Skip to content
BY 4.0 license Open Access Published by De Gruyter March 24, 2022

Synthesis and characterization of smart stimuli-responsive herbal drug-encapsulated nanoniosome particles for efficient treatment of breast cancer

  • Milad Akhlaghi , Mohammad Taebpour , Narges Nikoonahad Lotfabadi , Seyed Marteza Naghib EMAIL logo , Neda Jalili , Leila Farahmand , Bibi Fatemeh Haghiralsadat EMAIL logo , Mehdi Rahmanian EMAIL logo and Davood Tofighi
From the journal Nanotechnology Reviews

Abstract

Extraction from the herbs was performed using the Soxhlet method. Various formula was synthesized for niosomes containing the extracts through thin film synthesis technique, and the most efficient formulation was selected. Afterwards, physicochemical properties of niosomes, including size, polydispersity index (PDI), zeta potential, morphology, encapsulation efficacy (EE%), extract release rate, non-interactive action between the extracts and niosomes, antibacterial potential, and cellular uptake were assessed. Finally, the toxicity level of the niosomes to breast cancer cells was compared and their impact on the expression of p53 and MCL-1 was evaluated. Our data demonstrated that the synthesized niosomes were sensitive to the temperature and pH. Also, the niosomes containing Hedera Helix extract (Nio-HHE) sized 97.7 nm, with a zeta potential of −19.9 ± 6.7 mV, PDI of 0.35, and 58 ± 2.4% encapsulation efficacy showed more toxicity to the cancer cells than the niosomes of Glycyrrhiza glabra extract (Nio-GGE) with the size of 111 ± 8.5 nm, zeta potential of −23.5 ± 4.5 mV, PDI of 0.113, and 69 ± 1.2% encapsulation efficacy. The former system proved to have more antibacterial potential, and affect the expression of the oncogenes more than the latter. Meanwhile, both niosomal systems demonstrated an acceptable cellular uptake, and no chemical interaction with the extracts was observed. Furthermore, useful function of the synthesized niosomes was confirmed by morphological assessments. Our data confirmed that encapsulation of herbal extracts improves their anticancer and antibacterial potential. We concluded that Nio-HHE has more significant antitumor effects on breast cancer cells than Nio-GGE. Consequently, applying nano drug delivery systems based on herbal therapy could mitigate the side effects resulting from chemotherapy and radiotherapy, and offer promising perspectives for treatment of breast cancer.

1 Introduction

Medicinal plants and using them in the treatment of diseases have been considered by humans since the beginning of history [1]. Even today, many statistical studies have shown that the use of medicinal plants is increasing worldwide. The low side effects of medicinal plants compared to chemical drugs, their reasonable price, and their availability are among the factors that have augmented the use of these plants, their extracts, and essential oil all over the world. Recent studies confirm the beneficial usage of herbals in the treatment and prevention of diseases such as diabetes, atherosclerosis, neurological diseases, and cancer [2,3]. Ivy and Licorice are among herbals with many therapeutic properties.

Licorice, scientifically known as Glycyrrhiza glabra, which is mostly used orally, belongs to the Fabaceae family and is a species native to the Mediterranean region. Photochemical analysis shows that the chemical composition of this plant contains many compounds such as glycyrrhizin, 18β glycyrrhizic acid, glabrin A and B, or isoflavones that can be responsible for the antioxidant, antiinflammatory, antiviral, and antimicrobial properties of this plant. Numerous studies have also shown that the use of licorice is effective in the treatment of many diseases such as microbial/viral infections, cancer, skin inflammation, bronchitis, depression, kidney disease, and gastric and intestinal ulcers [4,5]. Ivy scientifically named Hedera helix, belongs to the Araliaceae family, is one of the evergreen plants that is found in most parts of the world and has been proven to be effective in treating many diseases. In the chemical analysis of the chemical composition of this plant, the presence of many compounds such as unsaturated sterols, tannins, saponins, alkaloids, and flavonoids have been identified that can be attributed to the therapeutic features of this plant. Hedera Helix has also been proven to improve the therapeutic process of diseases such as microbial and parasitic diseases, bronchitis, asthma, diabetes, depression, and cancer [6,7].

The use of plants and their extracts in the treatment of many diseases such as cancer is increasing and therefore has attracted the attention of researchers. Among these diseases, breast cancer, which is one of the most common cancers among women, is of particular importance. About 1.5 million women are diagnosed with breast cancer each year, which accounts for 25% of all cancers diagnosed in women and also is responsible for about 450,000 deaths yearly. Based on the World Health Organization reports, the occurrence of breast cancer in the world is expected to reach more than 3.2 million new cases by 2050 [8]. Many studies show the role of genetics and factors affecting genetics in addition to environmental factors on the onset and progression of breast cancer. p53 is a tumor suppressor protein whose mutation in its gene is one of the most common changes that can be seen in all cancers, including breast cancer. p53 mutations, in addition to increasing the risk of developing tumors, can also affect chemo-sensitivity in cancer cells [9]. MCL-1 is also one of the proteins whose expression is directly related to the development of breast cancer. Also, high expression of MCL-1, a member of the BCL-2 family of proteins that is involved in regulating the apoptotic pathway, reported in cancer cells causes chemical resistance and tumor development in cancer [10]. Therefore, it seems appropriate that regulating the expression of this gene can be considered as a useful way to treat this malignancy.

Surgical removal of the tumor, radiotherapy, and chemotherapy are still the most important methods for breast cancer treatment. The most important chemotherapeutic agents in breast cancer therapy include Docetaxel (DCX), Thioridazine (THZ), Disulfiram (DSF), and Camptothecin (CPT) [11]. However, the use of chemotherapy in the treatment of breast cancer is faced with problems such as resistance of cancer cells to chemotherapy agents, short half-life of these drugs in the circulatory system, and weak solubility of these drugs in water which decrease their cellular uptake by cell membranes. In addition to these challenges, the inability to differentiate normal cells with cancer cells by chemotherapy agents and the lack of targeted delivery of the drug to the target tissue and reduce the effect on non-target tissue, which in turn causes adverse unwanted side effects in the use of this drug, on the other hand, reveals the exigency for alternative treatment methods with lower side effects and easier biodegradability [12], such as the use of herbs and their essential oils and extracts.

Although the use of medicinal plants and their extracts also faces obstacles such as solubility and low half-life, nanotechnology as an interdisciplinary science is trying to overcome these challenges by designing novel strategies. Nanotechnology has attracted much attention in biomedicine and nanobiotechnology [13,14,15]. Designing and synthesizing nanoparticles that are sensitive to environmental stimuli such as niosomes and liposomes [16,17,18], is among these strategies [19,20,21,22,23]. Niosomes, which are formed from the accumulation of non-ionic surfactants in the aqueous medium, are capable of carrying hydrophilic and hydrophobic drugs due to their special chemical structure. The desirable properties of niosomes, such as their compatibility with the body, easy and low-cost design, and the controlled release of drug to the targeted tissue, are among the reasons that have attracted researchers’ attention to these nanoparticles as drug delivering nano-carriers [24,25]. So, the aim of this study was to synthesize and characterize nanoniosome containing Glycyrrhiza glabra (Nio-GGE) extract and niosome containing Hedera helix (Nio-HHE) extract in order to evaluate their cytotoxicity on breast cancer cells (MCF-7 and BT-474) and also to evaluate their effects on the expression of p53 and MCL-1 genes that are involved in the development of breast cancer and comparison of their antibacterial effects on Escherchia coli and Staphylococcus aureus in order to achieve an effective herbal based formulation for the treatment of breast cancer.

2 Methods

2.1 Extraction

The extract of Glycyrrhiza glabra was obtained from its root, while the extract of Hedera helix was obtained from its leaves. Before extracting, the type and species of plants were confirmed by the botanists of Yazd University. At first, 500 g of the mentioned parts of the plants were dried in ambient temperature, and were made into powder. Extraction was performed by Soxhlet method. First, 50 g of the powder obtained from each plant was compressed in a cartridge chamber and then placed in a Soxhlet column and by attaching a balloon containing 500 mL of ethanol (Merck, Germany) to the Soxhlet and installing a condenser and water inlet and outlet pipes, extraction was started and continued for 6 h to obtain plant extracts. Finally, in order to evaporate the solvent, the extract was dried away from sunlight at room temperature.

2.2 The maximum absorbance wavelength (λ max) and standard curves

In order to figure out the maximum absorbance wavelength of each extract, stock solution of each extract with a concentration of 1 mg/mL in PBS (Sigma, USA) and Isopropanol buffer (Merck, Germany) was prepared, and by using diluent method, various concentration of each extract from its stock solution were obtained. Then, the absorbance of each sample was measured by spectrophotometer (Epoch, USA) in the range of 200–800 nm and the wavelength which had the highest absorption in all sample was reported as λ max. The standard curves in PBS and Isopropanol buffer for each extract was plotted by measuring absorption of various concentration of each extract at λ max using spectrophotometer and the equation line of each curve was also calculated.

2.3 Synthesize of nanoniosomes containing extracts

To synthesize Nio-GGE and Nio-HHE and select the optimal formulations, the following steps were performed briefly:

  • synthesizing of nano-niosomes by thin film method,

  • evaluation and comparison of the effect of the type of non-ionic surfactants used in the synthesis of niosomes as well as different molar ratios of cholesterol:surfactant in the encapsulation efficiency (EE%) of niosomes,

  • selection of optimal formula from various formulations based on (EE%), extract release rate, and niosome size,

  • adding 5% distearoyl phosphoethanolamine-polyethylene glycol (DSPE-PEG) (Ludwigshafen, Germany) to enhance the stability of nanosystems and evaluate its effect,

  • adding 20% 1,2-dipalmitoyl-sn-glycero-3-phosphocholine phospholipid (DPPC) (Ludwigshafen, Germany) to induce temperature sensitivity as a stimulus for drug delivery to cancer tissue.

The thin film method was used to synthesize the niosomes to deliver the extracts to the cancer cells. In this study, different formulations of niosomes were synthesized to encapsulate each of the extracts and then the optimal formulation was selected from them. In summary, in the first stage, different ratios of cholesterol, non-ionic surfactants (Tween-60 and Span-60) (DaeJung Chemicals & Metals, South Korea), and DPPC were dissolved in solvent Chloroforms (Sigma, USA), according to Tables 1 and 2 for synthesizing Nio-GGE and Nio-HHE, respectively. The synthesizing of niosome by the thin film method has two phases. In the first or organic phase, cholesterol and surfactants using chloroform solvent were dissolved in a round bottom balloon. Then, in order to remove the solvent, the rotary instrument (Heidolph, Hei-VAP) at 50°C, 150 rpm, and vacuum conditions were used. After evaporation of chloroform as solvent, a thin lipid layer is formed at the bottom of the balloon. Finally, in order to ensure the removal of the solvent, the balloon was aerated with nitrogen gas for a few seconds and held at 4°C for 24 h. In the aqueous phase, Glycyrrhiza glabra extract (GGE) and Hedera helix extract (HHE) were inactively loaded into the niosomes. For this purpose, synthesized niosomes dissolved with GGE and HHE at a concentration of 2 mg/mL in PBS buffer at pH = 7 and were hydrated by a rotary apparatus at 60°C and 150 rpm. Reducing the size of the synthesized nanoniosomes were also done by using probe sonicator microtype (ChromeTech, UH1200B) with 60% power that was alternatively on for 15 s and off for 10 s every 45 min. Also, during the sonication, by placing the samples on the container containing ice, their temperature was prevented from increasing. Finally, the un-entrapped extracts were separated from the niosomes containing the extracts using a 12 kDa cut off dialysis bag.

Table 1

Type of surfactant, different molar ratios of non-ionic surfactant l/cholesterol, and type of lipids used in synthesizing different Nio-GGE formulations

Formula Tween60 (%) Span60 (%) Cholesterol (%) DPPC (%) DSPE:PEG (%)
F1 0 75 25 0 0
F2 0 70 30 0 0
F3 70 0 30 0 0
F4 0 70 30 10 0
F5 70 0 30 10 0
F6* 70 0 30 10 5

*Optimal formula.

2.4 Investigating the EE%

After separating un-entrapped extracts from niosomes, in order to destroy the membrane of niosomes for releasing encapsulated extract, niosome were lysed by using isopropanol buffer. Then, using spectrophotometric method at the maximum wavelength of each extract, the concentration of the encapsulated extract was calculated by the standard curve of the extract in isopropanol given by equation (1):

(1) EE % : Encapsulated extract in niosomes ( mg/mL ) / Total drug ( mg/mL ) × 100 .

2.5 Investigation of the in vivo release rate of extracts from nanoniosomes in response to temperature and pH stimuli

The temperature and pH of cancer cells (42°C and pH = 4.5 and 5.2) are different from the temperature and pH of normal cells (37°C and pH = 7.4), so temperature and pH can be used as stimulus to target drug delivery in cancer therapy. In this study, in order to investigate the release pattern of extracts from synthesized nanoparticles, the drug release pattern in different conditions in response to stimuli was investigated. To evaluate the release profile of the herbal extracts, 1 mL of the niosomes containing extracts was poured into a cellulose dialysis bag and placed in 10 mL of PBS buffer and stirred at different temperatures and pH (physiological conditions of the body and conditions of cancer cells). Buffer sampling was performed at 1, 2, 4, 6, 8, 24, and 48 h and the concentration of the extract in sample was evaluated by spectrophotometer at the maximum wavelength of each extract and standard curve of each extract at PBS buffer. It should be noted that after each sampling, the same amount of buffer was replaced with the same temperature and pH.

2.6 Investigation of size, dispersion index (PDI), and zeta potential

Using a nano-sizer (Brookhaven Instruments Corporation, Germany), the particle size distribution range as well as the peak particle size range were determined. The range of particle size distribution as well as the diameter of nanoparticles is determined using DLS, for which the Brookhaven Instruments Corporation (Germany) nanosayer was used. For this purpose, laser light was irradiated to the niosomes at a wavelength of 657 nm at angles 90° and 25°C. The zeta potential of nanosystems were measured using an instrument of Brookhaven Instruments Corporation (Germany) at 25°C. 2,000 µL of sample with a concentration of 0.1 mg/mL was used to determine the surface charge.

2.7 Microscopic analysis

Microscopic studies are always a reliable way to study the morphology and structure of nanoparticles. In this study, to analyze the morphology, 3D structure, and shape of nanoparticles, atomic force microscope (AFM) (Nanowizard II; JPK instruments; Germany), scanning electron microscope (SEM) (EM3200-KYKY, China), and Transmission electron microscopy (TEM) (FEI Tecnai 20, type Sphera, Lake Oswego, OR) were used. To prepare the sample for imaging by SEM, 5 µL of the sample was poured on a glass plate and after drying in air, it was covered with a thin layer of gold to provide electrical conductivity. To study the nanoparticles with AFM, the niosomes were diluted with water (1:1,000) and sonicated for 20 min in order to homogenize and reduce the size of the nanoparticles and after that the samples were placed on a mica sheet and imaging was performed. For TEM imaging, samples were placed on 200 Cu-coated grid and imaging was carried out by a cryogenic TEM.

2.8 FTIR evaluation

The functional groups of synthesized nanoniosomes were analyzed by infrared spectroscopy method and were compared with the functional groups of blank niosomes. In the infrared spectrum, two areas are mainly considered. The area from 1,550 to 4,000 cm−1 is the area where most of the bonding tensions occur. This area usually has a relatively small number of peaks, but many of its peaks identify functional groups. To ensure the absence of free drug and additives in the sample, the dialyzed sample was used and in order to reduce the humidity, the sample was placed in an oven at a temperature of approximately 60°C for about half an hour. Also, to investigate the non-interaction of the free form of extracts with niosomes, the spectrum of the extracts and blank niosome were assayed and compared with each other. To investigate the FTIR spectrum, 1 mg of the sample was first added to potassium bromide (KBr) in a ratio of 1:100, and then compressed under a pressure of 5–8 tons/cm, and after the cubes were compressed, the FTIR assay was carried out by FTIR spectrophotometer (Brucker, Germany) at a range of 400–4,000 cm−1.

2.9 Investigating the stability of synthesized nanoniosome

The poor stability of nanoniosomes over time is among main challenges in the application of nanoniosome as a drug delivery carrier. In this study, the EE% (for evaluating the extracts leakage), size maintaining, zeta potential, PDI, and stability of the synthesized nanoniosomes were evaluated in 180 days. For evaluating the stability, synthesized nanoniosomes were kept at 4°C for 180 days and the stability of the mentioned items was investigated at various time intervals by using spectrophotometry and DLS methods as previously described.

2.10 Evaluation of antimicrobial activity of synthesized nanoniosomes

The antibacterial activity of Nio-GGE and Nio-HHE were investigated and compared with free form of extracts, on Escherchia coli and Staphylococcus aureus bacteria obtained from Pasture institute of Iran, Tehran. Then, to evaluate the antibacterial activity of nanosystems, niosomes containing each extract and the free form of the extract were assayed by spotting from 2.5 to 100 µL of nanoparticle suspensions (20 mg/mL) on a Tryptone Soya Agar (TAS) (Merck, Germany) plate seeded with 107 cells/mL, S. aureus, and E. coli, and was incubated at 37°C for 24 h. As positive control, Amplicine and Tetracycline disks (Sigma, USA) were used to evaluate the antibacterial activity of the samples and finally to assay the antibacterial activity of the samples, the non-growth zone around each sample was studied.

2.11 Determination of minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC)

The minimum concentration of an antibacterial agent that is sufficient to prevent bacterial growth in the laboratory is called MIC). In this study, the broth dilution method was used to evaluate the MIC according to the Clinical and Laboratory Standard Institute guideline. In this method, successive dilutions of the niosome containing each extract and the free form of that extract were prepared separately in TSB (Tryptic soy broth; Merck, Germany). Bacterial suspensions were then added to each tube to obtain a final inoculation of 8 logs CFU/mL. Then, the tubes were incubated at 37°C for 24 h. The growth medium alone was a positive control and the growth medium containing bacteria without any antibacterial agent was used as a negative control. Finally, the MIC of each sample was examined using spectrophotometry method at wavelength of 620 nm and the lowest concentration of each sample that inhibited the growth of bacteria was reported as MIC. The lowest concentration of an antimicrobial agent that can kill 99% of the initial inoculated microorganism is called the MBC. In order to determine the MBC, after determining the MIC, 100 µL of suspension was separated from any tube in which bacterial growth was not observed and cultured on a TSA plate and incubated for 24 h at 37°C and after that the number of grown colonies was counted. The concentration at which the counts were reduced to one-thousandth of the initial suspension colonies was then reported as the MBC concentration.

2.12 Cell culture

In this study, MCF-7 and BT-474 cell lines were used as breast cancer cells and MCF-10A cell line was used as normal breast cells, all of which were prepared by Pasteur Institute of Iran and were cultured based on American Type Culture Collection (ATCC) cell culture protocol. Human foreskin fibroblastic (HFF) cell line as normal cells and control group were obtained from Stem cell biology research center, Yazd, Iran. All cells were cultured in Dulbecco’s modified Eagle Medium (DMEM) (Sigma, USA) containing 10% fetal bovine serum (FBS) (Sigma, USA) and penicillin-streptomycin (Gibco, Grand Island, NY). All cultures were performed under standard conditions (37°C, 95% humidity, and 5% CO2 to create and maintain physiological pH).

2.13 In vivo cellular uptake assay

Fluorescence microscopy technique was used to evaluate the cellular uptake of the synthesized nanoniosomes and also to evaluate the intracellular localization of penetrated nanoniosomes and the cellular uptake of nanoniosome in cancer cells compared to normal cells. MCF-7 cells as cancer cells and HFF cells as normal cells were seeded in a 6-wall plate (10 × 105 cells/well) and treated with niosomes containing the extracts and the free form of each extract for 6 h, then rinsed for 3 times using PBS buffer cells and 4% paraformaldehyde (Sigma, USA) solution was used to fix them. Fluorescence property was obtained by adding 0.1% mol of Dil (1,10-dioctadecyl-3,3,30,30-tetramethylindocarbocyanine Perchlorate) (Thermo Fisher Scientific Waltham, MA) to GGE and HHE. 4',6'-diamidino-2-phenylindole (DAPI) (Thermo Fisher Scientific Waltham, MA) (1 mg/mL) was also used to stain cell nuclei and cells’ imaging was done by fluorescence microscope (BX61, Olympus, Japan).

2.14 Cytotoxicity assay

The MTT assay was carried out to evaluate the cytotoxicity effect of the synthesized nanosystems in this study on cancer cells and normal cells. In this study, the effect of Nio-GGE toxicity on breast cancer cells was investigated and compared with its free form and also the cytotoxicity effect of Nio-HHE was evaluated and compared with its free form. Then, the toxicity of Nio-HHE and Nio-GGE on breast cancer cells was compared to select a drug with stronger anti-cancer effect. Finally, the toxicity of the synthesized nanosystems on normal breast cells (MCF 10A) and normal HHF cells was investigated in order to evaluate the side effect of the synthesized nanoniosomes on normal cells. After culturing the cells on 96-well plates and after reaching an approximate number of 10 × 104 cells/per well, the cells were treated with different concentrations (1,000, 750, 500, 250, 125, and 62.5) of each sample separately for 48 h and at the end of this stage, 20 µL of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT salt) (Sigma, USA) (concentration of 0.5 µL) was added to them and were incubated for 4 h. After the supernatant was separated, 150 µL of dimethyl sulfoxide (DMSO) (Sigma, USA) was added to each well to eliminate the formazan crystals and incubation was done for 30 min. The absorbance of each well at wavelength of 570 nm was then read by ELISA reader (Synergy HTX, BioTek, USA) and finally the cell viability rate was calculated.

2.15 Gene expression analysis

In order to investigate the effects of the synthesized niosomes on the expression of genes involved in breast cancer and compare with the effect of free form of extracts on the expression of these genes, the quantitative real-time PCR technique was applied. The genes studied in this study were p53 and MCL-1 genes. Also, β-actin gene was used as housekeeping gene (data normalize). After MCF-7 cells as cancerous cells, and MCF 10A as normal cells, were cultured and treated with Nio-GGE, Nio-HHE, GGE, HHE and blank niosome for 48 h. Total mRNA of each sample was isolated by RiboEx LS reagent (GeneAll, South Korea). cDNA was synthesized from 1 μg of isolated RNA by cDNA synthesized kit (Thermo-Fisher Scientific) according to kit protocol. Real-time PCR amplification was carried out by SYBR Premix Ex Taq II (Takara, Dalian) in 35 cycles based on the following:

• 1–95°C for 5 min, 2–95°C for 30 s, 3–60°C for 30 s, 4–72°C for 30 s, and 5–72°C for 5 min.

The sequences of primers used in RT and real-time PCR are listed in Table 3.

Table 2

Type of surfactant, different molar ratios of non-ionic surfactant l/cholesterol, and type of lipids used in synthesizing different Nio-HHE formulations

Formula Tween60 (%) Span60 (%) Cholesterol (%) DPPC (%) DSPE:PEG (%)
F1 0 75 25 0 0
F2 75 0 25 0 0
F3 0 75 25 10 0
F4 75 0 25 10 0
F5* 75 0 25 10 5

*Optimal formula.

Table 3

Primer sequence used in real-time PCR

Gene Forward primer 5′ to 3′ Reverse primer 5′ to 3′
p53 GGTTTCCGTCTGGGCTTCTT GGGCCAGACCATCGTATC
MCL-1 GCCTCCAGAGAAACGCAGTA TTTCCCGTAGCCAAGAGACG
β-actin AGACGCAGGATGGCATGGG GAGACCTTCAACACCCCAGCC

2.16 Western blot analysis

Bradford assay was used to assay the concentration of proteins. MCF-7 cells were first cultured in a cell culture flask and after reaching 80% confluence were treated with Nio-HHE, Nio-GGE, GGE, HHE, and empty niosomes for 48 h and were lysed by HES buffer (Sigma, USD). Then, 50 µg samples were applied to a 10% SDS-PAGE and then sent into a nitrocellulose membrane. The membranes were incubated for 2 h at 25°C in BSA 5% (Merck, German) in order to barricade non-specific binding. In the next step, the membranes were incubated for 12 h at 4°C with 1:1,000 diluted primary β-actin antibody. Then, by using 1:2,000 diluted horseradish peroxidase-conjugated secondary antibody at 25°C, blots were probed for 2 h and at the end, the visualization were done by applying a visualization system (Syngene GBOX Gel Documentation 680×) along with ECL kit (GE Healthcare).

2.17 Statistical analysis

In this study, all results were gathered with three repetitions and the values were displayed as mean value ± standard deviation for all three repetitions. Also, a student t-test was used to compare two independent groups and one-way ANOVA test was used to compare multiple samples. Data analysis was also performed using GraphPad PRISM version 8 software (GraphPad, San Diego, CA) and EXCEL.

3 Results

3.1 The maximum absorbance wavelength and standard curves

Based on Figure 1, it could be concluded that the λ max for GGE and HHE is at 310 nm and 350 nm, respectively (Figure 1a and d). Figure 1 also demonstrates the standard curves of GGE and HHE in PBS and Isopropyl buffer (Figure 1b, c, e, and f). These data were obtained by analyzing absorbance spectrum of each extract at the range of 200–800 nm. The results of this analysis show that the equation line in isopropyl buffer for GGE and HHE is Y = 0.001725X − 0.005714 and 0.005436X + 0.003732, respectively, and square value (R 2) is 0.9966 and 0.9993, respectively, while the equation line in PBS buffer for GGE and HHE is Y = 0.001219X – 0.003801 and Y = 0.004267X – 0.002001 and R 2 is 0.997 and 0.9968, respectively.

Figure 1 
                  The data obtained from spectrophotometry analysis. (a) Maximum wavelength of GGE is at 310 nm. (b) Standard curve of GGE in Isopropyl buffer, the equation line is Y = 0.001725X − 0.005714 and R
                     2 = 0.9966. (c) Standard curve of GGE in PBS buffer, the equation line is Y = 0.001219X − 0.003801 and R
                     2 = 0.997. (d) Maximum wavelength of HHE is at 350 nm. (e) Standard curve of HHE in Isopropyl buffer, the equation line is Y = 0.005436X + 0.003732 and R
                     2 = 0.9993. (f) Standard curve of HHE in PBS buffer, the equation line is Y = 0.004267X − 0.002001 and R
                     2 = 0.9968.
Figure 1

The data obtained from spectrophotometry analysis. (a) Maximum wavelength of GGE is at 310 nm. (b) Standard curve of GGE in Isopropyl buffer, the equation line is Y = 0.001725X − 0.005714 and R 2 = 0.9966. (c) Standard curve of GGE in PBS buffer, the equation line is Y = 0.001219X − 0.003801 and R 2 = 0.997. (d) Maximum wavelength of HHE is at 350 nm. (e) Standard curve of HHE in Isopropyl buffer, the equation line is Y = 0.005436X + 0.003732 and R 2 = 0.9993. (f) Standard curve of HHE in PBS buffer, the equation line is Y = 0.004267X − 0.002001 and R 2 = 0.9968.

3.2 Choosing the optimal formulation

Tables 4 and 5 show the different effects of molar ratio of surfactant; cholesterol, DPPC, and DSPE:PEG in EE%, maximum extract release in 72 h, and the size of nanoparticle in Nio-GGE and Nio-HHE, respectively. EE% and drug release from nanoparticles play critical roles in selecting the nanoparticle as a drug delivery carrier. Based on Table 1, F6 formula with EE% of 69 ± 1.2%, maximum release of 60.8% at 72 h, and size 111 nm is the optimal formula among other formulation for delivery of GGE to targeted tissue. According to Table 5, the optimal formula for delivery of HHE to cancerous tissue is F5 formula with the size of 97.7 nm, maximum release rate of 63.5 at 72 h, and EE% of 58 ± 2.4%. So, after selecting these formulations as optimal formulations, other analyses were performed on these nanoparticles.

Table 4

Size, EE%, and extract release rate at 24, 48, and 72 h for Nio-GGE

Formula code Size (nm) Encapsulation efficiency ( EE% mean value ± SD) Release % (24 h) Release % (48 h) Release % (72 h)
F1 98 26 ± 2.7 17.2 26.7 35.5
F2 108 24 ± 1.3 15.4 29.3 40.8
F3 95 47 ± 1.5 21.4 29.5 37.6
F4 112 44 ± 0.9 35.4 45.6 53.7
F5 68 58 ± 2.4 40.5 48.9 55.2
F6* 88.8 69 ± 1.2 48.7 54.9 60.8

*Optimal formula.

Table 5

Size, EE%, and extract release rate at 24, 48, and 72 h for Nio-HHE

Formula code Size (nm) Encapsulation efficiency (EE% mean value ± SD) Release % (24 h) Release % (48 h) Release % (72 h)
F1 82.5 17 ± 0.5 18.6 22.7 34.5
F2 105 31 ± 1.5 38.2 49.2 56.2
F3 96.2 30 ± 2.1 34.4 39.5 45.1
F4 76.4 57 ± 0.5 40.8 49.2 57.1
F5* 74.1 58 ± 2.4 46.1 50.9 63.5

*Optimal formula.

3.3 Size, PDI, and electrical charge of nanoparticles

Figure 2 illustrates the size, PDI, and zeta potential of optimal formula after extracting loading. DSPE-PEG reduces the size of nanoparticles and averts their accumulation by increasing the negative charge in nanoparticles and creating a stronger repulsive force. According to Figure 2, the Nio-GGE is anionic type with a zeta potential of −23.5 ± 4.5 mV (Figure 2b), a size of 111 ± 8.5 nm, and PDI of 0.113 (Figure 2a). Based on Figure 2, Nio-HHE is also anionic type with zeta potential of −19.9 ± 6.7 mV (Figure 2d), a PDI of 0.35, and a size of 97.7 nm (Figure 2c).

Figure 2 
                  Investigating the optimal formulation of synthesized niosomes after loading extract in terms of size and zeta potential. (a) Size of Nio-GGE. (b) Zeta potential of Nio-GGE. (c) Size of Nio-HHE. (d) Zeta potential of Nio-HHE. Data obtained from DLS analysis.
Figure 2

Investigating the optimal formulation of synthesized niosomes after loading extract in terms of size and zeta potential. (a) Size of Nio-GGE. (b) Zeta potential of Nio-GGE. (c) Size of Nio-HHE. (d) Zeta potential of Nio-HHE. Data obtained from DLS analysis.

3.4 Microscopy analysis

As mentioned previously, microscopic assay is the best way to examine the morphology of the nanosystems. In this study, AFM, SEM, and TEM were used to study the morphology of the nanoparticles. SEM (Figure 3a and c) shows a spherical shape with a homogeneous size of nanoparticles. The results of this analysis are consistent with the DLS results and confirm its data and, as was anticipated, confirmed the mean nanoparticle diameter measured by the DLS method. AFM data (Figure 3b and d) also show the appropriate 3D structure of the nanoparticles. In general, by examining the microscopic data, it can be concluded that the synthesized nanoparticles have a spherical shape, smooth and uniform surface, and do not have any aggregation. Images taken by TEM microscopy intelligibly show that the synthesized nanoparticles are spherical-elliptical in shape with a size of about 100 nm (Figure 4), which once again confirms the results of DLS.

Figure 3 
                  The microscopy imaging obtained by SEM and AFM; (a) SEM imaging of Nio-GGE. (b) AFM imaging of Nio-GGE. (c) SEM imaging of Nio-HHE. (d) AFM imaging of Nio-HHE.
Figure 3

The microscopy imaging obtained by SEM and AFM; (a) SEM imaging of Nio-GGE. (b) AFM imaging of Nio-GGE. (c) SEM imaging of Nio-HHE. (d) AFM imaging of Nio-HHE.

Figure 4 
                  Image taken by TEM from Nio-GGE. This image shows clearly two-layer structure of niosome and its spherical shape.
Figure 4

Image taken by TEM from Nio-GGE. This image shows clearly two-layer structure of niosome and its spherical shape.

3.5 Evaluation of the interaction between extracts and nanoparticles

FTIR spectrum were obtained in order to investigate the interaction between the extract and synthesized nanoniosome. The FTIR pattern (Figure 5) shows characteristic peaks in the range of 400–4,000 cm−1. The peak at 3,445 cm−1 in fact represents the hydroxyl bands vibrating in stretching and bending motion which repeated with slight difference in Nio-GGE and Nio-HHE, respectively, at 3,437 and 3,228 cm−1. The peak at 2,925 cm−1 is related to the symmetric and asymmetric stretching of carbon of CH3 group, which can be seen in the niosomes’ FTIR patterns after loading the extracts. Also, the peaks at 1,638 and 1,223 cm−1 show the stretching around the C═O axis and the bending at the CH2 axis in lipids and surfactants, which can be traced after loading the extract with a slight difference in the FTIR spectrum of the niosomes. The peak at 1,100 cm−1 is also due to the presence of Tween-60 and DSPE-PEG in the structure of niosomes, which indicates the stretching in the ester and ether groups of these compounds. Most peaks were repeated with a slight difference after the extracts were loaded into the niosomes, a slight difference due to intermolecular forces such as the hydrogen bond in Tween-60. Also, by examining the FTIR spectra, it can be concluded that the extracts and the nanoparticles have retained their chemical nature and integrity after loading, and no effective chemical interaction that changes their natural nature has occurred between them.

Figure 5 
                  FTIR spectra of (a) Nio-GGE and (b) Nio-HHE.
Figure 5

FTIR spectra of (a) Nio-GGE and (b) Nio-HHE.

3.6 The stability of nanoparticles

Figure 6 shows the stability of the synthesized niosomes after 180 days storage at −4°C. Based on Figure 6a, alteration in nanoparticle size during 180 days have been negligible, although nanoniosomes have increased slightly in size after 180 days so these data indicate that these particles are stable in terms of size. Examination of electrical charge changes in the nanoparticles (Figure 6b) also showed that the charge of the synthesized nanosystems tends to become more negative over time with a constant slope. Nanoparticle PDI has also increased over time, although this increase in Nio-HHE has occurred irregularly compared to Nio-GGE (Figure 6c). By examining the amount of extract loaded into the nanoparticles over 180 days, we concluded that GGE leakage from nanosystems occurred more rapidly than HHE leakage from nanosystems, which could be due to the chemical nature of GGE, which is more volatile than HHE (Figure 6d). Although this amount of nanoparticle leakage in this time period can be ignored. Overall, with this evaluation, it can be concluded that the synthesized niosomes have appropriate stability.

Figure 6 
                  The stability analysis of Nio-GGE and Nio-HHE: (a) in terms of size; (b) zeta potential; (c) PDI; and (d) drug leakage over 180 days storage at −4°C.
Figure 6

The stability analysis of Nio-GGE and Nio-HHE: (a) in terms of size; (b) zeta potential; (c) PDI; and (d) drug leakage over 180 days storage at −4°C.

3.7 Antibacterial activity

Antibacterial studies in this study showed that encapsulation of GGE and HHE increased their antibacterial properties. Also, by studying the antibacterial properties of Nio-GGE and Nio-HHE on E. coli and S. aureus bacteria, it can be concluded that Nio-HHE has stronger antibacterial properties compared to Nio-GGE. The results of this comparison are shown in Table 6.

Table 6

Results of antibacterial activity of Nio-HHE and Nio-GGE on E. coli and S. aureus bacteria and their comparison with free form of extracts

Microorganism type Antibacterial material type MIC (µg/mL) MBC (µg/mL) Diameter of zone of inhibition (mm)
Escherchia coli GGE 345 172.5 17
Escherchia coli Nio-GGE 172.5 86.25 14
Staphylococcus aureus GGE 690 345 9
Staphylococcus aureus Nio-GGE 345 172.5 12
Escherchia coli HHE 290 145 16
Escherchia coli Nio-HHE 145 72.5 21
Staphylococcus aureus HHE 290 145 12
Staphylococcus aureus Nio-HHE 145 72.5 19

3.8 Extract release profile

The results of extract release from nanoniosome after 72 h at various pH (4.5, 5.2, and 7.4) and temperatures are presented in Figure 7. Based on Figure 7, it can be concluded that increasing the temperature and decreasing the pH can increase the release of the drug from the nanoparticles, so that the highest release rate in Nio-GGE and Nio-HHE was related to the temperature of 42°C and pH = 4.5. However, by comparing the graphs, it can be concluded that the increase in temperature had a greater effect on drug release than the increase in pH. Accordingly, the highest rate of GGE release from niosomes in 72 h at 42°C and pH 4.5 and at 37°C and pH 7.2 was 71.3 and 50.1%, respectively. This rate was 76.2 and 48.2% for HHE, respectively. Therefore, considering these data, it can be concluded that the synthesized niosomes in this study had temperature-pH sensitivity. Responses of the synthesized nano-niosomes to different pH and temperatures also could be understood from Figure 7. According to this figure, the release of extracts from nanoniosomes occurred in a triphasic pattern, such that in the initial phase, the extract was burst released, in the second phase, it was released slowly, and in the third phase, release occurred close to zero.

Figure 7 
                  Thermo-pH sensivity release of extracts from nanoniosomes assay. (a) Release of GGE from nanosystem at constant temperature and variable pH. (b) Statistical analysis data of GGE release from nanosystems at constant temperature and variable pH. (c) Release of GGE from nanosystem at constant pH and variable temperature. (d) Statistical analysis data of GGE release from nanosystems at constant pH and variable temperature. (e) Release of HHE from nanosystem at constant temperature and variable pH. (f) Statistical analysis data of HHE release from nanosystem at constant temperature and variable pH. (g) Release of HHE from nanosystem at constant pH and variable temperature. (h) Statistical analysis data of HHE release from nanosystems at constant pH and variable temperature. *, **, ***:P-value <0.05.
Figure 7

Thermo-pH sensivity release of extracts from nanoniosomes assay. (a) Release of GGE from nanosystem at constant temperature and variable pH. (b) Statistical analysis data of GGE release from nanosystems at constant temperature and variable pH. (c) Release of GGE from nanosystem at constant pH and variable temperature. (d) Statistical analysis data of GGE release from nanosystems at constant pH and variable temperature. (e) Release of HHE from nanosystem at constant temperature and variable pH. (f) Statistical analysis data of HHE release from nanosystem at constant temperature and variable pH. (g) Release of HHE from nanosystem at constant pH and variable temperature. (h) Statistical analysis data of HHE release from nanosystems at constant pH and variable temperature. *, **, ***:P-value <0.05.

3.9 Cellular uptake

Figure 8 illustrates the successful delivery of Nio-GGE and Nio-HHE to MCF-7 and HFF cells. The in vivo transfection data was obtained by fluorescence microscopy. Based on Figure 8, the optimal nanoparticle had proper ability to deliver the drugs to the target tissue, which can confirm its efficiency in drug delivery to cancer cells.

Figure 8 
                  Intracellular delivery of niosomes containing extracts to MCF-7 cancerous cells and HFF normal cells. The nuclei is counterstained with DAPI (blue fluorescence). Dil dye is used in order to stain the phospholipid (green). Merge is the result of the integration of DAPI and Dil images which indicates the successful entry of nanoparticles into the cells.
Figure 8

Intracellular delivery of niosomes containing extracts to MCF-7 cancerous cells and HFF normal cells. The nuclei is counterstained with DAPI (blue fluorescence). Dil dye is used in order to stain the phospholipid (green). Merge is the result of the integration of DAPI and Dil images which indicates the successful entry of nanoparticles into the cells.

3.10 Cell viability study

Figure 9 demonstrates the cell viability rate of MCF-7, MCF 10A, BT-474, and HFF cell lines after treatment with different doses of GGE, Nio-GGE, HHE, Nio-HHE, and blank niosomes in 48 h. According to Figure 9, encapsulation of extracts in nanoniosome improves their therapeutic properties. The data of this analysis show that HHE IC50 on MCF-7 and BT-474 cancer cells was 264.3 µg/mL and 128 µg, respectively, while after encapsulation in nanoniosomes, Nio-HHE IC50 on these cells decreased to 180.1 and 62.89 µg/mL, respectively, which indicate decrease in the usage dose of the extract and thus increase in its anti-cancer properties after encapsulation. Similarly, the IC50 of GGE on MCF-7 and BT-474 cancer cells was 309.3 and 274.1 µg/mL, while the IC50 of Nio-GGE on these cells was 206.5  and 148 µg/mL, respectively. As mentioned, this decrease in IC50 of the extract indicates improvement in its anti-cancer properties after encapsulation. Also, in order to measure the cytotoxicity of GGE and HHE and compare it with the cytotoxicity of Nio-GGE and Nio-HHE on normal cells, normal HHF and MCF 10A cells were treated with different concentrations of extracts and niosomes containing extracts for 48 h. The results of this study showed the non-toxicity of extracts and nanosystems containing extracts on normal cells. Based on these results, it can be concluded that the use of plant extracts instead of conventional methods in the treatment of cancer can reduce the adverse side effects. To evaluate the compatibility of niosomes with normal body cells and to evaluate their non-toxicity on normal cells, different dilutions of blank niosomes were affected on HFF and MCF 10A normal cell lines, and the results demonstrate the non-toxicity of the synthesized nanosystems on normal cells. It can also be concluded from Figure 9 that Nio-HHE has much stronger anti-tumor properties than Nio-GGE.

Figure 9 
                  Cytotoxicity studies: (a) the cell viability rate (%) of MCF-7 cells after treatment with various concentrations of GGE and Nio-GGE in 48 h. (b) Comparative study of IC50 GGE and Nio-GGE on MCF-7 cancer cells. (c) The cell viability rate (%) of MCF-7 cells after treatment with various concentrations of HHE and Nio-HHE in 48 h. (d) Comparative study of IC50 HHE and Nio-HHE on MCF-7 cancer cells. (e) The cell viability rate (%) of BT-474 cells after treatment with various concentrations of GGE and Nio-GGE in 48 h. (f) Comparative study of IC50 GGE and Nio-GGE on BT-474 cancer cells. (g) The cell viability rate (%) of BT-474 cells after treatment with various concentration of HHE and Nio-HHE in 48 h. (h) Comparative study of IC50 HHE and Nio-HHE on BT-474 cancer cells. (i) The cell viability rate (%) of MCF 10A cells after treatment with various concentrations of GGE and Nio-GGE in 48 h. (j) The cell viability rate (%) of MCF 10A cells after treatment with various concentrations of HHE and Nio-HHE in 48 h. (k) The cell viability rate (%) of HFF cells after treatment with various diluent of blank niosome in 48 h. (l) The cell viability rate (%) of MCF 10A cells after treatment with various diluent of blank niosome in 48 h. ns: no significant difference. *, **, **: P-value <0.05.
Figure 9

Cytotoxicity studies: (a) the cell viability rate (%) of MCF-7 cells after treatment with various concentrations of GGE and Nio-GGE in 48 h. (b) Comparative study of IC50 GGE and Nio-GGE on MCF-7 cancer cells. (c) The cell viability rate (%) of MCF-7 cells after treatment with various concentrations of HHE and Nio-HHE in 48 h. (d) Comparative study of IC50 HHE and Nio-HHE on MCF-7 cancer cells. (e) The cell viability rate (%) of BT-474 cells after treatment with various concentrations of GGE and Nio-GGE in 48 h. (f) Comparative study of IC50 GGE and Nio-GGE on BT-474 cancer cells. (g) The cell viability rate (%) of BT-474 cells after treatment with various concentration of HHE and Nio-HHE in 48 h. (h) Comparative study of IC50 HHE and Nio-HHE on BT-474 cancer cells. (i) The cell viability rate (%) of MCF 10A cells after treatment with various concentrations of GGE and Nio-GGE in 48 h. (j) The cell viability rate (%) of MCF 10A cells after treatment with various concentrations of HHE and Nio-HHE in 48 h. (k) The cell viability rate (%) of HFF cells after treatment with various diluent of blank niosome in 48 h. (l) The cell viability rate (%) of MCF 10A cells after treatment with various diluent of blank niosome in 48 h. ns: no significant difference. *, **, **: P-value <0.05.

3.11 Gene expression

In this study, real-time PCR technique was used to evaluate the effect of GGE and HHE on the expression of p53 and MCL-1 genes as genes involved in the development of breast cancer and compare them with their encapsulated form. Figure 10 illustrates the effect on GGE, HHE, Nio-GGE, Nio-HHE, and blank niosome on the expression of p53 and MCL-1 genes in MCF-7 and MCF 10A cells. According to this figure, after treatment of MCF-7 cells with HHE, the expression of p53 and MCL-1 genes increased by 60% and decreased by 33% compared to the house keeping gene, respectively. While the expression of these genes after treatment of cancer cell with Nio-GGE compared to the control group increased by about 100% and decreased by 49.8%, respectively. Expression of p53 and MCL-1 genes in cancer cells after GGE treatment increased by 38% and decreased by 15% compared to the control group, respectively. While this rate after treatment of cells with Nio-HHE has reached to 86% increase in gene expression and 35% decrease in gene expression, respectively. Blank niosomes had no effect on the expression of these genes in breast cancer cells. On the other hand, by examining the effect of extracts and niosomes containing the extract on the expression of p53 and MCL-1 genes in normal MCF 10A cells, it was found that the extracts and niosomes of the extract had no effect on the expression of genes in normal cells. In other words, the results of this study consistent with the results of MTT assay show that niosomes containing extracts have no side effects on normal cells as non-target tissue.

Figure 10 
                  p53 and MCL-1 gene expression in MCF-7 cells as cancerous cells and in MCF 10A cells as normal cells. (a) Changes in expression of p53 gene after treatment with specific concentration of HHE and Nio-HHE and blank niosome. (b) Changes in expression of p53 gene after treatment with specific concentration of GGE and Nio-GGE and blank niosome. (c) Changes in expression of MCL-gene after treatment with specific concentration of HHE and Nio-HHE and blank niosome. (d) Changes in expression of MCL-1 gene after treatment with specific concentration of GGE and Nio-GGE and blank niosome; ns: no significant difference. *, **: P-value <0.05.
Figure 10

p53 and MCL-1 gene expression in MCF-7 cells as cancerous cells and in MCF 10A cells as normal cells. (a) Changes in expression of p53 gene after treatment with specific concentration of HHE and Nio-HHE and blank niosome. (b) Changes in expression of p53 gene after treatment with specific concentration of GGE and Nio-GGE and blank niosome. (c) Changes in expression of MCL-gene after treatment with specific concentration of HHE and Nio-HHE and blank niosome. (d) Changes in expression of MCL-1 gene after treatment with specific concentration of GGE and Nio-GGE and blank niosome; ns: no significant difference. *, **: P-value <0.05.

3.12 Western blot analysis

In order to investigate the specific effect of GGE, Nio-GGE, HHE, and Nio-HHE on the expression of p53 and MCL-1 proteins, MCF-7 cells were treated with extracts and niosomes containing extracts for 48 h. As displayed in Figure 11, the p53 proteins remarkably increase after treatment with extracts and niosomes containing extract, while the MCL-1 proteins are notably less after treatment. Also, by analyzing western blot data, it could be concluded that the expression of these proteins is more affected by niosomes containing extracts than their free form. Also, the effect of Nio-HHE compared to Nio-GGE on the expression of these proteins in treated cells is significantly higher, which indicates that Nio-HHE is outstandingly effective than Nio-GGE in the treatment of breast cancer.

Figure 11 
                  (a) Changes in expression of p53 protein in MCF-7 cells after treatment with GGE and Nio-GGE. (b) Changes in expression of p53 protein in MCF-7 cells after treatment with HHE and Nio-HHE. (c) Changes in expression of MCL-1 protein in MCF-7 cells after treatment with GGE and Nio-GGE. (d) Changes in expression of MCL-1 protein in MCF-7 cells after tratment with HHE and Nio-HHE. (e) Gel retardation assay to evaluate the genes expression in MCF-7 cells after treatment with GGE and Nio-GGE. (f) Gel retardation assay to evaluate the gene expression in MCF-7 cells after treatment with HHE and Nio-HHE. ns: no significant difference. *, **: P-value <0.05.
Figure 11

(a) Changes in expression of p53 protein in MCF-7 cells after treatment with GGE and Nio-GGE. (b) Changes in expression of p53 protein in MCF-7 cells after treatment with HHE and Nio-HHE. (c) Changes in expression of MCL-1 protein in MCF-7 cells after treatment with GGE and Nio-GGE. (d) Changes in expression of MCL-1 protein in MCF-7 cells after tratment with HHE and Nio-HHE. (e) Gel retardation assay to evaluate the genes expression in MCF-7 cells after treatment with GGE and Nio-GGE. (f) Gel retardation assay to evaluate the gene expression in MCF-7 cells after treatment with HHE and Nio-HHE. ns: no significant difference. *, **: P-value <0.05.

4 Discussion

Herbs and their extracts have been applied as effective treatments for different diseases. However, due to the inefficiency of current treatments and their side effects, namely chemotherapy, usage of these natural agents has been recently considered for the treatment of more complicated diseases such as cancer [26]. Many studies have confirmed the impact of herbal medicine in cancer treatment. In 2017, Telang et al. demonstrated that Dipsacus asperoides can induce apoptosis and arrest cell cycle in breast cancer cells through affecting RAS, PI3K, AKT, and RB signaling pathways [27]. Also, in 2021, research by Samatiwat et al. proved that the methanolic extract of Phyllanthus emblica induces apoptosis in KKU-452 cholangiocarcinoma cells [28]. On the other hand, using herbal medicine and their metabolites brings about many obstacles, to name a few, affecting untargeted tissues or having little impact on the targeted tissue, and in some cases, oxidation of some effective agents in the herbal extract; which in turn obviates the need for a novel strategy for using these compounds. Nanotechnology strives to open up new perspectives for fighting against cancer, the greatest and costliest health problem of the 21st century. This science tries to address the probable challenges and the side effects in cancer treatment through introducing innovative drug delivery systems [24]. Many studies have shown that encapsulation of herbal extract in nanoparticles leads to enhancement of their therapeutic potential. Alemi et al. in 2018 demonstrated that encapsulation of curcumin in PEGylated niosomes promotes its therapeutic effect on MCF-7 breast cancer cells [29]. Also, the study by Taebpour et al. in 2021 showed that the encapsulation of Silybum marianum in nano liposomes enhances its anticancer effects on SAOS bone cancer cells. In this study, nanoniosomes containing Glycyrrhiza glabra and Hedera helix extracts were synthesized and characterized, their anticancer and antimicrobial potential were compared, and their impact on the expression of p53 and MCL-2 was evaluated. Having some antitumor agents including bidesmoside (hederacoside c,b,a) and monodesmosides (α, β, and δ-hederin), Hedera helix along with aglycone hederagenin and also antioxidant compounds such as α-hederin and hederacoside-c can be a potential medicine for cancer [30]. Furthermore, since Licorice contains some antitumor compounds such as licochalcone, isoliquiritigenin, and licochalcone E (a new retrochalcone found in Licorice root), this herb can be applied as a potential anticancer drug [31]. In this research, we proved the anticancer effect of Licorice and Hedera helix extracts on breast cancer cells. As it was mentioned, using nanoparticles as drug delivery carriers gives rise to the enhancement of therapeutic potential of drugs. Meanwhile, many factors such as the type and amount of ingredients, EE%, size, electric charge, the rate of drug released from niosome, and stability of drug nanoparticles could determine the type of nanoparticle applied as a drug carrier [32]. In this study, the EE% of synthesized Nio-GGE and Nio-HHE was 58 ± 2.4 and 69 ± 1.2%, respectively. In fact, EE% refers to drug loading capacity of a nanoparticle, which has a direct correlation with the nanoparticle efficiency. This factor is determined by some elements including the type of surfactant, its ratio to cholesterol, and also the ratio between lipid and drug [33]. For instance, since Tween-60 features large hydrophobic head which creates stronger hydrogen bond with phenolic compounds in extracts, using this reagent compared with span-60 promotes EE%. In addition, having a longer hydrophobic tail, Tween-60 is more capable of trapping the extract than span-60 [33]. Furthermore, the cholesterol content in niosomes plays a leading role in EE%. High levels of cholesterol compete with hydrophobic drugs embedded in two-layer niosomes, causing extra membrane fluidity, and consequently, increases EE%. Nevertheless, using too much cholesterol in niosomes not only does not improve stability, but might make them unstable. Indeed, cholesterol should be kept in balance, since too high or too low cholesterol levels could have an adverse effect on EE% and rate of drug released from niosomes. Using phospholipids in the synthesis of niosomes can also improve their EE%. For instance, DPPC could promote EE% through creating hydrogen bonds with phenolic groups in the extract [34]. In addition, by increasing aqueous space, DSPE-PEG could enhance EE%. This component can also reduce the size of nanoparticles, improve their stability in circulatory system, and increase the solubility of hydrophobic drugs; which in turn enhances drug absorption by cells. Consequently, using DSPE-PEG is considered as a viable solution for instability of niosomes [35]. Our study demonstrated that in terms of size, electric charge, measures of dispersion, and drug release, our optimized formulations are stable for 180 days, which can be the consequence of using DSPE-PEG in the synthesis of niosomes. Drug release also plays a critical role in choosing an appropriate nanoparticle as a drug delivery system, and shows a direct correlation with the efficiency of the nanoparticle. In this research, the maximum drug release rate for Licorice extract from Nio-GGE was 60.8%, and for Nio-HHE was 63.5% in 72 h. As previously mentioned, Tween-60 has a longer hydrophobic tail than Span-60, which in turn could increase drug entrapment, and consequently, reduce drug release. As a result, it may be concluded that an increase in Tween-60/cholesterol ratio in niosome structure can result in a decrease in drug release rate; since not only does the high level of Tween-60 lowers drug release, but low levels of cholesterol also lead to an increase in the rate of drug release. In fact, cholesterol improves the flexibility of cell membrane by disturbing the regular structure of membrane and enhances drug release rate [36]. Also, having unsaturated alkyl chain, DPPC could increase the amount of drug released from nanoparticle, since this agent can improve mobility and flexibility of nanoparticles’ membrane [23]. One of the leading reasons for using nanoniosomes, such as PEG, is that they improve drug efficiency in targeted tissues. Indeed, niosomes can alter drug release rate in response to environmental stimuli such as temperature and pH, which in turn optimizes the use of drugs and reduces probable side effects. A rise in temperature gives rise to niosome membrane permeability, and as a result, drug release rate will rise. Using some compounds in the structure of nanoniosomes, namely PEG, reduces their sensitivity [37]. Acidic pH increases membrane permeability through creating a proton gradient in niosome membrane by protonization of amino groups in chemical components of extracts, and consequently, promotes drug release rate from nanoniosomes. Compared to normal cells, cancer cells have higher temperature and lower pH. These features facilitate differentiation between normal and cancer tissues for nanoniosomes containing drug. In other words, synthesis of pH- and temperature-sensitive niosomes is considered as a viable solution for the lack of differentiation between normal and cancer cells, the most common problem related to chemotherapy and radiotherapy, and could minimize the side effects of these therapies [38]. In this study, we managed to produce pH- and temperature-sensitive niosomes and demonstrated the positive impact of high temperature and low pH on the release of extracts from niosomes. Extract release witnessed a 20 and 30% rise in 42°C and a pH of 4.5 (cancer condition) than physiological condition (37°C and a pH of 7.2). In addition to the aforementioned factors, surface charge of niosomes is also taken into account. Theoretically, extra negative or positive zeta potential of nanoparticles results in their stability through electrostatic repulsion and prevention of aggregation. However, biologically, this sort of nanoparticles are characterized and cleared from body. The usual zeta potential for nanomedical systems ranges between −5 and 15 mV, as most cells show negative zeta potential. A negative zeta potential prevents non-specific interactions with blood components and minimizes the risk of opsonization [39]. In 2008, Zhang et al. demonstrated that the treatment of MCF-7 cells (zeta potential: 20.3 mV) using nanoparticles with the zeta potential of −13.5 resulted in a fall in the cells’ zeta potential by 4.2 mV in 30 min, and over a period of 24 h, reduced to −26.3 mV [40]. This study concluded that these changes resulted from the aggregation of nanoparticles with negative surface charge. TEM images and cellular uptake studies also confirmed the connection between cells and nanoparticles, and demonstrated that nanoparticles enter MCF-7 cells through endocytosis. In addition, endocytosis of negatively charged nanoparticles into cells with negative surface charge could be mediated by some proteins [39]. Therefore, the optimum range for negative charge of nanoparticles could be approximately equal to the surface charge of the targeted cells which would facilitate cell harvesting. In the mentioned research, Nio-HHE and Nio-GGE zeta potentials were −23.5 and −19.5, respectively, which were nearly equal to the zeta potential of normal cells. The size of the synthesized Nio-HHE and Nio-GGE in this research was 111 and 97.5 nm, and their PDI was 0.133 and 0.35, respectively. The size of niosomes is a critical factor as it influences distribution, stability, and excretion of these nanoparticles. Biodegradability of particles without having negative impacts on body must be considered, and it has an inverse relation with the size of the particles. Many studies have shown that huge particles are hardly removed from body. That is to say that liver, kidneys, and spleen are able to remove particles in the size of nearly 100, 6, and 250 nm, respectively [41]. Thus, optimum size and low PDI should be taken into consideration while choosing nanoparticles for drug delivery. The size of niosomes is influenced by several factors including the type of surfactant applied in niosome synthesis. Having a lower hydrocarbon chain volume compared to the hydrophilic surface, Tween-60 has a lower hydrophobic-lipophilic balance (HLB) than span-60. Although studies have demonstrated that the impact of cholesterol on the size of nanoparticles depends on the type of surfactant, high cholesterol could increase hydrophobicity of bilayer membrane of niosomes, and consequently, it reduces the surface free energy and the size of nanoparticles. However, in niosomes with the same content of cholesterol, surfactants with lower HLB produce smaller niosomes [42]. Antibacterial characteristics of Nio-HHE and Nio-GGE were also evaluated in this study, and it was proven that encapsulation of these herbal extracts enhances their antibacterial potential. Khatibi et al. in 2017 demonstrated that encapsulation of Zataria multiflora boiss extract promotes its antibacterial effect on E. coli and reduces its MIC and MBC on the bacteria than unencapsulated extract. Due to their small size, niosomes are able to be absorbed passively. As a result, they enhance the antibacterial effects of the encapsulated drug through reducing mass transfer resistances. The enhancement of antibacterial potential in encapsulated drugs than their free form could be related to the fact that these particles are biologically active. Thus, they can interact with bacteria and cause them to be absorbed through various mechanisms such as fusion, inter-membrane transfer, contact release, and phagocytosis, which in turn promotes their antibacterial features [43]. For instance, Sachetelli et al. in 2000 showed that antibiotic encapsulation through fusion of liposome and bacteria membrane increases membrane permeability for antibiotics, and also by inducing enzymatic hydrolysis between bacteria and liposome membrane, it enhances therapeutic potential of the drug and reduces antibiotic resistance [44]. Tomlinson et al. also proved that the fusion of the liposome containing horseradish peroxidase and E. coli leads to transferring the enzyme to the periplasmic space [45]. However, our findings showed that the antibacterial effect of HHE is stronger than GGE. Secondary metabolites with antibacterial and antioxidant properties, such as saponins, alkaloids, and flavonoids in Licorice confirm its antibacterial potential. Also, the antibacterial features of Hedera helix could be related to some active chemical compounds including hederacoside c, and α- and δ-hederin. This study aimed to enhance HHE and GGE anticancer potential by encapsulating them into niosomes which were synthesized on breast cancer cells. Many studies have confirmed the positive impact of encapsulation on the enhancement of therapeutic potential of drugs. Studies by Hemati et al. and Ghafari et al. proved that encapsulation of chemotherapy agents not only promotes their therapeutic effect, but reduces their side effects and also minimizes cell resistance to these medicines [46,47]. The results of MTT analysis showed that encapsulation of GGE and HHE extracts reduces their IC50 in MCF-7 and BT-474 cells. Comparing this data, it can be concluded that Nio-HHE has stronger anticancer effect than Nio-GGE which could be caused by the chemical nature of extracts. However, it requires more research to be proved. Furthermore, the impact of Blank-Nio on normal cells was investigated and it was proven that the synthesized niosomes did not have toxicity on normal cells and showed biocompatibility with human body. In addition, toxicity analysis of Nio-GGE and Nio-HHE on normal cells proved them as non-toxic nanoparticles, which is considered as the turning point of this research. In other words, cytotoxic assessments confirmed the effect of these nanoparticles on mitigating unpredicted side effects. As it was mentioned before, genes play a crucial role in cancer. This study investigated the impact of encapsulated and unencapsulated extracts of GGE and HHE on the expression of p53 and MCL-1, through which we strived to develop new perspectives for cancer treatment. Our findings showed that Nio-GGE could double the expression of p53 compared to control group, and increase it by 25% in comparison with the free form of the extract. However, these figures for Nio-HHE were nearly 75 and 32%, respectively. Also, this nanoparticle caused a decrease in the expression of MCL-1 by 0.4 and 0.2 compared to the control group and uncapsulated form. Meanwhile, none of the synthesized niosomes influenced MCF 10A gene expression profile, which could be the evidence for a decrease in side effects in using these drugs compared to chemotherapy agents.

5 Conclusion

Our main goal in this study was to introduce a new approach for cancer treatment by combining nanotechnology as modern science with herbal therapy as a traditional science so that we may be able to take a step towards improving the treatment process of one of the most common cancers in all over the world. In this study, we synthesized various formulations of nanoniosomes to deliver GGE and HHE to breast cancer cells, and from among them, we selected optimal formulations with higher EE% and more appropriate release rate of extract. The optimal formulation for GGE delivery to target tissue had EE% 69 ± 1.2% E, maximum release rate of 60.8% in 72 h, size of 111 nm, PDI of 0.113, and zeta potential of −23.5 ± 4.5 mV, while the optimal formulation for targeted delivery of HHE to breast cancer cells had an EE% of 58 ± 2.4%, a maximum release rate of 63.5% in 72 h, a size of 97.7 nm, a PDI of 0.35, and a zeta potential of −19.9 ± 6.7 mV. Physicochemical studies of the synthesized nanosystems showed that these niosomes are suitable in every way for the transfer of extracts to the target tissue. By examining cytotoxicity tests, we found that in addition to successfully penetrating into cells, nanosystems can effectively differentiate between normal and cancer cells to exert their toxicity effect on cancer cells and prevent unwanted side effects. Molecular tests have also shown that the extracts of these plants, with their biologically active substances, can exert their anti-tumor effects by interfering with more complex pathways in the cell, such as intracellular signaling pathways and cell cycle-regulating proteins. In addition, in this study, we perceive that encapsulation of plant extracts can not only increase their anti-tumor properties but can also improve their antibacterial properties. Totally, the results of this study showed that Nio-HHE has a higher toxicity to breast cancer cells than Nio-GGE and can be used as a more potent weapon in the treatment of this malignancy. However, this study, like other studies, had its deficiency, such as lack of investigation of the exact extracts’ action mechanisms on cancerous cells, lack of comparison of nanosystems with other drug nanocarriers, and lack of evaluation of the effect of the nanosystems in clinical stage which are highly recommended to interested researchers.

  1. Funding information: The authors state no funding involved.

  2. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Conflict of interest: The authors state no conflict of interest.

References

[1] Nikbakht A, Kafi M. The history of traditional medicine and herbal plants in Iran, VIII International People-Plant Symposium on Exploring Therapeutic Powers of Flowers. Greenery Nat. 2004;790:255–8.10.17660/ActaHortic.2008.790.37Search in Google Scholar

[2] Triantafyllidi A, Xanthos T, Papalois A, Triantafillidis JK. Herbal and plant therapy in patients with inflammatory bowel disease. Ann Gastroenterol Q Publ Hellenic Soc Gastroenterology. 2015;28(2):210.10.1155/2015/197167Search in Google Scholar

[3] Mansoori B, Mohammadi A, Doustvandi MA, Mohammadnejad F, Kamari F, Gjerstorff MF, et al. Photodynamic therapy for cancer: role of natural products. Photodiagnosis Photodynamic Ther. 2019;26:395–404.10.1016/j.pdpdt.2019.04.033Search in Google Scholar PubMed PubMed Central

[4] Sharma V, Katiyar A, Agrawal R. Glycyrrhiza glabra: chemistry and pharmacological activity. Sweeteners. 2018;87:87–100.10.1007/978-3-319-27027-2_21Search in Google Scholar

[5] Pastorino G, Cornara L, Soares S, Rodrigues F, Oliveira MBP. Liquorice (Glycyrrhiza glabra): a phytochemical and pharmacological review. Phytotherapy Res. 2018;32(12):2323–39.10.1002/ptr.6178Search in Google Scholar PubMed PubMed Central

[6] Lutsenko Y, Bylka W, Matlawska I, Darmohray R. Hedera helix as a medicinal plant. Herba Polonica. 2010;56(1):83–96.Search in Google Scholar

[7] Akhlaghi M, Taebpour M, Sharafaldini M, Javani O, Haghiralsadat BF, Oroojalian F, et al. Fabrication, characterization and evaluation of anti-cancer and antibacterial properties of nanosystems containing Hedera Helix aqueous extracts. Nanomed J. 2022;9(1):43–56.Search in Google Scholar

[8] Sun Y-S, Zhao Z, Yang Z-N, Xu F, Lu H-J, Zhu Z-Y, et al. Risk factors and preventions of breast cancer. Int J Biol Sci. 2017;13(11):1387.10.7150/ijbs.21635Search in Google Scholar PubMed PubMed Central

[9] Duffy MJ, Synnott NC, Crown J. Mutant p53 in breast cancer: potential as a therapeutic target and biomarker. Breast Cancer Res Treat. 2018;170(2):213–9.10.1007/s10549-018-4753-7Search in Google Scholar PubMed

[10] Young AI, Law AM, Castillo L, Chong S, Cullen HD, Koehler M, et al. MCL-1 inhibition provides a new way to suppress breast cancer metastasis and increase sensitivity to dasatinib. Breast Cancer Res. 2016;18(1):1–15.10.1186/s13058-016-0781-6Search in Google Scholar PubMed PubMed Central

[11] Núñez C, Capelo JL, Igrejas G, Alfonso A, Botana LM, Lodeiro C. An overview of the effective combination therapies for the treatment of breast cancer. Biomaterials. 2016;97:34–50.10.1016/j.biomaterials.2016.04.027Search in Google Scholar PubMed

[12] Nedeljković M, Damjanović A. Mechanisms of chemotherapy resistance in triple-negative breast cancer – how we can rise to the challenge. Cells. 2019;8(9):957.10.3390/cells8090957Search in Google Scholar PubMed PubMed Central

[13] Naghib SM, Zare Y, Rhee KY. A facile and simple approach to synthesis and characterization of methacrylated graphene oxide nanostructured polyaniline nanocomposites. Nanotechnol Rev. 2020;9(1):53–60.10.1515/ntrev-2020-0005Search in Google Scholar

[14] Kazemi F, Naghib SM, Mohammadpour Z. Multifunctional micro-/nanoscaled structures based on polyaniline: an overview of modern emerging devices. Mater Today Chem. 2020;16:100249.10.1016/j.mtchem.2020.100249Search in Google Scholar

[15] Kazemi F, Naghib SM, Zare Y, Rhee KY. Biosensing applications of polyaniline (PANI)-based nanocomposites: a review. Polym Rev. 2021;61(3):553–97.10.1080/15583724.2020.1858871Search in Google Scholar

[16] Khatibi SA, Misaghi A, Moosavy M-H, Basti AA, Koohi MK, Khosravi P, et al. Encapsulation of Zataria multiflora Boiss. Essential oil into nanoliposomes and in vitro antibacterial activity against Escherichia coli O157:H7. J Food Process Preservation. 2017;41(3):e12955.10.1111/jfpp.12955Search in Google Scholar

[17] Mirhosseini M, Shekari-Far A, Hakimian F, Haghiralsadat BF, Fatemi SK, Dashtestani F. Core-shell Au@Co-Fe hybrid nanoparticles as peroxidase mimetic nanozyme for antibacterial application. Process Biochem. 2020;95:131–8.10.1016/j.procbio.2020.05.003Search in Google Scholar

[18] Malekimusavi H, Ghaemi A, Masoudi G, Chogan F, Rashedi H, Yazdian F, et al. Graphene oxide-l-arginine nanogel: a pH-sensitive fluorouracil nanocarrier. Biotechnol Appl Biochem. 2019;66(5):772–80.10.1002/bab.1768Search in Google Scholar PubMed

[19] Abtahi NA, Naghib SM, Haghiralsadat F, Reza JZ, Hakimian F, Yazdian F, et al. Smart stimuli-responsive biofunctionalized niosomal nanocarriers for programmed release of bioactive compounds into cancer cells in vitro and in vivo. Nanotechnol Rev. 2021;10(1):1895–911.10.1515/ntrev-2021-0119Search in Google Scholar

[20] Yaghoubi F, Naghib SM, Motlagh NSH, Haghiralsadat F, Jaliani HZ, et al. Multiresponsive carboxylated graphene oxide-grafted aptamer as a multifunctional nanocarrier for targeted delivery of chemotherapeutics and bioactive compounds in cancer therapy. Nanotechnol Rev. 2021;10(1):1838–52.10.1515/ntrev-2021-0110Search in Google Scholar

[21] Abtahi NA, Naghib SM, Ghalekohneh SJ, Mohammadpour Z, Nazari H, Mosavi SM, et al. Multifunctional stimuli-responsive niosomal nanoparticles for co-delivery and co-administration of gene and bioactive compound: In vitro and in vivo studies. Chem Eng J. 2022;429:132090.10.1016/j.cej.2021.132090Search in Google Scholar

[22] Mazidi Z, Javanmardi S, Naghib SM, Mohammadpour Z. Smart stimuli-responsive implantable drug delivery systems for programmed and on-demand cancer treatment: An overview on the emerging materials. Chem Eng J. 2022;433:134569.10.1016/j.cej.2022.134569Search in Google Scholar

[23] Hemati M, Haghiralsadat F, Jafary F, Moosavizadeh S, Moradi A. Targeting cell cycle protein in gastric cancer with CDC20siRNA and anticancer drugs (doxorubicin and quercetin) co-loaded cationic PEGylated nanoniosomes. Int J Nanomed. 2019;14:6575–85.10.2147/IJN.S211844Search in Google Scholar PubMed PubMed Central

[24] Akhlaghi M, Ebrahimpour M, Ansari K, Parnian F, Zarezadeh Mehrizi M, Taebpour M. Synthesis, study and characterization of nano niosomal system containing Glycyrrhiza glabra extract in order to improve its therapeutic effects. N Cell Mol Biotechnol J. 2021;11(42):65–82.10.34172/ddj.2022.09Search in Google Scholar

[25] 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;2018:6847971.10.1155/2018/6847971Search in Google Scholar PubMed PubMed Central

[26] Meacham E, Orem J, Nakigudde G, Zujewski JA, Rao D. Exploring stigma as a barrier to cancer service engagement with breast cancer survivors in Kampala, Uganda. Psycho‐Oncology. 2016;25(10):1206–11.10.1002/pon.4215Search in Google Scholar PubMed

[27] Telang N, Nair HB, Wong G. Efficacy of Dipsacus asperoides (DA) in a model for triple negative breast cancer. Cancer Res. 2017;77(4 Suppl):4–13.10.1158/1538-7445.SABCS16-P4-13-04Search in Google Scholar

[28] Samatiwat P, Chankhonkaen P, Jaisin Y, Ratanachamnong P, Niwaspragrit C, Rungsiwiwut R, et al. Anticancer activity of the bark extract of Phyllanthus emblica on cholangiocarcinoma in vitro. J Basic Appl Pharmacology. 2021;1(1):60–71.Search in Google Scholar

[29] 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):1–20.10.1186/s12951-018-0351-4Search in Google Scholar PubMed PubMed Central

[30] Majester-Savornin B, Elias R, Diaz-Lanza A, Balansard G, Gasquet M, Delmas F. Saponins of the ivy plant, Hedera helix, and their leishmanicidic activity. Planta Medica. 1991;57(3):260–2.10.1055/s-2006-960086Search in Google Scholar PubMed

[31] Yoon G, Do Jung Y, Cheon SH. Cytotoxic allyl retrochalcone from the roots of Glycyrrhiza inflata. Chem Pharm Bull. 2005;53(6):694–5.10.1248/cpb.53.694Search in Google Scholar PubMed

[32] Akhlaghi M, Taebpour M, Sharafaldini M, Javani O, Haghiralsadat BF, Oroojalian F, et al. Fabrication, characterization and evaluation of anti-cancer and antibacterial properties of nanosystems containing Hedera Helix aqueous extracts. Nanomed J. 2022;9(1):43–56.Search in Google Scholar

[33] Mokhtar M, Sammour OA, Hammad MA, Megrab NA. Effect of some formulation parameters on flurbiprofen encapsulation and release rates of niosomes prepared from proniosomes. Int J Pharmaceutics. 2008;361(1–2):104–11.10.1016/j.ijpharm.2008.05.031Search in Google Scholar PubMed

[34] Gunay MS, Ozer AY, Erdogan S, Bodard S, Baysal I, Gulhan Z, et al. Development of nanosized, pramipexole-encapsulated liposomes and niosomes for the treatment of Parkinson’s disease. J Nanosci Nanotechnol. 2017;17(8):5155–67.10.1166/jnn.2017.13799Search in Google Scholar

[35] 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):3161–73.10.1016/j.apt.2021.05.047Search in Google Scholar

[36] Taymouri S, Varshosaz J. Effect of different types of surfactants on the physical properties and stability of carvedilol nano-niosomes. Adv Biomed Res. 2016;5:178781.10.4103/2277-9175.178781Search in Google Scholar

[37] Baranei M, Taheri RA, Tirgar M, Saeidi A, Oroojalian F, Uzun L, et al. Anticancer effect of green tea extract (GTE)-loaded pH-responsive niosome coated with PEG against different cell lines. Mater Today Commun. 2021;26:101751.10.1016/j.mtcomm.2020.101751Search in Google Scholar

[38] Tila D, Yazdani-Arazi SN, Ghanbarzadeh S, Arami S, Pourmoazzen Z. pH-sensitive, polymer modified, plasma stable niosomes: promising carriers for anti-cancer drugs. EXCLI J. 2015;14:21.Search in Google Scholar

[39] Honary S, Zahir F. Effect of zeta potential on the properties of nano-drug delivery systems-a review (Part 1). Tropical J Pharm Res. 2013;12(2):255–64.10.4314/tjpr.v12i2.19Search in Google Scholar

[40] Zhang Y, Yang M, Portney NG, Cui D, Budak G, Ozbay E, et al. Zeta potential: a surface electrical characteristic to probe the interaction of nanoparticles with normal and cancer human breast epithelial cells. Biomed microdevices. 2008;10(2):321–8.10.1007/s10544-007-9139-2Search in Google Scholar PubMed

[41] 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 IJPR. 2018;17(Suppl2):1.Search in Google Scholar

[42] Agarwal S, Bakshi V, Vitta P, Raghuram A, Udupa N. Effect of cholesterol content and surfactant HLB on vesicle properties of niosomes. Indian. J Pharm Sci. 2004;66(1):121.Search in Google Scholar

[43] Khatibi SA, Misaghi A, Moosavy MH, Basti AA, Koohi MK, Khosravi P, et al. Encapsulation of Zataria multiflora Boiss. Essential oil into nanoliposomes and in vitro antibacterial activity against Escherichia coli O157: H7. J Food Process Preservation. 2017;41(3):e12955.10.1111/jfpp.12955Search in Google Scholar

[44] Sachetelli S, Khalil H, Chen T, Beaulac C, Sénéchal S, Lagacé J. Demonstration of a fusion mechanism between a fluid bactericidal liposomal formulation and bacterial cells. Biochimica et Biophysica Acta (BBA)-Biomembranes. 2000;1463(2):254–66.10.1016/S0005-2736(99)00217-5Search in Google Scholar

[45] Tomlinson S, Taylor PW, Luzio JP. Transfer of phospholipid and protein into the envelope of gram-negative bacteria by liposome fusion. Biochemistry. 1989;28(21):8303–11.10.1021/bi00447a007Search in Google Scholar PubMed

[46] Hemati M, Haghiralsadat F, Yazdian F, Jafari F, Moradi A, Malekpour-Dehkordi Z. Development and characterization of a novel cationic PEGylated niosome-encapsulated forms of doxorubicin, quercetin and siRNA for the treatment of cancer by using combination therapy. Artif Cells, Nanomedicine, Biotechnol. 2019;47(1):1295–311.10.1080/21691401.2018.1489271Search in Google Scholar PubMed

[47] Ghafari M, Haghiralsadat F, Khanamani Falahati-pour S, Zavar, Reza J. Development of a novel liposomal nanoparticle formulation of cisplatin to breast cancer therapy. J Cell Biochem. 2020;121(7):3584–92.10.1002/jcb.29651Search in Google Scholar PubMed

Received: 2021-07-24
Revised: 2021-09-11
Accepted: 2022-02-17
Published Online: 2022-03-24

© 2022 Milad Akhlaghi et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 11.5.2024 from https://www.degruyter.com/document/doi/10.1515/ntrev-2022-0080/html
Scroll to top button