Skip to main content

Antineoplastic activity of Artemisia annua bio-united Fe3O4/CeO2 on 4T1 breast cancer cells: in vivo and in vitro

Abstract

Breast cancer has become the most prevalent malignancy among women, and it is estimated to comprise over 30% of all cancer cases diagnosed in women. This study evaluates the antitumor effects of a green-synthesized Fe3O4/CeO2 nanocomposite, utilizing Artemisia annua L. extract, against breast cancer. The viability assay was carried out using 100 to 1000 µg/mL of the as-prepared nanocomposites. The BALB/c mice were injected with 4T1 breast cancer cells and subsequently treated with 5 mg/kg body weight of the synthesized nanocomposites through intraperitoneal injection for two weeks. The in vitro studies on the cell viability and cytotoxicity of Fe3O4/CeO2 nanocomposites demonstrated superior effectiveness of the green-synthesized sample (AFC) compared to the chemically produced variant (FC) against 4T1 cells. Treatment with AFC significantly reduced tumor size in 4T1-injected BALB/c mice compared to untreated control. The IC50 results for FC (890.16 µg/mL) and AFC (490.21 µg/mL) corroborated these findings. Notably, the biointerface provided by the phytomolecules from A. annua extract played a crucial role in enhancing the cytotoxic effects of the nanocomposite. This research highlights the potential of green-synthesized Fe3O4/CeO2 nanocomposites as a safe and effective nanomedicine for targeting cancer cells while minimizing adverse effects on healthy cells.

Background

With a mortality rate of about ten million people per year, cancer and cancer-related diseases are among the leading causes of death all over the globe (Ferlay et al. 2020). Nevertheless, far too little information is available regarding the biology of metastasis (Riggio et al. 2021). Even though numerous antineoplastic agents with promising outcomes have been drafted over the last decades, many were abandoned pending clinical trials due to their potential toxicity and side effects (Dy and Adjei 2013). Cancer development is associated with several determinants, e.g., genetic, hormonal, lifestyle, cultural, social, and environmental factors (Manouchehri et al. 2022). According to the latest data in 2021, breast cancer has emerged as the most prevalent and widespread malignancy in women, and it is anticipated that it encloses over 30% of all cancer cases detected in women (Ferlay et al. 2020; Arnold et al. 2022). Presently, the conventional approaches to diagnose breast cancer involve procedures such as mammography, MRI scans, biopsies, and the examination of tumor tissue for traditional indicators such as estrogen and progesterone receptors (ER, PR), human epidermal growth factor type 2 receptor (HER2), proliferation index (Ki67), and cytokeratins (CK5/6, CK14, C19). In recent times, these diagnostic techniques have been enhanced by contemporary molecular methods, such as next-generation sequencing, in situ hybridization, and microRNA analysis, which serve to identify emerging molecular markers (Beňačka et al. 2022). The available breast cancer treatment options encompass traditional surgical procedures, chemotherapy, radiation therapy, hormonal therapy, as well as emerging medications designed to target specific biological markers and gene therapy (Waks and Winer 2019). Chemotherapy (CT) can be administered either before (known as neoadjuvant CT) or after (called adjuvant CT) the surgical procedure, during the maintenance phase, or even in a palliative setting for patients with metastatic tumors (Pellegrini et al. 2023). Although the therapies targeting the human epidermal growth factor receptor 2 (HER2), e.g., chemotherapy and radiation therapy, are effective in treating early-stage breast cancer, they are ineffective when dealing with advanced-stage cancers. This is because chemotherapy drugs generally fail to specifically target the cancer cells and unintentionally harm healthy cells (Davezac et al. 2023). Moreover, they can lead to significant harm to the central nervous system (known as chemobrain) and heart and blood vessels at high doses (Zajączkowska et al. 2019). For instance, the conventional chemotherapeutic agents used in cancer treatment such as mitoxantrone (MTX) and doxorubicin (DOX) can also cause heart-related side effects (Brandão et al. 2023). Additionally, patients undergoing chemotherapy are more susceptible to infections, which can lead to complications including colitis, mucositis, sepsis, and diarrhoea (Singh et al. 2023). On the other hand, the minimally invasive therapies, including laser ablation, cryotherapy, and radiofrequency ablation (RFA), are also introduced for early-stage breast cancer treatment. Currently, the most frequently employed therapies for breast cancer typically involve the combination of two drug treatments, such as doxorubicin (Adriamycin), docetaxel (Taxotere), and cyclophosphamide (TAC) (Brown et al. 2021).

In the realm of cancer research, one widely explored avenue is nanotechnology. Nanotechnology has yielded several nanomaterials with promising outcomes in their applications for cancer, encompassing cancer diagnosis, drug transport and delivery, targeted therapy, gene therapy, molecular imaging, and biomarker mapping. Nanotechnology not only allows for the precise targeting of neoplasms and cancerous cells, but also facilitates the detection of tumor sites and enhances the effectiveness of traditional treatment methods (Jin et al. 2020; Wang et al. 2023). Over the recent years, nanoparticles have been extensively studied for their role as carriers in drug delivery for cancer diagnosis and treatment. They have the potential to enhance the biological availability and effectiveness of barely soluble and unstable medications with prolonged circulation in the bloodstream, reduce overall toxicity, increase cellular uptake, and enable targeted and controlled drug bio-delivery via surface modifications (Farahani et al. 2021). The ongoing developments in the nanomaterial antitumor strategies based on metal ions are yielding exciting and emerging impacts. For now, they are giving rise to non-pharmaceutical theranostic agents wherein the metal ions that are integrated into the nanomaterial’s building block, substitute for the traditional medicines (Liu et al. 2023). In particular, magnetic nanoparticles (MNPs) are largely used for biomedical applications due to their exceptional magnetic features, compatibility with living systems, and lack of toxicity. In this line, a considerable portion of the research is focused on designing multifunctional theranostic MNPs due to their ability to precisely target the affected organs, enabling direct monitoring of the therapy effectiveness through imaging techniques (Freis et al. 2023; Chu et al. 2013).

Magnetite (Fe(II)Fe(III)2O4) NPs, the well-known superparamagnetic compound, display remarkable genotoxic and cytotoxic characteristics. In addition to cell necrosis, mitochondrial disruption, DNA fragmentation, and changes in the expression of oncogenes, Fe3O4 NPs can lead to oxidative stress upon yielding reactive oxygen species (ROS), which causes cell apoptosis (Yew et al. 2020). Likewise, ceria (CeO2) NPs are among the most potent agents for dealing with ROS-associated maladies, e.g., cancer. As per reports, cell proliferation can be promoted by Ce-incorporated mesoporous NPs without compromising the bioactivity (Tsamesidis et al. 2021). This is especially attributed to the existence of Ce3+ and Ce4+ in ceria, which can get switched between + 3 and + 4 upon pH alteration. Basically, when ceria NPs encounter tumor cells with an acidic pH, the oxidation state can be changed from + 3 to + 4, resulting in ROS production and thereupon, apoptosis is induced. However, the oxidation state can be converted from + 4 to + 3 under the neutral pH of the normal cells and assist the ROS elimination (Nourmohammadi et al. 2019).

Concerning the physical and chemical approaches for the synthesis of NPs, on the one hand, it typically applies high temperatures and pressures along with some type of radiation, on the other hand, employs toxic precursors which can pose risks and drive-up costs (Chormey et al. 2023). As a solution, the extracts and biological entities derived from various plants, algae, and fungi have been employed to biosynthesize various metallic NPs (Karunakaran et al. 2023). This method relies on the availability and abundance of plants and offers a safe, quick, and cost-effective alternative. The role of phytochemicals is especially significant in the synthesis of metal oxide NPs where they serve as reducing, capping, and/or stabilizing agents (Bayrami et al. 2019).

Sweet wormwood (Artemisia annua L.) has a long history of being used as a medicinal plant in Asia and Africa to treat malaria. This plant belongs to the Asteraceae family and its therapeutic properties are attributed to its large number of biologically active substances (Lang et al. 2019). The well-known medicinal compound of A. annua is artemisinin, which is a sesquiterpene lactone produced in the plant's trichomes (Fig. S1) (Krishna et al. 2008). Artemisinin possesses a crucial endoperoxide bridge that is essential for its bioactive properties (O’Neill and Posner 2004). The studies indicate that the anticancer potential of A. annua is not solely attributed to a single active phytomolecule, e.g., artemisinin but more to its entire collection of phytochemicals, which includes over 600 compounds, primarily coumarins, sesquiterpenoids, enzymes, flavonoids, and steroids (Dilshad et al. 2020; Shinyuy et al. 2023; Firestone and Sundar 2009; Isani et al. 2019). Since, there was a notable absence of research investigating the combined anticancer effects of A. annua with functional metal oxides such as Fe3O4 and CeO2, the present research investigated their synergistic potential for cancer treatment. This research addresses a critical gap in the literature and has the potential to advance both scientific understanding and clinical practice.

This study aimed to systematically investigate the therapeutic properties of Fe2O4/CeO2 nanocomposite prepared in the presence of A. annua extract on breast cancer and compare it with the Fe2O4/CeO2 nanocomposite without the action of Artemisia annua. At first, the prepared nanocomposites were characterized using XRD, TEM, SEM, DRS, FT-IR, and zeta potential analyses to explore their morphological, size, surface, structural, and optical features. Later, the orthotopic transplanted mouse models containing 4T1 breast cancer cells were used to assess the synergistic effects of combined Fe3O4 and CeO2 nanocomposite without (FC) and with A. annua extract (AFC) on the prevention and treatment of breast cancer in vitro and in vivo. Finally, the cytotoxicity of the AFC sample on the healthy mice was examined and antineoplastic efficiencies of the green-synthesized sample were assessed in relation to untreated mice.

Methods

Plant extract preparation

The dried A. annua plant was acquired from a local supplier in the Ardabil, Northwestern Iran, and the authenticity was confirmed by a botanist. The A. annua plant was carefully cleaned using deionized water to eliminate any dust and impurities. Subsequently, it was air-dried at room temperature for a period of 72 h and then ground into a fine powder. After this, 50 g of the resulting powder was blended with a 70% ethanol solution and agitated on a rotary shaker for 72 h. The resulting extract was then filtered using filter paper and preserved in a dark environment at 4 °C.

Green synthesis of Fe₃O₄/CeO₂

The environmentally friendly Fe2O4/CeO₂ green nanocomposite with an equal 1:1 ratio was prepared via a co-precipitation method (Hassandoost et al. 2019; Rahim Pouran et al. 2020). In detail, the mixture of CeO2 (1 g) and extract (20 mL) was introduced into a solution containing FeCl3.6H2O and FeCl2.4H2O. After stirring for 30 min under vacuum, the solution was subjected to reflux at 100 °C for 1 h. Subsequently, the resulting precipitate was separated and placed in an oven to dry for 24 h.

Physical–chemical characterization

The X-ray crystallography (XRD) patterns were obtained using an X-ray diffractometer apparatus (X'Pert Pro, Panalytical, Netherlands) with a scanning rate of 0.04°/s in the 2θ = 10°–80° Cu Kα radiation (k = 0.15406 nm), to verify the crystalline state of the samples. The Perkin Elmer Spectrum was employed to record FT-IR spectra in the range of 400–4000 cm−1 to determine the interaction between the phytochemicals and the metallic compound. UV–Vis DRS spectra were recorded by Scinco 4100 (Scinco, Korea) UV‐vis spectrophotometer from 300 to 800 nm using BaSO4 for baseline correction. Energy-dispersive X-ray spectroscopy (EDX) analysis was used for chemical analysis of the samples employing (TESCAN MIRA2, TESCAN, Czech Republic) detector. The transmission electron microscopy (TEM) micrographs were collected by HF3300 (Hitachi, Japan) to check the microscale morphology, size, and chemical composition of the samples. Data on the surface morphology, crystallinity, and elemental composition were obtained via SEM images (LEO 1430 VP, England). Zeta potential analysis (Malvern Instruments, Westborough, USA) was used to measure the electrical charge of the nanoparticles.

In vitro apoptosis assay

Cell culture

The 4T1 mouse breast cancer cell line was sourced from the Pasteur Institute, Iran, and maintained in Dulbecco’s Modified Eagle Medium (DMEM) together with heat-inactivated fetal bovine serum (FBS, 10%) and 100 IU/mL of penicillin in a humidified incubator with 5% CO2 at 37 °C.

Cell toxicity and viability assay (MTT assay)

To assess cell proliferation, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) or tetrazolium salt was used as the MTT assay. The assay was conducted using cells from passages 2 to 4. In brief, the 4T1 cells in active growth were trypsinized, collected using 0.25% trypsin–EDTA, and seeded in 96-well plates at a density of 10,000 cells per well. After a 24-h incubation, the cells were treated with varying concentrations of the samples (100, 250, 500, 750, 1000 µg/mL) in separate wells for each concentration. The cells were then further incubated in a 5% CO2 incubator for 24 h at 37 °C. The cell viability was determined via MTT (5 mg/mL) addition to each well and incubating for an additional 3–4 h at 37 °C. Then, dimethyl sulfoxide (DMSO, 100 mL) was used to dissolve the resulting formazan crystals, and the corresponding absorbance was recorded at 570 nm employing a spectrophotometer (Spectramax M5, Molecular Devices, Sunnyvale, CA, USA). Subsequently, the half-maximum inhibitory concentration (IC50) value was set for the 4T1 cells. It is significant to highlight that the preliminary testing of lower concentrations (below 100 µg/mL) revealed minimal cytotoxicity, with the 100 µg/mL concentration showing approximately 97% and 74% cell viability, indicating low toxicity of the nanocomposites at lower doses. Therefore, higher concentrations were used to assess the dose-dependent cytotoxic effects more clearly.

Flow cytometry (FCM)

Flow cytometry (FCM) remains the preferred method for investigating the apoptotic process concerning cell type, triggering mechanisms, and timing. To assess apoptosis, Annexin V-FITC was utilized as an indicator of phosphatidylserine exposure, and propidium iodide (PI) was employed to mark dead cells, using the APOAF Annexin V-FITC apoptosis detection kit from Sigma-Aldrich.

In vivo apoptosis assay

Animal

We procured nine four-week-old adult BALB/c female mice from the animal facility at the University of Tehran, Faculty of Medicine and Health Sciences. Throughout the experiments, the guidelines outlined in the National Institute of Health's (NIH) care and use manual for laboratory animals (No. 8023, revised 1978) were adhered to and duly authorized by the Ardabil Animal Care and Use Committee (IR.UMA.REC.1400.030). These mice were allowed to adapt to the laboratory conditions, maintained at a temperature of 24 ± 1 °C, and subjected to a 12-h dark–light cycle for a period of seven days before initiating the experiments.

Tumor induction

PBS (200 µL) was combined with the 4T1 cells (2 × 106) and injected into the fourth inguinal mammary fat pads of 4-week-old female BALB/c mice. In this experiment, we had three groups: Group 1 (n = 3) consisted of healthy mice that underwent surgery to investigate any potential toxicity and inflammation caused by the nanocomposites. Group 2 (n = 3) comprised mice with induced breast tumors using 4T1 cells (2 × 106) and given PBS as the control. Group 3 (n = 3) included mice-induced breast tumors via 4T1 cells (2 × 106) and served as the treatment group. Both Groups 1 and 3 received 5 mg/kg of green-synthesized nanocomposites by intraperitoneal injection for two weeks (Lu et al. 2014). Throughout the study, the animals were closely monitored twice a day (morning and afternoon) for signs of clinical abnormalities, including toxicity, inflammation, changes in body weight, and food consumption. The individual body weights of all test animals were recorded before dosing on days 0, 1, 7, and 14. Calipers were used for the measurement of tumors and the volume of tumors was calculated via the formula: V = 1/2 (width^2 × length) (Traynor et al. 2023).

Histopathology

When the tumors of the studied mice reached a size of 500–700 mm3, they were euthanized by CO2 inhalation in accordance with ethical guidelines. The collected breast tumor samples were sectioned into 0.5 cm2 pieces and preserved in 10% formalin for at least 48 h. Slides were prepared from these samples and stained with Harris hematoxylin and eosin (H&E) followed by examination under a Nikon light microscope (Tokyo, Japan) (Hölscher et al. 2023).

Statistical analysis

The experimental data were compared using two-way ANOVA and expressed as mean ± SE. The differences were considered significant at p < 0.05. All statistical analyses were conducted using SPSS software (version 20).

Fig. 1
figure 1

a XRD patterns, b FT-IR spectra, c UV–Vis DRS spectra, and d band-gap values of FC and AFC samples

Results and discussion

Characterization of materials

Figure 1a displays the XRD patterns of the synthesized Fe2O4/CeO2 (FC) and A. annua extract-mediated Fe2O4/CeO2 (AFC). For FC sample, the distinct diffraction peaks of Fe₃O₄ crystal are observed at 2θ values of 18.75°, 30.52°, 35.72°, 37.82°, 43.57°, 54.47°, 57.72°, 63.32°, 66.42°, 72.17°, and 74.92°. These peaks correspond to the (111), (220), (311), (222), (400), (422), (511), (440), (300), (620), and (533) lattice planes of cubic Fe3O4 crystal (JCPDS No. 01-075-0449), respectively. In addition, the peaks at 2θ values of 28.72°, 33.27°, 47.62°, 56.57°, 59.92°, 69.72°, 76.92°, and 79.47° attributed to the (111), (200), (220), (311), (222), (400), (331), and (420) planes signified the cubic fluorite lattice of cubic CeO2 (JCPDS No. 00-034-0394) (Hassandoost et al. 2019). A similar pattern was observed for the AFC sample and the peaks indicating the magnetite and ceria crystals were identified in the corresponding pattern with a small shift towards smaller 2θ values due to the presence of phytochemicals in the extract. Notably, in the case of the AFC, there is a significant reduction in the intensity of the peaks, indicating the presence of smaller particles (Franco et al. 2007). This suggests that the molecules from the A. annua extract have covered the surface of the Fe3O4/CeO2 nanocomposites, resulting in a reduction in particle size (Rahim Pouran et al. 2020). As per the calculated crystallite sizes of the FC and AFC samples (15.58 nm and 7.14 nm) using XRD data in the Debye–Scherrer equation, the reduction in the crystallite sizes upon extract employment can be strongly verified.

Fig. 2
figure 2

SEM images of a FC and b AFC samples, c size and d TEM image of AFC sample

FT-IR analysis was carried out to display the interaction of the phytochemicals of A. annua with Fe₃O₄ and CeO₂ via the identified functional groups on the surface of the samples (Fig. 1b). In the FC sample, the stretching vibration peaks of Fe–O and Ce–O bonds were detected at wavenumbers of 558 cm⁻1 and 464 cm⁻1, respectively (Ventruti et al. 2016; Garcia et al. 2020). These peaks are observed as an overlapped intense peak at about 590 cm⁻1 with a shoulder at 472 cm⁻1. Additional peaks related to the extract biomolecules were identified at 3756 cm⁻1, 3376 cm⁻1, 1608 cm⁻1, 1372 cm⁻1, and 1056 cm⁻1, respectively, assigned to the O‒H, N‒H, C=C, phenolic O‒H, and C‒O bonds vibrations. The energy-dispersive X-ray spectroscopy (EDX) was parallel with FT-IR analysis (Fig. S2).

UV–vis diffuse reflectance spectroscopy (UV–vis DRS) was employed to examine the optical characteristics of both samples. As depicted in Fig. 1c, it is apparent that the level of light absorption in the visible region was notably higher for the extract-mediated sample when compared to Fe2O4/CeO2 NC. As can be witnessed from the plotted Tauc (Fig. 1d), the band-gap energy of the AFC (1.78 eV) sample was significantly reduced compared to FC (2.62 eV), and its optical performance was greatly improved at the visible region, which can be mainly attributable to the large number of charge carriers from the extract phytomolecules (Khan et al. 2019).

Based on the characterization results, the A. annua extract appears to serve both as a reducing and capping agent in the synthesis of Fe₃O₄/CeO₂ nanocomposites. As per the UV–vis DRS analysis, the reduced band-gap energy and improved optical performance indicated that the extract could provide charge carriers, thus playing a reducing role. In addition to zeta potential results, FT-IR spectrum revealed the presence of functional groups from the extract, which are likely involved in stabilizing the nanoparticles, demonstrating its role as a capping agent (Rahim Pouran et al. 2020).

The SEM micrographs of FC and AFC samples exposed the clusters of spherical nanoparticles within the micrometer range (Fig. 2a–c). Due to the agglomeration of the particles, the observed average size for the AFC sample was around 42 nm in its SEM image. The TEM image of AFC displayed in Fig. 2d offers compelling evidence of the multitude of semi-spherical shapes and sizes. The TEM image further confirmed the particle size of AFC, and the provided scale bar indicated that the sizes were consistent with the XRD results, with smaller individual particles, though some aggregation was still apparent.

Fig. 3
figure 3

a Cell viability of FC and AFC samples in comparison to one another and b tamoxifen; and IC50 values of c FC and d AFC at varying concentrations [data are expressed as mean ± standard error of the mean (p < 0.05)]

Zeta potential measurements were also carried out, yielding values of 29.7 mV for FC and − 38.7 mV for AFC, as shown in Fig. S3. The zeta potential results indicated a significant difference in the surface charge properties of the two samples. The positive value of 29.7 mV for FC suggests a positively charged surface with good stability, as zeta potential values above + 30 mV or below − 30 mV typically indicate strong repulsive forces that help maintain dispersion stability. In contrast, the AFC sample has a zeta potential of − 38.7 mV, indicating a highly negatively charged surface. This more negative value implies that the nanoparticles in AFC are even more stable in suspension due to the increased electrostatic repulsion between particles, which minimizes aggregation. The negative charge of AFC is likely attributed to the presence of anionic groups from the A. annua extract, enhancing the overall stability of the nanocomposite.

As per the characterizing analysis, the phytomolecules in the extract played a capping role by stabilizing smaller nanoparticles (as per XRD results) and a reducing role by providing charge carriers that facilitated the reduction of metal ions, as evidenced by the increased light absorption in the visible region.

Anticancer activity

Studies on cell viability and cytotoxicity

The cell viability was examined using the MTT assay to test the cytotoxicity of FC and AFC samples on 4T1 cells. For this purpose, the growth inhibition percentage of 4T1 cells was compared to the untreated cells when exposed to varying doses of the samples (100, 250, 500, 750, and 1000 µg/mL). Figure 3a illustrates the concentration-dependent growth inhibition of 4T1 cells. As per the results, the increase in the nanocomposite concentration enhanced the cytotoxic efficacies in both FC and AFC. In order to calculate the half-maximal inhibitory concentrations (IC50) values of the samples, the fitted line obtained from the plotted dose–responses data was used to estimate the concentration at which the cell viability reached 50% (Iqbal et al. 2022). The IC50 values of 890.16 (µg/mL) and 490.21 (µg/mL) were determined within 24 h for FC and AFC, respectively. This not only indicates the time- and dose-dependency of the cytotoxic effects of the samples but also the higher effectiveness of AFC to FC against 4T1 cells (p < 0.05). Data on the effectiveness of the well-known therapy, tamoxifen (TAM), were also collected and provided for comparison purposes (IC50 = 87.63 µg/mL). It is significant to highlight that the preliminary testing of lower concentrations (below 100 µg/mL) revealed minimal cytotoxicity, with the 100 µg/mL concentration showing ~ 97% and 74% cell viability for FC and AFC nanocomposites, indicating their low toxicity at lower doses. Therefore, higher concentrations were used to assess the dose-dependent cytotoxic effects more clearly. This finding was in line with similar studies using lower concentrations of Fe3O4 and CeO2 (Ebadi et al. 2020; Marzi et al. 2013).

Flow cytometry (FCM)

The apoptosis caused by A. annua plant extract synthesized Fe3O4/CeO2 NC was further authenticated using Annexin V/propidium iodide (PI) assay for flow cytometry (Srichana et al. 2023). Figure 4 represents the flow cytometry profiles of the 4T1 cells’ cycle phase distribution. As expected, significant apoptosis took place in the 4T1 cells upon exposure to AFC for about 24 h (Fig. 4). The AFC-treated 4T1 cells showed early and late apoptosis with death rates of 85.2% and 6.35%, respectively. In contrast, no significant apoptosis was observed in the untreated cells.

Fig. 4
figure 4

a Flow cytometry analysis, b quadrangular plots representing the Annexin-V/PI expression in 4T1 cells upon exposure to c AFC (treated) and in the absence of AFC (untreated) (p < 0.05)

In vivo anticancer activity

Tumor size and body weight of BALB/c mouse models

More analyses were conducted to investigate whether the green-synthesized sample could effectively target the growth of the breast cancer cells in a living system where 4T1 cells-utilized BALB/c mice were employed as a xenograft model. Throughout the study, the healthy mice treated with AFC (via intraperitoneal injections at a dose of 5 mg/kg and for a duration of 2 weeks) did not exhibit any fatalities or signs of toxicity (Fig. 5). On the other hand, in the context of the antitumor investigation, the tumor size in mice treated with AFC was notably smaller than the untreated mice following the 2-week treatment period (p < 0.05, Fig. 5). The results strongly disclosed that the administration of AFC did not result in any discernible toxicity, as evidenced by the unchanged body weight of the studied mice.

Fig. 5
figure 5

The body weights and tumor sites of the AFC-treated and untreated mice (data are presented as mean ± SD (n = 3), (mm3) (*p < 0.05))

Histopathology analysis

To compare the histopathological alterations in the tumors of untreated 4T1-injected mice with those of mice treated with AFC, breast tissues were collected, fixed, and subjected to hematoxylin and eosin (H&E) staining. Upon closer examination, higher magnification images of the untreated 4T1-injected mice breast tumors predominantly displayed viable tumor cells, characterized by pleomorphic vesicular nuclei and prominently hyperchromatic nucleoli (Fig. 6a). In contrast, higher magnification images of the AFC-treated 4T1 breast tumors revealed extensive areas of necrosis and late apoptosis, evident through the increased presence of apoptotic cells, cellular debris, and nuclear fragments (Fig. 6b).

Fig. 6
figure 6

Hematoxylin and eosin (H&E) staining of breast tumors (magnification: 100 ×): a untreated tumors, b AFC-treated tumors; apoptotic cells (red arrow), cellular debris (green arrow), and nuclear dust (white arrow)

The significant antineoplastic activity of A. annua-mediated Fe₃O4/CeO2 nanocomposite on 4T1 breast cancer cells can be explained as follows: Fe₃O₄ nanoparticles are capable of producing localized heating due to their magnetic property. This temperature rise can selectively destroy cancer cells, as they are more susceptible to heat than normal cells (Malehmir et al. 2023). Beyond this, magnetite can facilitate the generation of ROSs, which triggers oxidative stress in cancer cells and ultimately induces apoptosis (Paunovic et al. 2020). It is worth noting that the magnetic properties of Fe3O4 nanoparticles make them ideal candidates for drug delivery systems, enabling the controlled release of drugs directly to tumor sites with the help of an external magnetic field, thereby limiting damage to healthy tissue (Blazkova et al. 2013). CeO2 nanoparticles, on the other hand, possess the unique ability to shift between Ce3⁺ and Ce4⁺ oxidation states. This property allows them to neutralize excessive ROS in healthy cells while simultaneously promoting ROS production in cancer cells, thereby causing oxidative stress and triggering cell death through apoptosis. This selective ROS regulation is central to the anticancer activity of ceria (Selvaraj et al. 2024). Furthermore, CeO₂ nanoparticles can imitate the behavior of certain enzymes, such as superoxide dismutase and catalase, disrupting the redox balance in cancer cells and leading to their death through oxidative stress (Tang et al. 2023). Upon the combination of Fe3O4 and CeO2, their ability to regulate oxidative stress is amplified, creating a synergistic effect. This combined action enhanced their therapeutic potential and resulted in increased cancer cell apoptosis and overall efficacy in cancer treatment.

Moreover, as per the findings of the in vivo and in vitro studies, it can be strongly deduced that the extract phytomolecules have a prominent role in enhancing the antiproliferative efficacy of the studied nanocomposite. The extract of A. annua significantly boosts the anticancer effectiveness of nanocomposites, attributed to its abundant bioactive phytomolecules (Lang et al. 2019). These compounds can trigger apoptosis in cancer cells by elevating oxidative stress and interfering with cell cycle progression (Cao et al. 2019). Several studies have reported the antitumor action of artemisinin and its derivatives as one of the frequently studied secondary metabolites of A. annua due to its preventive effects on inflammation and cell damage resulting from necrosis (Crespo-Ortiz and Wei 2012; Slezakova and Ruda-Kucerova 2017). Moreover, the green route of synthesizing nanostructures supplies a safe and biocompatible approach to preparing therapeutically active alternatives. The extract functions as a reducing agent during the synthesis of nanoparticles, enhancing their stability. Its phytomolecules also act as capping agents, which not only improve biocompatibility, but also promote the selective targeting of tumor tissues (Villagrán et al. 2024). This dual function of A. annua extract, as both a therapeutic agent and a stabilizing component, highlights its critical role in the as-prepared nanocomposite for effective breast cancer treatment.

Conclusion

In this study, we successfully synthesized Fe3O4/CeO2 magnetic nanocomposites using both chemical (FC) and A. annua extract (AFC). FT-IR and EDX analyses confirmed the presence of Fe₃O₄ and CeO₂ in both samples, while AFC also incorporated phytomolecules from the extract. AFC demonstrated smaller particle size (7.14 nm) compared to FC (15.58 nm), more uniform morphology, and enhanced optical properties (band-gap energy reduced to 1.78 eV compared to 2.62 eV for FC), which can be attributed to the dual capping and reducing effects of the A. annua extract. The in vitro cytotoxicity assays on 4T1 breast cancer cells demonstrated the superior anticancer efficacy of AFC over FC, with IC50 values of 490.21 µg/mL for AFC and 890.16 µg/mL for FC. On the other hand, the flow cytometry analysis showed that AFC induced substantial apoptosis in 4T1 cells after 24 h of exposure, supporting its enhanced cytotoxicity. The significant roles of the extract at the nanoscale level contributed to these cellular responses by modulating the surface properties and bioactivity of the nanocomposite. The in vivo studies exhibited no signs of toxicity or fatalities in the AFC-treated healthy mice, while tumor size in AFC-treated mice significantly decreased compared to the untreated control group. Histopathological analysis confirmed notable antineoplastic effects in AFC-treated mice, characterized by an increased number of apoptotic cells, cellular debris, and nuclear fragments in breast tissue samples. This further highlighted the efficacy of AFC in selectively targeting cancer cells without causing harm to healthy tissues. It should be underlined that the dual capping and reducing roles of A. annua extract, along with its anticancer phytomolecules, enhanced both the nanocomposite properties and the therapeutic efficacy of AFC against cancer cells. The combined anticancer effects of Fe₃O₄, CeO₂, and the A. annua extract emphasize their potential as a synergistic therapeutic strategy in breast cancer treatment. Future studies are encouraged to explore its application in combination therapies and investigate its underlying mechanisms of action.

Availability of data and materials

No datasets were generated or analysed during the current study.

References

  • Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, Vignat J, Gralow JR, Cardoso F, Siesling S et al (2022) Current and future burden of breast cancer: global statistics for 2020 and 2040. The Breast 66:15–23

    Article  PubMed  PubMed Central  Google Scholar 

  • Bayrami A, Alioghli S, Rahim Pouran S, Habibi-Yangjeh A, Khataee A, Ramesh S (2019) A facile ultrasonic-aided biosynthesis of ZnO nanoparticles using Vaccinium arctostaphylos L. leaf extract and its antidiabetic, antibacterial, and oxidative activity evaluation. Ultrasonics Sonochem 55:57–66

    Article  CAS  Google Scholar 

  • Beňačka R, Szabóová D, Guľašová Z, Hertelyová Z, Radoňák J (2022) Classic and new markers in diagnostics and classification of breast cancer. Cancers 14(21):5444

    Article  PubMed  PubMed Central  Google Scholar 

  • Blazkova I, Nguyen HV, Dostalova S, Kopel P, Stanisavljevic M, Vaculovicova M, Stiborova M, Eckschlager T, Kizek R, Adam V (2013) Apoferritin modified magnetic particles as doxorubicin carriers for anticancer drug delivery. Int J Mol Sci 14(7):13391–13402

    Article  PubMed  PubMed Central  Google Scholar 

  • Brandão SR, Reis-Mendes A, Duarte-Araújo M, Neuparth MJ, Rocha H, Carvalho F, Ferreira R, Costa VM (2023) Cardiac molecular remodeling by anticancer drugs: doxorubicin affects more metabolism while mitoxantrone impacts more autophagy in adult CD-1 male mice. Biomolecules 13(6):921

    Article  PubMed  PubMed Central  Google Scholar 

  • Brown T, Sykes D, Allen AR (2021) Implications of breast cancer chemotherapy-induced inflammation on the gut, liver, and central nervous system. Biomedicines 9(2):189

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Cao Y, Feng Y-H, Gao L-W, Li X-Y, Jin Q-X, Wang Y-Y, Xu Y-Y, Jin F, Lu S-L, Wei M-J (2019) Artemisinin enhances the anti-tumor immune response in 4T1 breast cancer cells in vitro and in vivo. Int Immunopharmacol 70:110–116

    Article  CAS  PubMed  Google Scholar 

  • Chormey DS, Zaman BT, Borahan Kustanto T, Erarpat Bodur S, Bodur S, Tekin Z, Nejati O, Bakırdere S (2023) Biogenic synthesis of novel nanomaterials and their applications. Nanoscale 15(48):19423–19447

    Article  CAS  PubMed  Google Scholar 

  • Chu M, Shao Y, Peng J, Dai X, Li H, Wu Q, Shi D (2013) Near-infrared laser light mediated cancer therapy by photothermal effect of Fe3O4 magnetic nanoparticles. Biomaterials 34(16):4078–4088

    Article  CAS  PubMed  Google Scholar 

  • Crespo-Ortiz MP, Wei MQ (2012) Antitumor activity of artemisinin and its derivatives: from a well-known antimalarial agent to a potential anticancer drug. J Biomed Biotechnol 2012:247597

    Article  PubMed  Google Scholar 

  • Davezac M, Meneur C, Buscato M, Zahreddine R, Arnal J-F, Dalenc F, Lenfant F, Fontaine C (2023) The beneficial effects of tamoxifen on arteries: a key player for cardiovascular health of breast cancer patient. Biochem Pharmacol 214:115677

    Article  CAS  PubMed  Google Scholar 

  • De Marzi L, Monaco A, De Lapuente J, Ramos D, Borras M, Di Gioacchino M, Santucci S, Poma A (2013) Cytotoxicity and genotoxicity of ceria nanoparticles on different cell lines in vitro. Int J Mol Sci 14(2):3065–3077

    Article  PubMed  PubMed Central  Google Scholar 

  • Dilshad E, Ismail H, Khan MA, Cusido RM, Mirza B (2020) Metabolite profiling of Artemisia carvifolia Buch transgenic plants and estimation of their anticancer and antidiabetic potential. Biocatal Agric Biotechnol 24:101539

    Article  Google Scholar 

  • Dy GK, Adjei AA (2013) Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA Cancer J Clin 63(4):249–279

    Article  PubMed  Google Scholar 

  • Ebadi M, Buskaran K, Bullo S, Hussein MZ, Fakurazi S, Pastorin G (2020) Synthesis and cytotoxicity study of magnetite nanoparticles coated with polyethylene glycol and sorafenib-zinc/aluminium layered double hydroxide. Polymers (Basel) 12(11):2716

    Article  CAS  PubMed  Google Scholar 

  • Farahani M, Moradikhah F, Shabani I, Soflou RK, Seyedjafari E (2021) Microfluidic fabrication of berberine-loaded nanoparticles for cancer treatment applications. J Drug Deliv Sci Technol 61:102134

    Article  CAS  Google Scholar 

  • Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor A, Bray F (2021) Cancer statistics for the year 2020: an overview. Int J Cancer 149:778–789

    Article  CAS  Google Scholar 

  • Firestone GL, Sundar SN (2009) Anticancer activities of artemisinin and its bioactive derivatives. Expert Rev Mol Med 11:e32

    Article  PubMed  Google Scholar 

  • Franco F, Cecila JA, Pérez-Maqueda LA, Pérez-Rodríguez JL, Gomes CSF (2007) Particle-size reduction of dickite by ultrasound treatments: effect on the structure, shape and particle-size distribution. Appl Clay Sci 35(1):119–127

    Article  CAS  Google Scholar 

  • Freis B, Ramirez MDLA, Kiefer C, Harlepp S, Iacovita C, Henoumont C, Affolter-Zbaraszczuk C, Meyer F, Mertz D, Boos A et al (2023) Effect of the size and shape of dendronized iron oxide nanoparticles bearing a targeting ligand on MRI, magnetic hyperthermia, and photothermia properties from suspension to in vitro studies. Pharmaceutics 15(4):1104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Garcia IM, Leitune VCB, Takimi AS, Bergmann CP, Samuel SMW, Melo MA, Collares FM (2020) Cerium dioxide particles to tune radiopacity of dental adhesives: microstructural and physico-chemical evaluation. J Funct Biomater 11(1):7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Hassandoost R, Pouran SR, Khataee A, Orooji Y, Joo SW (2019) Hierarchically structured ternary heterojunctions based on Ce3+/Ce4+ modified Fe3O4 nanoparticles anchored onto graphene oxide sheets as magnetic visible-light-active photocatalysts for decontamination of oxytetracycline. J Hazard Mater 376:200–211

    Article  CAS  PubMed  Google Scholar 

  • Hölscher DL, Bouteldja N, Joodaki M, Russo ML, Lan Y-C, Sadr AV, Cheng M, Tesar V, Stillfried SV, Klinkhammer BM et al (2023) Next-generation morphometry for pathomics-data mining in histopathology. Nat Commun 14(1):470

    Article  PubMed  PubMed Central  Google Scholar 

  • Iqbal H, Razzaq A, Khan NU, Rehman SU, Webster TJ, Xiao R, Menaa F (2022) pH-responsive albumin-coated biopolymeric nanoparticles with lapatinab for targeted breast cancer therapy. Biomater Adv 139:213039

    Article  PubMed  Google Scholar 

  • Isani G, Bertocchi M, Andreani G, Farruggia G, Cappadone C, Salaroli R, Forni M, Bernardini C (2019) Cytotoxic effects of Artemisia annua L. and pure artemisinin on the D-17 canine osteosarcoma cell line. Oxid Med Cell Longev 2019:1615758

    Article  PubMed  PubMed Central  Google Scholar 

  • Jin C, Wang K, Oppong-Gyebi A, Hu J (2020) Application of nanotechnology in cancer diagnosis and therapy—a mini-review. Int J Med Sci 17(18):2964

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Karunakaran G, Sudha KG, Ali S, Cho EB (2023) Biosynthesis of nanoparticles from various biological sources and its biomedical applications. Molecules 28(11):4527

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Khan MM, Saadah NH, Khan ME, Harunsani MH, Tan AL, Cho MH (2019) Potentials of Costus woodsonii leaf extract in producing narrow band gap ZnO nanoparticles. Mater Sci Semicond Process 91:194–200

    Article  CAS  Google Scholar 

  • Krishna S, Bustamante L, Haynes RK, Staines HM (2008) Artemisinins: their growing importance in medicine. Trends Pharmacol Sci 29(10):520–527

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Lang SJ, Schmiech M, Hafner S, Paetz C, Steinborn C, Huber R, Gaafary ME, Werner K, Schmidt CQ, Syrovets T et al (2019) Antitumor activity of an Artemisia annua herbal preparation and identification of active ingredients. Phytomedicine 62:152962

    Article  CAS  PubMed  Google Scholar 

  • Liu C, Guo L, Wang Y, Zhang J, Fu C (2023) Delivering metal ions by nanomaterials: turning metal ions into drug-like cancer theranostic agents. Coord Chem Rev 494:215332

    Article  CAS  Google Scholar 

  • Lu J, Zhao W, Huang Y, Liu H, Marquez R, Gibbs RB, Li J, Venkataramanan R, Xu L, Li S et al (2014) Targeted delivery of Doxorubicin by folic acid-decorated dual functional nanocarrier. Mol Pharm 11(11):4164–4178

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Malehmir S, Esmaili MA, Khaksary Mahabady M, Sobhani-Nasab A, Atapour A, Ganjali MR, Ghasemi A, Moradi Hasan-Abad A (2023) A review: hemocompatibility of magnetic nanoparticles and their regenerative medicine, cancer therapy, drug delivery, and bioimaging applications. Front Chem. 11

  • Manouchehri E, Taghipour A, Ebadi A, Homaei Shandiz F, Latifnejad Roudsari R (2022) Understanding breast cancer risk factors: is there any mismatch between laywomen perceptions and expert opinions. BMC Cancer 22(1):309

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Nourmohammadi E, Khoshdel-sarkarizi H, Nedaeinia R, Sadeghnia HR, Hasanzadeh L, Darroudi M, Kazemi oskuee R (2019) Evaluation of anticancer effects of cerium oxide nanoparticles on mouse fibrosarcoma cell line. J Cell Physiol 234(4):4987–4996

    Article  CAS  PubMed  Google Scholar 

  • O’Neill PM, Posner GH (2004) A medicinal chemistry perspective on artemisinin and related endoperoxides. J Med Chem 47(12):2945–2964

    Article  CAS  PubMed  Google Scholar 

  • Paunovic J, Vucevic D, Radosavljevic T, Mandić-Rajčević S, Pantic I (2020) Iron-based nanoparticles and their potential toxicity: focus on oxidative stress and apoptosis. Chem Biol Interact 316:108935

    Article  CAS  PubMed  Google Scholar 

  • Pellegrini M, Merlo FD, Agnello E, Monge T, Devecchi A, Casalone V, Montemurro F, Ghigo E, Sapino A, Bo S (2023) Dysgeusia in patients with breast cancer treated with chemotherapy—a narrative review. Nutrients 15(1):226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Rahim Pouran S, Bayrami A, Mohammadi Arvanag F, Habibi-Yangjeh A, Darvishi Cheshmeh Soltani R, Singh R, Abdul Raman AA, Chae KH, Khataee A, Kang HK (2020) Biogenic integrated ZnO/Ag nanocomposite: surface analysis and in vivo practices for the management of type 1 diabetes complications. Colloids Surf B Biointerfaces 189:110878

    Article  CAS  PubMed  Google Scholar 

  • Riggio AI, Varley KE, Welm AL (2021) The lingering mysteries of metastatic recurrence in breast cancer. Br J Cancer 124(1):13–26

    Article  PubMed  Google Scholar 

  • Selvaraj S, Chauhan A, Radhakrishnan A, Rana G, Dutta V, Batoo KM, Ghotakar S (2024) Cerium oxide nanoparticles and their polymeric composites: advancements in biomedical applications. J Inorg Organometall Polym Mater. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10904-024-03263-5

    Article  Google Scholar 

  • Shinyuy LM, Loe GE, Jansen O, Mamede L, Ledoux A, Noukimi SF, Abenwie SN, Ghogomu SM, Souopgui J, Robert A et al (2023) Secondary metabolites isolated from Artemisia afra and Artemisia annua and their anti-malarial, anti-inflammatory and immunomodulating properties-pharmacokinetics and pharmacodynamics: a review. Metabolites 13(5):613

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Singh NK, Beckett JM, Kalpurath K, Ishaq M, Ahmad T, Eri RD (2023) Synbiotics as supplemental therapy for the alleviation of chemotherapy-associated symptoms in patients with solid tumours. Nutrients 15(7):1759

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Slezakova S, Ruda-Kucerova J (2017) Anticancer activity of artemisinin and its derivatives. Anticancer Res 37(11):5995–6003

    CAS  PubMed  Google Scholar 

  • Srichana T, Thawithong E, Nakpheng T, Paul PK (2023) Flow cytometric analysis, confocal laser scanning microscopic, and holotomographic imaging demonstrate potentials of levofloxacin dry powder aerosols for TB treatment. J Drug Deliv Sci Technol 84:104464

    Article  CAS  Google Scholar 

  • Tang JLY, Moonshi SS, Ta HT (2023) Nanoceria: an innovative strategy for cancer treatment. Cell Mol Life Sci 80(2):46

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Traynor S, Terp MG, Nielsen AY, Guldberg P, Jakobsen M, Pedersen PG, Gammelgaard OL, Pedersen CB, Pedersen MT, Rattenborg S et al (2023) DNA methyltransferase inhibition promotes recruitment of myeloid-derived suppressor cells to the tumor microenvironment through induction of tumor cell-intrinsic interleukin-1. Cancer Lett 552:215982

    Article  CAS  PubMed  Google Scholar 

  • Tsamesidis I, Gkiliopoulos D, Pouroutzidou GK, Lymperaki E, Papoulia C, Reybier K, Perio P, Paraskevopoulos KM, Kontonasaki E, Theocharidou A (2021) Effect of artemisinin-loaded mesoporous cerium-doped calcium silicate nanopowder on cell proliferation of human periodontal ligament fibroblasts. Nanomaterials 11(9):2189

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ventruti G, Ventura GD, Bellatreccia F, Lacalamita M, Emanuela S (2016) Hydrogen bond system and vibrational spectroscopy of the iron sulfate fibroferrite, Fe(OH)SO4·5H2O. Eur J Mineral 28(5):943–952

    Article  CAS  Google Scholar 

  • Villagrán Z, Anaya-Esparza LM, Velázquez-Carriles CA, Silva-Jara JM, Ruvalcaba-Gómez JM, Aurora-Vigo EF, Rodríguez-Lafitte E, Rodríguez-Barajas N, Balderas-León I, Martínez-Esquivias F (2024) Plant-based extracts as reducing, capping, and stabilizing agents for the green synthesis of inorganic nanoparticles. Resources 13(6):70

    Article  Google Scholar 

  • Waks AG, Winer EP (2019) Breast cancer treatment: a review. JAMA 321(3):288–300

    Article  CAS  PubMed  Google Scholar 

  • Wang Y, Iqbal H, Ur-Rehman U, Zhai L, Yuan Z, Razzaq A, Lv M, Wei H, Ning X, Xin J et al (2023) Albumin-based nanodevices for breast cancer diagnosis and therapy. J Drug Deliv Sci Technol 79:104072

    Article  CAS  Google Scholar 

  • Yew YP, Shameli K, Miyake M, Ahmad Khairudin NBB, Mohamad SEB, Naiki T, Lee KX (2020) Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles and biomedical applications in targeted anticancer drug delivery system: a review. Arab J Chem 13(1):2287–2308

    Article  CAS  Google Scholar 

  • Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J (2019) Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci 20(6):1451

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful for the support provided by the University of Mohaghegh Ardabili.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

Ziba Ahmadi (Z.A) Abolfazl Bayrami (A.B) Saber Zahri (S.Z) Shima Rahim Pouran (S.RP) Aziz Habibi-Yangjeh (A.HY) Z.A Did the experimental work. A.B and S.Z were supervisors. S.RP and A.HY were the project advisors. All authors except Z.A completed the methodology. Z.A and S.RP wrote the main manuscript text. All authors except Z.A reviewed the manuscript. A.B, S.Z, and A.HY provided resources.

Corresponding authors

Correspondence to Abolfazl Bayrami or Shima Rahim Pouran.

Ethics declarations

Ethics approval and consent to participate

The guidelines outlined in the National Institute of Health's (NIH) care and use manual for laboratory animals (No. 8023, revised 1978) were adhered to throughout the experiments and duly authorized by the Ardabil Animal Care and Use Committee (IR.UMA.REC.1400.030).

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ahmadi, Z., Bayrami, A., Zahri, S. et al. Antineoplastic activity of Artemisia annua bio-united Fe3O4/CeO2 on 4T1 breast cancer cells: in vivo and in vitro. Cancer Nano 15, 53 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12645-024-00294-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12645-024-00294-y

Keywords