, and antimicrobial assessment. AAPS PharmSciTech. 2020;21:1-16. . ;:. Google Scholar
  198. El-Sayed MM, Hussein AK, Sarhan HA, Mansour HF. Flurbiprofen-loaded niosomes-in-gel system improves the ocular bioavailability of flurbiprofen in the aqueous humor. Drug Dev Ind Pharm. 2017;43(6):902-910. . ;:. Google Scholar
  199. Aboali FA, Habib DA, Elbedaiwy HM, Farid RM. Curcumin-loaded proniosomal gel as a biofreindly alternative for treatment of ocular inflammation: In vitro and in vivo assessment. Int J Pharm. 2020;589:119835. . ;:. Google Scholar
  200. Puras G, MartiĚnez-Navarrete G, Mashal M, Zarate J, Agirre M, Ojeda E, et al. Protamine/DNA/niosome ternary nonviral vectors for gene delivery to the retina: the role of protamine. Mol Pharm. 2015;12(10):3658-3671. . ;:. Google Scholar
  201. Eid HM, Naguib IA, Alsantali RI, Alsalahat I, Hegazy AM. Novel chitosan-coated niosomal formulation for improved management of bacterial conjunctivitis: A highly permeable and efficient ocular nanocarrier for azithromycin. J Pharm Sci. 2021;110(8):3027-3036. . ;:. Google Scholar
  202. Li Q, Li Z, Zeng W, Ge S, Lu H, Wu C, et al. Proniosome-derived niosomes for tacrolimus topical ocular delivery: in vitro cornea permeation, ocular irritation, and in vivo anti-allograft rejection. Eur J Pharm Sci. 2014;62:115-123. . ;:. Google Scholar
  203. Ojeda E, Puras G, Agirre M, Zarate J, Grijalvo S, Eritja R, et al. The influence of the polar head-group of synthetic cationic lipids on the transfection efficiency mediated by niosomes in rat retina and brain. Biomaterials. 2016;77:267-279. . ;:. Google Scholar
  204. Ojeda E, Puras G, Agirre M, Zárate J, Grijalvo S, Pons R, et al. Niosomes based on synthetic cationic lipids for gene delivery: the influence of polar head-groups on the transfection efficiency in HEK-293, ARPE-19 and MSC-D1 cells. Org Biomol Chem. 2015;13(4):1068-1081. . ;:. Google Scholar
  205. Javadzadeh Y, Yaqoubi S. Chapter 20 - Therapeutic nanostructures for pulmonary drug delivery. In: Andronescu E, Grumezescu AM, editors. Nanostructures for Drug Delivery. Amsterdam: Elsevier; 2017. p. 619-638. . ;:. Google Scholar
  206. Kleinstreuer C, Feng Y, Childress E. Drug-targeting methodologies with applications: A review. World J Clin Cases. 2014;2(12):742. . ;:. Google Scholar
  207. Ruge CA, Kirch J, Lehr C-M. Pulmonary drug delivery: from generating aerosols to overcoming biological barriers—therapeutic possibilities and technological challenges. Lancet Respir Med. 2013;1(5):402-413. . ;:. Google Scholar
  208. Kunda NK, Somavarapu S, Gordon SB, Hutcheon GA, Saleem IY. Nanocarriers targeting dendritic cells for pulmonary vaccine delivery. Pharm Res. 2013;30(2):325-341. . ;:. Google Scholar
  209. Shoyele SA, Cawthorne S. Particle engineering techniques for inhaled biopharmaceuticals. Adv Drug Deliv Rev. 2006;58(9-10):1009-1029. . ;:. Google Scholar
  210. Mohamad Saimi NI, Salim N, Ahmad N, Abdulmalek E, Abdul Rahman MB. Aerosolized Niosome Formulation Containing Gemcitabine and Cisplatin for Lung Cancer Treatment: Optimization, Characterization and In Vitro Evaluation. Pharmaceutics. 2021;13(1):59. . ;:. Google Scholar
  211. OFEV. OFEV® (nintedanib) for IPF, SSc-ILD & chronic fibrosing ILDs with a progressive phenotype.: OFEV; 2022. . ;:. Google Scholar
  212. K Shukla S, Nguyen V, Goyal M, Gupta V. Cationically modified inhalable nintedanib niosomes: enhancing therapeutic activity against non-small cell lung cancer. Nanomedicine. 2022;17(13):935-958. . ;:. Google Scholar
  213. Xu X, Zhu Y. Curcumin inhibits human non-small cell lung cancer xenografts by targeting STAT3 pathway. Am J Transl Res. 2017;9(8):3633–3641. . ;:. Google Scholar
  214. Deng X, Chen C, Wu F, Qiu L, Ke Q, Sun R, et al. Curcumin inhibits the migration and invasion of non-small cell lung cancer cells through radiation-induced suppression of epithelial–mesenchymal transition and soluble E-cadherin expression. Technol Cancer Res Treat. 2020;19:1533033820947485. . ;:. Google Scholar
  215. Samadizadeh S, Arabi MS, Yasaghi M, Salimi V, Tabarraei A, Moradi A, et al. Anti-inflammatory effects of curcumin-loaded niosomes on respiratory syncytial virus infection in a mice model. J Med Microbiol. 2022;71(4). . ;:. Google Scholar
  216. Kwatra S, Taneja G, Nasa N. Alternative routes of drug administration-transdermal, pulmonary & parenteral. Indo Glob Res J Pharm Sci. 2012;2(4):409-426. . ;:. Google Scholar
  217. Marianecci C, Paolino D, Celia C, Fresta M, Carafa M, Alhaique F. Nonionic surfactant vesicles in pulmonary glucocorticoid delivery: characterization and interaction with human lung fibroblasts. J Control Release. 2010;147(1):127-135. . ;:. Google Scholar
  218. Henning A, Hein S, Schneider M, Bur M, Lehr CM. Pulmonary drug delivery: medicines for inhalation. Handbook of experimental pharmacology. 2010(197):171-192. . ;:. Google Scholar
  219. Zhuang X, Qi Y, Wang M, Yu N, Nan F, Zhang H, et al. mRNA vaccines encoding the HA protein of influenza A H1N1 virus delivered by cationic lipid nanoparticles induce protective immune responses in mice. Vaccines. 2020;8(1):123. . ;:. Google Scholar
  220. Böttger R, Pauli G, Chao P-H, Fayez NA, Hohenwarter L, Li S-D. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev. 2020;154:79-101. . ;:. Google Scholar
  221. Sharma V, Anandhakumar S, Sasidharan M. Self-degrading niosomes for encapsulation of hydrophilic and hydrophobic drugs: an efficient carrier for cancer multidrug delivery. Mater Sci Eng C. 2015;56:393-400. . ;:. Google Scholar
  222. Temprom L, Krongsuk S, Thapphasaraphong S, Priperm A, Namuangruk S. A novel preparation and characterization of melatonin loaded niosomes based on using a ball milling method. Mater Today, Commun. 2022;31:103340. . ;:. Google Scholar
  223. Yaghoobian M, Haeri A, Bolourchian N, Shahhosseni S, Dadashzadeh S. The impact of surfactant composition and surface charge of niosomes on the oral absorption of repaglinide as a BCS II model drug. Int J Nanomedicine. 2020;2020:8767-8781. . ;:. Google Scholar
  224. Barani M, Reza Hajinezhad M, Sargazi S, Zeeshan M, Rahdar A, Pandey S, et al. Simulation, in vitro, and in vivo cytotoxicity assessments of methotrexate-loaded pH-responsive nanocarriers. Polymers. 2021;13(18):3153. . ;:. Google Scholar
  225. Barani M, Hajinezhad MR, Zargari F, Shahraki S, Davodabadi F, Mirinejad S, et al. Preparation, characterization, cytotoxicity and pharmacokinetics of niosomes containing gemcitabine: In vitro, in vivo, and simulation studies. J Drug Deliv Sci Technol. 2023;84:104505. . ;:. Google Scholar
  226. De A, Venkatesh N, Senthil M, Sanapalli BKR, Shanmugham R, Karri VVSR. Smart niosomes of temozolomide for enhancement of brain targeting. Nanobiomedicine. 2018;5:1849543518805355. . ;:. Google Scholar
  227. Liu F-r, Jin H, Wang Y, Chen C, Li M, Mao S-j, et al. Anti-CD123 antibody-modified niosomes for targeted delivery of daunorubicin against acute myeloid leukemia. Drug Deliv. 2017;24(1):882-890. . ;:. Google Scholar
  228. Muzzalupo R, Mazzotta E. Do niosomes have a place in the field of drug delivery? Expert Opin Drug Deliv. 2019;16(11):1145-1147. . ;:. Google Scholar
  229. Dhapte-Pawar V, Kadam S, Saptarsi S, Kenjale PP. Nanocosmeceuticals: Facets and aspects. Future Sci OA. 2020;6(10):FSO613. . ;:. Google Scholar
  230. Aparajay P, Dev A. Functionalized niosomes as a smart delivery device in cancer and fungal infection. Eur J Pharm Sci. 2022;168:106052. . ;:. Google Scholar
  231. Alemi A, Zavar Reza J, Haghiralsadat F, Zarei Jaliani H, Haghi Karamallah M, Hosseini SA, et al. Paclitaxel and curcumin coadministration in novel cationic PEGylated niosomal formulations exhibit enhanced synergistic antitumor efficacy. J Nanobiotechnology. 2018;16:1-20. . ;:. Google Scholar
  232. Agarwal S, Mohamed MS, Raveendran S, Rochani AK, Maekawa T, Kumar DS. Formulation, characterization and evaluation of morusin loaded niosomes for potentiation of anticancer therapy. RSC Adv. 2018;8(57):32621-32636. . ;:. Google Scholar


Author's Affiliation
Article Details

Issue: Vol 27 No 3 (2024)
Page No.: 3507-3534
Published: Sep 30, 2024
Section: Section: HEALTH SCIENCES
DOI: https://doi.org/10.32508/stdj.v27i3.4420

 Copyright Info

Creative Commons License

Copyright: The Authors. This is an open access article distributed under the terms of the Creative Commons Attribution License CC-BY 4.0., which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

 How to Cite
Nguyen, M. H., Le, T. H. N., Nguyen, T. P. T., Le, T. N. T., Nguyen, T. Y. N., Nguyen, K. A., Pham, T. T., & Le, M. T. (2024). Niosomes: recent advances and applications in targeted drug delivery. Science and Technology Development Journal, 27(3), 3507-3534. https://doi.org/https://doi.org/10.32508/stdj.v27i3.4420

 Cited by



Article level Metrics by Paperbuzz/Impactstory
Article level Metrics by Altmetrics

 Article Statistics
HTML = 0 times
PDF   = 0 times
XML   = 0 times
Total   = 0 times