Next Article in Journal
Dynamic Susceptibility Contrast Magnetic Resonance Imaging with Carbon-Encapsulated Iron Nanoparticles Navigated to Integrin Alfa V Beta 3 Receptors in Rat Glioma
Previous Article in Journal
In Situ TEM Observation of Electric Field-Directed Self-Assembly of PbS and PbSe Nanoparticles
Previous Article in Special Issue
Comparison In Vitro Study on the Interface between Skin and Bone Cell Cultures and Microporous Titanium Samples Manufactured with 3D Printing Technology Versus Sintered Samples
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Green Synthesis of Chitosan-Coated Selenium Nanoparticles for Paclitaxel Delivery

1
Laboratory of Materials, Crystal Chemistry and Applied Thermodynamics, Faculty of Sciences of Tunis, University of Tunis El Manar, El Manar II, Tunis 2092, Tunisia
2
Department of Chemistry, College of Science, University of Anbar, Ramadi 31001, Iraq
3
Laboratory of Transmission, Control and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, 13 Place Pasteur, 1002 Tunis le Belvedere, Tunis 2092, Tunisia
4
Preparatory Institute for Engineering Studies of Tunis, University of Tunis, Jawaharlal Nehru Street, 1089, Montfleury, Tunis 2092, Tunisia
5
Institute for Inorganic and Materials Chemistry, Department of Chemistry and Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Greinstrasse 6, 50939 Köln, Germany
*
Authors to whom correspondence should be addressed.
Nanomaterials 2025, 15(16), 1276; https://doi.org/10.3390/nano15161276
Submission received: 27 June 2025 / Revised: 30 July 2025 / Accepted: 14 August 2025 / Published: 18 August 2025

Abstract

Selenium nanoparticles (Se NPs) were synthesized from Na2SeO3 using Foeniculum vulgare (fennel) seed extract as mild sustainable reductant, coated with chitosan (Ch), and loaded with Paclitaxel (PTX). The PTX release from the Se@Ch–PTX NPs and their cytotoxicity against MDA-MB-231 breast cancer cells was studied in view of an application as drug delivery platform. Thermogravimetric analysis (TGA) showed the thermal stability of the NPs up to 300 °C. UV–vis absorption and Fourier transform IR (FT-IR) spectroscopy allowed to trace surface species originating from the F. vulgare extract on the Se NPs, while the surface of the Se@Ch–PTX NPs is characterized from Ch and PTX functionalities. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) showed approximate spherical shaped NPs with sizes ranging from 10 to 40 nm. Zeta potential measurements showed a clear distinction between the −39 mV found the Se NPs and +57 mV for the Ch–PTX coated NPs. The NPs showed good biocompatibility with red blood cells (RBCs) in hemolytic activity assays, exhibiting no hemolytic effects at concentrations ranging from 50 to 400 µg/mL. In vitro release studies showed a sustained and pH-responsive release pattern with a maximum release of about 80% within 22 h for Se@Ch–PTX at pH = 3.5. The Se@Ch–PTX NPs showed high antiproliferative activity against MDA-MB-231 cells with an IC50 value of 12.3 µg/mL compared to about 36 for PTX and 52 µg/mL for the Se NPs. The reactive oxygen species (ROS) activity as studied through DPPH scavenging showed higher values for the Se@Ch–PTX NPs compared to the Se NP.

Graphical Abstract

1. Introduction

Breast cancer is the most common type of cancer among women worldwide, posing a significant public health threat with millions of cases annually [1,2,3]. Established treatments, including surgery, radiotherapy, and chemotherapy, face significant challenges, including severe side effects, limited selectivity in targeting cancer cells, and the development of drug resistance [4,5].
Paclitaxel (PTX) is a very prominent chemotherapeutic agent used in breast cancer treatment [6,7,8,9,10]. Initially discovered in the bark of the Pacific yew tree, PTX is known for its high cytotoxicity against cancer cells [9]. However, its use is associated with serious side effects, including neurotoxicity, poor bioavailability, and adverse effects on healthy cells, which limit its therapeutic efficacy [11,12]. Consequently, enhancing the efficiency of PTX and thus minimizing side effects through reduced doses is an important objective in cancer therapy research [13,14].
Nanoparticles (NPs) represent an innovative and promising approach as drug-delivery systems in cancer treatment, as NP can be functionalized to enhance solubility and bioavailability, and even allow the targeting of cancer cells [15,16,17,18,19]. Recent studies have shown that selenium NPs (Se NPs) are not only good candidates for NP-based drug delivery [19,20,21,22,23,24,25,26,27,28,29,30], but that Se additionally possesses anticancer properties by promoting oxidative stress in cancer cells through the production of reactive oxygen species (ROS) and thus inducing apoptosis [20,22,23,24]. Additionally, Se NPs are biocompatible and exhibit relatively low toxicity toward healthy cells, making them good candidates for cancer therapy [23,24].
A number of studies has thus used Se NPs for PTX delivery [25,26,27,28,29,30,31]. In a recent study, the nanocomposite Se@β-CD–FA–PTX was produced from Se NPs, β-cyclodextrin (β-CD) and folic acid (FA), using a layer-by-layer assembly method, and loaded with PTX. This system is capable of entering cancer cells through folate receptor-mediated endocytosis, enabling targeted intracellular drug delivery [27]. In another study, Se NPs conjugated with hyaluronic acid and loaded with PTX (Se@HA–PTX) were studied and showed antiproliferative activities against A549 lung cancer cells by enhancing cellular uptake through HA receptor-mediated mechanisms [28]. Furthermore, Se NPs were loaded with PTX using Pluronic F-127 as a stabilizer and their anticancer activity was evaluated against various cancer cell lines (A549, MCF-7, HeLa, HT29) [29]. The induction of G2/M phase cell cycle arrest and the promotion of apoptosis via mechanisms involving mitochondrial membrane potential disruption increased reactive oxygen species (ROS) production, and caspase activation was studied and discussed as possible mode of action [29].
In a number of reports, Se NPs for various purposes were produced from SeO2 or selenite using biological material as mild sustainable reductant [23,26,29,30,31,32,33,34,35,36,37,38,39,40,41,42]. FT-IR and UV–vis absorption spectroscopy of thus produced Se NPs clearly show that the reducing biomaterials have left a plethora of functional groups on the Se surface as protecting ligands. These surface ligands probably contribute also to the biological activities of such Se NPs [30,31,32,33,34,35,37,42].
On the other hand, an important strategy in NP-based drug delivery is the use of defined polymeric materials that allow blending with drug molecules and slow, controlled release of drug molecules through slow degradation of the surface polymer [43]. Chitosan, a natural polymer derived from chitin, is known for its biocompatibility and non-toxic properties [44,45,46]. Its positive charge facilitates strong interactions with negatively charged cancer cell membranes, potentially enhancing cellular uptake [46,47,48].
Herein we report on the develop of a PTX drug-delivery system based on chitosan-covered Se NP, Se@Ch–PTX. We used the aqueous extract of Foeniculum vulgare (fennel) seeds to produce Se NPs, covered them with chitosan and loaded them with PTX (Scheme 1). We studied the PTX-release, the antiproliferative activity against MDA-MB-231 breast cancer cells, and the ROS activity. F. vulgare seed extract was chosen as mild reductant. Fennel represents as important medicinal plant widely used in traditional medicine for its therapeutic benefits [49,50,51,52,53,54]. It contains bioactive compounds such as anethole, fenchone, and estragole, which showed antioxidant, anti-inflammatory, and antibacterial properties [51,52]. Given its antioxidant properties, F. vulgare extract should be suitable as bio-reductant for the production of Se NPs and at the same time provide bio-active surface ligands [52,54].

2. Results and Discussion

2.1. Synthesis

Red Se NPs were synthesized from Na2SeO3 and F. vulgare seed extract as mild and sustainable (green) reductant at 30 °C in 60 min with a yield of 87%. The NPs were stabilized using phosphate-buffered saline (PBS) solution at pH = 7.4. The chitosan-coated NPs (Se@Ch) were synthesized by adding a chitosan glacial acetic acid mixture to the Se NPs. Paclitaxel (PTX) was loaded to the Se@Ch NP by stirring them in a PTX DMSO solution. The Se@Ch–PTX NPs were produced in 0.8 g quantity and can be stored in de-aerated PBS solution (pH = 7.4) under the exclusion of light for at least one week.

2.2. Analytic and Spectroscopic Characterization

The FT-IR spectrum of the F. vulgare extract (Figure S1 in the Supplementary Materials) shows characteristic bands at 3450, 2963, 1626, and 1123 cm−1 that have been previously reported for such materials [55]. The FT-IR of the Se NPs are quite similar to those of the extract, in keeping with our intention that characteristic components of the extract cover the Se NPs as stabilizing ligands. In the FT-IR spectrum of the Se@Ch–PTX, a marked shift of the broad band centered around 3400 cm−1 and a broad band at around 600 cm−1 point to the presence of Ch and PTX on the particles. Unfortunately, the spectrum does provide any information about the way PTX is loaded onto the Se@Ch NPs and if F. vulgare components are still present in the Ch–PTX shell around the Se NPs.
In the UV–vis absorption spectrum (Figure S2) the F. vulgare extract shows a broad band peaking at 245 nm. The Se NPs are characterized by a maximum at 280 nm shoulders at 360, 450, and 600 nm, in keeping with previous reports [56,57,58]. For the Se@Ch–PTX NPs three distinct absorptions were recorded at 269, 355, and 448 nm, which represents a slight variation in the Se NP absorption and is probably due to the Ch–PTX functionalization [59,60].
The powder X-ray diffractogram of the Se@Ch–PTX NPs (Figure S3) shows the characteristic signals of highly crystalline Se in its hexagonal structure, in line with previous reports [39,40,61] and in agreement with the standard [61]. Using the Debye–Scherrer equation, the nanoparticle size of the synthesized particles was calculated to 33.6 nm. Additional peaks indicate the presence of PTX and chitosan in the coating, but also do not provide any information on the binding of PTX and Ch on the Se surface.
The Thermogravimetric Analysis (TGA) of the Se@Ch–PTX NPs shows a subtle weight loss up to 300 °C indicating loss of water molecules attached to the surface (Figure S4). From 300 to 420 °C the NPs underwent a loss of more than 50% of their weight, leaving a material that is stable up to 640 °C and then loses further 30% weight up to 750 °C, leaving a residual of 11% of the original mass.
The Zeta potential of the Se NPs was measured to −38.29 mV (Figure S5), in line with the presence of weakly bound surface ligands from the extract. Functionalization with chitosan and PTX led to a Zeta potential of +56.66 mV, which is probably due to the surface of the particles being completely covered by chitosan and PTX. The same behavior has previously been reported for Se@β-CD–FA@PTX (+30 mV) and Se@β-CD–FA NPs (+37 mV; FA = folate, β-CD = β-cyclodextrin), in contrast to pristine Se NPs (−25 mV) [27].

2.3. Scanning Electron (SEM) and Transmission Electron (TEM) Microscopy

The SEM of the Se NPs (Figure 1a) showed approximately spherical shape NPs separated with clear boundaries, a rough granular surface, and an average size of 25.4 ± 9.3 nm. The TEM (Figure 1b) confirmed the spherical shape of the nanoparticles, with particle sizes ranging from 10 to 40 nm.
For the Se@Ch–PTX NPs, a far more pronounced spherical morphology was observed in the SEM (Figure 2a), with a markedly smaller size distribution ranging from 20 to 30 nm. The TEM (Figure 2b) confirms their spherical shape and size distribution. The transparent layer surrounding the particles represents likely the chitosan/PTX covering shell, confirming the complete coverage of the Se NP concluded from the Zeta potential measurements.
The analysis of the NP materials show that chitosan effectively coated the surface of the Se NPs, resulting in the formation of a stable core@shell structure as reported before [62]. Furthermore, chitosan contributes to nanoparticle stabilization against agglomeration through its highly positive Zeta potential of +56.66 mV. This observation aligns with previous reports demonstrating that chitosan significantly enhances the colloidal stability of metal-based nanoparticles through surface charge modification and steric protection [63,64].

2.4. In Vitro Drug Release

A rather controlled release of up to 80% PTX from Se@Ch–PTX NPs was found within 22 h at pH = 3.5 (Figure 3a). At higher pH = 7.4, and 9.0, the release was slower and lower of up to 62% at pH = 7.4 and 57% at pH = 9.
The release kinetics showed high conformity with the Korsmeyer–Peppas model (Figure 3d), with correlation coefficients R2 exceeding 0.98 (Table 1). This model is characteristic of drug release through a combined mechanism involving both diffusion and degradation of the biopolymer coating [43,53,65]. The agreement to the Higuchi model (Figure 3c) is only slightly lower. This model is applicable if diffusion plays the primary role [37]. In contrast to this, the zero-order release model (Figure 1a) showed quite moderate suitability, with R2 values ranging between 0.85 and 0.9, reflecting good control over the release rate over time and sustained drug release and also indicates system stability [38]. The first-order release model (Figure 1b) gave the poorest fit, confirming that drug release is influenced not only by the remaining drug concentration but also by factors such as surface composition and coating nature [66].
The release rates of our materials are markedly lower compared to those of very related Se@Ch–PTX NPs, for which a release of about 80% after 55 h at pH 5.5 and about 15% after 55 h at pH 7.4 was reported [26]. The reason for the higher release lies very probably in the much larger size of these NPs of about 140 nm, compared with the approximately 30 nm of our NPs. An interesting comparison was drawn for C-peptide-conjugated solid lipid nanoparticles as carriers for PTX [67]. While the C-peptide–SLN–PTX NPs showed controlled release of about 80% after 60 h at pH 5.5, the SLM-PTX material delivered 80% in 50 h, and PTX NPs within 5 h. These two studies and others show a strong dependence of the release on pH [26,27,28,67], thus confirming our findings.

2.5. Hemolysis Test

Treatment of red blood cells (RBCs) with the Se@Ch–PTX NPs showed only marginal hemolysis at 400 µg/mL (Figure 4a).
In contrast to this, a 100 µg/mL concentration of Triton-X-100 resulted in 100% hemolysis, which agrees with the membrane-disrupting nature of this detergent. Furthermore, aPTT (activated partial thromboplastin time) and PT (prothrombin time) assays showed that concentrations of Se@Ch–PTX NPs, ranging from 50 to 400 µg/mL did not cause any significant change (Figure 4b). The coagulation times remained within the normal physiological ranges for both PT (9.4–12.5 s) and aPTT (25.1–36.5 s), according to a reference [68].

2.6. Antiproliferative Properties

The anti-proliferative activity of the Se and Se@Ch–PTX NPs were evaluated in vitro on the MDA-MB-231 triple-negative breast cancer cell line using the MTT assay (Figure 5). The cell viability decreased with increasing concentrations of the Se@Ch–PTX NPs showing superior activity over PTX and over the Se NPs (Figure 5a). The IC50 value for Se@Ch–PTX of 14.3 µg/mL (Figure 5b, Table 2) recorded after 24 h is markedly higher than that of the components PTX (36.2 µg/mL) and Se NPs (52.4 µg/mL). It is noteworthy, that PTX was applied in equivalent mg amounts as the NPs in these experiments, which leads to a far higher overall PTX-content for the pure PTX material compared with the Se@Ch–PTX NPs. Which means that the use of the Se@Ch-based release system is superior to the application of pure PTX. The efficient release from the Se@Ch–PTX NPs aligns to the morphological changes observed in microcopy (Figure S6). Our findings agree very well with those of the quite similar Se NP-based PTX-release system Se@β-CD–FA@PTX (FA = folate, β-CD = β-cyclodectrin) [27], which showed markedly improved antiproliferative activity over the Se@β-CD–FA component or the Se NPs prepared from Na2SeO3 and ascorbic acid. The same has previously been reported for C-peptide-conjugated solid lipid NPs with an IC50 value of 1.0 µg/mL (Table 2) [67]. Our system is less active than this system, but can easily compete with other Se-based NP delivery systems (Table 2). Generally, Table 2 demonstrates that suitable coating of the Se NPs before loading them with PTX or co-loading other organic material improves the release and cytotoxicity of the NP materials.
Our findings and those of others (Table 2) also reveal that unloaded Se NPs show some antiproliferative activities. Presumably, the Se NPs contribute to the generation of reactive oxygen species (ROS), which play a key role in cytotoxic activity by overcoming the antioxidant defense mechanisms in tumor cells [24,27,28,29,58,69,70]. In our case, the activity might as well be enhanced by flavonoid and phenolic compounds present on the surface of the Se NPs originating from the F. vulgare seed extract. Therefore, the lower Se-content of the Se NPs compared with the Se@Ch–PTX (we used equivalent mg amounts) will probably not play a significant role. Our assumption aligns well with use of certain plant extracts leads to the formation of small-sized Se NPs, significantly enhancing cellular toxicity [33,34,42]. As many reported Se NPs for drug-delivery were synthesized using plant extracts or other biomaterials as reductants, the possibility that the biomaterials are far from being just “innocent” reductants requires a comparative study using drug-loaded Se NP produced using different biomaterials. We are planning such a study for the future.

2.7. Antioxidant Activities

The antioxidant potential of the Se NPs and the Se@Ch–PTX NPs was evaluated through a DPPH assay, using ascorbic acid as reference compound. All three samples showed concentration-dependent scavenging of DPPH free radicals (Figure 6). At the highest concentration of 50 mg/mL, ascorbic acid showed the highest activity with about 81% followed by the Se@Ch–PTX with 76% and the Se NPs with 71% (Table 3). Although ascorbic acid showed superior activity, the NPs demonstrated promising antioxidant potential, particularly Se@Ch–PTX, which consistently outperformed the Se NPs across all tested concentrations. This enhanced performance is further supported by the IC50 values, with the Se@Ch–PTX NPs showing higher activity (34 mg/mL) than the Se NPs (39 mg/mL), though being still less active than ascorbic acid (22 mg/mL) (Table 3).
Our values agree largely with those of similar materials in previous reports [25,27,42,57,69].
Overall, the chitosan-coating of our Se@Ch–PTX NPs seems to play a pivotal role in drug loading and release of PTX. This is probably due to the presence of amino and hydroxyl functional groups, which facilitate the encapsulation of PTX via electrostatic and hydrophobic interactions. These interactions not only improve drug loading efficiency but might also contribute to a sustained release profile as has been pointed out before [59,60,62,63,64]. Collectively, the multifunctional role of chitosan includes nanoparticle stabilization, enhanced drug-loading capacity, and favorable surface characteristics that improve drug release. Potentially, chitosan also improves cellular uptake, but we have not studied this point herein.

3. Materials and Methods

3.1. Materials

Sodium selenite Na2SeO3 (>90%RT, Sigma Aldrich, Merck, Darmstadt, Germany), Paclitaxel (PTX, C47H51NO14, MW = 853.91 g/mol; Merck, Darmstadt, Germany), chitosan ((C6H11O4N)n, 684 kDa, deacetylation approx. 84%, Merck, Darmstadt Germany), Triton-X-100 (Sigma Aldrich), and dimethyl sulfoxide (DMSO, C2H6SO, Merck) were used as purchased. F. vulgare seeds were purchased from local markets in the city of Habbaniyah, Iraq. The seeds were carefully dry-cleaned, washed with distilled water, and carefully dried.

3.2. Preparation of F. vulgare Seed Extract

In accordance with a previous report [54,70], 100 g of the F. vulgare seeds were ground into a fine powder using an electric grinder (Moulinex AR1100, Groupe SEB, Écully, France), mixed with 250 mL of distilled water, and heated to 45 °C for 4 h. The mixture was left to cool to room temperature and filtered using a Whatman filter paper No. 1.

3.3. Synthesis of Se NPs

Adopting reported procedures [35,36], we initially tested different volumes of F. vulgare seed extract for the NP production (1, 2, 3, and 5 mL) and monitored the speed of the reaction by UV–vis absorption and the completeness of the reaction by analyzing the yields. The optimized conditions were 2 mL of freshly prepared F. vulgare seed extract added slowly to 2 g (11.5 mmol) of sodium selenite dissolved in 15 mL H2O. The mixture was stirred thoroughly for 60 min at 30 °C, followed by centrifugation for 15 min at 10,000 rpm. The NPs were washed with distilled water and then dried at 50 °C, resulting in 810 mg (10 mmol, 87%) red Se NP material. The NPs immediately were suspended in a phosphate-buffered saline (PBS) solution (pH = 7.4) using ultrasound and directly used for coating and loading.

3.4. Preparation of the Chitosan-Coated Se NPs—Se@Ch

In initial experiments, we prepared chitosan solutions in glacial acetic acid with varying contents (0.2%, 0.5%, 1%, and 2% v/v) and studied the NP stability and Zeta potentials. The optimum condition was a 1% solution, stirred for 90 min to ensure complete homogeneity. Then, 1 g of Se NPs were added with continuous stirring at room temperature for 10 h. After centrifugation, the supernatant was discarded and the NP precipitate washed twice with water.

3.5. Loading with Paclitaxel (PTX)

In initial loading experiments were prepared solutions of different concentrations of PTX in DMSO (10 mg, 25 mg, and 40 mg in 5 mL DMSO) to find the optimum conditions. The speed and the efficiency of the loading was tested through the PTX content in the supernatant solutions using HPLC. The optimum conditions were 25 mg Paclitaxel (PTX) dissolved in 5 mL DMSO and added dropwise at a rate of 1 mL/min to the Se@Ch NPs. The mixture was left stirring for 16 h, then the solution was centrifuged at 10,000 rpm for 20 min. The supernatant was discarded and the resulting precipitate was washed three times with 5 mL of distilled water and finally air-dried. The last supernatant was free from PTX as confirmed through HPLC. The Se@Ch–PTX samples were stored at room temperature for further analysis and characterization.

3.6. Instrumentation

Field emission scanning electron microscopy (FESEM) was carried out using a FESEM MIRA3 TESCAN-XMU (Tescan, Brno, Czech Republic, set at 20 kV. Transmission electron microscopy (TEM) images were obtained using a JEM-1230 instrument (JEOL, Akishima, Tokyo, Japan) with an acceleration voltage of 200 kV. Powder X-ray diffraction was performed using a Bruker AXSD8 instrument (D8 FOCUS 2200 V Bruker AXS, Rheinhausen, Germany), employing Cu Kα radiation with a 2θ range from 20° to 80°. Diffuse reflectance spectra (DRS) at a resolution of 4 cm−1 were recorded using Perkin Elmer 1750 FTIR spectrophotometer (PerkinElmer, Waltham, MA, USA). The Zeta potential was measured using a Malvern Zetasizer Nano (Malvern Panalytical, Malvern, UK). For sample preparation, the Se@Ch–PTX NPs were dispersed in distilled water at a concentration of 4 mg/mL and sonicated for 15 min. Then, approximately 900 µL of the dispersion was injected into the DTS1070 cells, and Zeta potential measurements were conducted at 25 °C. The collected data were analyzed using Zetasizer Software version 8.02. Thermogravimetric Analysis (TGA) was performed using a SETSYS Evolution TGA-DTA/DSC instrument (Setaram Instrumentation, Caluire-et-Cuire, France) on powdered samples.

3.7. In Vitro PTX Release

A suspension of 10 mg of the NPs was prepared in 10 mL of buffer solution for each pH = 3.5, 7.4, and 9.0. The mixture was transferred to dialysis tubes (SERVA Electrophoresis GmbH, Heidelberg, Germany) with a pore size of 25 inches (MWCO = 12,000–14,000 Da). The dialysis tubes were immersed in 50 mL of the corresponding buffer solution in a water bath maintained at 37 °C, with continuous stirring at 100 rpm. Samples of 1 mL were taken at 0, 1, 2, 4, 6, 8, 12, 18, and 24 h. After each sample collection, the same volume of fresh buffer solution was added to maintain the volume. The PTX concentration in the collected samples was analyzed using an Agilent 1260 Infinity II High-Performance Liquid Chromatography (HPLC) (Agilent, Santa Clara, CA, USA). A C18 column (250 mm × 4.6 mm; 5 μm) was employed. Detection was carried out at 227 nm. The mobile phase consisted of an 80:20 (v/v) mixture of MeOH and water, the flow rate was 1 mL/min, ensuring efficient separation of the sample components [71]. A 20 μL volume of each sample was injected into the system using an auto-injector. The drug release percentage was calculated by comparing the concentration of PTX in the analyzed samples to the total amount of drug loaded onto the NPs (determined using HPLC). All measurements were conducted in triplicate.

3.8. Hemolytic Activity Assay

The hemolytic activity was evaluated using a previously reported method [72]. A fresh human blood sample was collected from three healthy donors (age 24–30 years) and prepared by centrifugation at 800× g for 10 min, followed by three washes with normal saline. The red blood cells (RBCs) were suspended in normal saline (10% v/v), and 200 μL of this suspension was mixed with 200 μL of each concentration of the NPs (25, 50, 100, 200, and 400 μg/mL). The mixture was incubated at 37 °C for 60 min. Then, the samples were centrifuged at 10,000 rpm for 5 min. A total of 100 μL of each sample was transferred to a 96-well plate to measure absorbance at 540 nm using an Agilent BioTek Synergy H1 microplate reader (Agilent, Santa Clara, CA, USA). Triton-X-100 and normal saline were used as positive and negative controls, respectively. The hemolytic activity of the NPs was calculated using Equation (1).
%Hemolysis = (absorbance of NPs − absorbance of negative control)/(absorbance of positive control − absorbance of negative control) × 100

3.9. Coagulation Time Assay

The effect of Se@Ch–PTX NPs on coagulation time was assessed through aPTT (activated partial thromboplastin time) and PT (prothrombin time) assays [73]. Briefly, fresh blood samples were collected from healthy donors (aged 25–30 years) and centrifuged at 2500× g for 15 min to obtain platelet-poor plasma. A total of 100 µL of Se@Ch–PTX NPs at concentrations of 50, 100, 200, and 400 µg/mL was dispersed in 900 µL of the platelet-poor plasma and incubated for 30 min at 37 °C. Then, the suspension of each sample was centrifuged at 18,000× g for 5 min for both assays. Coagulation time was measured using the Sysmex CA-7000 analyzer (Sysmex Corporation, Kobe, Japan) for both aPTT and PT tests. The results were presented as mean ± standard error and compared to the control samples (normal saline).

3.10. Antiproliferative Activity

The MDA-MB-231 human breast cancer cell line was obtained from the Pasteur Institute of Tunis. Cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% antibiotic–antimycotic solution. Cell cultures were maintained under standard incubation conditions at 37 °C in a humidified atmosphere containing 5% CO2, with routine sub-culturing to ensure viability and genetic stability. in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% antibiotic–antimycotic solution. Cell cultures were maintained under standard incubation conditions at 37 °C in a humidified atmosphere containing 5% CO2, with routine sub-culturing to ensure viability and genetic stability. Once the cells reached approximately 70–80% confluency, they were harvested for use in cytotoxicity assays. Cells were seeded into 96-well microplates at a density of 1 × 105 cells per well and incubated for 24 h to allow for proper attachment [74,75]. Subsequently, the cells were treated with serial concentrations of Se@Ch–PTX NPs, Se NPs, and PTX ranging from 3.12 to 200 µg/mL, followed by 72 h incubation under identical conditions. To assess the antiproliferative activity, a standard MTT assay was employed. Following treatment, the medium was carefully removed, and 25 µL of MTT solution (5.5 mg/mL) was added to each well. Plates were wrapped in aluminum foil to protect from light and incubated for an additional 4 h. Subsequent to incubation, the MTT reagent was aspirated, and 100 µL of dimethyl sulfoxide (DMSO) was added to each well to solubilize the resulting formazan crystals. Absorbance was measured at 570 nm and 650 nm using a UV–vis microplate reader (BioTek Synergy H1, Agilent Technologies, Santa Clara, CA, USA). Untreated wells were designated as the negative control group for reference. Cell viability (%) was calculated using Equation (2):
Cell viability (%) = [(OD570 − OD650)_test ÷ (OD570 − OD650)_control] × 100
The half maximal inhibitory concentration (IC50), representing the concentration required to inhibit 50% of cell growth, was determined using Equation (3):
Inhibition (%) = [(OD_control − OD_test) × 100] ÷ OD_control
All treatments were performed in triplicate. Results were expressed as mean ± standard deviation (SD). Statistical analysis was carried out using SPSS version 25.0 (IBM Corp., Armonk, NY, USA), and differences were considered statistically significant at p < 0.05. The cell morphology of MDA-MB-231 cells was studied using a CKX53 phase-contrast light microscope (Olympus, Tokyo, Japan).

3.11. Antioxidant Activity Assay Using DPPH

The colorimetric method based on the compound DPPH (1,1-diphenyl-2-picrylhydrazyl) was employed to evaluate the antioxidant activity NPs, following a previously reported protocol [76]. The samples were prepared through serial dilutions, in which equal volumes of the sample and MeOH were mixed to obtain the following concentrations: 5, 10, 20, 30, 40, and 50 mg/L. Then, 1 mL of DPPH solution (c = 0.135 mM) was added to each sample. Ascorbic acid was used as a reference. After incubating the samples in the dark at 298 K for 30 min, the absorbance was measured at a wavelength of 517 nm using a UV–vis absorption spectrophotometer (UV-2600i, Shimadzu, Japan). The percentage of free radical scavenging (antioxidant activity) was calculated using Equation (4), with the DPPH solution in MeOH serving as the control.
% Radical Scavenging = ((A_control − A_sample)/A_control) × 100
This procedure was based on previously published methods with only minor modifications [77]. To determine the half-maximal inhibitory concentration (IC50), an exponential curve was used to illustrate the relationship between sample concentration and the remaining percentage of DPPH• free radicals [78].

3.12. Statistical Analysis

Data are presented as mean ± standard error (SE). A one-way analysis of variance (ANOVA) was employed to assess differences between group means. A p-value of less than 0.05 was considered to indicate statistical significance.

4. Conclusions

Selenium nanoparticles (Se NPs) were synthesized from Na2SeO3 using Foeniculum vulgare (fennel) seed extract as mild sustainable reductant, coated with chitosan (Ch), and loaded with Paclitaxel (PTX). The PTX release from the Se@Ch–PTX NPs and the cytotoxicity against MDA-MB-231 cells was studied in view of an application as drug delivery platform for the treatment of breast cancer. Thermogravimetric Analysis (TGA) showed thermal stability of the NPs up to 300 °C, the UV–vis absorption and Fourier-transform IR spectroscopy allowed to trace surface species originating from the F. vulgare extract on the Se NP, while the surface of the Se@Ch–PTX NPs seems to be dominated by functionalities originating from Ch and PTX. This agrees with the positive Zeta potential of +57 mV for the Ch/PTX coated NPs in contrast to the −39 mV found the Se NPs. The NPs showed good biocompatibility with red blood cells in hemolytic activity assays, exhibiting no hemolytic effects at concentrations ranging from 50 to 400 µg/mL. In vitro release studies indicated a sustained and pH-responsive release pattern with a maximum release of about 80% within 22 h for Se@Ch–PTX at pH = 3.5, which is perfect for medical application. The Se@Ch–PTX NPs showed high antiproliferative activity against MDA-MB-231 with an IC50 value of 12.3 µg/ML compared to about 38 for PTX and 55 for the Se NPs. The reactive oxygen species (ROS) activity as studied through DPPH scavenging showed higher values for the Se@Ch–PTX NPs compared to the Se NP. The modeling of the release kinetics showed high conformity with the Korsmeyer–Peppas model, which is characteristic of drug release through a combined mechanism involving both the diffusion and degradation of the biopolymer coating. Thus, the synthesis of the Se NPs using F. vulgare extract is a reliable and robust method. The NPs were coated with chitosan and this cover impregnated with PTX. Compared with other Se NP-based drug delivery systems for PTX, Se@Ch–PTX is superior in its stability and controlled release. The high antiproliferative activity of Se@Ch–PTX is mainly due to the PTX release. However, comparison with other work showed that the Se NP carrier also contributed through production of ROS. Our study also illustrates the beneficial role of chitosan in nanoparticle stabilization, drug-loading capacity, and surface characteristics that are likely to improve drug-release. Furthermore, when using biomaterials as reductants for the production of such Se NPs, remainders of these materials were found as surface-protecting ligands on the NPs and are probably not entirely removed during subsequent processes such as coating or drug-loading. Thus, these ligands/species might also contribute to the biological activities of such NPs and could thus be selected on purpose. Unfortunately, there are not enough reports so far to evaluate this interesting approach. But this might be an interesting area for future work.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nano15161276/s1. The materials contain Figure S1: FT-IR spectra of PTX, Se NPs, F. vulgare seed extract, and Se@Ch–PTX NPs. Figure S2: UV–vis absorption spectra of F. vulgare seed extract, Se NPs, and Se@Ch-PTX NPs. Figure S3: Powder X-ray diffractograms of Se NPs, Se@Ch-PTX NPs, and chitosan with assigned reflexes for Se (hexagonal; JCPDS file number 06-0362). Figure S4: TGA of the Se@Ch-PTX NPs. Figure S5: Zeta potential measurements of (a) Se NPs and (b) Se@Ch-PTX NPs. Figure S6: Morphological changes in MDA-MB-231 breast cancer cells after treatment with a concentration of 200 mg/mL for 72 h using PTX, Se NPs, and the Se@Ch–PTX NPs.

Author Contributions

M.Y.A.-D.: investigation, formal analysis, data curation, visualization. M.M.: software, methodology, data curation. H.C.: supervision, resources, methodology, funding acquisition, writing—original draft, writing—review and editing, A.K.: supervision, methodology, visualization, writing—original draft, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and was approved by the Institutional Review Board of the participating center, the Saleh Azeiz Institute (SAI) (n°2023-1618).

Informed Consent Statement

Informed consent was obtained from all patients involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Acknowledgments

The University of Cologne is acknowledged for general support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hasan, M.M.U.; Hoq, M.I.; Tanju, R.I.; Jakaria, M.; Sayeed, M.A. Breast Cancer Awareness, Screening Practices, and Perceived Barriers Among Female Undergraduate Students: An Institution-Based Cross-Sectional Study. Cancer Rep. 2025, 8, e70187. [Google Scholar] [CrossRef]
  2. Qureshi, M.A.; Khan, M.Y.; Imran, A.; Maqsood, Q.; Hussain, N.; Ali, S.W. Revolutionizing Breast Cancer Care: Cutting-Edge Breakthroughs and Future Frontiers in Precision Medicine. In Breast Cancer Treatment: An Interdisciplinary Approach; Rezaei, N., Ed.; Springer: Cham, Switzerland, 2024; pp. 115–141. [Google Scholar] [CrossRef]
  3. Shirzad, M.; Shaban, M.; Mohammadzadeh, V.; Rahdar, A.; Fathi-karkan, S.; Hoseini, Z.S.; Najafi, M.; Kharaba, Z.; Aboudzadeh, M.A. Artificial Intelligence-Assisted Design of Nanomedicines for Breast Cancer Diagnosis and Therapy: Advances, Challenges, and Future Directions. BioNanoScience 2025, 15, 354. [Google Scholar] [CrossRef]
  4. Shokoohi, M.; Sedaghatshoar, S.; Arian, H.; Mokarami, M.; Habibi, F.; Bamarinejad, F. Genetic Advancements in Breast Cancer Treatment: A Review. Discov. Oncol. 2025, 16, 127. [Google Scholar] [CrossRef]
  5. Ma, P.; Luo, Z.; Li, Z.; Lin, Y.; Li, Z.; Wu, Z.; Ren, C.; Wu, Y.L. Mitochondrial Artificial K+ Channel Construction Using MPTPP@5F8 Nanoparticles for Overcoming Cancer Drug Resistance via Disrupting Cellular Ion Homeostasis. Adv. Healthc. Mater. 2024, 13, 2302012. [Google Scholar] [CrossRef]
  6. Abouzeid, H.A.; Kassem, L.; Liua, X.; Abuelhana, A. Paclitaxel Resistance in Breast Cancer: Current Challenges and Recent Advanced Therapeutic Strategies. Cancer Treat. Res. Commun. 2025, 43, 100918. [Google Scholar] [CrossRef]
  7. Elshaer, M.; Howley, B.V.; Howe, P.H. ARIH1 Inhibition Promotes Microtubule Stability and Sensitizes Breast Cancer Cells to Microtubule-Stabilizing Agents. Cancers 2025, 17, 782. [Google Scholar] [CrossRef]
  8. Huang, H.; Kung, F.-L.; Huang, Y.-W.; Hsu, C.-C.; Guh, J.-H.; Hsu, L.-C. Sensitization of Cancer Cells to Paclitaxel-Induced Apoptosis by Canagliflozin. Biochem. Pharmacol. 2024, 223, 116140. [Google Scholar] [CrossRef] [PubMed]
  9. Kumar, S.; Arora, A.; Pant, V.; Guchhait, S.; Kumar, R.; Mathur, D.; Singh, B.K. Advances in Drug Delivery Systems for Lipophilic Drug Paclitaxel: Developments, Challenges, and Opportunities (A Review). Russ. J. Bioorg. Chem. 2024, 50, 1752–1782. [Google Scholar] [CrossRef]
  10. Singh, S.; Pal, K. Polyphenol Modified CuO Nanorods Capped by Kappa-Carrageenan for Controlled Paclitaxel Release in Furnishing Targeted Chemotherapy in Breast Carcinoma Cells. Int. J. Biol. Macromol. 2024, 255, 127893. [Google Scholar] [CrossRef]
  11. Sati, P.; Sharma, E.; Dhyani, P.; Attri, D.C.; Rana, R.; Kiyekbayeva, L.; Büsselberg, D.; Samuel, S.M.; Sharifi-Rad, J. Paclitaxel and Its Semi-Synthetic Derivatives: Comprehensive Insights into Chemical Structure, Mechanisms of Action, and Anticancer Properties. Eur. J. Med. Res. 2024, 29, 90. [Google Scholar] [CrossRef] [PubMed]
  12. Gralewska, P.; Gajek, A.; Marczak, A.; Rogalska, A. Targeted Nanocarrier-Based Drug Delivery Strategies for Improving the Therapeutic Efficacy of PARP Inhibitors Against Ovarian Cancer. Int. J. Mol. Sci. 2024, 25, 8304. [Google Scholar] [CrossRef] [PubMed]
  13. Hertz, D.L.; Joerger, M.; Bang, Y.-J.; Mathijssen, R.H.; Zhou, C.; Zhang, L.; Gandara, D.; Stahl, M.; Monk, B.J.; Jaehde, U. Paclitaxel Therapeutic Drug Monitoring-International Association of Therapeutic Drug Monitoring and Clinical Toxicology Recommendations. Eur. J. Cancer 2024, 202, 114024. [Google Scholar] [CrossRef] [PubMed]
  14. Al-Kofahi, T.; Altrad, B.; Amawi, H.; Aljabali, A.A.; Abul-Haija, Y.M.; Obeid, M.A. Paclitaxel-Loaded Niosomes in Combination with Metformin: Development, Characterization and Anticancer Potentials. Ther. Deliv. 2024, 15, 109–118. [Google Scholar] [CrossRef] [PubMed]
  15. Abdelkarim, E.A.; Elsamahy, T.; El Bayomi, R.M.; Hussein, M.A.; Darwish, I.A.; El-tahlawy, A.S.; Alahmad, W.; Darling, R.J.; Hafez, A.E.-S.E.; Sobhi, M.; et al. Nanoparticle-Driven Aquaculture: Transforming Disease Management and Boosting Sustainable Fish Farming Practices. Aquacult. Int. 2025, 33, 288. [Google Scholar] [CrossRef]
  16. Lunawat, A.K.; Thakur, S.; Kurmi, B.D.; Gupta, G.D.; Patel, P.; Raikwar, S. Revolutionizing Cancer Treatment: The Role of Chitosan Nanoparticles in Therapeutic Advancements. J. Drug Deliv. Sci. Technol. 2024, 96, 105661. [Google Scholar] [CrossRef]
  17. Maghimaa, M.; Sagadevan, S.; Suryadevara, P.R.; Sudhan, H.H.; Burle, G.S.R.; Ruokolainen, J.; Nelson, V.K.; Kesari, K.K. Cytotoxicity and Targeted Drug Delivery of Green Synthesized Metallic Nanoparticles Against Oral Cancer: A Review. Inorg. Chem. Commun. 2025, 173, 113806. [Google Scholar] [CrossRef]
  18. Hossain, A.; Rayhan, M.T.; Mobarak, M.H.; Rimon, M.I.H.; Hossain, N.; Islam, S.; Al Kafi, S.A. Advances and Significances of Gold Nanoparticles in Cancer Treatment: A Comprehensive Review. Results Chem. 2024, 8, 101559. [Google Scholar] [CrossRef]
  19. Waqar, M.A. A Comprehensive Review on Recent Advancements in Drug Delivery via Selenium Nanoparticles. J. Drug Target. 2025, 33, 157–170. [Google Scholar] [CrossRef]
  20. Sonkusre, P.; Cameotra, S.S. Biogenic Selenium Nanoparticles Induce ROS-Mediated Necroptosis in PC-3 Cancer Cells Through TNF Activation. J. Nanobiotechnol. 2017, 15, 43. [Google Scholar] [CrossRef]
  21. He, L.; Javid Anbardan, Z.; Habibovic, P.; van Rijt, S. Doxorubicin-and Selenium-Incorporated Mesoporous Silica Nanoparticles as a Combination Therapy for Osteosarcoma. ACS Appl. Nano Mater. 2024, 7, 25400–25411. [Google Scholar] [CrossRef]
  22. Nie, S.; He, X.; Sun, Z.; Zhang, Y.; Liu, T.; Chen, T.; Zhao, J. Selenium Speciation-Dependent Cancer Radiosensitization by Induction of G2/M Cell Cycle Arrest and Apoptosis. Front. Bioeng. Biotechnol. 2023, 11, 1168827. [Google Scholar] [CrossRef]
  23. Nag, S.; Kar, S.; Mishra, S.; Stany, B.; Seelan, A.; Mohanto, S.; Kamaraj, C.; Subramaniyan, V. Unveiling Green Synthesis and Biomedical Theranostic Paradigms of Selenium Nanoparticles (SeNPs)-A State-of-the-Art Comprehensive Update. Int. J. Pharm. 2024, 622, 124535. [Google Scholar] [CrossRef] [PubMed]
  24. Umapathy, S.; Pan, I.; Issac, P.K.; Kumar, M.S.K.; Giri, J.; Guru, A.; Arockiaraj, J. Selenium nanoparticles as neuroprotective agents: Insights into molecular mechanisms for Parkinson’s disease treatment. Mol. Neurobiol. 2025, 62, 6655–6682. [Google Scholar] [CrossRef] [PubMed]
  25. Li, J.; Gu, Y.; Zhang, W.; Bao, C.-Y.; Li, C.-R.; Zhang, J.-Y.; Liu, T.; Li, S.; Huang, J.-X.; Xie, Z.-G. Molecular Mechanism for Selective Cytotoxicity Towards Cancer Cells of Diselenide-Containing Paclitaxel Nanoparticles. Int. J. Biol. Sci. 2019, 15, 1755–1770. [Google Scholar] [CrossRef] [PubMed]
  26. Menon, S.; Jayakodi, S.; Yadav, K.K.; Somu, P.; Isaq, M.; Shanmugam, V.K.; Chaitanyakumar, A.; Basavegowda, N. Preparation of Paclitaxel-Encapsulated Bio-Functionalized Selenium Nanoparticles and Evaluation of their Efficacy against Cervical Cancer. Molecules 2022, 27, 7290. [Google Scholar] [CrossRef]
  27. Gong, G.; Fu, B.; Ying, C.; Zhu, Z.; He, X.; Li, Y.; Xuan, Q.; Huang, Y.; Lin, Y.; Li, Y. Targeted delivery of paclitaxel by functionalized selenium nanoparticles for anticancer therapy through ROS-mediated signaling pathways. RSC Adv. 2018, 8, 39957–39966. [Google Scholar] [CrossRef]
  28. Zou, J.; Su, S.; Chen, Z.; Liang, F.; Zeng, Y.; Cen, W.; Zhang, X.; Xia, Y.; Huang, D. Hyaluronic acid-modified selenium nanoparticles for enhancing the therapeutic efficacy of paclitaxel in lung cancer therapy. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3456–3464. [Google Scholar] [CrossRef]
  29. Bidkar, A.P.; Sanpui, P.; Ghosh, S.S. Efficient induction of apoptosis in cancer cells by paclitaxel-loaded selenium nanoparticles. Nanomedicine 2017, 12, 2641–2651. [Google Scholar] [CrossRef]
  30. Tugarova, A.V.; Vetchinkina, E.P.; Loshchinina, E.A.; Burov, A.M.; Nikitina, V.E.; Kamnev, A.A. Reduction of selenite by Azospirillum brasilense with the formation of selenium nanoparticles. Microb. Ecol. 2014, 68, 495–503. [Google Scholar] [CrossRef]
  31. Ibrahim, I.M.; Ebid, W.M.; Sayed, A.M.E. Enhancing the Structure, Optical, and Antimicrobial Advancements of Starch/Chitosan Blend Through Green-Synthesized SeO2 Nanoparticles and Their Application for Ras Cheese Packaging. Food Bioprocess. Technol. 2025, 18, 5572–5588. [Google Scholar] [CrossRef]
  32. Ikram, M.; Javed, B.; Raja, N.I.; Mashwani, Z.-U.-R. Biomedical potential of plant-based selenium nanoparticles: A comprehensive review on therapeutic and mechanistic aspects. Int. J. Nanomed. 2021, 16, 249–268. [Google Scholar] [CrossRef]
  33. Pyrzynska, K.; Sentkowska, A. Biosynthesis of selenium nanoparticles using plant extracts. J. Nanostruct. Chem. 2022, 12, 467–480. [Google Scholar] [CrossRef]
  34. Puri, A.; Mohite, P.; Ansari, Y.; Mukerjee, N.; Alharbi, H.M.; Upaganlawar, A.; Thorat, N. Plant-derived selenium nanoparticles: Investigating unique morphologies, enhancing therapeutic uses, and leading the way in tailored medical treatments. Mater. Adv. 2024, 5, 3602–3628. [Google Scholar] [CrossRef]
  35. Keshtmand, Z.; Khademian, E.; Jafroodi, P.P.; Abtahi, M.S.; Yaraki, M.T. Green synthesis of selenium nanoparticles using Artemisia chamaemelifolia: Toxicity effects through regulation of gene expression for cancer cells and bacteria. Nano Struct. Nano Objects 2023, 36, 101049. [Google Scholar] [CrossRef]
  36. Alagesan, V.; Venugopal, S. Green synthesis of selenium nanoparticle using leaves extract of Withania somnifera and its biological applications and photocatalytic activities. Bionanoscience 2019, 9, 105–116. [Google Scholar] [CrossRef]
  37. Hussain, A.; Lakhan, M.N.; Hanan, A.; Soomro, I.A.; Ahmed, M.; Bibi, F.; Zehra, I. Recent progress on green synthesis of selenium nanoparticles—A review. Mater. Today Sustain. 2023, 23, 100420. [Google Scholar] [CrossRef]
  38. Zhong, B.; Xu, W.; Wu, H.; Xian, W.; Gong, M.; Wu, Z. Differences in slow-release characteristics and release kinetics of three selenium nanoparticles from different synthesis strategies: Revealing the advantages synthesized by Lactiplantibacillus plantarum. Food Biosci. 2024, 62, 105307. [Google Scholar] [CrossRef]
  39. Yassein, A.S.; Elamary, R.B.; Alwaleed, E.A. Biogenesis, characterization, and applications of Spirulina selenium nanoparticles. Microb. Cell Fact. 2025, 24, 39. [Google Scholar] [CrossRef]
  40. Prabhu, K.; Mohanraj, K.; Kannan, S.; Barathan, S.; Sivakumar, G. Effect of pH, L-arginine concentration, and aging time on selenium nanostructures. Synth. React. Inorg. Met. Org. Nano Met. Chem. 2014, 44, 383–388. [Google Scholar] [CrossRef]
  41. Cruz, L.Y.; Wang, D.; Liu, J. Biosynthesis of selenium nanoparticles, characterization and X-ray induced radiotherapy for the treatment of lung cancer with interstitial lung disease. J. Photochem. Photobiol. B 2019, 191, 123–127. [Google Scholar] [CrossRef] [PubMed]
  42. Sentkowska, A.; Konarska, J.; Szmytke, J.; Grudniak, A. Herbal polyphenols as selenium reducers in the green synthesis of selenium nanoparticles: Antibacterial and antioxidant capabilities of the obtained SeNPs. Molecules 2024, 29, 1686. [Google Scholar] [CrossRef]
  43. Ferrari, R.; Sponchioni, M.; Morbidelli, M.; Moscatelli, D. Polymer nanoparticles for the intravenous delivery of anticancer drugs: The checkpoints on the road from the synthesis to clinical translation. Nanoscale 2018, 10, 22701–22719. [Google Scholar] [CrossRef] [PubMed]
  44. Kozma, M.; Acharya, B.; Bissessur, R. Chitin, chitosan, and nanochitin: Extraction, synthesis, and applications. Polymers 2022, 14, 3989. [Google Scholar] [CrossRef]
  45. Peter, S.; Lyczko, N.; Gopakumar, D.; Maria, H.J.; Nzihou, A.; Thomas, S. Chitin and chitosan based composites for energy and environmental applications: A review. Waste Biomass Valorization 2021, 12, 4777–4804. [Google Scholar] [CrossRef]
  46. Youssef, Y.A.; Tammam, S.N.; Elshenawy, B.M.; Ilyas, S.; Gad, A.A.; Farag, K.S.; Mathur, S.; Abdel-Kader, R.M. Peptide-loaded chitosan nanoparticles improve mitochondrial and cognitive functions via inhibition of Aβ-ABAD interaction in Alzheimer’s disease. Eur. J. Pharmaceut. Biopharmaceut. 2025, 214, 114778. [Google Scholar] [CrossRef]
  47. Aibani, N.; Rai, R.; Patel, P.; Cuddihy, G.; Wasan, E.K. Chitosan nanoparticles at the biological interface: Implications for drug delivery. Pharmaceutics 2021, 13, 1686. [Google Scholar] [CrossRef] [PubMed]
  48. Herdiana, Y.; Wathoni, N.; Gozali, D.; Shamsuddin, S.; Muchtaridi, M. Chitosan-based nano-smart drug delivery system in breast cancer therapy. Pharmaceutics 2023, 15, 879. [Google Scholar] [CrossRef]
  49. Sharma, H.; Yang, H.; Sharma, N.; An, S.S.A. Multivalent Neuroprotective Activity of Elettaria cardamomum (Cardamom) and Foeniculum vulgare (Fennel) in H2O2-Induced Oxidative Stress in SH-SY5Y Cells and Acellular Assays. Pharmaceutics 2024, 18, 2. [Google Scholar] [CrossRef]
  50. Beyazen, A.; Dessalegn, E.; Mamo, W. Phytochemical screening and biological activities of leaf of Foeniculum vulgare (Ensilal). World J. Agric. Sci. 2017, 13, 1–10. [Google Scholar] [CrossRef]
  51. Rafieian, F.; Amani, R.; Rezaei, A.; Karaça, A.C.; Jafari, S.M. Exploring fennel (Foeniculum vulgare): Composition, functional properties, potential health benefits, and safety. Crit. Rev. Food Sci. Nutr. 2024, 64, 6924–6941. [Google Scholar] [CrossRef]
  52. Vella, F.M.; Pignone, D.; Laratta, B. The Mediterranean Species Calendula officinalis and Foeniculum vulgare as Valuable Source of Bioactive Compounds. Molecules 2024, 29, 3594. [Google Scholar] [CrossRef]
  53. Molaei-Kordabad, N.; Alizadeh-Salteh, S.; Ghanbari-Jahromi, M.; Saber, M. In vitro study on anticancer effect of Dodder grown on fennel (Foeniculum vulgare) and camelthorn (Alhagi maorurum) against human cancer cells lines. J. Herbal Med. 2024, 43, 100819. [Google Scholar] [CrossRef]
  54. Saleem, M.; Noor, S.; Naqvi, S.T.Q.; Muhammad, S.A. Green Synthesis of Copper Nanoparticles using Foeniculum Vulgare Seed Extract and Evaluation of their Biological Activities. Int. J. Res. Appl. Sci. Eng. Technol. 2024, 12, 269–279. [Google Scholar] [CrossRef]
  55. Hussein, H.J.; Hadi, M.Y.; Hameed, I.H. Study of chemical composition of Foeniculum vulgare using Fourier transform infrared spectrophotometer and gas chromatography-mass spectrometry. J. Pharmacogn. Phytother. 2016, 8, 60–89. [Google Scholar] [CrossRef]
  56. Sentkowska, A.; Pyrzynska, K. Catechins and Selenium Species—How They React with Each Other. Molecules 2023, 28, 5897. [Google Scholar] [CrossRef]
  57. Sentkowska, A.; Pyrzyńska, K. The influence of synthesis conditions on the antioxidant activity of selenium nanoparticles. Molecules 2022, 27, 2486. [Google Scholar] [CrossRef]
  58. Salah, M.; Elkabbany, N.A.; Partila, A.M. Evaluation of the cytotoxicity and antibacterial activity of nano-selenium prepared via gamma irradiation against cancer cell lines and bacterial species. Sci. Rep. 2024, 14, 20523. [Google Scholar] [CrossRef]
  59. Gosala, R.; Subramanian, R.; Subramanian, B. Modulating drug delivery with nano-selenium capped by chitosan reverse micelles for anticancer potential. J. Drug Deliv. Sci. Technol. 2025, 108, 106860. [Google Scholar] [CrossRef]
  60. Hassan, M.G.; Hawwa, M.T.; Baraka, D.M.; El-Shora, H.M.; Hamed, A.A. Biogenic selenium nanoparticles and selenium/chitosan-nanoconjugate biosynthesized by Streptomyces parvulus MAR4 with antimicrobial and anticancer potential. BMC Microbiol. 2024, 24, 21. [Google Scholar] [CrossRef]
  61. Kumar, A.; Sevonkaev, I.; Goia, D.V. Synthesis of selenium particles with various morphologies. J. Colloid Interface Sci. 2014, 416, 119–123. [Google Scholar] [CrossRef]
  62. Derakhshan-Sefidi, M.; Bakhshi, B.; Rasekhi, A. Thiolated chitosan nanoparticles encapsulated nisin and selenium: Antimicrobial/antibiofilm/anti-attachment/immunomodulatory multi-functional agent. BMC Microbiol. 2024, 24, 257. [Google Scholar] [CrossRef]
  63. Herdiana, Y.; Febrina, E.; Nurhasanah, S.; Gozali, D.; Elamin, K.M.; Wathoni, N. Drug Loading in Chitosan-Based Nanoparticles. Pharmaceutics 2024, 16, 1043. [Google Scholar] [CrossRef] [PubMed]
  64. Mikušová, V.; Mikuš, P. Advances in Chitosan-Based Nanoparticles for Drug Delivery. Int. J. Mol. Sci. 2021, 22, 9652. [Google Scholar] [CrossRef]
  65. Santadkha, T.; Skolpap, W.; Thitapakorn, V. Diffusion Modeling and in vitro release kinetics studies of curcumin−loaded superparamagnetic nanomicelles in cancer drug delivery system. J. Pharm. Sci. 2022, 111, 1690–1699. [Google Scholar] [CrossRef] [PubMed]
  66. Nakayama, M.; Okano, T.; Miyazaki, T.; Kohori, F.; Sakai, K.; Yokoyama, M. Molecular design of biodegradable polymeric micelles for temperature-responsive drug release. J. Control. Release 2006, 115, 46–56. [Google Scholar] [CrossRef]
  67. Rahdari, T.; Mahdavimehr, M.; Ghafouri, H.; Ramezanpour, S.; Ehtesham, S.; Asghari, S.M. Advancing Triple-Negative Breast Cancer Treatment through Peptide Decorated Solid Lipid Nanoparticles for Paclitaxel Delivery. Sci. Rep. 2025, 15, 6043. [Google Scholar] [CrossRef]
  68. Kamenska, T.; Abrashev, M.; Georgieva, M.; Krasteva, N. Impact of polyethylene glycol functionalization of graphene oxide on anticoagulation and haemolytic properties of human blood. Materials 2021, 14, 4853. [Google Scholar] [CrossRef] [PubMed]
  69. Zhai, X.; Zhang, C.; Zhao, G.; Stoll, S.; Ren, F.; Leng, X. Antioxidant capacities of the selenium nanoparticles stabilized by chitosan. J. Nanobiotechnol. 2017, 15, 4. [Google Scholar] [CrossRef]
  70. Chen, J.; Ding, J.; Li, D.; Wang, Y.; Wu, Y.; Yang, X.; Chinnathambi, A.; Salmen, S.H.; Alharbi, S.A. Facile preparation of Au nanoparticles mediated by Foeniculum vulgare aqueous extract and investigation of the anti-human breast carcinoma effects. Arab. J. Chem. 2022, 15, 103479. [Google Scholar] [CrossRef]
  71. Yadav, P.K.; Verma, S.; Chauhan, D.; Yadav, P.; Tiwari, A.K.; Saklani, R.; Gupta, D.; Rana, R.; Shah, A.A.; Verma, S. Simultaneous estimation of paclitaxel and bortezomib via LC-MS/MS: Pharmaceutical and pharmacokinetic applications. Nanomedicine 2024, 19, 1995–2010. [Google Scholar] [CrossRef]
  72. Chahardoli, A.; Qalekhani, F.; Shokoohinia, Y.; Fattahi, A. Biological and catalytic activities of green synthesized silver nanoparticles from the leaf infusion of Dracocephalum kotschyi Boiss. Glob. Chall. 2021, 5, 2000018. [Google Scholar] [CrossRef]
  73. Yang, J.-Y.; Bae, J.; Jung, A.; Park, S.; Chung, S.; Seok, J.; Roh, H.; Han, Y.; Oh, J.-M.; Sohn, S.; et al. Surface functionalization-specific binding of coagulation factors by zinc oxide nanoparticles delays coagulation time and reduces thrombin generation potential in vitro. PLoS ONE 2017, 12, e0181634. [Google Scholar] [CrossRef]
  74. Wang, C.-H.; Yang, J.-M.; Guo, Y.-B.; Shen, J.; Pei, X.-H. Anticancer Activity of Tetrandrine by Inducing Apoptosis in Human Breast Cancer Cell Line MDA-MB-231 In Vivo. J. Evid. Based Complement. Altern. Med. 2020, 2020, 6823520. [Google Scholar] [CrossRef]
  75. Ravikumar, S.; Fredimoses, M.; Gnanadesigan, M. Anticancer property of sediment actinomycetes against MCF-7 and MDA-MB-231 cell lines. Asian Pac. J. Trop. Biomed. 2012, 2, 92–96. [Google Scholar] [CrossRef] [PubMed]
  76. Alamri, A.A.; Alanazi, N.A.H.; Mashlawi, A.M.; Shommo, S.A.; Akeel, M.A.; Alhejely, A.; Sulieman, A.M.E.; Salama, S.A. Chemical Composition of Anabasis articulata, and Biological Activity of Greenly Synthesized Zinc Oxide Composite Nanoparticles (Zn-NPs): Antioxidant, Anticancer, and Larvicidal Activities. Agronomy 2024, 14, 1742. [Google Scholar] [CrossRef]
  77. Hamrita, B.; Emira, N.; Papetti, A.; Badraoui, R.; Bouslama, L.; Ben Tekfa, M.-I.; Hamdi, A.; Patel, M.; Elasbali, A.M.; Adnan, M. Phytochemical Analysis, Antioxidant, Antimicrobial, and Anti-Swarming Properties of Hibiscus sabdariffa L. Calyx Extracts: In Vitro and In Silico Modelling Approaches. Evid. Based Complement. Altern. Med. 2022, 2022, 1252672. [Google Scholar] [CrossRef] [PubMed]
  78. Shojaee, M.S.; Moeenfard, M.; Farhoosh, R. Kinetics and stoichiometry of gallic acid and methyl gallate in scavenging DPPH radical as affected by the reaction solvent. Sci. Rep. 2022, 12, 8765. [Google Scholar] [CrossRef]
Scheme 1. Schematic of the synthesis of Se@Ch–PTX NPs using F. vulgare seed extract.
Scheme 1. Schematic of the synthesis of Se@Ch–PTX NPs using F. vulgare seed extract.
Nanomaterials 15 01276 sch001
Figure 1. SEM (a) and TEM (b) of the Se NPs.
Figure 1. SEM (a) and TEM (b) of the Se NPs.
Nanomaterials 15 01276 g001
Figure 2. SEM (a) and TEM (b) of the Se@Ch–PTX NPs.
Figure 2. SEM (a) and TEM (b) of the Se@Ch–PTX NPs.
Nanomaterials 15 01276 g002
Figure 3. Kinetic models for release of PTX from Se@Ch–PTX NPs: (a) zero-order; (b) first order; (c) Higuchi; and (d) Korsmeyer–Peppas model of kinetics.
Figure 3. Kinetic models for release of PTX from Se@Ch–PTX NPs: (a) zero-order; (b) first order; (c) Higuchi; and (d) Korsmeyer–Peppas model of kinetics.
Nanomaterials 15 01276 g003
Figure 4. (a) Hemolytic activity of the Se@Ch–PTX NPs (a) compared with Triton-X-100. (b) Results of the PT (green bars) and aPPT assays (red bars) on Se@Ch–PTX NPs. Control for PT and aPPT = saline.
Figure 4. (a) Hemolytic activity of the Se@Ch–PTX NPs (a) compared with Triton-X-100. (b) Results of the PT (green bars) and aPPT assays (red bars) on Se@Ch–PTX NPs. Control for PT and aPPT = saline.
Nanomaterials 15 01276 g004
Figure 5. (a) Cell viability of MDA-MB-231 cells in the presence of PTX, Se NPs, and Se@Ch–PTX NPs at increasing concentrations. (b) IC50 values after 24 h treatment. All treatments were performed in triplicate. Results were expressed as mean ± standard deviation (SD).
Figure 5. (a) Cell viability of MDA-MB-231 cells in the presence of PTX, Se NPs, and Se@Ch–PTX NPs at increasing concentrations. (b) IC50 values after 24 h treatment. All treatments were performed in triplicate. Results were expressed as mean ± standard deviation (SD).
Nanomaterials 15 01276 g005
Figure 6. Antioxidant activities of Se NPs, Se@Ch–PTX NPs, and ascorbic acid in DPPH radical scavenging. All experiments were performed in triplicate. Results were expressed as mean ± standard deviation (SD).
Figure 6. Antioxidant activities of Se NPs, Se@Ch–PTX NPs, and ascorbic acid in DPPH radical scavenging. All experiments were performed in triplicate. Results were expressed as mean ± standard deviation (SD).
Nanomaterials 15 01276 g006
Table 1. Release rate constant (K) and regression coefficient (R2) for different release models a.
Table 1. Release rate constant (K) and regression coefficient (R2) for different release models a.
pHZero-OrderFirst-OrderHiguchiKorsmeyer-Peppas
K0R2K1R2KHR2KR2
9.01.850.900.01640.81180.98350.98350.38350.9905
7.41.540.890.01630.80380.98020.98020.38250.9887
3.52.210.850.01760.73930.96180.96180.42870.9695
a For the release of PTX from Se@Ch–PTX NPs (see Figure 3).
Table 2. Comparison of reported IC50 values of Se NP formulations against cancer cells a.
Table 2. Comparison of reported IC50 values of Se NP formulations against cancer cells a.
Delivery System b [Incubation]IC50 (µg/mL) [Cell Line] cReference
Se@Ch–PTX NPs [24 h]14.3 [MDA-MB-231]this work
PTX/Se NPs [24]36.2/52.4 [MDA-MB-231]this work
PTX@Se NPs b [48 h or 72 h]2.98 [MCF-7], 0.79 [HeLa][25]
Se@Ch–PTX NPs [24 h]30.0 [HeLa][26]
Se@HA–PTX d [24 h]4 [A549][28]
Se@PTX [24 h]8 [A549][28]
Se@PTX e [48 h]13.8 [A549], 5.4 [MCF-7], 8.7 [HeLa], 4.8 [HT-29][29]
Se NPs [48 h]28.1 [A549], 12.2 [MCF-7], 25.3 [HeLa], 10.9 [HT-29][29]
C-peptide–SLN–PTX f [24 h]1.2 [4T1] e, 1.0 [MDA-MB-231][67]
SLN–PTX [24 h]3.4 [4T1], 4.0 [MDA-MB-231][67]
C-peptide/PTX [24 h]10.7/8.9 [4T1] 9.8/8.3 [MDA-MB-231][67]
a In each case an MTT assay was used. b Diselenide-containing PTX NPs. c MCF-7 is a breast cancer cell line, 4T1 is a triple-negative breast cancer cell line, HeLa is a cervical cancer cell line, HT-29 is a colon cancer cell line. d Prepared from Na2SeO3, ascorbic acid, hyaluronic acid (HA). e Prepared from SeO2, ascorbic acid, pluronic F-127. f C-peptide-conjugated solid lipid nanoparticles, prepared from stearic acid, Precirol, Tween 20, lecithin, and Poloxamer 407.
Table 3. DPPH-scavenging activity (%) and IC50 (mg/mL) values a.
Table 3. DPPH-scavenging activity (%) and IC50 (mg/mL) values a.
Concentrations (mg/mL)Ascorbic AcidSe@Ch–PTX NPsSe NPs
5080.64 ± 1.5675.91 ± 1.2770.63 ± 1.36
4072.12 ± 1.3770.25 ± 0.8960.78 ± 1.32
3064.22 ± 1.9165.15 ± 0.7154.52 ± 1.28
2055.64 ± 1.3558.96 ± 0.5646.42 ± 0.98
1046.81 ± 1.2850.85 ± 0.4739.12 ± 0.54
538.94 ± 1.0845.12 ± 0.4830.36 ± 0.28
2.531.47 ± 1.1237.56 ± 0.5123.21 ± 0.38
IC50 (mg/mL) b22.0333.7239.44
F value c1.91 ***1.52 ***1.63 ***
a Values significance at probability level of 0.001 (*** p < 0.001). b IC50 = half-maximal inhibitory concentration. c F value = ratio between the variance caused by differences between groups (between-group variance) and the variance caused by differences within each group (within-group variance).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Al-Darwesh, M.Y.; Manai, M.; Chebbi, H.; Klein, A. Green Synthesis of Chitosan-Coated Selenium Nanoparticles for Paclitaxel Delivery. Nanomaterials 2025, 15, 1276. https://doi.org/10.3390/nano15161276

AMA Style

Al-Darwesh MY, Manai M, Chebbi H, Klein A. Green Synthesis of Chitosan-Coated Selenium Nanoparticles for Paclitaxel Delivery. Nanomaterials. 2025; 15(16):1276. https://doi.org/10.3390/nano15161276

Chicago/Turabian Style

Al-Darwesh, Mouhaned Y., Maroua Manai, Hammouda Chebbi, and Axel Klein. 2025. "Green Synthesis of Chitosan-Coated Selenium Nanoparticles for Paclitaxel Delivery" Nanomaterials 15, no. 16: 1276. https://doi.org/10.3390/nano15161276

APA Style

Al-Darwesh, M. Y., Manai, M., Chebbi, H., & Klein, A. (2025). Green Synthesis of Chitosan-Coated Selenium Nanoparticles for Paclitaxel Delivery. Nanomaterials, 15(16), 1276. https://doi.org/10.3390/nano15161276

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop