Maryam
Roostaee
a,
Atefeh
Derakhshani
b,
Hadiseh
Mirhosseini
c,
Elmira
Banaee Mofakham
d,
Sonia
Fathi-Karkan
ef,
Shekoufeh
Mirinejad
g,
Saman
Sargazi
*gh and
Mahmood
Barani
*i
aDepartment of Chemistry, Faculty of Sciences, Vali-e-Asr University of Rafsanjan, Rafsanjan, Iran. E-mail: maryamroostaee3@gmail.com
bDepartment of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran. E-mail: atefe.drkhshani93@gmail.com
cDepartment of Chemistry, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran. E-mail: mirhoseinihadis@yahoo.com
dDepartment of Nanotechnology and Advanced Materials Research, Materials & Energy Research Center, Karaj, Iran. E-mail: e.banaee@merc.ac.ir
eNatural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran. E-mail: Soniafathi92@gmail.com
fDepartment of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd 9414974877, Iran. E-mail: Soniafathi92@gmail.com
gCellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran. E-mail: sgz.biomed@gmail.com; shokufemn@gmail.com; Tel: +989103161030
hDepartment of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
iMedical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 7616913555, Iran. E-mail: mahmoodbarani7@gmail.com
First published on 7th December 2023
Nanoniosome-based drug codelivery systems have become popular therapeutic instruments, demonstrating tremendous promise in cancer therapy, infection treatment, and other therapeutic domains. An emerging form of vesicular nanocarriers, niosomes are self-assembling vesicles composed of nonionic surfactants, along with cholesterol or other amphiphilic molecules. This comprehensive review focuses on how nanosystems may aid in making anticancer and antibacterial pharmaceuticals more stable and soluble. As malleable nanodelivery instruments, the composition, types, preparation procedures, and variables affecting the structure and stability of niosomes are extensively investigated. In addition, the advantages of dual niosomes for combination therapy and the administration of multiple medications simultaneously are highlighted. Along with categorizing niosomal drug delivery systems, a comprehensive analysis of various preparation techniques, including thin-layer injection, ether injection, and microfluidization, is provided. Dual niosomes for cancer treatment are discussed in detail regarding the codelivery of two medications and the codelivery of a drug with organic, plant-based bioactive compounds or gene agents. In addition, niogelosomes and metallic niosomal carriers for targeted distribution are discussed. The review also investigates the simultaneous delivery of bioactive substances and gene agents, including siRNA, microRNA, shRNA, lncRNA, and DNA. Additional sections discuss the use of dual niosomes for cutaneous drug delivery and treating leishmanial infections, Pseudomonas aeruginosa, and Mycobacterium tuberculosis. The study concludes by delineating the challenges and potential routes for nanoniosome-based pharmaceutical codelivery systems, which will be useful for nanomedicine practitioners and researchers.
An emerging form of vesicular nanocarriers, niosomes, are self-assembling vesicles composed of nonionic surfactants, along with cholesterol or other amphiphilic molecules.20,21 These vesicles have a bilayer hydrophobic membrane and a central cavity containing an aqueous phase, allowing them to encase both hydrophilic and hydrophobic substances.22–24 Niosomes are typically formulated with nonionic surfactants, cholesterol, a charge inducer, and a hydration medium, and are primarily composed of amphiphilic nonionic surfactants with a polar head and nonpolar tail.25 Nonionic surfactants are more stable, compatible, and less toxic than anionic, cationic, and amphoteric surfactants due to their lack of charge.26 By inhibiting p-glycoprotein,27 these agents improve the absorption and delivery of numerous anticancer medications, such as doxorubicin (DOX),28 curcumin (CUR),29 morusin,30 hydrocortisone steroids,31 HIV-protease inhibitors such as ritonavir,32 as well as beta-blockers.33
Niosome rigidity can be increased by the incorporation of cholesterol into the bilayer structure to decrease membrane permeability, improve membrane stability, and decrease membrane permeability.34 Electrostatic repulsion inhibits coalescence via charge inducers in niosome preparations, contributing to the stability of the niosomes.35 When niosomes are constructed, dicetyl phosphate and phosphatidic acid are among the most commonly used negatively charged inducers, while stearyl amine and stearylpyridinium chloride are frequently used positively charged inducers.36 Usually, phosphate buffered saline (PBS) is used for niosome hydration, and their pH changes depending on the solubility of the drug.23
Niosomes are divided into three classes with corresponding references based on the size of their compartments.37 It is possible to categorize niosomes into three types according to the size of their vesicles, such as multilamellar vesicles bigger than 0.05 mm, large unilamellar vesicles bigger than 0.10 mm, and small unilamellar vesicles bigger than 0.025 mm.38 The production of niosomes involves a variety of techniques, such as the microfluidization technique, reverse phase evaporation technique, ether injection technique, trans-membrane pH gradient (inside acidic) technique, bubble method, supercritical carbon dioxide fluid method, and ball milling technique.27,39 Niosomes have different physicochemical properties influenced by different variables.40 The additives and surfactants used in the niosome preparation affect their stability and permeability.41 By altering how surfactants are assembled into vesicles, the hydration temperature, volume, and duration of niosome hydration can affect their morphologies.42 A slow release of the medication is caused by difficulty eluting fluid from vesicles, and a rapid release is caused by the mechanical disintegration of vesicles under osmotic stress.27,37,43
Nanoniosomes, liposomes, and micelles share a common ground in their role as carriers for drug delivery and therapeutic applications, yet each possesses distinct characteristics. Nanoniosomes exhibit enhanced stability and improved drug encapsulation efficiency compared with conventional liposomes.44 They harness the advantages of both liposomes and micelles, combining the structural integrity of liposomes with the amphiphilic properties of micelles. While liposomes consist of phospholipid bilayers and micelles form single-layered structures, nanoniosomes strike a balance between these, offering a versatile platform for targeted drug delivery, promising both stability and increased bioavailability.45 Their innovative nature propels advancements in pharmaceuticals, offering a potential breakthrough in precision medicine and therapeutics.46
Numerous advantages of niosomes, such as their capacity to transport multiple drugs at once and reduced systemic toxicity of the drugs in cancer treatments, have prompted researchers to use these nano-carriers for the codelivery of drugs.47,48 Combination therapy can lead to stronger synergistic effects and greater efficacy.49 It reduces the risk of severe adverse effects, so it would be more effective than single-agent therapy in inhibiting cancer growth.50 Niosomes enable the combination of anticancer therapeutics, including chemotherapy, genes, herbal remedies, photodynamic sensitizers, inhibitors of small molecules, classic antineoplastics, and small interfering RNAs (siRNA) in order to block tumor resistance to multiple drug cytotoxicity (MDR) and effectively regulate multiple signaling pathways.51–57 Using anticancer agents with various mechanisms of action concurrently enhances the therapeutic effects by encompassing more than one mechanism of action, thereby increasing the efficacy of the treatment.35
Developing innovative, personalized pharmaceutical products based on novel niosomes will require further research and investigation into formulation, preparation, and modification methods. A concise description of the composition, varieties, and preparation techniques of niosomes is presented in this article, along with examples of how niosomes can be used for targeted delivery, and the simultaneous delivery of several substances. Examples include the codelivery of drug and gene agents, two gene agents, bioactive compounds, and genes, antioxidants, drug niogelosomes, and other therapeutic applications, including in infections, cutaneous drug delivery, and others.
Non-ionic surfactants | Surfactant examples |
---|---|
a Sorbitan fatty acid esters. b Polyoxyethylene sorbitan fatty acid esters. | |
Alkyl esters: | |
(i) Spansa | Span 20, Span 40, Span 60, Span 65, Span 80, Span 85 |
(ii) Tweensb | Tween 20, Tween 60, Tween 60, Tween 40, Tween 80, Tween 85 |
Alkyl ethers: | |
(i) Alkyl glycerol ethers | Hexadecyl diglycerol ether |
(ii) Polyoxyethylene glycol alkyl ethers (Brij) | Brij30, Brij52, Brij72, Brij76, Brij78 |
Crown ethers | Bola |
Alkyl amides: | |
(i) Glycosides | C-glycoside derivative surfactant |
(ii) Alkyl polyglucoside | Octyl-decyl polyglucoside |
Fatty alcohols or fatty acids | |
(i) Fatty alcohols | Stearyl alcohol, acetyl alcohol, myristyl alcohol |
(ii) Fatty acids | Stearic acid, palmitic acid, myristic acid |
Block copolymer: (i) Pluronic | Pluronic L64, Pluronic 105 |
Lipidic components: cholesterol | |
l-α-Soya phosphatidyl choline | |
Charged molecule: | |
Negative charge | Dicetyl phosphate, phosphatidic acid, lipoamino acid, dihexadecyl phosphate |
Positive charge | Stearyl amine, stearyl pyridinium chloride |
The production of bilayer vesicles is affected by the critical packing parameter (CPP), the hydrophilic–lipophilic balance (HLB) scale, the constituents’ chemical composition, and the gel liquid transmission temperature. Longer alkyl chains increase the entrapment efficiency of non-ionic surfactants. The Tween series of surfactants containing cholesterol and an extended alkyl chain with an enlarged hydrophilic moiety has the highest entrapment efficiency for water-soluble medications. The HLB value of a surfactant is essential for controlling drug entrapment in the vesicle it creates. The CPP value of a surfactant can be calculated using the length and volume of the non-polar component and the head area of the polar component. Fig. 1 illustrates how CPP parameters can be used to predict the type of vesicle that will form, and eqn (1) can be used to calculate the CPP. It is anticipated that the surfactant property will play a significant role in the in vivo performance of these drug delivery methods, as it is a crucial component that affects the physicochemical properties of niosomes, such as particle size, two-layer strength, drug release, and consistency.62
![]() | (1) |
![]() | ||
Fig. 1 Schematic representation of an amphiphile: a0, hydrophilic head group area: v, hydrophobic chain volume; and lc, hydrophobic chain length. Reproduced from ref. 63 with permission from [MDPI], copyright [2022]. |
![]() | ||
Fig. 2 Types of niosomes based on their size or number of lamellar layers. Reproduced from ref. 27 with permission from [Society of Pharmaceutical Tecnocrats], copyright [2021]. |
Additional niosome varieties include bola surfactant-niosomes, proteasomes, aspasomes, niogelosomes, and vesicle-in-water-and-oil systems (v/w/o). To develop novel niosomal formulations, new surfactants have been created. Nonionic surfactants known as bola-form amphiphiles consist of two identical aza-crown ether groups that function as polar centers and are connected by a prolonged alkyl chain. When they are in close proximity to cholesterol, they can form colloidal systems.58 Proniosomes are produced through straightforward formulation processes that can be avoided by retaining the composition and known properties of niosomes. They are surfactant-coated carrier granules that can be rehydrated in aqueous dietary materials just before use to produce niosomes. Proniosomal granules have several advantages over their liquid counterparts, including a reduced risk of bacterial contamination and easier administration and transport.72 Aspasomes are novel nanovesicles composed of one or more ascorbyl palmitate (AP) layers. These bilayer vesicles are advantageous for transdermal drug delivery because they improve percutaneous absorption and stability. Niogelosomes are double-delivery systems (consisting of niosomes and gel) that differ from conventional niosomes in terms of their release characteristics and pharmacokinetics. Researchers are creating niosomal gel for a variety of applications.35 A vesicle-in-water-in-oil (v/w/o) combination describes the aqueous suspension of niosomes emulsified in another oil component. It is possible to alter the characteristics of the surfactant used to produce the vesicles, the surfactant or combination of surfactants used to immobilize the emulsion, and the composition of the oil component to produce systems with variable limits for drugs or antigens and variable release properties.73
Utilizing the transmembrane pH gradient method, Kalaiselvi et al. synthesized clove bud oil (CBO)-loaded niosomes with cholesterol and Span 20, 60, and 80 surfactant types. Niosome aggregation was stopped by the addition of dicetyl phosphate. SEM, FT-IR, and zeta potential examinations were used to analyze the produced niosomes’ appearance, functional characteristics, size, and charge. The zeta potential was used to look at the durability of CBO-loaded niosomes and the mean dimension of the particles. The generated CBO-loaded niosomes had a median particle size of 417 nm, and the manufactured niosomes showed excellent stability, according to the data. SEM analysis was used to assess the dimensions and form of the created CBO-loaded niosomes. The results demonstrated the presence of CBO-loaded niosomes in their distinct spherical form. Particle aggregation was absent from the flat surfaces of the CBO-loaded niosomes.82 Furthermore, Jeno et al. produced niosomes encased in Indian plant-based oils by using the transmembrane gradient of pH strategy. Utilizing SEM, FT-IR, zeta-sizer, and zeta-potential, the obtained niosomes were examined and described. Eucalyptus, neem, and rosemary oils were discovered to contain niosomes with average sizes of 869.64 nm, 693.25 nm, and 912.36 nm, respectively. This suggests more stability. Neem oil was used to create the niosomes, and SEM examination of those niosomes showed similar-sized NPs with a circular structure as well.83
Composition of niosome | En-capsulated agent | Preparation method | Niosome size (nm) | Key feature | Ref. |
---|---|---|---|---|---|
TFH; thin-film hydration, EIM: ethanol injection method, BM: ball milling, TPT: topotecan, ACM: acemetacin, ALS: alendronate sodium, MB: methylene blue, EE: encapsulation efficiency. | |||||
Span 60, cholesterol, Cremophor® ELP or Lauroglycol® 90 | Methylene blue | TFH | 292.4 | Production of stable niosomes with optimal lipid content for optimal EE | 93 |
Span 60, TWEEN 60, chloroform, dihexadecyl phosphate chloride, and Carbopol 934 | Fusidic acid micropowder | TFH | 377.2–725.4 | The produced fusidic acid-laden niosomes improved the penetration of fusidic acid via the skin | 75 |
Span 60, Tween 60, Tween 80, cholesterol and diethyl ether | Alendronate sodium | EIM | 99.6–464.3 | Successfully prepared ALS niosomes for transdermal delivery to offer the prolonged release of ALS and reduce GI negative impacts, and create a replacement production for oral ALS administration | 77 |
Cholesterol, Span 60, diethyl ether | Acemetacin | EIM | 315.23 | Examination of the utilization of 131I-ACM niosomes as a potentially effective dual anticancer therapy to combine the chemotherapeutic impacts of ACM and the radiotherapeutic impacts of 131I in a single treatment cycle | 78 |
Span 60, cholesterol, chloroform and DSPE-PEG(2000) maleimide | Topotecan | Microfluidization | 128.5 | Improving anti-glioma treatment with tLyp-1-functionalized TPT-loaded niosomes | 80 |
Span 20, 60, and 80, cholesterol, chloroform | Clove bud oil | Transmembrane pH gradient | 417 | The larvicidal higher performance of the tested mosquito species with the clove bud oil-loaded niosomes and employing these niosomes as possible mosquito repellents | 82 |
Span 60, chloroform, dicetyl phosphate (DCP), and cholesterol | Indian herb oils (neem, eucalyptus, and rosemary) | Transmembrane pH gradient | 912.36, 693.25, and 869.64 | Niosomes that include neem oil and those made from Indian plant oils may be used instead of commercial mosquito larvicides and have higher rates of larval death | 83 |
Span 80, theophylline powder, Tween 80, cholesterol | Theophylline | Supercritical carbon dioxide fluid | 160 and 171 | Getting longer theophylline release time of about 5-fold | 85 |
Span 80, cholesterol, stearylamine | Ovalbumin | Reverse phase evaporation | ∼300 nm | Increasing the niosomal hydrogel platform's antigen delivery in comparison with niosomal elastic gel and taking into account niosomal hydrogel as an excellent transcutaneous antigen delivery carrier | 87 |
Span 60, cholesterol ≥92.5%, melatonin ≥98%, and phosphate-buffered saline (PBS) | Melatonin | BM | 533–266 | Exploration of the BM approach as a potentially effective way to create niosomes for the encapsulation of poorly soluble medicines with better drug release profiles | 89 |
Compounds | Niosome fabrication method | Surface functionalities | In vitro/in vivo models | IC50 value (μg mL−1) | Cancer type | Key observation | Ref. |
---|---|---|---|---|---|---|---|
DDS: drug delivery system, NLC: niosomes loaded with (letrozole + cyclophosphamide), NLCPFA: surface-functionalized with a folic-acid-targeting moiety, NCE: cisplatin and epirubicin-loaded niosome, FPNCE: FA-PEGylated niosomal cisplatin and epirubicin, N-Far + Gin: farnesol–gingerol-loaded niosome, NGC: niosome-loaded (cisplatin + gemcitabine). | |||||||
1. Tamoxifen | Box–Behnken design | NA | MCF-7 | NA | Breast | - Sustained release | 57 |
2. Doxorubicin | NA | - Robust drug-excipient compatibility | |||||
Free doxorubicin + free tamoxifen | Free doxorubicin + free tamoxifen (0.15 ± 0.3) | ||||||
DDS: doxorubicin + tamoxifen niosome | DDS (0.01 ± 0.3) | ||||||
1. Letrozole | Thin-layer hydration method | Folic acid | MDA-MB-231 | 48 h: Let (167.47), cyclo (184.91), Let + cyclo (85.76), NLC (47.84), NLCPFA (31.13) | Breast | ↑Caspase-3↑Caspase-9 | 102 |
2. Cyclophosphamide | 72 h: Let (99.48), cyclo (127.29), Let + cyclo (59.22), NLC (37.53), NLCPFA (23.18) | ||||||
DDS: (NLC) (NLCPFA) | SKBR3 | 48 h: Let (147.71), cyclo (109.62), Let + cyclo (67.85), NLC (36.67), NLCPFA (24.92)72 h: Let (75.77), cyclo (88.33), Let + cyclo (31.01), NLC (28.08), NLCPFA (20.94) | ↓Cyclin-D↓Cyclin-E↓MMP-2↓MMP-9 | ||||
1. Epirubicin2. Cisplatin | Thin-layer hydration method | Folic acid | 4T1 | 48 h: CIS (96.56), EPI (135.63), CIS + EPI (87.47), NCE (66.19), FPNCE (50.3) | Breast | ↑Bax↑Caspase3↑Caspase9↑Mfn1 | 103 |
Poly ethylene glycol | SKBR3 | 72 h: CIS (78.35), EPI (88.85), CIS + EPI (55.24), NCE (48.18), FPNCE (37.2) | ↓Bcl2↓Drp1↓MMP-2 | ||||
DDS: (FPNCE) | BALB/c mice | 48 h: CIS (135.20), EPI (188.16), CIS + EPI (98.43), NCE (79.96), FPNCE (64.08)72 h: CIS (91.35), EPI (155.21), CIS + EPI (76.63), NCE (57.24), FPNCE (44.82) | ↓MMP-9Reduction in: invasion, mitosis index, pleomorphism in BALB/c miceSevere hyperchromicity observation in BALB/c mice | ||||
1. Farnesol2. GingerolDDS: N-Far + Gin | Box–Behnken design | NA | MCF7 | 48 h: niosome (1263.45), Gin (986.9), Far (192.46), Gin + Far (131.99), N-Gin + Far (25.56) | Breast | ↑P21↑Bax↑CASP-3↑CASP-9 | 104 |
SKBR3 | 72 h: niosome (1166.31), Gin (583.65), Far (125.267), Gin + Far (95.27), N-Gin + Far (19.563)48 h: niosome (2230.06), Gin (986.495), Far (233.628), Gin + Far (165.09), N-Gin + Far (44.01)72 h: niosome (1097.27), Gin (958.434), Far (183.005), Gin + Far (134.227), N-Gin + Far (26.58) | ↓BCL2↓HER2↓CDK4↓CCND1↓CCNE- High antioxidant potency- Improved cellular uptake efficiency | |||||
1. Rifampicin2. Ceftriaxone | Ecological probe sonication method | NA | NA | NA | NA | - Improved drug release and entrapment | 105 |
- Faster in vitro drug release rates | |||||||
1. Cisplatin2. Gemcitabine | Heating method (optimized by D-optimal mixture design) | NA | MRC5 | 72 h: niosome (>500), CIS (<1.56), Gem (<1.56), Gem + Cis (<1.56), NGC (280) | Lung | - Improved drug release and entrapment | 106 |
A549 | 72 h: niosome (>500), CIS (66), Gem (<1.56), Gem + Cis (<1.56), NGC (46) |
In another study for the breast cancer therapy field, Sahrayi and co-workers have developed nanoscale niosome-based transporters as targeted delivery systems.102 The developed platform is composed of dual-loaded niosomes with letrozole (Let) and cyclophosphamide (Cyclo) (NLC), and folic acid (FA) functionalized on the surface (NLCPFA) and can activate apoptosis, reduce Bcl-2 expression, and increase Bax expression by targeting the PTEN/AKT/P53 signaling pathway.107 Niosome NPs contain an equal amount of both drugs, and formulation optimization features have been investigated using various surfactant-cholesterol molar proportions of 1:
1 and 2
:
1 and lipid-drug molar proportions of 10
:
1 and 20
:
1, that affected the EE.108 EE values of samples improved by 2–3% for each drug at the greater surfactant:cholesterol and lipid:drug molar proportions. It was suggested that the thicker bilayer could have greater encapsulation potential for hydrophobic drugs,109–111 and also the presence of FA may increase the EE. Additionally, the mean size of the modified NPs was lower than that of the NLC. According to the CI, both drugs have considerable synergistic effects against MDA-MB-231 and SKBR3, but the NLCPFA group had a higher cytotoxic effect on the cancer cells, which was probably caused by the interaction of FA-coated niosomes with folate receptors on breast cancer cells, leading to the active transportation of niosomes within cells.112,113 Based on IC50 values (Table 3), a synergistic effect was evident when both drugs were administered, which had a far more toxic effect than each single drug for both cell lines. Furthermore the cytotoxicity effect was enhanced by the addition of FA to the functionalized niosome, perhaps due to folate receptor-mediated endocytosis, which increased the cellular uptake of drugs. FA plays a vital role in cell growth, differentiation, repair, and host defence, as demonstrated in Fig. 4A. NLCPFA significantly enhanced total apoptosis in both cancerous cells, and cell cycle analysis in Fig. 4B and C showed a strong synergic effect and niosomal co-loaded delivery of both drugs causing a shift toward the sub-G1 phase in each cancerous cell. NLCPFA produced a greater change in the effectiveness of the drugs.102 This study showed that, along with the synergistic effect of dual-drug niosomes, coated FA can inhibit the activity of breast cancer cells by promoting the apoptosis of both MDA-MB-231 and SKBR3 cells.
![]() | ||
Fig. 4 (A) Analysis of the total apoptosis of cancer cell lines (MDA-MB-231 and SKBR3) after treatment by free drugs and various niosomal structures. NLCPFA significantly increases total apoptosis in cancerous cells treated in increments of 21.9% for MDA-MB-231 and 24.5% for SKBR3, as opposed to NLC formulations. The cell cycle analysis graphs of (B) MDA-MB-231 and (C) SKBR3 cancer cells after treatment with a variety of niosomal and free drug formulations. Reproduced from ref. 102 with permission from [MDPI], copyright [2021]. |
In another study on the effects of FA-coated niosomes and the codelivery of cisplatin (CIS) and epirubicin (EPI), two chemotherapeutic drugs for breast cancer treatment, Moammeri and his colleagues designed an FA-PEGylated niosome (FPNCE) to produce a functionalized DDS based on co-loaded niosomes to increase endocytosis and finally injected it into BALB/c mice (Fig. 5A).103 The formulation was pH-responsive because hypoxia in cancerous cells and tumor tissue usually causes a decrease in pH value. CIS is an effective hydrophilic drug for human cancer treatment because it can cause apoptosis and DNA damage in cancerous cells.114 EPI, which has reduced cardiotoxicity and is a semisynthetic analog of DOX, is a hydrophobic functional drug used to treat breast cancer. EPI can inhibit topoisomerase II and plays a vital role in cancer therapy by inhibiting DNA replication and lipid peroxidation.115,116 The in vitro release profile results indicated that the release of drugs from NCE and FPNCE samples at a pH of 5.4 is remarkably higher than the release at pH 7.4, indicating that the designed DDSs are pH-responsive. This can be attributed to the electrostatic interaction between the positively charged drugs and PEG chains, and the ionization state at physiological pH. Additionally, the obtained data indicated that the delayed drug release of FA-PEGylation caused greater drug aggregation in target cells and that an acidic pH ruptured the niosome structure, which led to enhanced toxicity.117 The increase in vesicle size and poly-dispersity index (PDI), as well as the reduction rate in EE for the FPNCE, was lower than that for the NCE throughout the shelf life. Therefore, the data show that FA-PEGylation can also stabilize the DDS formulation because the polymer-coated niosomes decreased systematic phagocytosis.54 Based on the in vivo results shown in Fig. 5B and C, the number of tumors decreased significantly in BALB/c mouse in FPNCE-treated tumors and the anticancer efficacy of FPNCE also significantly mitigated tumor size and enhanced body weight mass, as FA inhibited the expression of anti-apoptotic genes such as Bcl2. Following FPNCE treatment, invasion, mitosis, and nuclear polymorphism were reduced. Local drug delivery, cytotoxicity efficacy, and apoptotic body creation showed that nano-niosome FA-PEGylated was an appropriate nano-based delivery system for dual drug delivery.103 The addition of FA to the surface of niosomes has been shown to induce apoptosis in breast cancerous cell lines, as indicated by the results of the last two studies. The amount of FA receptors on the surface of normal cells is less than that on cancerous cells, so it can be hypothesized that the surface modification of niosomes can play a key role in the cellular uptake of the drugs and targeted delivery. Table 3 summarizes the details of the studies on dual delivery niosomal systems.
![]() | ||
Fig. 5 (A) Illustration of a diagram showing the simultaneous injection of CIS and EPI into a BALB/c mouse. (B) Mass of the animal body during the course of therapy compared with days (i), and the mouse body weight heat map (ii). The weight of the mouse after 20 days of treatment (iii). (C) The heat mapping of the volume of the tumor (ii) and the observation of tumor volume during therapy versus time (i). The volume of the tumor on day 20 of treatment (iii). (D) Malignant breast tumors are examined under a microscope; the malignant control, CIS + EPI, NCE, and FPNCE are stained with H&E. Nuclear polymorphism is shown by arrowheads, and cancer tissue by arrows (magnification 400×; scale bar equals 50 μm). Reproduced from ref. 103 with permission from [American Chemical Society], copyright [2022]. |
Recent research has aimed to formulate and characterize an advanced system of pH-responsive drug administration based on farnesol–gingerol-loaded (N-Far/Gin) niosomes (without surface modification) as a new method for breast cancer treatment.104 N-Far/Gin showed outstanding biocompatibility with the control human foreskin fibroblast (HFF) cells, indicating remarkable cytotoxicity against SKBR3 and MCF7 cell lines. Similar to other loaded niosomal DDSs, endocytosis is the penetration method of entrapped drugs into cancerous cells, and internalization is facilitated by the hydrophobic surface and smaller size of the niosome. N-Far/Gin causes apoptosis in SKBR3 and MCF7 cells through the synergistic effect of the drugs, as reported by apoptosis tests, by increasing the expression of anti-apoptotic genes (Table 3).104
To produce another niosomal formulation for co-loading treatment using the ecological probe sonication approach, Khan et al.105 utilized rifampicin (Rif), a biopharmaceutics classification system (BCS) class II drug with low water solubility, and ceftriaxone sodium (Cfx), a BCS class III drug with poor permeability.118,119 Owing to the undesired free-form characteristics of these drugs, they have been chosen as appropriate candidates for DDS encapsulation. To increase niosome function as nano-carriers, a combination of non-ionic surfactants from Pluronic L121 and Span 60 was considered for the niosome structure. By adding two or more non-ionic amphiphiles, more stable, monodisperse, and smaller niosomes with improved drug release profiles and a high EE% can be achieved.120 Cellular uptake can be improved by smaller niosomes, which results in greater anticancer effectiveness.
A combination of CIS and gemcitabine (GEM) for lung cancer therapy via aerosolization against normal (MRC5) and cancerous (A549) lung cell lines was studied by Mohamad Saimi et al.106 The results from this study showed that the optimized formulation of niosomes with a low dose of CIS and Gem (NGC), small particle size, and three-month stability can be applied for cancer therapy. CIS and Gem have shown good interaction in a dual delivery experiment for lung cancer cell treatments, with the potential to be more effective towards cancerous cell lines and the ability to decrease side effects in comparison with systemic drug release due to their low toxicity in neutral blood conditions.121 The EE% results demonstrated that CIS entrapment efficiency was greater than that of Gem. This was caused by the polarity of the drugs, where CIS was less polarized than Gem. As a result, CIS can easily cross the lipid bilayer membrane and Gem is expected to pass passively through the lipid bilayer membrane and become entrapped in the aqueous core.106 The ability to inhale the niosome formulation is a patient-friendly method that has the potential for further lung cancer treatment studies. Such research is based on the low surface tension of the formulations and high electrostatic charge, which can result in a high aerosol output.
![]() | ||
Fig. 6 Cellular uptake images of MG-63, KG-1, and SaOs2 osteosarcoma cancerous cell lines, treated with free DOX and CUR and drugs entrapped into LipoNio carriers. For staining the nuclei, DAPI was applied. DOX and CUR have red and green fluorescence, respectively. (A) The comparison between the cellular uptake of free drugs after 8 h-treatment in the KG-1 and MG-63 cell lines; (B) cellular uptake results of MG-63 and KG-1 cells treated with LipoNio-DOX–CUR; (C) cellular uptake of SaOs2 cells treated with LipoNio-DOX–CUR for 3 and 8 hours; (D) cellular uptake of SaOs2 and KG-1 cells treated with LipoNio-DOX–CUR for 3 hours; (E) typical features on merging. Treated cells with entrapped drugs have shown more violet and turquoise blue color intensity than free drug-treated cells. Entrapped drugs could penetrate cells by endocytosis, while the diffusion mechanism transported free drug molecules. (F) and (G) The drug release profiles in PBS at pH 7.4, 6.5, 5.4, and a temperature of 42 °C. Reproduced from ref. 54 with permission from [Royal Society of Chemistry], copyright [2017]. |
Compounds | Niosome fabrication method | Surface functionalities | In vitro/in vivo models | IC50 value (μg mL−1) | Cancer type | Key observation | Ref. |
---|---|---|---|---|---|---|---|
DDS: drug delivery system, PEG: polyethylene glycol, FA: folic acid. | |||||||
1. DOX | Probe sonication method | NA | MCF-7 | NA | Breast | - Sustained drug release | 129 |
2. PTX | PC3-MM2 | Prostate | - Improved antiproliferative effects in time- and dose-dependent manner | ||||
1. PTX | Thin film hydration | NA | HT-29 | PTX (19.98) | Colorectal | - Extended drug release | 130 |
2. OXP | OXP (23.56) | - Increase the cellular uptake of both drugs | |||||
- Increased cytotoxic and apoptosis activity | |||||||
1. CUR | Thin film hydration | PEG | U87 | 48 h: DOX + CUR (0.96) PEGNIO/DOX + CUR (0.9) PEGNIO/DOX + CUR/t-Lyp-1 (0.76) | Glioblastoma | - Increased cellular interaction | 126 |
2. DOX | Modified with tumor-homing and penetrating peptide (tLyp-1) | - Enhanced cytotoxicity | |||||
DDS: PEGNIO/DOX + CUR/t-Lyp-1 | |||||||
1. CUR | Thin film and pH-gradient technique | DSPE-mPEG | Saos-2 | NA | Osteosarcoma | - Improved drug loading capacity, anti-proliferative | 54 |
2. DOX | MG-63 | ||||||
DDS: (LipoNiosomal) | KG-1 | ||||||
1. CUR | Thin-layer hydration | NA | MCF-7 | CUR (63.12) | Breast | ↑Bax | 127 and 128 |
2. TMX | TMX (41.98) | ↑p53 | |||||
CUR + TMX | CUR + TMX (36.12) | ↓Bcl2 | |||||
DDS: nano (CUR + TMX) | Nano (CUR + TMX) (20.68) | - Controlled release at physiological pH | |||||
- Burst release at acidic pH | |||||||
1. Gem | Handjani-Vila and film hydration methods | NA | AsPC-1 | NA | Pancreatic | - Improved the efficacy of drug combination with a 9-fold increase in cytotoxicity | 131 |
2. Tocotrienols | SW 1990 | ||||||
Panc 10.05 | |||||||
BxPC-3 | |||||||
1. CUR | Thin-layer hydration | FA | MCF-7 | NA | Breast | ↓Cyclin-D | 47 |
2. Let | PEG | MDA-MB-231 | ↓Cyclin-E | ||||
↓Bcl2 | |||||||
↑Bax | |||||||
↑Caspase3 | |||||||
↑Caspase9 | |||||||
↑p53 | |||||||
- pH-dependent release behavior | |||||||
1. Artemisinin (Art) | NA | NA | SW480 | DDS (10.39) | Colorectal | ↑Bax | 132 |
2. CUR | ↑p53 | ||||||
DDS: Cur-Art-loaded NioNps | ↑Fas | ||||||
↓Cyclin-D1 | |||||||
↓Bcl2 | |||||||
↓Rb | |||||||
- Appropriate drug release | |||||||
1. Let | Thin-layer hydration | FA | MDA-MB-231 | 48 h: Let + AA (422.67), Let + niosome (255.5), AA + niosome (791.76) | Breast | ↓Cyclin-D | 133 |
2. Ascorbic acid (AA) | PEG | SKBR3 | 48 h: Let + AA (298.32), Let (457.5), AA (819.4), Let-AA-niosome (75) | ↓MMP-2 | |||
DDS: folate-PEGylated Let-AA-niosome | MCF-10A | ↓MMP-9 | |||||
↑Caspase3 | |||||||
↑Caspase9 |
The other herbal product, paclitaxel (PTX), an essential antineoplastic drug, is extracted from Taxus brevifolia bark. PTX shows effective chemotherapeutic and cytotoxic potential against many types of cancer. However, the overall therapeutic effects of PTX are restricted due to poor water solubility and low therapeutic index.134,135 To overcome these limitations and improve the therapeutic potency of cancer therapies, Alemi et al.95 developed a DDS to co-administer PTX and CUR in PEGylated niosomal formulations for increased effectiveness in MCF-7 cells. The system remarkably decreased MCF-7 cell growth to a greater extent than the free PTX/CUR combination (Table 4). The combination of PTX with niosomes and another anticancer drug, oxaliplatin (OXP), has been studied by El-Far et al.130 to encapsulate drugs in an optimized niosome and develop their therapy results against colorectal cancer. Delivering dual drug-loaded niosomes altered the release profile compared with that of their free drugs, as they revealed a widened drug release, causing a reduction in their toxicity. The encapsulation of OXP and PTX into the niosome significantly increased their cytotoxicity and apoptosis activity by up to two-or threefold compared with the free drugs. Adding D-tocopheryl polyethylene glycol 1000 succinate (TPGS) to the niosome preparation can affect the apoptotic activity of drug-loaded niosomes through different mechanisms, such as the participation of TPGS in the inhibition and destruction of the mitochondrial respiratory complex136 and the induction of DNA damage or oxidation of proteins, enzymes, and lipids, which lead to cell destruction.137 In other words, TPGS could increase bioavailability and reverse MDR.136,138 PTX was also co-encapsulated with DOX and delivered via niosomes to study the DDS in MCF-7 and PC3-MM2 cancer cell lines. The niosomes revealed a high EE and a synergistic effect due to the presence of both drugs, which can overcome MDR.129 Let is co-administrated with a supplementary and natural drug ascorbic acid (AA) by Bourbour and her colleagues133 to decrease the side effects of Let, and both are loaded on a similar folate-PEGylated niosome47 (i.e., folate-PEGylated Let-AA-niosomes). It was found that the remarkable anti-cancer activity of the niosomal DDS was due to the dual-loading of Let and AA. In addition, investigations have shown that PEG has no cytotoxic potential. Thus, the increase in cytotoxicity activities in cancerous cells by the FA-Let-AA-niosome is due to the release of both loaded drugs inside the cells (good cellular uptake) and not to the components of the niosomal formulation itself.139,140 In Fig. 7, the invasion (A) and (D), migration (B) and (E), and scratch (C) results are demonstrated. The FA-LA-niosome and FA-niosome effectively inhibited migration in both cancerous cells, which could potentially lead to FA cytotoxicity by upregulating Bax, PTEN, and p53, as previously mentioned in FA-surface-functionalized studies.
![]() | ||
Fig. 7 (A) and (D) Invasion, (B) and (E) migration, and (C) and (F) scratch results (i) microscope images (ii) for the MDA-MB-231 cells and SKBR3 cells (control) after treatment with an aqueous solution of (Let + AA), FA-Let-AA-niosome, FA functionalized bare niosomes (FA-niosome), and bare niosome. Magnification: 10×. Bar chart showing the average number of cells that invaded through the pores during the Matrigel experiment. The results are shown concerning values measured in cells with no treatment (blank group) and depict average values ±SD from at least three independent experiments. The inhibition in the SKBR3 cells was higher than in the MBR-MB-231 cell line. Reproduced from ref. 133 with permission from [Royal Society of Chemistry], copyright [2022]. |
FA-functionalized niosomes have been optimized and co-loaded with CUR and Let as a promising DDS for breast cancer cell therapy.47 Both drugs are anticipated to exist chiefly in the niosome lipid bilayer, depending on their hydrophobic nature. These results revealed that the niosomal formulation utilizing a lipid/drug proportion of 10 was considerably smaller than the same formulations with a lipid/drug proportion of 20. A greater amount of lipids in the niosomal formulation could form larger NPs and a thicker lipid bilayer.141 In addition, the mean size of niosomes was significantly increased, which was confirmed by drug encapsulation.47 Owing to the presence of FA in the formulation, the niosomal formulation had significant inhibitory effects on both the MDA-MB-231 and MCF-7 cell lines, which is consistent with the results of Sahrayi et al.102 The details of recent studies on the codelivery of drugs and herbal-based molecules are listed in Table 4.
In this regard, to enhance the inherent therapeutic efficacy of siRNAs, it has been demonstrated that nano-DDS improves the stability of siRNA, increases selectivity to the target, prevents premature degradation, and enables fast in vivo clearance of siRNAs. One of the most interesting delivery systems in the study of niosomes and siRNAs is the possibility of loading various drug molecules into niosomes to increase the efficacy of cancer treatments. To prove this, Hemati and her colleagues developed cationic PEGylated niosomes that could co-encapsulate siRNA and DOX against cell division cycle 20 (CDC20).51,53 At the cell-cycle checkpoint (G2/M), CDC20 is a regulatory protein that interacts with an anaphase-promoting complex/cyclosome (APC/C), initiating anaphase. In cancerous cells, CDC20 upregulation has been shown to result in increased cell proliferation, tumor initiation, and cancer development;149 hence, CDC20 siRNA can suppress cell growth and target the G2/M phase. The first study was conducted in three cancerous cell lines: human gastric cancer (AGS), human adenocarcinoma prostate cancer (PC3), and MCF7. The drug release profile showed sustained release, and cellular uptake data via endocytosis revealed that the number of cancerous cells treated with Nio-DOX was higher than that in the group treated with the free drug. Additionally, it has a key function in delivering the combination of DOX and siRNA through cell membranes, compared with free drugs that penetrate through a diffusion mechanism. The results indicated that the dual encapsulation of drugs and siRNA in cationic PEGylated niosomes showed an enhanced anti-cancer potential against the death of tumor cells.51
The second study by the same team focused on gastric cancer treatment with CDC20 siRNA and anticancer drug molecules to target cell cycle proteins, as previously mentioned.53 Quercetin (QC) is an herbal drug and chemo-sensitizer. It can also prevent tumor proliferation by activating the intrinsic path of apoptosis and blocking the phase S during the progression of the cellular cycle.150,151 DOX/QC/siRNA was loaded into a different niosomal formulation. The various molar ratios of dioleoyl-3-trimethylammonium propane (DOTAP) were evaluated to optimize the niosomes for advancing the loaded siRNA. The DOTAP addition leads to a reduction in the vesicle size, which was discovered by Campbell et al.,152 and PDI, which is related to the reciprocal repulsion force existing among the particles with the same charge in a suspension system.153–155 Also, the cellular uptake results indicated that the DOTAP addition improved the DOX/QC/siRNA transfection potential.153,154 This resulted in cationic lipids that improved the optimized niosome transfection potential. Apoptosis analysis revealed that there was a remarkable increase in the cell death rate when DOX and siRNA were co-delivered due to a synergistic effect on apoptosis induction. These results demonstrated that the synergistic antitumor activities and controlled release of siRNA/anticancer drug-encapsulated niosomes could downregulate the expression of CDC20 with high efficiency, resulting in a synergistic therapeutic reaction, ultimately causing gastric cancer elimination.53
Many studies have shown that the addition of DOTAP improves the niosome characteristics and the release of loaded drugs and nucleotide acids. The DOTAP presence in gene and bioactive compound niosomal formulations to boost transfection efficacy and stability has been followed by Abtahi and her colleagues.156 MicroRNA-34, a member of the p53 network, is one of the tumor-inhibiting microRNAs. A potential target of miR-34a in breast cancer is regulating the type of silent mating information 2 homolog 1 (SIRT1) oncogene.157 Down-regulation of miR-34a in various tumors has been proved. Many studies have revealed the potential of miR-34a in cancer therapy by targeting nano-DDS. Based on the results (Fig. 8), after two months of storage, the vesicle size, PDI, and percentage of EE of the optimized niosomal structure demonstrated no remarkable changes in comparison with the synthesized samples, which verifies the physical stability of niosome-CUR for up to two months (Fig. 8A). To investigate the impact of extended storage on miR-34a leakage from the outer surface of the niosomes, samples were stored at 4 °C for up to four months. Fig. 8B demonstrates that even after four months, the structures were stable with the lowest leakage. The reason for this stability is the presence of DOTAP in the niosomal formulations. In a previous study,106 the stability of niosomes was measured for three months, but in the formulation, sodium dodecyl sulfate (SDS) was used instead of DOTAP. To stabilize the niosomes, the particle size is reduced, and the negatively charged zeta potential is increased, which leads to electrostatic repulsion between the niosome particles and prevents aggregation. Furthermore, the graph in Fig. 8C illustrates that DOTAP is not cytotoxic. Consequently, the incorporation of DOTAP into niosomes has both physical and physiological benefits. According to the tumor growth graph (in vivo), the niosome codelivery system could increase the tumor inhibition activity and present a synergetic therapeutic impact higher than that of co-delivering or free structures of both miR-34a and CUR (Fig. 8D and E). The results from the curve (Fig. 8F) indicated that the mice treated with a co-delivery system (loaded with miR-34a and CUR) showed a slight change in body weight. The data showed that niosomal codelivery did not enhance the toxicity compared with niosome-miR-34a and niosome-CUR.156
![]() | ||
Fig. 8 (A) Physical stability of niosome-CUR after 15, 30, 45, and 60 days, (B) time-dependent stability of miR-34a loaded niosomes, (C) cytotoxic effect of DOTAP addition to the niosome structure, (D) impact of different structures on mouse weight, measured every 3 days, (E) effect of different structures on the tumor size (cm3) of the mouse models of breast cancer (cases treated with the structures for 21 days. A digital vernier caliper measured the tumor volumes. In the 2nd and 3rd weeks after therapy tumor volumes were remarkably less than that of the control groups), (F) mouse body weight graph (data were presented as the mean ± SD, and P < 0.05 was considered a significant statistical difference). Reproduced from ref. 156 with permission from [Elsevier], copyright [2022]. |
Similarly, the impact of DOTAP on the niosomal delivery system, transfection efficiency in MCF-7 cells, vesicle size, and PDI were investigated in a recent study by Abtahi and co-workers.158 miR-34a was loaded alone on novel PEGylated niosomes in different formulations. Following the addition of 5–15% DOTAP, the zeta potential increased, and there was a reduction in the PDI and average size. Also, the findings indicated that the presence of PEG on the niosome surface along with DOTAP resulted in increased stability and reduced average diameter. The in vivo study revealed that the body weight of niosomal miR-34a-treated mice decreased slightly after three weeks. These findings are consistent with those of niosome-miR-34a-CUR-delivery in their previous study.156
![]() | ||
Fig. 9 Fluorescent microscopic images of the uptake of free miRNAs, bare niosomes, and co-loaded DDS 3 hours after transfection. miRNAs co-loaded niosomes showed a greater cellular uptake rate and higher turquoise-blue and purple intensity compared with free miRNA-treated cells. The average fluorescence intensity and the greatest percentage of cellular uptake belong to miR15a-/miR16-1 loaded niosomes. DAPI was used to stain cell nuclei (blue), and 4% paraformaldehyde was utilized to fix transfected cells (960 magnification). Fluorescence intensities were calculated by ImageJ and plotted as mean fluorescence intensity. *p < 0.05. Reproduced from ref. 52 with permission from [Springer], copyright [2021]. |
Another study on the dual-gene-loaded niosome field was conducted by Gharbavi and his co-workers.169 NANOG, a homeobox protein, is an important transcription factor and has an essential involvement in stem cell mitosis. Furthermore, it has been discovered that it is a key player in maintaining the carcinogenic properties of various cancer types and glioblastomas.170,171 Additionally, by upregulating the multidrug resistance mutation (MDR1), NANOG can increase chemoresistance, which makes it an optimistic target for cancer treatment.172 The decoy oligodeoxynucleotides (decoy ODNs), a class of nucleic acid-based drugs, are short double-stranded DNA molecules. This therapeutic method can be a safe, efficient, and promising approach for eliminating cancerous cells.173 Previously, BSA-coated niosomes (NISM@B) were synthesized through a thin-layer hydration approach by the same researchers, and the obtained data revealed that NISM@B has efficient therapeutic agent binding potency, high uptake efficiency, and low cytotoxicity.174 In this regard, NANOG decoy ODNs were loaded into NISM@B to form NISM@B-decoy ODN complexes (NISM@B-DEC) to study the capacity of the delivery system for the cancerous cell line U87. A steady DDS was successfully prepared with a regular spherical form and narrow size distribution with acceptable zeta-potential results and biocompatibility. The ODNs released from the DDS demonstrated a controlled and pH-responsive profile as the best model for explaining the ODN release pattern. Finally, NISM@ B-DEC efficiently reduced tumor formation, was taken up by the U87 cell line, and markedly inhibited cell growth.169 According to a small number of studies, this codelivery system may be promising for cancer treatment. However, due to the complicated method and gene loading, the obtained studies are insufficient and warrant further investigation.
Recently, a new study aimed to design a novel smart nano-DDS with anticancer and antibacterial properties that can be used for lung cancer treatment.180 This DDS is prepared from a niosomal encapsulating CUR as an anticancer agent and is embedded in a chitosan gel structure containing Rose Bengal (RB) as a dye with photodynamic antibacterial capability. The fabricated system exhibited a high EE% for both drugs and temperature- and pH-sensitive release profiles. The antibacterial activity of carriers was evaluated using the colony formation technique and was applied against a Gram-negative (E. coli) and a Gram-positive (S. aureus) bacterium. RB inhibited the antibacterial effects against both bacterial strains, but prepared DDS only showed bacterial inhibition and antibacterial activity against E. coli. The designed DDS might be acceptable for lung cancer treatment because Gram-negative bacteria could promote cancer development alongside the synergetic effect of the two drugs and other good properties.180 Another study presented an advanced dual-loaded delivery system for intravesical application prepared from CUR/gentamicin sulfate (GS)-loaded niosomes embedded in thermo-responsive in situ gels (CUR/GS-co-loaded niosomes).55 The main portion of the chosen gel was Poloxamer 407, with adequate mucoadhesive and reversible gelation (depending on temperature) properties. The thermosensitive behavior of the in situ gel DDSs has been investigated by oscillation tests in time sweep mode. The test is applied to discover the required time for transition from a liquid state (room temperature 25 °C) to gel form (body temperature 37 °C) after administration in the body. This transition initiated within 20 seconds, whereas the gel state appeared after 35 seconds. This is a typical gel-dominated behavior by the elastic portion and shows a sufficient transition time in the body. The results depicted that adding niosomes to in situ gels did not change the physical stability; only a small enhancement in size (less than 10%) was observed due to polymer deposition on the niosomal surface. The results showed that the CUR/GS compound in the niosomal-embedded gel was more conserved at the end of the study compared with those in niosomes without gel.55
To evaluate chitosan hydrogel, in another experiment, chitosan-coated niosome (ChN) was utilized to increase the bioavailability of both boswellic acid (BA) and CUR drugs.181 Many studies have suggested different medicinal effects of BA, which include anti-carcinogen, anti-inflammatory, anti-arthritis, and memory impairment features. However, there are disadvantages, including low solubility and low bioavailability.182 The characteristic analysis of samples showed that at low levels of chitosan, the average particle size of ChN was reduced compared with bare niosomes (BN). It has been suggested that ultrasound processing after the adhesion of chitosan to the outer layer of niosomes caused this reduction. Moreover, increasing the concentration of chitosan enhanced the average vesicle size. The creation of a thicker chitosan coat and chitosan bridges among particulates caused an increase in particle size at higher chitosan levels.181 In advanced technologies, the prototyping method and nanotechnology have opened novel cancer cell treatment areas.183 The emergence of the 3D-printing method has produced numerous updated possibilities for polymer hydrogel-based drug administration approaches, particularly for cancer therapy.184,185 3D-printing prepares controllable and organized scaffolds with excessive precision, which might give rise to the prolonged and mediated release of various agents and molecules, such as proteins and different types of drugs in a possible and acceptable procedure.186 Hosseini et al. recently presented a novel DDS based on 3D-printed gelatin–alginate biodegradable scaffolds incorporating paclitaxel-loaded niosomes (Nio-PTX@GT-AL) for breast cancer therapy.183 Due to the presence of alginate, the printed scaffolds are pH-sensitive DDSs for cancer treatment. According to the mechanical properties (tensile and compressive states) of Nio@GT-AL and Nio-PTX@GT-AL, the existence of drug-containing niosomes leads to a slight enhancement in modulus and strength in both the compressive and tensile states, which may lead to prolonged stability. The activity of caspase-3, caspase-8, and caspase-9 has been demonstrated in Fig. 10. Treated cells with free-PTX, Nio-PTX, Nio@GT-AL, and Nio-PTX@GT-AL markedly enhanced the activity of the three caspases. However, niosome-containing samples, mostly Nio-PTX@GT-AL, have improved the activity of caspase considerably more than free-PTX.183
![]() | ||
Fig. 10 The comparison of activity of caspase-3, caspase-8, and caspase-9 in niosome-containing scaffold therapies and free-drug (PTX). An enhancement in the activity and expression of three caspases can be observed as an index of the capability of the cancer therapy system. (A) Caspase-3; (B) caspase-8; and (C) caspase-9. Nio-PTX@GT-AL has distinctly shown the most increase in the activity of all three types of caspase, leading to a 5-fold improvement in comparison with the control group (P-value = P, *P < 0.05, **P < 0.01, ***P < 0.001). Reproduced from ref. 183 with permission from [Elsevier], copyright [2023]. |
Various parameters must be considered when designing an efficient dual delivery niosome system. The use of material combinations, such as surfactants, is necessary to translate the niosome formulation method. The characteristics of these materials include size, morphology, stability, PDI, EE, zeta potential, and even cytotoxicity, and gene induction, such as DOTAP, as previously mentioned. The therapeutic impact of these materials is both dual and effective. Also, drug properties, such as hydrophobic and hydrophilic properties, as well as their molecular size, play a crucial role in achieving a high EE%. Surface modification is another solution to optimize niosome characteristics. (i) Using polymers with different molecular weights and characteristics has a considerable effect on niosome properties. The stability of niosomes is facilitated by PEGylation, a biocompatible biomaterial. Niogelsomes, such as chitosan, can also cause sustained drug release or aid in targeted delivery. (ii) Various agents, such as FA (vitamin B9), may have dual impacts in optimizing DDS by increasing EE and biological effects by attaching cancer cell receptors to increase endocytosis. Therefore, there are different methodologies to prepare targeted DDSs along with the synergistic effects of dual drugs. To examine the efficacy and therapeutic dose of dual drugs, it is necessary to conduct more in vivo experiments because the pharmacokinetic and pharmacodynamic effects are measurable in a body condition.
Psoriasis is an immune-mediated skin disorder. Methotrexate is often the first line of treatment, but its low solubility and systemic toxicity can limit its use. Yang et al. addressed this issue by using ceramide-based niosomes (cerosomes) to dual load methotrexate and nicotinamide (NIC) to reduce toxicity and increase treatment efficiency.188 The combination of methotrexate and nicotinamide can synergistically influence psoriasis treatment. Nicotinamide's hydrotropic and anti-inflammatory properties can enhance the solubility and efficacy of methotrexate and decrease the production of pro-inflammatory factors. In their study, cerosomes were used to deliver methotrexate and nicotinamide to deeper skin layers. This reduced the skin thickness and the expression of pro-inflammatory cytokines in psoriatic skin. In vitro and in vivo permeation studies showed that the cerosomes significantly increased methotrexate and nicotinamide permeation and retention in the skin. Methotrexate/nicotinamide cerosomes demonstrated potent anti-proliferative effects on HaCaT cells irritated by lipopolysaccharide by arresting the cell cycle at the S phase and inducing apoptosis. In imiquimod (IMQ)-induced psoriatic mouse model, using methotrexate/nicotinamide niosomes improved skin lesions compared with the oral administration of methotrexate. This approach also reduced the spleen index and epidermal thickness while down-regulating the expression levels of pro-inflammatory cytokine mRNA (Fig. 11).
![]() | ||
Fig. 11 (A) A schematic illustration of the procedure for the animal model and treatment of IMQ-induced psoriasis. (B) Photographs of the dorsal epidermis of rodents treated with various formulations for 1, 3, 5, and 7 days. (C) Throughout the investigation, each group's physical weight changed. (D) PASI of epidermis on days 1, 3, 5, and 7 (&&p = 0.01). (E) The scores are added together and reported as cumulative scores. Spleen index following seven days of therapy. No statistically significant difference between the normal and model groups; ***P < 0.001 between the MTX (oral) and model groups; ***P < 0.001 among the normal and histological groups. Reproduced from ref. 188 with permission from [Elsevier], copyright [2021]. |
Nevertheless, the application of dual-niosomes, a specific category of drug delivery system, faces significant obstacles, specifically in the context of cutaneous drug delivery. A significant concern pertains to the efficacy of drug delivery systems, as certain medications may accumulate prior to permeating their intended site or fail to reach the epidermis. Moreover, the assertions regarding the effectiveness of dual-niosomes have yet to undergo replication or peer review by other scientists, which presents a substantial barrier to their acceptability. Additionally, the potential systemic toxicity of particular medications, such as methotrexate, may impose restrictions on their usage. While there have been suggestions to enhance treatment effectiveness and mitigate toxicity through the use of ceramide-based niosomes, the extent of their potential adverse effects and long-term ramifications has yet to be investigated.
In another study, Khan and colleagues examined the encapsulation of ceftriaxone sodium and rifampicin in niosomes using the eco-friendly probe sonication procedure for tuberculosis treatment.105 The optimized formulations showed very high drug entrapment efficiencies for ceftriaxone sodium and rifampicin, with values exceeding 96%. Additionally, some drug release studies revealed that the niosomal formulations had faster in vitro ceftriaxone sodium and rifampicin release rates than pure ceftriaxone sodium and pure rifampicin.
Sohrabi et al. prepared and characterized chitosan gel-embedded moxifloxacin-loaded niosomes as a potential drug carrier for topical antimicrobial delivery.189 The designed moxifloxacin niosomal gel hybrid system showed sustainable release behaviors. The antibacterial efficacy of various formulations was studied by evaluating the minimal concentrations for inhibition, minimal concentrations for a bactericidal effect, and an assay of agar diffusion by PA and Staphylococcus aureus (SA). The results of antimicrobial experiments exhibited that niosomal formulation played a remarkable role in increased anti-PA efficacy. Chitosan gel-embedded moxifloxacin niosomes could guarantee an extended local retention and better effectiveness due to high drug loading, prolonged drug release, and bioadhesive behaviors. Therefore, the hybrid system has potential as an efficient novel platform for the controlled delivery of antimicrobial agents.
Targhi and co-workers have investigated the synergistic effect of silver/copper NPs and CUR against PA and SA.190 The niosomal formulations and their hydrogel counterparts were evaluated to study their antibacterial activity against SA and PA using disk diffusion, and assays for the minimum concentration for an inhibitory effect, and minimum concentration for a bactericidal effect. Furthermore, to study the anti-biofilm effect of the samples, anti-biofilm assay and biofilm-associated gene expression by real-time PCR have been used. The niosomal formulations exhibited high drug entrapment efficiencies with a prolonged release of drug patterns over 72 hours. A crystal violet assay was used to study the anti-biofilm performance of the materials. The results of the crystal violet assay displayed that CUR-AgNP-loaded niosomes, CUR-CuNP-loaded niosomes, CUR-AgNP-loaded niosomal hydrogel, and CUR-CuNP-loaded niosomal hydrogel significantly reduced biofilm formation in the strains compared with free forms of metal NPs and CUR. Interestingly, compared with niosomal formulations, hydrogel-embedded niosomal formulations enhanced the inhibition zone and significantly decreased the minimal inhibitory and bactericidal concentrations. The cytotoxicity of the samples was analyzed by the tetrazolium (MTT) assay. The findings indicated that CUR-loaded niosomes exhibited the most negligible toxicity compared with the CUR-AgNP-loaded niosomal hydrogel and CUR-CuNP-loaded niosomal hydrogel at the same concentration. Moreover, the formulations of CUR-AgNP-loaded niosomes and CUR-CuNP-loaded niosomes displayed higher cytotoxicity than their hydrogel-incorporated counterparts (Fig. 12). It is worth noting that niosomal CUR demonstrated lower cytotoxicity than free CUR, possibly attributed to variances in intracellular trafficking or diverse cellular uptake mechanisms between niosomal and free forms.
![]() | ||
Fig. 12 (A) S. aureus (left) and Pseudomonas aeruginosa (PA) (right) inhabit drug-free and drug-loaded zones. Left side: (a) pure curcumin, (b) curcumin-AgNP-laden niosomal hydrogel, curcumin-CuNP-containing niosomes, (d) copper NPs, (e) curcumin-laden niosomes, (f) niosomes laden with curcumin-AgNPs, (g) NPs of silver, and (h) curcumin-CuNP-infused niosomal hydrogel. The right side: (a) curcumin-AgNP-laden niosomal hydrogel, (b) copper NPs, (c) curcumin-CuNP-laden niosomal hydrogel, (d) curcumin-laden niosomes, (e) silver NPs, (f) free curcumin, curcumin-AgNP-laden niosomes, as well as curcumin-CuNP-containing niosomes. (B) In vitro cytotoxicity of HFF cells. FreeNio: niosomes that have been loaded with curcumin, Cur-AgNPs-Nio: niosomes that have been loaded with curcumin and AgNPs, Cur-CuNPs: niosomes that have been loaded with curcumin and CuNPs, niosomal hydrogel containing curcumin-CuNPs is known as MGel-Cur-AgNPs-Nio. Results are presented as mean, and standard deviation, with n = 3. ***P < 0.001, **P < 0.01, *P < 0.05. Reproduced from ref. 190 with permission from [Elsevier], copyright [2021]. |
In other research, Mostafavi et al. reported selenium and Glucantime-loaded niosomes as a novel niosomal formulation against Leishmania tropica.192 The efficacy of dual drug-loaded niosomes was compared with their simple forms alone. For evaluation of the effectiveness of novel niosomal formulations, an in vitro MTT test, gene expression profile, and intra-macrophage model were used. The results showed that no cytotoxicity effect has appeared for niosomal formulations of selenium and Glucantime, alone and in combination, and simple forms of selenium and Glucantime. Furthermore, niosomal formulations of the forms of selenium and Glucantime exhibited more inhibitory effects than the simple forms of selenium and Glucantime. Also, the levels of gene expression of interleukin (IL-10) reduced, while the level of IL-12 and metacaspase enhanced. Bahraminejad et al. prepared a dual zirconium/tioxolone niosomal formulation via a film hydration procedure, and its leishmanicidal performance against amastigotes and promastigotes was evaluated by MTT and flow cytometry methods.193 Furthermore, apoptosis, gene expression levels, superoxide dismutase activity, the production of reactive oxygen species, and nitrite generation were studied to evaluate the mechanism of action niosomal formulation. The niosomal formulation of zirconium/tioxolone exhibited no cytotoxic effect. It increased the interleukin (IL)-12 expression level and inducible nitric oxide synthase and remarkably decreased the IL-10 gene expression level, which confirms the immunomodulatory role of zirconium/tioxolone niosomes.
Hakimi Parizi et al. reported niosomal formulations of tioxolone coupled with benzoxonium chloride against Leishmania tropica.194 This research evaluated the leishmanicidal activity against amastigotes and promastigotes, apoptosis, gene expression levels of free tioxolone and benzoxonium chloride, and dual niosomal formulations of tioxolone and benzoxonium chloride. The niosomal formulations of tioxolone and benzoxonium chloride were more efficient than Glucantime on promastigotes and amastigotes. Furthermore, the toxicity of the prepared niosomal formulation was remarkably less than Glucantime. Also, the flow cytometry test on the dual niosome of tioxolone and benzoxonium chloride exhibited a higher number of early apoptotic events as the principal mode of action. Furthermore, the niosomal formulations enhanced the IL-12 expression level and metacaspase genes and reduced the IL-10 gene expression level. Anjum et al. designed AmB/pentamidine (PTM) niosomes and optimized them by the Box–Behnken design method.195 The optimized AmB/PTM niosomes were incorporated into the chitosan. The as-synthesized AmB/PTM/niosome/gel increases the retention time of nanocarriers at the infection site and enables AmB/PTM niosomes to penetrate layers of skin. To analyze the release pattern of AmB and pentamidine, their in vitro release from AmB/PTM niosomes and AmB/PTM/niosome/gel was performed. The results showed a prolonged release property compared with their respective drug solution. The results of the ex vivo permeation study showed a greater percentage inhibition, higher skin penetration, and lower IC50 against the promastigotes of AmB/PTM niosomes, confirming its better antileishmanial activity.
Bacterial resistance to multiple antibiotics is a significant concern in the field of global public health. This resistance causes antibacterial treatments to fail and infectious diseases to develop. Innovative approaches have been investigated to address bacterial infections. These include the co-administration of D-cycloserine and ethionamide, the encapsulation of ceftriaxone sodium and rifampicin within niosomes, and the utilization of chitosan gel-embedded niosomes laden with moxifloxacin for topical antimicrobial delivery. Notwithstanding this, the efficacy and enduring consequences of these methodologies remain incompletely comprehended.199 The management of diseases such as MT, PA, SA, and leishmaniasis represents an additional obstacle. Although niosomes and combination therapies have been suggested as potential approaches to address the issue of antibiotic resistance exhibited by biofilm-forming bacteria and drug-resistant MT, PA, and SA, respectively, the effectiveness of these treatments is still being studied.200 The existing therapeutic approaches for leishmaniasis, a prevalent health concern induced by diverse species of leishmania, are accompanied by several limitations: exorbitant expenses, severe adverse effects, drug resistance, suboptimal treatment efficacy, prolonged therapy duration, and pharmacological inadequacies. The injection of amphotericin B (AmB) and Glucantime into niosomes for the treatment of cutaneous leishmaniasis has been the subject of research. However, the precise inhibitory effects of these treatments remain to be determined.201 Finally, antimicrobial resistance poses a significant therapeutic challenge for Klebsiella pneumoniae. Although scientists have loaded niosomes with gentamicin and ethylenediaminetetraacetic acid (EDTA) to examine their biofilm inhabitation and antibacterial activity against Klebsiella pneumoniae strains, the antimicrobial and anti-biofilm inhibitory effects of these niosomal formulations have not yet been validated.202
Porkoláb et al. examined the loading of alanine and glutathione in niosomes to deliver a protein cargo into cultured cells of the neurovascular unit.206 In this study, the results demonstrated that using alanine/glutathione niosomal formulations led to increased cargo delivery into the neurovascular unit and astroglial cells. Moreover, when metabolic and endocytic inhibitors were employed, it was observed that the cellular uptake of niosomal formulations was partially mediated through endocytosis. Among the alanine transporter genes, the small neutral amino acid transporter SNAT2 (SLC38A2) exhibited high expression in all cell types except for SH-SY5Y neurons. The neutral amino acid transporter genes ASCT1 (SLC1A4) and ASCT2 (SLC1A5), as well as SNAT1 (SLC38A1), showed moderate expression levels. On the other hand, SNAT5 (SLC38A5) expression was found to be low-to-moderate in all tested cell types.
In another report, Mészáros et al. loaded glutathione and solute carrier ligands, including alanine and glucopyranose, into niosomes.207 Loading targeting ligands on the niosomal formulations enhanced the cargo molecule uptake in cultured brain endothelial cells. The cellular uptake depends on temperature and is reduced by endocytosis blockers and metabolic inhibitors. The niosomal formulations enhanced the plasma membrane fluidity, revealing the fusion of nanovesicles with endothelial cell membranes. The application of cell-penetrating peptides like TAT ((AYGRKKRRQRRR)) for cellular transportation improvement has been widely considered. Yadavar-Nikravesh et al. loaded an anti-HIV drug (tenofovir) into a PEGylated niosomal formulation by a thin-film hydration procedure.208 After that, the TAT peptide is embedded in the niosomal formulation. The as-prepared niosomal formulation exhibited a good entrapment efficiency of 75 ± 2.516%. The MTT assay was employed to analyze the cytotoxic effects of different formulations, namely the PEGylated NP, tenofovir-loaded PEGylated NP, free drug, tenofovir-loaded TAT-conjugated NP, and TAT peptide. All the treatments demonstrated concentration-dependent cytotoxicity on HeLa cells. Notably, the tenofovir-loaded TAT-conjugated NP exhibited a higher cytotoxic effect than the tenofovir-loaded PEGylated NP. Furthermore, the inhibitory effects of the samples against HIV-infected HeLa cells were investigated at various non-toxic dosages. The anti-Scr HIV-1 effects of all treatments followed a concentration-dependent pattern. Combining both the tenofovir-loaded TAT-conjugated NP and the tenofovir-loaded PEGylated NP demonstrated more potent inhibitory effects than free tenofovir at different concentrations. However, it was observed that the tenofovir-loaded TAT-conjugated NP exhibited weaker anti-HIV-1 effects than the tenofovir-loaded PEGylated NP. The results obtained from this study revealed a lower anti-Scr HIV effect, higher cytotoxicity, and improved release of tenofovir for the tenofovir-loaded TAT-conjugated PEGylated NP when compared with the PEGylated NP.
It is critical to emphasize that niosome stability is an initial consideration. Due to the aggregation, fusion, and leakage of the encapsulated drug that can occur as a result of their fluid bilayer structure, their shelf life, storage conditions, and biological activity may be adversely affected.209 Expanding the manufacturing capacity of niosomes poses an additional obstacle. The preparation methods commonly employed are intended for laboratory-scale operations and may not be viable for large-scale manufacturing on account of cost, yield, and reproducibility concerns.209 Targeting represents an additional substantial obstacle in the application of niosomes. Although medications can be passively targeted to specific tissues or organs, it may not always be possible to deliver them specifically and exclusively to the intended cells or receptors.210 Lastly, niosome toxicity is a significant concern. The concentration, charge, and structure of particular surfactants utilized in niosomes may lead to cytotoxic, hemolytic, or irritant effects. As a result, a meticulous selection and optimization of surfactants and their mixtures are required.211
Despite extensive research in the field of dual-niosomes as a controlled DDS to treat a wide range of diseases due to multiple advantages such as chemical stability, biocompatibility, and biodegradability, the application of targeted niosome formulations towards clinical usage has been limited. The different affinity of the drugs towards the niosomes is one of the main challenges for the dual-functional drug niosomes.205 Therefore, the choice of amphiphilic molecules to prepare niosomes is significant because it provides various structural compartments to interact with the hydrophobic and hydrophilic drugs.215 Leakage of drugs, a low EE, the short shelf life of the formulation, aggregation, fusion, hydrolysis of encapsulated drugs, and carrier stability are the other limitations that hinder the use of dual-niosomes as potential DDSs. Membrane filtration and heat sterilization are unsuitable for dual-niosomes. So, these challenges require further study to prepare commercially dual-functional drug niosomes. There are two methods to improve the dual-niosome stability at ambient conditions.42 Dispersing dual-niosomes in a viscous gel is applied either to decrease the rapid leakage of the loaded drugs from niosomal formulations or to decrease the effect of a burst release perceived with dual-niosomes.216 However, the release of drugs from this system may be complex because the molecules of drugs have to be released from the bilayer membranes and diffuse via the viscous gel.
Another procedure for better physical stability is using freeze-drying or spray-drying methods to convert the dual-niosomal liquid dispersion to a powder form. This approach increases the physical stability of the dual-niosomes and can also decrease the oxidative instability of oxidizable drug molecules by reducing the formation of hydroxyl free radicals. Sterilization of lipid-based products such as niosomes is a challenging effort. Steam sterilization and dry heat are inappropriate for lipid-based DDSs, as this may lead to an extensive leakage of drug molecules from the bilayer vesicles. Furthermore, membrane filtration is unsuitable for drug delivery formulations larger than the pore size of the bacterial filters. Fabrication of dual-niosomes under gas (ethylene oxide), gamma irradiation, and aseptic conditions can be suitable, as minimum heat is produced within the sterilization procedure. To sterilize the dual-niosomes, all the raw materials, including drug solutions, organic lipids, and buffer, are passed from the bacterial filters, and the final formulation is prepared under aseptic conditions. Gamma irradiation could be used for a packaged product and is applicable for heat-sensitive drugs due to its high penetration power. The sterilization of dual-niosome formulations with gamma irradiation is a useful method, and thus the influence of gamma illumination on the physical stability of dual-niosomes can be a potential investigative line for future work.
The building components of dual-niosomes are surfactants, which play critical roles in the preparation and properties of these DDSs. The usage of surfactants can lead to some degree of toxicity. However, no specific studies have been conducted to investigate the toxicity of these formulations in animal models, particularly in long-term investigations. It is crucial to address this research gap and thoroughly examine the long-term tolerability of surfactant-based systems before they can be applied in clinical settings. Furthermore, there is a lack of sufficient attention given to exploring the potential applications of amphiphilic molecules, which possess biological activity or can serve as targetable ligands in bilayer vesicles. However, a few compounds with dual properties like biological activity and amphiphilicity may be available. So, preparing these amphiphilic compounds that could be converted into bilayer vesicles for the simultaneous delivery of two drugs is effective.
The development of appropriate dual-niosomes requires deliberation. The formulation of niosomes is influenced by a variety of factors, including the surfactant employed, the variables of production, and the method of preparation. Most presently employed, clinically acceptable nanotherapies have a simple structure and composition that lends itself well to mass production methods. The preponderance of dual-niosomes has been produced using conventional methods, such as reverse-phase evaporation, ether injections, and thin-film rehydration techniques. This method has a number of disadvantages, including the need to remove organic solvents, expensive costs, and a lengthy process. The laboratory production and analysis of dual-niosomes are typically performed on a milliliter scale. These small quantities are adequate for both in vitro and in vivo research. Dual-niosomes may be challenging to construct on a large scale due to the impossibility of mass-producing dual-drug carriers using current production methods. Therefore, it is essential to establish an efficient and cost-effective method for mass-producing dual-niosomes that are uniform in size and possess clinically acceptable properties. A comprehensive summary of the primary benefits, drawbacks, and possible remedies of nanosomal drug delivery systems is presented in Table 5. This entails an exhaustive examination of the advantages and disadvantages of these systems, including potential toxicity and difficulties in large-scale production, in addition to the benefits they provide, such as enhanced drug stability and targeted delivery. The table additionally examines possible approaches to surmount these obstacles, providing an impartial viewpoint on the implementation of nanosomal drug delivery systems in therapeutic contexts.
Challenge and future direction | Limitations | Solutions | Ref. |
---|---|---|---|
Drug affinity towards niosome | Variability in drug affinity towards niosomes | Selection of amphiphilic molecules for niosome preparation to interact with hydrophobic and hydrophilic drugs | 217 and 218 |
Leakage of drugs | Low encapsulation efficiency (EE) | Incorporating dual-niosomes in a viscous gel to reduce rapid leakage and burst release, or utilizing freeze-drying or spray-drying methods to convert liquid dispersion to powder form | 219 and 220 |
Formulation stability | Aggregation, fusion, hydrolysis, and short shelf life | Further research on stability enhancement methods such as freeze-drying, spray-drying, and sterility through gamma irradiation or aseptic conditions | 221 and 222 |
Toxicity of surfactants | Lack of toxicity studies | Thorough investigation of long-term tolerability of surfactant-based systems in animal models before clinical application | 209 |
Mass production | Infeasibility of mass-producing dual-drug carriers using current methods | Establishment of an efficient and cost-effective method for mass-producing dual-niosomes with uniform size and clinically acceptable properties | 36 |
This journal is © The Royal Society of Chemistry 2024 |