Next Article in Journal
Structure–Optical Properties and Sustainability Assessment of Carbon Dots Derived from Laurus nobilis Leaves
Previous Article in Journal
Modulating Photodissociation and Photobleaching via Plasmon Resonance to Enhance Light-Induced Nitric Oxide Release
Previous Article in Special Issue
Super-Resolution Microscopy in the Structural Analysis and Assembly Dynamics of HIV
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Usefulness of Nanoparticles in the Fight Against Esophageal Cancer: A Comprehensive Review of Their Therapeutic Potential

by
Gabriel Tchuente Kamsu
* and
Eugene Jamot Ndebia
*
Department of Human Biology, Faculty of Medicine and Health Sciences, Walter Sisulu University, Mthatha 5100, South Africa
*
Authors to whom correspondence should be addressed.
Appl. Nano 2025, 6(3), 18; https://doi.org/10.3390/applnano6030018
Submission received: 24 July 2025 / Revised: 21 August 2025 / Accepted: 27 August 2025 / Published: 1 September 2025
(This article belongs to the Collection Review Papers for Applied Nano Science and Technology)

Abstract

Esophageal squamous cell carcinoma (ESCC) accounts for the majority of esophageal cancers worldwide, with a poor prognosis and increasing resistance to conventional treatments. Faced with these limitations, nanoparticles (NPs) are attracting growing interest as innovative therapeutic agents capable of improving specificity and efficacy and reducing systemic toxicity. This study critically examines the pharmacological effects, mechanisms of action, and toxicity profiles of different metallic or organic nanoparticles tested on ESCC cell lines. Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) 2020 guidelines were followed by a meticulous literature search of Google Scholar, Web of Science, PubMed/Medline, and Scopus databases to achieve this goal. The results show that the anti-tumor properties vary according to the type of nanoparticle (copper(II) oxide (CuO), silver (Ag), gold (Au), nickel(II) oxide (NiO), nano-curcumin, etc.), the synthesis method (chemical vs. green), and the biological activity assessment method (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Bromodeoxyuridine (BrdU), Cell Counting Kit-8 (CCK8) assays, etc.). NPs derived from green synthesis, such as those based on Moringa oleifera, Photinia glabra, or pomegranate bark, exhibit moderate cytotoxic activity (50% inhibitory concentration (IC50) between 92 and 500 µg/mL) but show good tolerance on normal cells. In contrast, chemically synthesized NPs, such as Cu(II) complexes with 1,3,5-benzenetricarboxylic acid (H3btc) or 1,2,4-triazole (Htrz), show lower IC50 (34–86 µM), indicating more marked cytotoxicity towards cancer cells, although data on their toxicity are sometimes lacking. In addition, multifunctional nanoparticles, such as gold-based nano-conjugates targeting Cluster of Differentiation 271 (CD271) or systems combined with doxorubicin, show remarkable activity with IC50 below 3 µM and enhanced tumor selectivity, positioning them among the most promising candidates for future clinical application against ESCC. The most frequently observed mechanisms of action include induction of apoptosis (↑caspases, ↑p53, ↓Bcl-2), oxidative stress, and inhibition of proliferation. In conclusion, this work identifies several promising nanoparticles (silver nanoparticles derived from Photinia glabra (PG), gold-based nano-immunoconjugates targeting CD271, and silver–doxorubicin complexes) for future pharmaceutical exploitation against ESCC. However, major limitations remain, such as the lack of methodological standardization, insufficient in vivo and clinical studies, and poor industrial transposability. Future prospects include the development of multifunctional nanocomposites, the integration of biomarkers for personalized targeting, and long-term toxicological assessment.

1. Introduction

Esophageal cancer (EC) is among the most aggressive and lethal gastrointestinal cancers, ranking seventh worldwide in terms of incidence and sixth in terms of mortality [1]. Among its histological forms, esophageal squamous cell carcinoma (ESCC) accounts for approximately 90% of cases, with a marked prevalence in Asia and Sub-Saharan Africa [2]. Its exponential progression gives cause for concern, as the number of deaths is likely to double by 2030 according to GLOBOCAN forecasts if nothing is done [1,3]. Conventional therapeutic approaches (surgery, chemotherapy, radiotherapy, or their combination) have significant limitations, due in particular to late diagnosis, 5-year survival rates of less than 20%, systemic toxicity of the agents administered, and the emergence of therapeutic resistance [4,5].
Faced with these challenges, nanotechnology has emerged in recent years as an innovative and promising therapeutic alternative in the field of oncology [6]. Nanoparticles (NPs), whether metallic (gold, silver, copper), based on oxides, polymers, or lipids, offer multiple pharmacological advantages. Their advantages include improved solubility of active ingredients, targeted and controlled release, and above all, increased permeability and retention (EPR effect) at the tumor level, enabling preferential targeting of cancer cells while reducing exposure of healthy tissue [7,8]. Moreover, some NPs possess intrinsic antitumor activity via the generation of reactive oxygen species (ROS), the alteration of mitochondrial function, or the induction of apoptosis, making them potential therapeutic agents in their own right, alone or in combination with other treatments [9,10].
However, despite these advantages, their clinical use in the treatment of esophageal cancer remains limited. This is due in particular to a still incomplete understanding of their mechanisms of action, variability in biocompatibility, problems of selectivity towards tumor cells, and heterogeneous toxicity profiles, particularly towards non-cancerous cells [11,12]. In addition, parameters such as the method of synthesis (chemical vs. biological), the size and surface area of the particles, and their specific functionalization have a strong influence on their biological behavior, underlining the need for a standardized and rational approach to their development.
Against this backdrop, this review provides a critical and exhaustive analysis of the pharmacological effects, mechanisms of action, and toxicity of nanoparticles investigated in the treatment of esophageal cancer. By drawing on recent data from the scientific literature, our aim is to identify the most promising nanoparticles for future clinical application, while highlighting the current limitations and research prospects in this fast-growing field.

2. Methodology

2.1. Data Sources, Search Strategy, and Eligibility Criteria

Original studies published before July 2024 were systematically retrieved from Google Scholar, Medline/PubMed, Scopus, and Web of Science, and critically evaluated in accordance with the PRISMA 2020 guidelines (see PRISMA checklist Table S1) [13]. The review protocol was registered in the Open Science Framework (OSF) associated project: osf.io/a4h5j (Registration DOI: https://doi.org/10.17605/OSF.IO/4FYS9). The search terms included “nanoparticles” OR “nanodrugs” OR “Nanoparticles drugs” AND “anti-esophageal cancer” OR “anti-esophageal squamous cell carcinoma” OR “anti-esophageal adenocarcinoma”. Studies were included if they evaluated, in primary or secondary objectives, the anti-esophageal cancer activity of nanoparticles. This review includes only original research articles. Studies centered on other classes of metabolites were not considered. Review papers, conference proceedings, and editorial pieces were also excluded. The selection process did not impose any limitations regarding the language or publication date of the studies.

2.2. Selection Process and Data Collection

To enhance the organization of the selection and review process, search results were initially imported into Endnote to eliminate duplicates, after which they were transferred to the Rayyan platform for further screening [14]. The authors (E.J.N., G.T.K.) independently screened for the titles and abstracts. After that, a second independent selection was conducted by looking through the complete texts of the articles that had been retained after the first review. Any disagreement was resolved through discussion. Information on nanoparticles, structural characteristics, anti-esophageal cancer activities, mechanism of action, and toxicity of nanoparticles was extracted from the included studies. Regarding biological properties, data such as IC50 values and therapeutic doses were independently retrieved by the authors for each study.

2.3. Risk of Bias and Certainty Assessment

Publication bias was assessed to verify whether the included studies predominantly reported positive results regarding the use of nanoparticles against EC. This assessment was carried out qualitatively by examining the trend of published results and the diversity of sources. Additionally, the level of confidence in the results of the included studies was assessed qualitatively, taking into account methodological rigor, consistency of results between studies, and relevance of sources.

2.4. Synthesis Methods

Data synthesis and analysis followed a structured approach, beginning with a general overview of the included studies, followed by their classification to enable deeper insight. A comprehensive summary table was developed to visually present the main findings. As this is a purely systematic review, a narrative synthesis was performed to integrate and interpret the results [15].

3. Results

3.1. Search Outcomes and Studies Characteristics

The systematic search process resulted in the selection of 20 studies on 20 distinct types of NPs evaluated for their anticancer effects on ESCC cell lines (see PRISMA 2020 flow diagram, Figure 1). Table 1 highlights the wide diversity of nanoparticles, both in terms of their chemical composition and the synthesis methods used. It includes nanoparticles produced by so-called “green” synthesis processes, using plant or microbial extracts [16,17,18,19,20,21,22,23,24,25,26], but also chemically synthesized NPs [27,28,29,30,31]. Some studies are notable for their evaluation of multifunctional nanoparticles or nanoparticles formulated as combined systems, such as nano-immuno-conjugates targeting the CD271 marker [32] or nanoparticles loaded with doxorubicin [25,33]. Analysis of the table also reveals the significant influence of several experimental parameters, particularly the synthesis method, the cell density seeded, and the techniques used to measure biological activity. Indeed, cell density varies significantly from one study to another, oscillating between 1 × 103 and 5 × 105 cells/mL. In terms of cytotoxicity assessment, the MTT assay, BrdU, CCK8, or Resazurin tests are the most used methods [18,28].
Table 1. Description of included studies.
Table 1. Description of included studies.
Type of NanoparticlesTypes of ActivitiesCancer LinesCell Seeded DensityBiological Activities(Country)
References
Copper oxide nanoparticles (CuO NPs) synthesized using viable cells, cell lysate supernatant, and protein extracts of Vibrio sp. VLCCytotoxicity activity by the MTT assayKYSE3010,000 cells/well (KYSE30) and 8000 cells/well (HDF)IC50 = 13.96 mg/L(Iran) [16]
Gold nanoparticles synthesized using Moringa oleifera leaf extractCytotoxicity activity by the MTT assaySNO cells20,000 cells/wellIC50 = 92.01 µg/mL(South Africa) [17]
Cu (II) nanoparticles synthesized using 1,3,5-benzenetricarboxylic acid (H3btc)Cytotoxicity activity by the MTT assayKYSE150, EC109, and EC87122 × 105 cells/wellKYSE150 (IC50 = 86 μM), EC109 (IC50 = 73 μM), and EC8712 (IC50 = 78 μM)(China) [27]
Cu (II) nanoparticles synthesized using 1,2,4-triazole (Htrz)2 × 105 cells/wellKYSE150 (IC50 = 48 μM), EC109 (IC50 = 42 μM), and EC8712 (IC50 = 34 μM)
Nanoparticle colloidal dispersion synthesized using curcumin (nano-curcumin)Cell proliferation was measured using a BrdU incorporation assayOE33, and OE195 × 105 cells/mLOE33 (20%) and OE19 (17%) when treated with 50 mM(The Netherlands) [18]
Copper nanoparticles (Cu NPs) synthesized using Mentha piperita aqueous extractCytotoxicity activity by the MTT assayKYSE-270, OE33, and ESO265 × 104 cells (per square centimeter)IC50 of the Cu NPs were 241, 278, and 240 mg/mL against KYSE-270, OE33, and ESO26(China) [19]
Nickel nanoparticles (NiONPs) synthesized using Calendula officinalis leaf aqueous extractCytotoxicity activity by the MTT assayFLO-1, ESO26, OE33, and KYSE-270
normal esophageal cell line HUVEC
7 × 103FLO-1 (IC50 = 380 µg/mL), ESO26 (IC50 = 263 µg/mL), OE33 (IC50 = 229 µg/mL), and KYSE-270 (IC50 = 251 µg/mL)(China) [20]
Silver nanoparticles synthesized using pomegranate peel extractCytotoxicity activity by the MTT assayKYSE-30, KYSE-50, KYSE-70, KYSE-110, KYSE-270, OE33, ESO26, and FLO-1/KYSE-30 (IC50 = 487 μg/mL), KYSE-50 (IC50 = 500 μg/mL), KYSE-70 (IC50 = 435 μg/mL), KYSE-110 (IC50 = 461 μg/mL), KYSE-270 (IC50 = 285 μg/mL), OE33 (IC50 = 338 μg/mL), ESO26 (IC50 = 253 μg/mL), and FLO-1 (IC50 = 288 μg/mL)(Iran) [21]
Silver nanoparticles synthesized using aqueous Photinia glabra fruit extract (PG-Ag NPs) Cytotoxicity activity by the MTT assay Eca-109/IC50 less than 20 µg/mL(China) [22]
Gold nanoparticles green-synthesized by Rhus coriaria L. fruit aqueous extractCytotoxicity activity by the MTT assay FLO-1, ESO26, OE33, and KYSE-270
Normal esophageal cell line HUVEC
1 × 103 cellKYSE-270 (IC50 = 226 mg/mL), OE33(IC50 = 213 mg/mL), ESO26(IC50 = 267 mg/mL), and FLO-1(IC50 = 294 mg/mL)(China) [23]
Multifunctional nanoparticles co-loaded with AdriamycinCell viability and Cell cycle and apoptosisKYSE510 and Adriamycin-resistant KYSE510 (KYSE510K)1 × 104Inhibit the growth of cancer cells and tumor development by reducing drug efflux by ESCC cells and promoting apoptosis in mice(China) [34]
Gel-nano systemsCytotoxicity activity by the cell counting kit-8 (CCK8) assayTE-1 and KYSE-150 3000 cells/well for CCK8 assay and 10,000 cells/well for apoptosisBoost T-cell immunity and restore p53 activity in mice(China) [28]
Cerium oxide nanoparticlesCell Viability Assay by Resazurin assayYM125,000 cells/wellIC50s = 630 μM after 48 h(Iran) [29]
Gold nano-immuno-conjugate (NIC)Cell viability
Cytotoxicity activity by the MTT assay
HKESC-1 cell line5 × 1052.333 µM ≤ IC50 ≤ 2.998 µM(South Africa) [32]
Copper (Cu) nanoparticlesCytotoxicity activity by the MTT assay OE33, KYSE-270, and ESO26/OE33 (IC50 = 241 mg/mL), ESO26 (IC50 = 278 mg/mL), and KYSE-270 (IC50 = 240 mg/mL) (China) [30]
Gold nanoparticles mediated by potato starchCytotoxicity activity by the MTT assay (KYSE-30 and FLO-1) and normal cells (HUVEC)3 × 103 cellsKYSE-30 (IC50 = 125 μg/mL) and FLO-1 (IC50 = 176 μg/mL)(India) [31]
Silver nanoparticles synthesized using peel of pomegranate Cytotoxicity activity by the MTT assay KYSE-30, KYSE-50, KYSE-70, KYSE-110, KYSE-270, OE33, ESO26 and FLO-1 cell and normal (HUVEC) 410 × 3 cells in 100 microlitersKYSE-30 (IC50 = 487 µg/mL), KYSE-50 (IC50 = 500 µg/mL), KYSE-70 (IC50 = 435 µg/mL), KYSE-110 (IC50 = 461 µg/mL), KYSE-270 (IC50 = 285 µg/mL), OE33 (IC50 = 338 µg/mL), ESO26 (IC50 = 253 µg/mL), and FLO-1 (IC50 = 288 µg/mL)(China) [24]
Hypericum perforatum-loaded nanoparticles Cytotoxicity activity by the MTT assay KYSE-30 (Cat No: 94072011)5000 cells/mLDox NPs (IC50 = ~0.04–0.06 mg/mL) and HP-NPs (IC50 = ~0.6–0.7 mg/mL)(Iran) [25]
Silver-nanoparticles-enhanced doxorubicin Cytotoxicity activity by the MTT assay OE33/IC50 = 2.399 ±1.39 μM(Egypt) [33]
Au NPs decorated over sodium lignosulfonate (NaLS) by using Cydonia oblonga extractCytotoxicity activity by the MTT assay FLO-1, ESO26, OE33, and KYSE-270105FLO-1 (IC50 = 181 µg/mL), ESO26 (IC50 = 130 µg/mL), OE33 (IC50 = 205 µg/mL), and KYSE-270 (IC50 = 133 µg/mL)(China) [26]
HDF: human dermal fibroblast; EAC: esophageal adenocarcinoma; ESCC: esophageal squamous cell carcinoma.

3.2. Anti-Esophageal Cancer Activities of Green-Synthesized Nanoparticles

3.2.1. Copper Oxide Nanoparticles (CuO NPs) Synthesized from Vibrio Luminescent Strain C (Vibrio sp. VLC)

Microorganisms, acting as biofactories, are commonly used for the synthesis of metallic nanoparticles [35]. The products obtained through this method are generally non-toxic, particularly when synthesized using the cell lysate supernatant (CLS) approach, which employs proteins from luminescent bacteria (Vibrio sp.) to reduce Cu2+ ions into CuO nanoparticles [36]. CuO NPs were synthesized by the reduction of copper sulfate and copper nitrate using Vibrio sp. VLC acts as a reducing agent. Polydispersed CuO NPs had a crystallite size of approximately 8.83 nm. Despite their multiple biological properties (antimicrobial, antioxidant, etc.) [37], these biologically synthesized nanoparticles from Vibrio sp. have shown activity against the ESCC cell line KYSE30 (IC50 = 13.96 mg/L) [16]. However, the mechanism through which this nanoparticle exerts its anti-ESCC effect remains unknown to date. Conversely, although no robust toxicological data are available, Nakhaeepour et al. [16] reported low in vitro toxicity on human dermal fibroblast (HDF) cells, with an IC50 value of 48.88 mg/L.

3.2.2. Gold Nanoparticles Synthesized from Moringa oleifera

Renowned for their broad spectrum of biological activities, Moringa oleifera-based nanoparticles are commonly formulated to enhance their pharmacological effects for various therapeutic applications [38]. These nanoparticles have demonstrated antibacterial, antioxidant, anti-inflammatory, hepatoprotective, and anticancer properties [39]. Polydispersed NPs had a crystallite size of approximately 10–20 nm. In this context, Tiloke et al. [17] reported anti-ESCC activity of gold nanoparticles synthesized from M. oleifera leaf extracts against the SNO esophageal cancer cell line, with an IC50 value of 92.01 µg/mL. The anti-ESCC effects of these Moringa-derived nanoparticles are mediated by the induction of apoptosis, through increased activity of caspases-9 and -3/7, elevated levels of p53, SRp30a, Bcl-2-associated X protein (Bax), Second mitochondria-derived activator of caspases/Direct IAP Binding protein with Low pI (Smac/DIABLO), and the 24kDa fragment of Poly(ADP-ribose) polymerase-1 (PARP-1), as well as phosphatidylserine externalization and mitochondrial depolarization. Moreover, they reduce the expression of anti-apoptotic and oncogenic proteins such as B-cell lymphoma 2 (Bcl-2), Heat shock protein 70 (Hsp70), S-phase kinase-associated protein 2 (Skp2), F-box and WD repeat domain-containing 7 alpha (Fbw7α), c-Myc, and adenosine triphosphate (ATP) levels [17]. Although no comprehensive toxicological study has been conducted on M. oleifera nanoparticles to date, Belliraj et al. [40] reported their in vitro hepatoprotective effects on liver cell lines.

3.2.3. Colloidal Nanoparticle Dispersion Synthesized with Curcumin (Nano-Curcumin)

Curcumin is a polyphenolic pigment found in the rhizome of Curcuma longa [41]. Its properties have been widely studied, demonstrating beneficial effects against various diseases, including antibacterial, antifungal, antioxidant, anti-inflammatory, antimutagenic, and anticancer activities [42]. However, to enhance its efficacy, various types of nanoparticle formulations have been developed [43,44], and among them, only the colloidal nano-curcumin dispersion has been evaluated so far on ESCC cell lines. Milano et al. [18] reported the antiproliferative effect of curcumin nanoparticles on human esophageal adenocarcinoma cell (EAC) lines OE33 and OE19, reducing their proliferation by approximately 20% and 17%, respectively, following treatment with a 50 mM concentration. However, these nanoparticles do not appear to induce apoptosis in these cell lines, and their precise mechanism of action against ESCC remains unclear [18]. In vitro, nano-curcumin was found to be non-toxic to the normal esophageal epithelial cell line HET-1A [18]. Although no specific toxicological studies on nano-curcumin have been reported, curcumin itself has been established and approved as non-toxic by the JECFA (Joint FAO/WHO Expert Committee on Food Additives) and the EFSA (European Food Safety Authority) [45].

3.2.4. Copper Nanoparticles (Cu NPs) Synthesized from Mentha piperita

Mentha piperita L. is a widely consumed species across the globe, used in medicinal, industrial, and culinary contexts [46]. It is well-known for its antimicrobial, antioxidant, antispasmodic, anthelmintic, and acetylcholinesterase inhibitory properties [47]. To enhance its therapeutic efficacy, various nanoparticle formulations have been developed, demonstrating excellent wound-healing properties and potent antibacterial activity [48,49,50,51]. Green synthesis of AuNPs was performed by mixing 100 mL HAuCl4·H2O (1 × 10−3 M) with 200 mL R. coriaria fruit extract (20 mg/mL) in a 1:10 ratio, yielding polydispersed nanoparticles with a crystallite size of 19–24 nm. Among these, copper nanoparticles (Cu NPs) synthesized using aqueous extract of M. piperita (40 µg/mL) combined with Cu(NO3)2·3H2O have been specifically evaluated against ESCC cell lines. Zhuang et al. and Zang [19,30] reported antiproliferative effects of these Cu NPs on KYSE-270, OE33, and ESO26 cell lines, with IC50 values of 241, 278, and 240 mg/mL, respectively. Although no specific toxicological studies on M. piperita-derived Cu NPs have been reported, Mentha piperita extracts themselves have been established and approved as non-toxic by the American College of Toxicology [52].

3.2.5. Nickel Nanoparticles (NiONPs) Synthesized from Calendula officinalis

Nanoparticles derived from the medicinal plant Calendula officinalis have recently garnered increasing attention due to their remarkable antioxidant, antibacterial, and anticancer properties [53,54,55]. They were synthesized by mixing 30 mL of Calendula officinalis extract (40 µg/mL) with 30 mL of 0.3 M Cu(NO3)2·3H2O, followed by agitation for 24 h at 60 °C, yielding quasi-spherical gold nanoparticles with an average size of 25–30 nm. To date, only nickel nanoparticles (NiONPs) synthesized from C. officinalis have been investigated as a potential alternative therapy for esophageal cancer [20]. These nanoparticles have demonstrated significant cytotoxic activity against several EC cell lines, including FLO-1 (IC50 = 380 µg/mL), ESO26 (IC50 = 263 µg/mL), OE33 (IC50 = 229 µg/mL), and KYSE-270 (IC50 = 251 µg/mL) [20]. However, the exact mechanisms underlying their anticancer effects remain unknown. Toxicological assessments suggest that nickel and silver nanoparticles derived from C. officinalis extracts are non-toxic to normal human endothelial cells (HUVECs) [20,55]. Nevertheless, further dedicated toxicological studies are warranted, although previous toxicity evaluations of C. officinalis extract alone in rats and mice have indicated a relatively safe profile [56].

3.2.6. Silver Nanoparticles Derived from Pomegranate Peel Extract

Silver nanoparticles synthesized from pomegranate peel extract have recently gained increasing attention due to their exceptional properties, including antioxidant, anti-inflammatory, antibacterial, and anticancer activities [57,58]. The resulting polydispersed nanoparticles had a crystallite size of ~45.55 nm. To date, only silver nanoparticles derived from this extract have been investigated as a potential therapeutic alternative for the treatment of EC [21]. These nanoparticles have demonstrated notable cytotoxic effects against multiple EC cell lines, including KYSE-30 (IC50 = 487 µg/mL), KYSE-50 (IC50 = 500 µg/mL), KYSE-70 (IC50 = 435 µg/mL), KYSE-110 (IC50 = 461 µg/mL), KYSE-270 (IC50 = 285 µg/mL), OE33 (IC50 = 338 µg/mL), ESO26 (IC50 = 253 µg/mL), and FLO-1 (IC50 = 288 µg/mL). The IC50 values for these cell lines ranged from 253 to 500 µg/mL [21]. However, the underlying mechanisms of their anticancer effects on ESCC remain unclear, and comprehensive toxicological data are still lacking.

3.2.7. Silver Nanoparticles from Photinia glabra (PG) Fruit Extract

Traditionally valued for their medicinal properties (such as anthelmintic, anti-dysenteric, anti-hemorrhoidal, and anti-jaundice effects) [59], PG fruit extracts have recently attracted growing interest, particularly in the form of silver nanoparticles (AgNPs). Silver nitrate solution was reduced using aqueous PG fruit extract to form spherical PG-Ag nanoparticles, sized approximately 39–77 nm. These Photinia glabra-based nanoparticles represent an innovative approach in the fight against various human diseases, including EC. Namulinda et al. [22] reported a remarkable antiproliferative activity of these AgNPs against the ESCC line Eca-109, with an IC50 value below 20 µg/mL. However, the underlying mechanisms of action against ESCC, as well as their toxicological profiles, remain poorly understood and require further investigation.

3.2.8. Gold Nanoparticles Green-Synthesized from Rhus coriaria L. Fruit Extract

Known for its well-documented biological activities (including antioxidant, anti-inflammatory, hypoglycemic, and hypolipidemic properties) [60], Rhus coriaria has been employed as an active component in various types of nanoparticles with broad pharmacological applications [61,62,63]. Green synthesis of AuNPs was performed by mixing 100 mL HAuCl4·H2O (1 × 10−3 M) with 200 mL R. coriaria fruit extract (20 mg/mL) in a 1:10 ratio, yielding polydispersed nanoparticles with a crystallite size of 19–24 nm. Among them, gold nanoparticles synthesized from the aqueous extract of Rhus coriaria represent the only type tested so far against both ESCC and EAC cell lines. These nanoparticles exhibited antiproliferative activity against FLO-1, ESO26, OE33, and KYSE-270 cell lines, with IC50 values of 226, 213, 267, and 294 mg/mL, respectively [23]. While the specific mechanism of action against ESCC remains unclear, recent findings by Mongy and Shalaby [63] suggest that Rhus coriaria-based nanoparticles may exert their anticancer effects through the modulation of apoptosis and metastasis-related genes.

3.2.9. Gold Nanoparticles Mediated by Potato Starch

Gold nanoparticles mediated by potato starch are synthesized through the mixing of HAuCl4 (1.5 mM) with potato starch (1% w/v) [31,64]. Quasi-spherical PS-Au nanoparticles (25–30 nm) were prepared by adding 1.5 mM HAuCl4 to 1% PS solution, adjusting to pH 11, and sonication at 60 °C for 30 min. These nanoparticles have demonstrated various biological activities, notably antioxidant and anticancer effects [65]. Specifically, their anti-ESCC activity was reported by Liu et al. [31] on the KYSE-30 and FLO-1 cell lines, with IC50 values of 125 and 176 µg/mL, respectively. Although the exact mechanisms of action on ESCC cells and the comprehensive toxicological profile of these nanoparticles remain unknown, Li et al. [65] reported an IC50 value greater than 1000 µg/mL on normal primary human umbilical vein endothelial cells (HUVEC), suggesting low toxicity.

3.2.10. Sodium Lignosulfonate (NaLS) Nanoparticles from Cydonia oblonga

These nanoparticles are synthesized through a combined process involving HAuCl4, sodium lignosulfonate (NaLS), and aqueous extract of Cydonia oblonga. Polydispersed NPs had a crystallite size of approximately 15–25 nm. Their antioxidant and anti-ESCC activities were investigated by Lin et al. [26], who reported antiproliferative effects against the FLO-1, ESO26, OE33, and KYSE-270 cell lines, with IC50 values of 181, 130, 205, and 133 µg/mL, respectively. Although the precise mechanisms of action on ESCC cells and comprehensive toxicological data remain unknown, Lin et al. [26] observed IC50 values greater than 1000 µg/mL on normal primary human umbilical vein endothelial cells (HUVEC), suggesting low cytotoxicity to normal cells.

3.3. Anti-Esophageal Cancer Activities of Chemically Synthesized NPs

3.3.1. Cerium Oxide Nanoparticles (CeO2 NPs)

Cerium oxide nanoparticles consist of a cerium core surrounded by an oxygen atom lattice [66]. These nanoparticles are well known for their ability to facilitate targeted drug delivery [67] as well as their multiple pharmacological effects, including antioxidant [68,69], anti-inflammatory [70], genoprotective [71], anti-obesity [72], neuroprotective [73], and anticancer properties [74]. Regarding their anticancer activity, an antiproliferative effect was reported by Javid et al. [29] on the ESCC YM1 cell line, with an IC50 of 630 µM after 48 h of incubation. Although the exact mechanism of action on ESCC cells remains unclear, Gunasekaran et al. [74] demonstrated that these nanoparticles induce oxidative stress in lung cancer cell lines. Toxicological studies in vitro have revealed high toxicity, showing over 80% inhibition at very low concentrations in bioluminescence assays, and a 50% lethal concentration (LC50) of 0.012 mg/mL in Daphnia magna mortality tests [75]. However, in vivo studies indicate that cerium oxide nanoparticles have no significant effects on biochemical markers of renal and hepatic function [76].

3.3.2. Cu2(btc)(trz)3-NPs and Cu5(Hbtc)4(trz)2(H2O)4-NPs

These Cu(II)-based nanoparticles are synthesized respectively using 1,3,5 benzenetricarboxylic acid (H3btc) and 1,2,4-triazole (Htrz). The study by Zhu et al. [27] reported their potential applicability in the treatment of esophageal cancer. Cu2(btc)(trz)3-NPs exhibited cytotoxic activity against KYSE150, EC109, and EC8712 esophageal squamous carcinoma cell lines, with IC50 values of 48 µM, 42 µM, and 34 µM, respectively. In contrast, Cu5(Hbtc)4(trz)2(H2O)4-NPs showed IC50 values of 86 µM, 73 µM, and 78 µM on the same cell lines [27]. These findings suggest a higher cytotoxic potency for the [Cu2(btc)(trz)] complex compared to its Hbtc-based counterpart.

3.4. Anti-Esophageal Cancer Activities of Multifunctional and Combined System Nanoparticles

3.4.1. Gold Nano-Immuno-Conjugates (NIC)

Nano-immuno-conjugates (NIC) are synthesized through the combination of AlPcS4Cl-AuNPs and AlPcS4Cl-AuNPs conjugated with anti-cluster of differentiation (Anti-CD271) antibodies via chemical reactions and physical interactions [32]. The chemical reactions involve covalent bonding of the anti-CD antibody to the gold nanoparticles (AuNPs), while the physical interactions are based on electrostatic forces between the components [77]. The resulting nanoparticles had a size of 7.7 ± 0.8 nm. These gold nano-immuno-conjugates offer several advantages, including applications in diagnostics, drug delivery, cancer therapy, and treatment of various diseases [78,79]. Among their anticancer properties, the anti-ESCC effect of NIC has been demonstrated by Didamson et al. [32] on the HKESC-1 cell line, with IC50 values ranging from 2.333 to 2.998 µM. However, the underlying mechanisms of these effects on ESCC lines and the toxicological profile remain poorly understood.

3.4.2. Silver Nanoparticles Enhanced Doxorubicin

This nanoparticle is synthesized through the combination of silver nitrate and doxorubicin (Dox) via citrate reduction, resulting in a final silver concentration of 5 × 10−3 mol/L [33]. Silver nanoparticles (~15 ± 1.5 nm) were prepared at 5 × 10−3 M via citrate reduction of 0.0850 g AgNO3 in 100 mL double-distilled water. This formulation enhances the intracellular delivery of Dox and improves its anticancer therapeutic index [80,81,82]. It has been applied to enhance doxorubicin efficacy in OE33 EC cells, reducing the IC50 from 8.0 µM with free doxorubicin to 2.399 µM with the silver nanoparticle–doxorubicin complex [33]. In vitro toxicological studies of this silver–doxorubicin nanoparticle formulation (20 µM AgNPs/0.3 µM Dox) revealed no significant effects on cell migration, apoptosis, or the expression of Bax, Bcl-2, and P53 in normal cardiac cell lines (H9c2) [80].
Table 2 provides a summary of the biological activities, toxicological profiles, and mechanisms of action explored to date for nanoparticles tested on EC cell lines.

4. Discussion

This study aimed to identify nanoparticles (NPs) with potential effects against ESCC, assess their toxicological profiles, and explore therapeutic perspectives. A total of twenty nanocomplexes exhibiting anti-ESCC activity were identified. The data analyzed highlight the growing scientific interest in NPs as potential therapeutic agents for the treatment of this aggressive form of esophageal cancer. The included studies revealed a remarkable diversity in nanoparticle types, synthesis approaches, cell lines used, and biological outcomes observed.
A global comparative analysis allowed for the classification of these NPs into three main categories: green-synthesized nanoparticles (derived from plant or microbial extracts), chemically synthesized nanoparticles, and combined or multifunctional formulations, which incorporate active pharmaceutical ingredients or specific targeting systems. According to the criteria established by Kamsu and Ndebia [83], the most promising NPs are those with an IC50 below 20 µg/mL or µM, a threshold below which cytotoxicity is considered pharmacologically significant. In this regard, three nanocomplexes clearly stand out: silver nanoparticles derived from Photinia glabra (IC50 < 20 µg/mL on the Eca-109 line), gold nano-immunoconjugates targeting the CD271 receptor (IC50 = 2.3–2.9 µM), and the silver–doxorubicin complex (IC50 = 2.4 µM on OE33). The IC50 values of gold nano-immunoconjugates and the silver–doxorubicin complex are very similar, indicating comparable potency in inhibiting esophageal cancer cell proliferation. When compared to conventional chemotherapeutic agents, these nanoparticle-based systems may offer similar efficacy with the potential for improved selectivity and reduced toxicity. Conversely, some formulations, such as copper NPs derived from Mentha piperita (IC50 > 240 mg/mL) or gold NPs from Rhus coriaria (IC50 > 200 mg/mL), showed significantly lower activity and are considered “poorly active” according to the same threshold.
The type of metal used also influences anti-ESCC activity. Metal-based NPs, such as silver or copper, generally exert cytotoxic effects through the generation of reactive oxygen species (ROS), leading to oxidative stress and apoptosis [84]. In contrast, gold nanoparticles can be functionalized to specifically target cancer cells, enhancing their efficacy while reducing toxicity to healthy cells [85]. Metal-based NPs (e.g., Ag/Cu) induce reactive oxygen species (ROS)-mediated cell killing by releasing ions, whereas gold nanoparticles can be functionalized for active targeting (e.g., CD271 antibody), which improves tumor selectivity and reduces non-specific toxicity via the enhanced permeability and retention (EPR) effect and internalization.
Regarding the mechanisms of action, only a few studies have clarified the molecular pathways involved in the anticancer effects. Gold NPs from Moringa oleifera were shown to induce apoptosis by increasing the expression of p53 and caspases 3/7 and 9, and by downregulating oncogenic proteins such as Bcl-2, Hsp70, and c-Myc [17]. Gold-based nano-immunoconjugates appear to act through selective targeting of CD271-expressing cells, promoting their internalization [32]. Meanwhile, cerium oxide NPs (CeO2), although weakly active on YM1 cells (IC50 = 630 µM), are known to induce strong oxidative stress, as demonstrated in lung cancer models [74]. For most plant-derived NPs, however, the precise molecular mechanisms remain poorly understood, limiting their current pharmacological application.
It is also important to note that the variability in reported activity is not solely attributable to the chemical nature of the nanoparticles but also strongly depends on methodological parameters. Cell density, which varied widely among studies (from 1 × 103 to 5 × 105 cells/mL), directly impacts the effective exposure of cells to NPs. Higher densities can lead to increased consumption of nutrients and oxygen, altering cellular responses [86]. Additionally, cytotoxicity assessment methods (such as MTT, CCK-8, BrdU, or Resazurin assays) differ in sensitivity and specificity, which contributes to variations in IC50 values, even across studies using the same cell lines. Future research should aim to standardize experimental conditions, particularly cell density, exposure time, and assay methods, to ensure more rigorous comparison and reproducibility.
Finally, the issue of nanoparticle safety is crucial for therapeutic development [87]. In this context, several green-synthesized NPs show great promise. Gold NPs synthesized from Rhus coriaria and NaLS-AuNPs exhibited IC50 values above 1000 µg/mL on HUVEC, indicating low cytotoxicity. Similarly, formulations based on curcumin and silver NPs from Photinia glabra were non-toxic to normal cells tested, including HET-1A and HUVEC lines [18,22]. On the other hand, cerium oxide NPs, despite showing limited anticancer activity, displayed significant in vitro toxicity, with over 80% inhibition in luminescence assays, although in vivo results were more reassuring [75,76].
Moreover, the mechanism of action is closely related to toxicity. NPs that induce apoptosis via the mitochondrial pathway, such as silver NPs, may be effective but pose a risk to healthy cells due to ROS production [88]. In contrast, functionalized NPs targeting cancer cells specifically, such as gold-based systems, offer greater safety by minimizing off-target effects [89]. These findings highlight the urgent need for systematic and comprehensive toxicological evaluations, including normal cell models and in vivo assays, to ensure an acceptable benefit/risk balance in the therapeutic use of nanoparticles.

5. Limitations and Future Perspectives

Despite their promising pharmacological activity against ESCC, several limitations remain. These include the reproducibility of synthesis methods, variability in biological responses across different cell lines, and the need for standardized experimental protocols. Industrial translation of NPs remains particularly challenging. Current synthesis approaches, especially green or plant-mediated methods, are subject to batch-to-batch variability due to differences in plant extracts, complicating standardization. Large-scale production further encounters critical issues related to purification, sterilization, and compliance with Good Manufacturing Practice standards, all of which can increase production costs. To overcome these challenges, future research should prioritize scalable and reproducible green synthesis techniques, such as microfluidics or continuous-flow systems, which allow precise control over particle size, morphology, and uniformity. Integrating nanoparticles into drug-device combination delivery systems may also facilitate clinical translation while maintaining safety and efficacy. Addressing these industrial and translational bottlenecks is essential to fully exploit the therapeutic potential of NPs against ESCC. Looking forward, the development of multifunctional nanoparticles capable of specific cancer cell targeting, therapeutic delivery, and real-time monitoring of treatment efficacy represents a key objective. In parallel, comprehensive studies on long-term toxicity and environmental impact are crucial to ensure the safety of these nanoparticles before their widespread clinical application.

6. Conclusions

In summary, this review highlights the significant potential of nanoparticles as innovative therapeutic agents in the treatment of ESCC. Among the different categories studied, certain formulations stand out for their remarkable cytotoxic efficacy, particularly silver nanoparticles derived from Photinia glabra, gold-based nano-immunoconjugates targeting CD271, and silver–doxorubicin complexes. However, the diversity of experimental methods and the lack of sufficient toxicological data still limit their evaluation for clinical application. Therefore, it is essential to continue research by harmonizing evaluation protocols and deepening toxicological studies to ensure safety and efficacy, thus paving the way for new therapeutic strategies against ESCC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/applnano6030018/s1.

Author Contributions

The conceptualization, writing, and preparation of this manuscript were carried out equally by G.T.K. and E.J.N. All authors have read and agreed to the published version of the manuscript.

Funding

We gratefully acknowledge the financial support provided by the Chemical Industries Education and Training Authority (CHIETA) and the Medical Research Council (MRC) for this study.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This study did not involve the creation or analysis of new data; therefore, data sharing does not apply.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Liu, C.Q.; Ma, Y.L.; Qin, Q.; Wang, P.H.; Luo, Y.; Xu, P.F.; Cui, Y. Epidemiology of esophageal cancer in 2020 and projections to 2030 and 2040. Thorac. Cancer 2023, 1, 3–11. [Google Scholar] [CrossRef] [PubMed]
  3. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  4. Pennathur, A.; Gibson, M.K.; Jobe, B.A.; Luketich, J.D. Oesophageal Carcinoma. Lancet 2013, 381, 400–412. [Google Scholar] [CrossRef] [PubMed]
  5. Lagergren, J.; Smyth, E.C.; Cunningham, D.; Lagergren, P. Oesophageal Cancer. Lancet 2017, 390, 2383–2396. [Google Scholar] [CrossRef]
  6. Ammar, M.M.; Ali, R.; Abd Elaziz, N.A.; Habib, H.; Abbas, F.M.; Yassin, M.T.; Maniah, K.; Abdelaziz, R. Nanotechnology in Oncology: Advances in Biosynthesis, Drug Delivery, and Theranostics. Discov. Onc. 2025, 16, 1172. [Google Scholar] [CrossRef]
  7. Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for Cancer Therapy. Nat. Nanotechnol. 2007, 2, 751–760. [Google Scholar] [CrossRef] [PubMed]
  8. Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-Based Medicines: A Review of FDA-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016, 33, 2373–2387. [Google Scholar] [CrossRef]
  9. Zhang, L.; Li, X.; Yue, G.; Guo, L.; Hu, Y.; Cui, Q.; Wang, J.; Tang, J.; Liu, H. Nanodrug systems for therapy and diagnosis of esophageal cancer. Front. Bioeng. Biotechnol. 2023, 11, 1233476. [Google Scholar] [CrossRef]
  10. Siddiqi, K.S.; Husen, A. Green Synthesis, Characterization and Uses of Palladium/Platinum Nanoparticles. Nanoscale Res. Lett. 2018, 13, 482. [Google Scholar] [CrossRef]
  11. Bondarenko, O.; Mortimer, M.; Kahru, A.; Feliu, N.; Javed, I.; Kakinen, A.; Lin, S.; Xia, T.; Song, Y.; Davis, T.P.; et al. Nanotoxicology and Nanomedicine: The Yin and Yang of Nano-Bio Interactions for the New Decade. Nano Today 2021, 39, 101184. [Google Scholar] [CrossRef]
  12. Sharifi, S.; Behzadi, S.; Laurent, S.; Forrest, M.L.; Stroeve, P.; Mahmoudi, M. Toxicity of nanomaterials. Chem. Soc. Rev. 2012, 41, 2323–2343. [Google Scholar] [CrossRef]
  13. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  14. Ndebia, E.J.; Kamsu, G.T. Harnessing the Power of Natural Terpenoid Compounds against Esophageal Squamous Cell Carcinoma: A Systematic Review. Future Pharmacol. 2025, 5, 21. [Google Scholar] [CrossRef]
  15. Kamsu, G.T.; Ndebia, E.J. Uncovering Risks Associated with Smoking Types and Intensities in Esophageal Cancer within High-Prevalence Regions in Africa: A Comprehensive Meta-Analysis. Cancer Epidemiol. Biomark. Prev. 2024, 33, 874–883. [Google Scholar] [CrossRef]
  16. Nakhaeepour, Z.; Mashreghi, M.; Matin, M.M.; NakhaeiPour, A.; Housaindokht, M.R. Multifunctional CuO Nanoparticles with Cytotoxic Effects on KYSE30 Esophageal Cancer Cells, Antimicrobial and Heavy Metal Sensing Activities. Life Sci. 2019, 234, 116758. [Google Scholar] [CrossRef] [PubMed]
  17. Tiloke, C.; Phulukdaree, A.; Anand, K.; Gengan, R.M.; Chuturgoon, A.A. Moringa oleifera gold nanoparticles modulate oncogenes, tumor suppressor genes, and caspase-9 splice variants in A549 cells. J. Cell. Biochem. 2015, 117, 2302–2314. [Google Scholar] [CrossRef]
  18. Milano, F.; Mari, L.; van de Luijtgaarden, W.; Parikh, K.; Calpe, S.; Krishnadath, K.K. Nano-Curcumin Inhibits Proliferation of Esophageal Adenocarcinoma Cells and Enhances the T Cell Mediated Immune Response. Front. Oncol. 2013, 3, 137. [Google Scholar] [CrossRef]
  19. Zhuang, X.; Kang, Y.; Zhao, L.; Guo, S. Design and Synthesis of Copper Nanoparticles for the Treatment of Human Esophageal Cancer: Introducing a Novel Chemotherapeutic Supplement. J. Exp. Nanosci. 2022, 17, 274–284. [Google Scholar] [CrossRef]
  20. Zhang, Y.; Mahdavi, B.; Mohammadhosseini, M.; Rezaei-Seresht, E.; Paydarfard, S.; Qorbani, M.; Karimian, M.; Abbasi, N.; Ghaneialvar, H.; Karimi, E. Green Synthesis of NiO Nanoparticles Using Calendula officinalis Extract: Chemical Characterization, Antioxidant, Cytotoxicity, and Anti-Esophageal Carcinoma Properties. Arab. J. Chem. 2021, 14, 103105. [Google Scholar] [CrossRef]
  21. Fu, Y.; Li, J.; Almasi, M. Pomegranate Peel Extract-Mediated Green Synthesis of Silver Nanoparticles: Evaluation of Cytotoxicity, Antioxidant, and Anti-Esophageal Cancer Effects. ChemistrySelect 2023, 8, e202204841. [Google Scholar] [CrossRef]
  22. Namulinda, T.; Bao, L.L.; Kwetegyeka, J.; Gumula, I.; Yan, Y.J.; Chen, Z.L. Antibacterial and Anticancer Activities of Green-Synthesized Silver Nanoparticles Using Photinia glabra Fruit Extract. Nanomedicine 2023, 18, 987–1002. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, J.; Zangeneh, A.; Zangeneh, M.M.; Guo, B. Antioxidant, Cytotoxicity, Anti-Human Esophageal Squamous Cell Carcinoma, Anti-Human Caucasian Esophageal Carcinoma, Anti-Adenocarcinoma of the Gastroesophageal Junction, and Anti-Distal Esophageal Adenocarcinoma Properties of Gold Nanoparticles Green Synthesized by Rhus coriaria L. Fruit Aqueous Extract. J. Exp. Nanosci. 2020, 15, 202–216. [Google Scholar] [CrossRef]
  24. Fan, P. Green Formulation, Chemical Characterization, Anti-Esophageal Cancer, Cytotoxicity and Antioxidant Potentials of Silver Nanoparticles Containing Plant Extract. SSRN Electron. J. 2022, 19. Available online: https://papers.ssrn.com/sol3/papers.cfm?abstract_id=4238345 (accessed on 10 July 2025).
  25. Amjadi, I.; Mohajeri, M.; Borisov, A.; Hosseini, M.S. Antiproliferative Effects of Free and Encapsulated Hypericum perforatum L. Extract and Its Potential Interaction with Doxorubicin for Esophageal Squamous Cell Carcinoma. J. Pharmacopunct. 2019, 22, 102–108. [Google Scholar] [CrossRef]
  26. Lin, X.; Liu, Y.; Pan, J.; Li, M.; Wang, J.; Fang, G.; Shang, Y. Chemical Characterization, Antioxidant and Anti-Esophageal Cancer Properties of Au NPs over Sodium Lignosulfonate Formulated by Cydonia oblonga. Inorg. Chem. Commun. 2024, 160, 111985. [Google Scholar] [CrossRef]
  27. Zhu, K.; Chen, L.; Jin, X.; Qu, C. Two Cu(II) Coordination Polymers Based on Benzene-1,3,5-Tricarboxylate and 1,2,4-Triazolide Ligands: Their Crystal Structures and Application of Nanoparticles in Anti-Esophageal Cancer Activity Evaluation. Struct. Chem. 2019, 30, 1485–1494. [Google Scholar] [CrossRef]
  28. Gong, K.; Lin, J.; Chen, X.; Duan, Y.; Zhang, J.; Yu, J.; Wang, J.; Sun, R.; Li, J.; Duan, Y. Thermosensitive Gel-Nano System against Esophageal Cancer via Restoring p53 Activity and Boosting T-Cell Immunity. J. Control. Release 2024, 371, 111–125. [Google Scholar] [CrossRef]
  29. Javid, H.; Hashemy, S.I.; Heidari, M.F.; Esparham, A.; Gorgani-Firuzjaee, S. The Anticancer Role of Cerium Oxide Nanoparticles by Inducing Antioxidant Activity in Esophageal Cancer and Cancer Stem-Like ESCC Spheres. Biomed. Res. Int. 2022, 2022, 3268197. [Google Scholar] [CrossRef]
  30. Zang, K. Nanotechnology-Enhanced Chemoradiotherapy Using Copper and Gold Nanoparticles for Esophageal Cancer. J. Angiother. 2024, 8, 1–9. [Google Scholar] [CrossRef]
  31. Liu, M.; Xue, X.; Karmakar, B.; Eltantawy, W.; El-Kott, A.F.; El Nashar, E.M.; Abd-Ella, E.M. Sonochemical Synthesis of Gold Nanoparticles Mediated by Potato Starch: Its Performance in the Treatment of Esophageal Cancer. Open Chem. 2024, 22, 20230193. [Google Scholar] [CrossRef]
  32. Didamson, O.C.; Chandran, R.; Abrahamse, H. Synthesis, Characterisation, and Anti-Tumour Activity of Nano-Immuno-Conjugates for Enhanced Photodynamic Therapy of Oesophageal Cancer Stem Cells. Biomed. Pharmacother. 2024, 181, 117693. [Google Scholar] [CrossRef] [PubMed]
  33. Moawad, M.; Youssef, A.M.; Elsherbeni, S.A.E.; Fahmy, A.M.; El-Ghannam, G. Silver Nanoparticles Enhanced Doxorubicin Treatment for Improving Their Efficacy against Esophageal Cancer Cells. Egypt. J. Chem. 2024, 67, 505–512. [Google Scholar] [CrossRef]
  34. Zhang, X.; Wang, M.; Feng, J.; Qin, B.; Zhang, C.; Zhu, C.; Liu, W.; Wang, Y.; Liu, W.; Huang, L.; et al. Multifunctional Nanoparticles Co-Loaded with Adriamycin and MDR-Targeting siRNAs for Treatment of Chemotherapy-Resistant Esophageal Cancer. J. Nanobiotechnol. 2022, 20, 166. [Google Scholar] [CrossRef]
  35. Ghorbani, H.R.; Mehr, F.P.; Poor, A.K. Extracellular Synthesis of Copper Nanoparticles Using Culture Supernatants of Salmonella typhimurium. Orient J. Chem. 2015, 31, 527–529. [Google Scholar] [CrossRef]
  36. Gahlawat, G.; Choudhury, A.R. A Review on the Biosynthesis of Metal and Metal Salt Nanoparticles by Microbes. RSC Adv. 2019, 9, 12944–12967. [Google Scholar] [CrossRef] [PubMed]
  37. Rehana, D.; Mahendiran, D.; Kumar, R.S.; Rahiman, A.K. Evaluation of Antioxidant and Anticancer Activity of Copper Oxide Nanoparticles Synthesized Using Medicinally Important Plant Extracts. Biomed. Pharmacother. 2017, 89, 1067–1077. [Google Scholar] [CrossRef]
  38. Devkota, H.P.; Gaire, B.P.; Hori, K.; Subedi, L.; Adhikari-Devkota, A.; Belwal, T.; Paudel, K.R.; Jha, N.K.; Singh, S.K.; Chellappan, D.K.; et al. The Science of Matcha: Bioactive Compounds, Analytical Techniques and Biological Properties. Trends Food Sci. Technol. 2021, 118, 735–743. [Google Scholar] [CrossRef]
  39. Perumalsamy, H.; Balusamy, S.R.; Sukweenadhi, J.; Nag, S.; MubarakAli, D.; Farh, M.E.-A.; Vijay, H.; Rahimi, S. A Comprehensive Review on Moringa oleifera Nanoparticles: Importance of Polyphenols in Nanoparticle Synthesis, Nanoparticle Efficacy and Their Applications. J. Nanobiotechnol. 2024, 22, 71. [Google Scholar] [CrossRef]
  40. Belliraj, T.S.; Nanda, A.; Ragunathan, R. In-Vitro Hepatoprotective Activity of Moringa oleifera Mediated Synthesis of Gold Nanoparticles. J. Chem. Pharm. Res. 2015, 7, 781–788. [Google Scholar]
  41. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
  42. Ciuca, M.D.; Racovita, R.C. Curcumin: Overview of Extraction Methods, Health Benefits, and Encapsulation and Delivery Using Microemulsions and Nanoemulsions. Int. J. Mol. Sci. 2023, 24, 8874. [Google Scholar] [CrossRef]
  43. Gera, M.; Sharma, N.; Ghosh, M.; Huynh, D.L.; Lee, S.J.; Min, T.; Kwon, T.; Jeong, D.K. Nanoformulations of Curcumin: An Emerging Paradigm for Improved Remedial Application. Oncotarget 2017, 8, 66680–66698. [Google Scholar] [CrossRef]
  44. Morsy, M.K.; Al-Dalain, S.Y.; Haddad, M.A.; Diab, M.M.E.; Abdeen, A.; Ibrahim, S.F.; Shukry, M.; Fericean, L.; Ghamry, H.I.; Abdelaziz, M.; et al. Curcumin Nanoparticles as a Natural Antioxidant and Antimicrobial Preservative against Foodborne Pathogens in Processed Chicken Fingers. Front. Sustain. Food Syst. 2023, 7, 1267075. [Google Scholar] [CrossRef]
  45. Kocaadam, B.; Şanlier, N. Curcumin, an Active Component of Turmeric (Curcuma longa), and Its Effects on Health. Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef]
  46. Gholamipourfard, K.; Salehi, M.; Banchio, E. Mentha piperita Phytochemicals in Agriculture, Food Industry and Medicine: Features and Applications. S. Afr. J. Bot. 2021, 141, 183–195. [Google Scholar] [CrossRef]
  47. McKay, D.L.; Blumberg, J.B. A Review of the Bioactivity and Potential Health Benefits of Peppermint Tea (Mentha piperita L.). Phytother. Res. 2006, 20, 619–633. [Google Scholar] [CrossRef]
  48. Valencia, G.A.; Vercik, L.C.O.; Ferreira, L.G.; Llanos, J.H.R.; Vercik, A. Synthesis and Characterisation of Gold Nanoparticles Using Mentha piperita Leaf Extract: A Green, Non-Toxic and Rapid Method. Int. J. Nano Biomater. 2015, 5, 181–192. [Google Scholar] [CrossRef]
  49. Hussain, Z.; Jahangeer, M.; Sarwar, A.; Ullah, N.; Aziz, T.; Alharbi, M.; Alshammari, A.; Alasmari, A.F. Synthesis and Characterization of Silver Nanoparticles Mediated by the Mentha piperita Leaves Extract and Exploration of Its Antimicrobial Activities. J. Chil. Chem. Soc. 2023, 68, 5865–5870. [Google Scholar] [CrossRef]
  50. Aftab, R.; Akbar, F.; Afroz, A.; Asif, A.; Khan, M.R.; Rehman, N.; Zeeshan, N. Mentha piperita Silver Nanoparticle-Loaded Hydrocolloid Film for Enhanced Diabetic Wound Healing in Rats. J. Wound Care 2024, 33, xlviii–lx. [Google Scholar] [CrossRef] [PubMed]
  51. Yousaf, R.; Zain, M.; Ijaz, U. Green Synthesis and Evaluation of Mentha piperita Mediated Iron Nanoparticles for In Vitro Antibacterial Activity. J. Xi’an Shiyou Univ. 2024, 20, 653–663. Available online: https://papers.ssrn.com/sol3/papers.cfm?abstract_id=4970247 (accessed on 12 July 2025).
  52. American College of Toxicology (ACT). Final Report on the Safety Assessment of Mentha piperita (Peppermint) Oil, Mentha piperita (Peppermint) Leaf Extract, Mentha piperita (Peppermint) Leaf, and Mentha piperita (Peppermint) Leaf Water. Int. J. Toxicol. 2001, 20 (Suppl. 3), 61–73. [Google Scholar] [CrossRef]
  53. Olfati, A.; Kahrizi, D.; Balaky, S.T.J.; Sharifi, R.; Tahir, M.B.; Darvishi, E. Green Synthesis of Nanoparticles Using Calendula officinalis Extract from Silver Sulfate and Their Antibacterial Effects on Pectobacterium carotovorum. Inorg. Chem. Commun. 2021, 125, 108439. [Google Scholar] [CrossRef]
  54. Wei, X.; Liu, Y.; El-kott, A.E.; Ahmed, A.E.; Khames, A. Calendula officinalis-Based Green Synthesis of Titanium Nanoparticle: Fabrication, Characterization, and Evaluation of Human Colorectal Carcinoma. J. Saudi Chem. Soc. 2021, 25, 101343. [Google Scholar] [CrossRef]
  55. Xu, Y.; Mahdavi, B.; Zangeneh, M.M.; Zangeneh, A.; Qorbani, M.; Paydarfard, S. Calendula officinalis Green-Mediated Silver Nanoparticles: Formulation, Characterization and Assessment of Colorectal Cancer Activities. Arch. Med. Sci. 2021. [Google Scholar] [CrossRef]
  56. International Journal of Toxicology (IJT). Final Report on the Safety Assessment of Calendula officinalis Extract and Calendula officinalis. Int. J. Toxicol. 2001, 20 (Suppl. 2), 13–20. [Google Scholar] [CrossRef] [PubMed]
  57. Alnehia, A.; Al-Odayni, B.; Al-Sharabi, A.; Al-Hammadi, A.H.; Saeed, W.S. Pomegranate Peel Extract-Mediated Green Synthesis of ZnO-NPs: Extract Concentration-Dependent Structure, Optical, and Antibacterial Activity. J. Chem. 2022, 2022, 9647793. [Google Scholar] [CrossRef]
  58. Azmat, F.; Safdar, M.; Ahmad, H.; Khan, M.R.J.; Abid, J.; Naseer, M.S.; Aggarwal, S.; Imran, A.; Khalid, U.; Zahra, S.M.; et al. Phytochemical Profile, Nutritional Composition of Pomegranate Peel and Peel Extract as a Potential Source of Nutraceutical: A Comprehensive Review. Food Sci. Nutr. 2024, 12, 661–674. [Google Scholar] [CrossRef]
  59. Plants For A Future (PFAF). Photinia glabra-(Thunb.) Franch. & Sav; Plants For A Future: Dawlish, UK, 2025; Available online: https://pfaf.org/user/Plant.aspx?LatinName=Photinia+glabra (accessed on 8 July 2025).
  60. Abu-Reidah, I.M.; Jamous, R.M.; Ali-Shtayeh, M.S. Phytochemistry, Pharmacological Properties and Industrial Applications of Rhus coriaria L. (Sumac). Jordan J. Biol. Sci. 2014, 7, 233–244. [Google Scholar] [CrossRef]
  61. Gur, T. Green Synthesis, Characterizations of Silver Nanoparticles Using Sumac (Rhus coriaria L.) Plant Extract and Their Antimicrobial and DNA Damage Protective Effects. Front. Chem. 2022, 10, 968280. [Google Scholar] [CrossRef]
  62. Rahimzadeh, C.Y.; Barzinjy, A.A.; Mohammed, A.S.; Hamad, S.M. Green Synthesis of SiO2 Nanoparticles from Rhus coriaria L. Extract: Comparison with Chemically Synthesized SiO2 Nanoparticles. PLoS ONE 2022, 17, e0268184. [Google Scholar] [CrossRef] [PubMed]
  63. Mongy, Y.; Shalaby, T. Green Synthesis of Zinc Oxide Nanoparticles Using Rhus coriaria Extract and Their Anticancer Activity against Triple-Negative Breast Cancer Cells. Sci. Rep. 2024, 14, 13470. [Google Scholar] [CrossRef]
  64. Janik, M.; Khachatryan, K.; Khachatryan, G.; Krystyjan, M.; Oszczęda, Z. Comparison of Physicochemical Properties of Silver and Gold Nanocomposites Based on Potato Starch in Distilled and Cold Plasma-Treated Water. Int. J. Mol. Sci. 2023, 24, 2200. [Google Scholar] [CrossRef]
  65. Li, M.; Shi, P.; Xu, C.; Ren, J.; Qu, X. Cerium Oxide Caged Metal Chelator: Anti-Aggregation and Anti-Oxidation Integrated H2O2-Responsive Controlled Drug Release for Potential Alzheimer’s Disease Treatment. Chem. Sci. 2013, 4, 2536–2542. [Google Scholar] [CrossRef]
  66. Dhall, A.; Self, W. Cerium Oxide Nanoparticles: A Brief Review of Their Synthesis Methods and Biomedical Applications. Antioxidants 2018, 7, 97. [Google Scholar] [CrossRef]
  67. Li, J.; Feizipour, S.; Delirezh, N.; Sheikhzadeh, S.; Hobbenaghi, R.; Amraii, S.A.; Khorasani, F.; Hemmati, S.; Joshani, Z.; Kamangar, S.A.; et al. Green Synthesis of Gold Nanoparticles Using Potato Starch as a Phytochemical Template, Green Reductant and Stabilizing Agent and Investigating Its Cytotoxicity, Antioxidant and Anti-Ovarian Cancer Effects. Inorg. Chem. Commun. 2023, 155, 111002. [Google Scholar] [CrossRef]
  68. Tisi, A.; Pulcini, F.; Carozza, G.; Mattei, V.; Flati, V.; Passacantando, M.; Antognelli, C.; Maccarone, R.; Delle Monache, S. Antioxidant Properties of Cerium Oxide Nanoparticles Prevent Retinal Neovascular Alterations In Vitro and In Vivo. Antioxidants 2022, 11, 1133. [Google Scholar] [CrossRef] [PubMed]
  69. Khan, M.; Sohail; Raja, N.I.; Asad, M.J.; Mashwani, Z.-U.-R. Antioxidant and Hypoglycemic Potential of Phytogenic Cerium Oxide Nanoparticles. Sci. Rep. 2023, 13, 4514. [Google Scholar] [CrossRef]
  70. Corsi, F.; Deidda Tarquini, G.; Urbani, M.; Bejarano, I.; Traversa, E.; Ghibelli, L. The Impressive Anti-Inflammatory Activity of Cerium Oxide Nanoparticles: More Than Redox? Nanomaterials 2023, 13, 2803. [Google Scholar] [CrossRef] [PubMed]
  71. Sadhu, A.; Ghosh, I.; Moriyasu, Y.; Mukherjee, A.; Bandyopadhyay, M. Role of cerium oxide nanoparticle-induced autophagy as a safeguard to exogenous H2O2-mediated DNA damage in tobacco by-2 cells. Mutagenesis 2018, 33, 161–177. [Google Scholar] [CrossRef] [PubMed]
  72. El-Seidy, A.M.A.; Elbaset, M.A.; Ibrahim, F.A.A.; Abdelmottaleb Moussa, S.A.; Bashandy, S.A. Nano Cerium Oxide and Cerium/Zinc Nanocomposites Characterization and Therapeutic Role in Combating Obesity via Controlling Oxidative Stress and Insulin Resistance in Rat Model. J. Trace Elem. Med. Biol. 2023, 80, 127312. [Google Scholar] [CrossRef]
  73. Rzigalinski, B.A.; Carfagna, C.S.; Ehrich, M. Cerium Oxide Nanoparticles in Neuroprotection and Considerations for Efficacy and Safety. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, e1444. [Google Scholar] [CrossRef]
  74. Gunasekaran, N.K.; Nazario Bayon, N.; Tumkur, P.P.; Prabhakaran, K.; Hall, J.C.; Ramesh, G.T. Anticancer Activity of Cerium Oxide Nanoparticles Towards Human Lung Cancer Cells. Nanomanufacturing 2025, 5, 6. [Google Scholar] [CrossRef]
  75. García, A.; Espinosa, R.; Delgado, L.; Casals, E.; González, E.; Puntes, V.; Barata, C.; Font, X.; Sánchez, A. Acute Toxicity of Cerium Oxide, Titanium Oxide and Iron Oxide Nanoparticles Using Standardized Tests. Desalination 2011, 269, 136–141. [Google Scholar] [CrossRef]
  76. Khorrami, M.B.; Sadeghnia, H.R.; Pasdar, A.; Ghayour-Mobarhan, M.; Riahi-Zanjani, B.; Hashemzadeh, A.; Zare, M.; Darroudi, M. Antioxidant and Toxicity Studies of Biosynthesized Cerium Oxide Nanoparticles in Rats. Int. J. Nanomed. 2019, 14, 2915–2926. [Google Scholar] [CrossRef]
  77. Crous, A.; Abrahamse, H. Effective Gold Nanoparticle-Antibody-Mediated Drug Delivery for Photodynamic Therapy of Lung Cancer Stem Cells. Int. J. Mol. Sci. 2020, 21, 3742. [Google Scholar] [CrossRef]
  78. Didamson, O.C.; Chandran, R.; Abrahamse, H. A Gold Nanoparticle Bioconjugate Delivery System for Active Targeted Photodynamic Therapy of Cancer and Cancer Stem Cells. Cancers 2022, 14, 4558. [Google Scholar] [CrossRef] [PubMed]
  79. Prakashan, D.; Shrikrishna, N.S.; Byakodi, M.; Nagamani, K.; Gandhi, S. Gold Nanoparticle Conjugate-Based Lateral Flow Immunoassay (LFIA) for Rapid Detection of RBD Antigen of SARS-CoV-2 in Clinical Samples Using a Smartphone-Based Application. J. Med. Virol. 2023, 95, e28416. [Google Scholar] [CrossRef]
  80. Saeidi, J.; Dolatabadi, S.; Esfahani, M.B.; Saeidi, M.; Mohtashami, M.; Mokhtari, K.; Ghasemi, A. Anticancer Potential of Doxorubicin in Combination with Green-Synthesized Silver Nanoparticle and Its Cytotoxicity Effects on Cardio-Myoblast Normal Cells. Anti-Cancer Agents Med. Chem. 2021, 21, 1842–1849. [Google Scholar] [CrossRef]
  81. Gołuński, G.; Konkel, K.; Bełdzińska, P.; Bury, K.; Zakrzewski, M.; Butowska, K.; Sądej, R.; Piosik, J. Influence of Silver Nanoparticles’ Size on Their Direct Interactions with Doxorubicin and Its Biological Effects. Sci. Rep. 2024, 14, 18544. [Google Scholar] [CrossRef]
  82. Le-Thi, P.; Trung-Nguyen, D.; An-Nguyen-Huu, T.; Tran, Q.; Truong, M.; Thanh-Hang, N.; Quyen-Tran, N.; Dong-Park, K. Hyaluronic Acid-Coated Silver Nanoparticles Releasing Doxorubicin for Combinatorial Antitumor Therapy. J. Ind. Eng. Chem. 2025, 142, 431–440. [Google Scholar] [CrossRef]
  83. Kamsu, G.T.; Ndebia, E.J. Usefulness of Natural Phenolic Compounds in the Fight against Esophageal Cancer: A Systematic Review. Future Pharmacol. 2024, 4, 626–650. [Google Scholar] [CrossRef]
  84. Devaraji, M.; Thanikachalam, P.V.; Elumalai, K. The Potential of Copper Oxide Nanoparticles in Nanomedicine: A Comprehensive Review. Biotechnol. Notes 2024, 5, 80–99. [Google Scholar] [CrossRef]
  85. Peng, L.; Gao, Z.; Liang, Y.; Guo, X.; Zhang, Q.; Cui, D. Nanoparticle-Based Drug Delivery Systems: Opportunities and Challenges in the Treatment of Esophageal Squamous Cell Carcinoma (ESCC). Nanoscale 2025, 17, 8270–8288. [Google Scholar] [CrossRef]
  86. Udekwu, K.I.; Parrish, N.; Ankomah, P.; Baquero, F.; Levin, B.R. Functional Relationship between Bacterial Cell Density and the Efficacy of Antibiotics. J. Antimicrob. Chemother. 2009, 63, 745–757. [Google Scholar] [CrossRef] [PubMed]
  87. Wolfram, J.; Zhu, M.; Yang, Y.; Shen, J.; Gentile, E.; Paolino, D.; Fresta, M.; Nie, G.; Chen, C.; Shen, H.; et al. Safety of Nanoparticles in Medicine. Curr. Drug Targets 2015, 16, 1671–1681. [Google Scholar] [CrossRef] [PubMed]
  88. He, J.; Ma, Y.; Niu, X.; Pei, J.; Yan, R.; Xu, F.; Ma, J.; Ma, X.; Jia, S.; Ma, W. Silver Nanoparticles Induce Endothelial Cytotoxicity through ROS-Mediated Mitochondria-Lysosome Damage and Autophagy Perturbation: The Protective Role of N-Acetylcysteine. Toxicology 2024, 502, 153734. [Google Scholar] [CrossRef] [PubMed]
  89. Huang, H.; Liu, R.; Yang, J.; Dai, J.; Fan, S.; Pi, J.; Wei, Y.; Guo, X. Gold Nanoparticles: Construction for Drug Delivery and Application in Cancer Immunotherapy. Pharmaceutics 2023, 15, 1868. [Google Scholar] [CrossRef]
Figure 1. PRISMA 2020 flow diagram for the selection of studies.
Figure 1. PRISMA 2020 flow diagram for the selection of studies.
Applnano 06 00018 g001
Table 2. Comparison of nanoparticles tested against esophageal cancer cells.
Table 2. Comparison of nanoparticles tested against esophageal cancer cells.
NanoparticlesOrigin/SynthesisEC Cell Lines TestedIC50 (µg/mL or µM)Mechanism of ActionToxicity (Normal Cells or Organism)
CuO NPsVibrio sp. VLCKYSE3013.96 mg/L/HDF: IC50 = 48.88 mg/L)
Au NPsMoringa oleiferaSNO92.01 µg/mLApoptosis (↑Caspases, p53, ↓Bcl-2, ATP…)Hepatoprotection effect (in vitro)
NaLS-Au NPsCydonia oblonga + NaLSFLO-1, ESO26, OE33, KYSE-270181, 130, 205, 133 µg/mL/IC50 > 1000 µg/mL on HUVEC cells
Nano-curcuminCurcuma longaOE33, OE1920% and 17% inhibition at 50 mM/Non-toxic on HET-1A cells
Cu NPsMentha piperitaKYSE-270, OE33, ESO26241, 278, 240 mg/mL//
NiO NPsCalendula officinalisFLO-1, ESO26, OE33, KYSE-270380, 263, 229, 251 µg/mL/Non-toxic on HUVEC cells
Ag NPsPomegranate peelKYSE30-270, OE33, ESO26, FLO-1253–500 µg/mL//
Ag NPsPhotinia glabraEca-109<20 µg/mL//
Au NPsRhus coriariaFLO-1, ESO26, OE33, KYSE-270226–294 mg/mLApoptosis/
CeO2 NPsChemical synthesisYM1630 µMOxidative stressHighly toxic in vitro; low toxicity in vivo
Cu2(btc)(trz)3-NPs Chemical synthesisKYSE150, EC109, EC871248, 42, 34 µM//
Cu5(Hbtc)4(trz)2(H2O)4-NPsChemical synthesis86, 73, 78 µM//
Au NPsPotato starchKYSE-30, FLO-1125, 176 µg/mL/IC50 > 1000 µg/mL on HUVEC cells
Gold NICAuNPs + antibody CD271HKESC-12.3–2.9 µM//
Ag-Dox NPsAgNPs + doxorubicinOE332.399 µM↑Dox efficacy, ↓migrationNo harmful effect on H9c2 cells
NIC: nano-immuno-conjugate; NPs: nanoparticles; CuO: copper(II) oxide; Ag: silver; Au: gold; NiO: nickel(II) oxide; IC50: 50% inhibitory concentration; H3btc: 1,3,5-benzenetricarboxylic acid; Htrz: 1,2,4-triazole; CD271: Cluster of Differentiation 271; Vibrio sp. VLC: Vibrio luminescent strain C; PG: Photinia glabra; NaLS: sodium lignosulfonate; HUVEC: human umbilical vein endothelial cells; Dox: doxorubicin; HDF: human dermal fibroblast; ↑: increase expression; ↓: decrease expression.
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

Kamsu, G.T.; Ndebia, E.J. Usefulness of Nanoparticles in the Fight Against Esophageal Cancer: A Comprehensive Review of Their Therapeutic Potential. Appl. Nano 2025, 6, 18. https://doi.org/10.3390/applnano6030018

AMA Style

Kamsu GT, Ndebia EJ. Usefulness of Nanoparticles in the Fight Against Esophageal Cancer: A Comprehensive Review of Their Therapeutic Potential. Applied Nano. 2025; 6(3):18. https://doi.org/10.3390/applnano6030018

Chicago/Turabian Style

Kamsu, Gabriel Tchuente, and Eugene Jamot Ndebia. 2025. "Usefulness of Nanoparticles in the Fight Against Esophageal Cancer: A Comprehensive Review of Their Therapeutic Potential" Applied Nano 6, no. 3: 18. https://doi.org/10.3390/applnano6030018

APA Style

Kamsu, G. T., & Ndebia, E. J. (2025). Usefulness of Nanoparticles in the Fight Against Esophageal Cancer: A Comprehensive Review of Their Therapeutic Potential. Applied Nano, 6(3), 18. https://doi.org/10.3390/applnano6030018

Article Metrics

Back to TopTop