Open Access

Targeted gold nanoparticles for ovarian cancer (Review)

  • Authors:
    • Wenjuan He
    • Fuyuan Yang
    • Keming Chen
    • Qingsong Zeng
  • View Affiliations

  • Published online on: October 3, 2024     https://doi.org/10.3892/ol.2024.14723
  • Article Number: 589
  • Copyright: © He et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Among all malignant gynecological tumors, ovarian cancer (OC) has the highest mortality rate. OC is often diagnosed at advanced and incurable stages; however, early diagnosis can enable the use of optimized and personalized treatments. Intensive research into the synthesis and characterization of gold nanoparticles (AuNPs) has been performed with the aim of developing innovative materials for use in biological and photothermal therapies for OC. AuNPs can be chemically modified and functionalized by binding to a variety of organic compounds and biomolecules, such as peptides, antibodies and therapeutic agents, via simple synthetic processes. They are particularly suitable for use as carriers for drug delivery. In the present review, the synthesis and characteristics of AuNPs are summarized, and their potential in OC therapy are discussed.

Introduction

Cancer is among the most serious ailments endangering human health, ranking second only to cardiovascular disease (1). Ovarian cancer (OC) is a heterogeneous group of malignancies of the fallopian tubes, ovaries and abdominal cavity (2). It is the fifth deadliest and eighth most common cancer affecting females worldwide (3). Conventional cancer treatments for OC include radiation, surgery, traditional chemotherapy and invasive catheters (4). Epithelial OC (EOC) is the most common type of gynecological malignancy (5). Despite great breakthroughs in EOC therapy, patients frequently experience chemotherapeutic resistance and disease relapse within five years, highlighting that improved therapeutic options are necessary (6). Therefore, the accurate targeting of treatment is important for improving patient prognosis.

Nanoparticles (NPs) typically range in diameter from 1 to 100 nm, with small NPs composed of only a few to several hundred atoms (7,8). Nanomaterials (NMs) are widely utilized in material science and nanotechnology due to their unique properties, which differ from those of conventional materials. The appeal of NPs for medical purposes lies in their special and significant features, including a surface-to-mass proportion, quantum nature and the capability of adsorbing and transporting other compounds, including proteins, drugs and probes (9). The morphologies of NPs can be highly varied and are indicative of their distinctive characteristics (10). The nanoscale dimensions of NPs render them suitable for biolabeling by enabling interaction with biomolecules at both the surface and intracellular levels, generating valuable signals and specific targets for diagnostic and therapeutic applications (11). Due to their unique features, NPs are highly valuable in various applications, including tissue engineering, biomarker identification and drug delivery systems (12). The value of NPs for medical purposes may be attributed to various features, including their surface-to-mass proportion, quantum nature and ability to adsorb and transport other compounds, including proteins, drugs and probes. As studies on NMs have become more prevalent, metal NPs have been evaluated for a broad range of uses, including electronics, catalysis and sensing (9,13,14). Functionalized NPs, particularly those derived from metal NPs, have the potential to serve as valuable biological probes for a range of uses, including organic chemistry research tools, bioassays, clinical diagnosis and cancer treatment (15). In addition, the use of NPs for drug encapsulation is viewed as a promising and effective approach for drug delivery (11).

Among different inorganic NPs, gold NPs (AuNPs) are actively studied for their different biomedical applications. This is mainly due to their stability, simple and easy synthesis, low-cost preparation techniques, size-controllable synthesis, biocompatibility, relatively easy surface modification and low toxicity properties (1618). In the present study, the features of AuNPs, their potential in OC therapy and their contributions to tumor treatment are reviewed.

AuNPs

Properties of AuNPs

AuNPs are ideal carriers as they can be functionalized or modified with various chemical groups and are inert to biological systems (19,20). Owing to their biocompatibility and ability to be surface modified with biocompatible molecules, AuNPs can be engineered to minimize undesirable immune responses, such as antibody production (2123). Furthermore, AuNPs are resistant to oxidation and can be synthesized by controlled crystallization methods, which provide AuNPs with precise morphologies and advantageous size distributions (14).

The electronic and optical properties of AuNPs can be modulated by altering their shape, size, aggregation state and surface chemistry (14). When AuNPs are used as nanocarriers in various applications, chemical modification is necessary. It is crucial that the surface functionalization of the AuNPs is appropriate for the intended usage, for example, to improve their stability and biocompatibility while preventing aggregation (24). The primary purposes of the surface modification of AuNPs include: i) Stabilizing the AuNPs by the attachment of ligands to the AuNP surface, ii) enabling additional functionalization reactions through the bonding of linkers to the AuNP surface, and iii) facilitating further functionalization or bioconjugation by directly immobilizing functional ligands and biomolecules on the AuNP surface, thereby expanding their application range (2429) (Table I).

Table I.

Surface modification of AuNPs.

Table I.

Surface modification of AuNPs.

First author, yearSurface modification methodMechanismFunction(Refs.)
Ielo et al, 2021Secondary modification‘Place exchange’ of a thiol ligandIntroduces various functionalities that may react via condensation(24)
Xiao et al, 2018Physical sorptionPhysical sorption of ligands or biomolecules on AuNP surfaces driven by electrostatic and hydrophobic interactionsModifies the AuNP charge state and the degree of immobilization of functional molecules(26)
De Luca et al, 2018Dative bonding and formation of self-assembled monolayersThiolated ligands densely bond to AuNP surfaces to produce self-assembled monolayersStable capping of the AuNPs prevents the coupling of other ligands and biomolecules(27)
Boyer et al, 2010Polymer coatingNeutral polymers or charged polymers are used to coat the AuNP surfacesSteric repulsion or repulsive electrostatic interactions, respectively, improve the colloidal stability of the AuNPs(28)
Presnova et al, 2014Bioaffinity immobilization of ligandsSynthesis of chemically stable protein-@AuNP conjugates by immobilization of affinity-bound biomolecules and ligandsAllows biotinylated nucleic acids, antibodies and aptamers to be immobilized(29)

[i] AuNP, gold nanoparticles.

Synthesis of AuNPs

Approaches for synthesizing AuNPs can be categorized as either ‘top-down’ or ‘bottom-up’ methods (30,31) (Fig. 1). Top-down methods typically involve producing NPs by reducing the size of bulk materials. This serves as the basis for most physical approaches, including pulsed laser ablation, plasma arc discharge, evaporation-condensation, spray pyrolysis, ball milling, vapor and gas phase processes, and lithographic techniques (32,33). However, the unfinished surface structure of the resulting NPs represents a disadvantage (34). Another constraint of these top-down methods is their high costs, as a substantial quantity of energy is needed to maintain high-temperature and high-pressure conditions (24). Bottom-up methods are those that generate NPs from smaller components, including atoms and molecules, and include chemical synthesis techniques, including microemulsion, coprecipitation, chemical reduction, microwave-assisted synthesis, electrochemical, sonochemical, solvothermal and thermal decomposition methods. Green biological synthesis methods also fall into this category. Compared with physical and chemical methods, the use of whole organisms or biological molecules to synthesize NPs offers notable advantages. Biological synthesis methods are nontoxic and relatively sustainable, providing a comparatively environmentally friendly approach to NP synthesis (30,35).

The synthesis of AuNPs by biological methods can be a relatively straightforward process that does not require high temperature or pressure. The procedure generally involves the dropwise addition of a biological extract, such as that from bacteria, fungi and/or plants, into a solution of HAuCl4 salt with thorough mixing to initiate AuNP synthesis (36). Subsequent formulation of the AuNPs consists of two primary phases: In the first phase, the gold precursor, typically in the form of an aqueous gold salt solution, is reduced to form AuNPs using a reducing agent, such as citrate. In the second phase, the AuNPs are stabilized by the introduction of a capping agent, which prevents the agglomeration of the metallic NPs (37).

The use of toxic reducing agents and the gases produced by the process of producing NPs are harmful to humans and the environment. Therefore, safer, nontoxic and environmentally friendly methods for the generation of NPs have been devised, with the use of reducing agents obtained from plant materials, including leaves, roots, flowers and seeds (38). Modification of the reaction time, pH, reaction temperature and fungal biomass can improve the efficiency of the fungal synthesis of AuNPs (39). Commonly employed methods for the characterization of AuNPs include atomic force microscopy, X-ray powder diffraction, scanning electron microscopy, dynamic light scattering, high-resolution transmission electron microscopy, zeta potential, energy dispersive spectroscopy, Fourier transform infrared spectroscopy and ultraviolet (UV)-visible spectroscopy (40). Fig. 2 schematically illustrates various methods for the synthesis, optimization, characterization and conjugation of therapeutic agents with AuNPs. The existing synthesis approaches often involve costly and low-yield purification processes, such as differential centrifugation, to obtain NPs (41). Therefore, the development of nonpoisonous, eco-friendly and clean sustainable synthesis procedures with high yields and low cost is critical (42).

Synthesis and stabilization procedures for precious metal-based NPs using plant extracts are regarded to be safe, economical, eco-friendly and green (43). In one study, AuNP formation was realized at ambient temperature by mixing thyme extract with gold salts. The reaction could be scaled by the adjustment of various reaction conditions, particularly temperature (44). A number of other studies have also synthesized AuNPs by methods using plant extracts or other biological materials, such as fungi and bacteria, to reduce metal salts and obtain bio-friendly, stable metal-based NPs (45,46). For example, the microbial synthesis of AuNPs was first reported in 1980, with the use of Bacillus subtilis (47).

Table II presents some other examples of AuNPs that have been synthesized (6,4863).

Table II.

Examples of AuNP synthesis and characterization.

Table II.

Examples of AuNP synthesis and characterization.

First author/s, yearNameMorphologySize, nmIngredients Characterization(Refs.)
Lee et al, 2022HA-AuDEN-DoxSpherical<2HAuCl4, G6-NH2 dendrimer solution, NaBH4, HA, DoxUV-vis, fluorescence spectroscopy, TEM, DLS and zeta potential(49)
Lee et al, 2020Dox-DNA-AuNP-13HAuCl4, sodium citrate, DNA, sodium chloride, sodium phosphate buffer, DoxUV-vis, zeta potential, DLS and fluorescence spectroscopy(48)
Kotcherlakota et al, 2017Au-TR-DX-siSpherical105HAuCl4, NaBH4, Dox, erbB2 siRNA, TRUV-vis(50)
Kotcherlakota et al, 2019Au-C225-p53DNASpherical52.3±2.6HAuCl4, C225, polyethylene imine, p53 DNAUV-vis(51)
Piktel et al, 2021Peanut-shaped AuNPsPeanut-like 60.00±4.24×30±3.49Cetrimonium bromide, HAuCl4, AgNO3, NaBH4, ascorbic acidUV-vis(6)
Piktel et al, 2021AuP@CSA-131Peanut-like60±5×30±3.5CTAB, HAuCl4, NaBH4, AgNO3, MHDAHAADF-STEM, Fourier-transform Raman spectroscopy and TGA(52)
Jabir et al, 2020LG/LGC-11/13HAuCl4, NaBH4, GSH, linalool/CALNNUV spectrophotometry, SEM and TEM(53)
Asl et al, 2023AuNPsSpherical15.1±3.7HAuCl4, NaOH, dimethyl sulfoxide, Satureja rechingeri Jamzad aqueous leaf extractXRD, FTIR, UV-vis, TEM, SEM, EDX, DLS and zeta potential(54)
Xiong et al, 2014AuNPs-20HAuCl4 trihydrate, trisodium citrate, NaBH4DLS and zeta potential(55)
Kip et al, 2022AuNCsCone-shaped100HAuCl4, o-phenetidine, hexaneUV-vis, DLS and STEM(56)
Patra et al, 2010Au-PSH-CP-FA-5HAuCl4, NaBH4, CP, FA, tritiated FA, [3H]thymidineUV-vis, TEM and ICP(57)
Borghei and Hosseinkhani, 2022Wh@AuNPs--HAuCl4, whey, 3,3′,5,5′-tetramethylbenzidineUV-vis(58)
Wang et al, 201415P-PPy-NPs--AuNPs, pyrrole aqueous solution, SDS, acidic (NH4)2S2O8 solution, EDCUV-vis and TEM(59)
Shen et al, 2022RHMH18@AuD NPs-Varies with reaction time and HAuCl4 concentrationRHMH18 protein, HAuCl4, NaOH, phosphate-buffered saline, DTXTEM(60)
Van de Broek et al, 2011Branched AuNPsBranched60.4±9.7HAuCl4, sodium citrate, BSPP, H2O2, NaOH, HCl, HNO3, anti-HER2UV-vis(61)
Geng et al, 2011Glu-AuNPs-14.37±2.49HAuCl4, NaBH4, sodium citrate, PEG, GluTEM and XPS(62)
Cui et al, 2017 GNP-NHN=Dox-mPEG-179.0±7.5HAuCl4, hydrazine hydrate, DCC, DMAP, TFA, NPC, mPEG, Dox HCl, NaBH4FTIR and NMR(63)

[i] AuNP/GNP, gold nanoparticle; HA, hyaluronic acid; AuDEN, dendrimer encapsulated AuNPs; Dox/DX, doxorubicin; G6-NH2, amine-terminated poly(amidoamine); UV-vis, ultraviolet-visible spectrophotometry; TEM, transmission electron microscopy; DLS, dynamic light scattering; TR, bifunctional recombinant fusion protein TRAF(C); si/siRNA, small interfering RNA; C225, cetuximab; CSA-131, cationic steroid antibiotic 131; CTAB, cetyl trimethylammonium bromide; MHDA, 16-mercaptohexadecanoic acid; HAADF, high-angle annular dark-field; STEM, scanning TEM; TGA, thermogravimetric analysis; LG, linalool-AuNP; LGC, linalool-AuNP-CALNN peptide; GSH, glutathione; SEM, scanning electron microscopy; XRD, X-ray diffraction; FTIR, Fourier-transform infrared; EDX, energy-dispersive X-ray spectroscopy; PSH, mercapto-PEG of molecular weight 2,000; CP, cisplatin; FA, folic acid; ICP, inductively coupled plasma; Wh, whey; 15P, SHSWHWLPNLRHYAS protein; PPy, polypyrrole; NPs, nanoparticles; SDS, sodium dodecyl sulfate; EDC, 1-ethyl-3-[(3-dimethylamino)propyl]carbodiimide; AuDNPs, DTX-loaded AuNPs; DTX, docetaxel; BSPP, bis(p-sulfonatophenyl)phenylphosphine dihydrate dipotassium salt; Glu, 1-thio-β-glucose; PEG, polyethylene glycol; XPS, X-ray photoelectron spectroscopy; mPEG, modified PEG; DCC, dicyclohexylcarbodiimide; DMAP, 4-dimethylaminopyridine; TFA, trifluoroacetic acid; NPC, N-hydroxysuccinimide propionate; NMR, nuclear magnetic resonance.

Application of AuNPs

The utilization of nanosized materials has facilitated a number of advances in biological applications such as biomedicine. These advances include antitumor activity and drug delivery (64), fluorescent biological labeling, gene delivery, tissue engineering, protein detection, contrast enhancement magnetic resonance imaging, DNA probing, hyperthermia treatment, phagokinetic research and cell or molecular filtration leveraging biological interactions (11).

AuNPs are among the most commonly used materials for diagnostics, bioimaging and cancer therapy due to their inherent stability and low cytotoxicity (65,66). AuNPs of various shapes, such as nanoshells, nanorods, nanocages, nanostars, nanospheres and branched AuNPs, have been manufactured and investigated (67,68) (Fig. 3). For example, the hollow structure of gold nanocages provides a high capacity for loading various types of payloads, while the payload can be rapidly loaded and released through pores in the walls. These features are particularly attractive for drug delivery and controlled release (69,70). Specific applications of AuNPs include their use as contrast agents in medical imaging and as drug carriers for gene delivery (71). AuNPs have been extensively adopted in drug delivery due to their chemical inertness, biocompatibility and ease of functionalization (72,73). In nanomedicine, AuNPs are non-toxic at the doses utilized for drug delivery (74). The high affinity of AuNPs for thiols, polymers and amines allows the introduction of reactive molecules that can be employed for targeting, including peptides, antibodies, carbohydrates and aptamers, and for the conjugation of therapeutic agents, including radionuclides, drugs, photosensitizers, genes and small interfering RNAs (75).

It has been suggested that AuNPs are redox active and noncytotoxic, as they can reduce reactive oxygen and nitrite species without inducing the secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and other inflammatory factors, which makes then ideal nanomedicine candidates (76). Ben Haddada et al (77) demonstrated that AuNPs prepared using Hubertia ambavilla are nonpoisonous to human skin fibroblasts and can scavenge free radicals and protect fibroblasts and dermal cells against UV-A radiation-induced damage. In addition, Taratummarat et al (78) reported that spherical AuNPs of diameter 20–30 nm are nontoxic to mice, and exhibit anti-inflammatory properties. It has been reported that positively charged particles are more toxic to bacteria than are negatively charged or neutral particles, indicating that surface charge affects the toxicity of AuNPs (79). However, other studies have reported conflicting findings, with one reporting that positively charged AuNPs exhibited no toxicity to human epithelial cells (80), while another reported that both positively and negatively charged AuNPs were toxic to human keratinocytes (81). The reason for such differences may be the various physicochemical features of different NPs, and the lack of a standardized method to verify toxicity (82). Moreover, Shukla et al (76) suggested that AuNPs do not elicit an initial immunological response or induce the production of the proinflammatory cytokines IL-1β and TNF-α until a high concentration of 100 µM is reached. In addition, Ghosh et al (83) reported that AuNPs do not trigger complement activation. Due to their lack of toxicity and immunogenicity, AuNPs are an ideal choice for drug delivery scaffolds. Moreover, the ability to functionalize AuNPs renders them highly promising vehicles for drug delivery applications (8487). Currently, nanomedicine is advancing the development of novel therapeutic and diagnostic tools, including biosensors for biomolecule detection, tumor chemotherapeutics, and gene or drug delivery. Owing to their good biocompatibility and small particle size, AuNPs are promising candidates for biological applications (88).

There are many methods for the treatment of OC, which vary in their advantages and disadvantages (Fig. 4). Importantly, AuNPs can be used for OC treatment. In one study, in vitro experiments demonstrated that AuNPs successfully induced autophagy and apoptosis in SK-OV-3 cells via reactive oxygen species (ROS)-mediated pathways, indicating their potential as new nanotherapeutics (6). AuNPs hold great promise in diagnostic and therapeutic medicinal applications.

Drug delivery systems targeting OC

The large surface-to-volume ratio and good biocompatibility of AuNPs, together with the ability to synthesize AuNPs with varied morphological characteristics and surface chemistries, render AuNPs very suitable for use as drug delivery vehicles (89). AuNPs coupled with targeted molecules can precisely deliver tumor-targeting drugs via both passive and active targeting mechanisms (90,91).

Passive targeting involves the nonspecific accumulation of NPs in tumors due to the specific characteristics of the tumor microenvironment (92,93). In passive targeting, AuNPs primarily exploit the enhanced permeability and retention effect. Drugs attached to AuNPs can selectively accumulate in tumor tissues and persist for an extended period due to vascular leakage and compromised lymphatic drainage, respectively (94,95). Although a meta-analysis of 117 studies (96) on nanodrug delivery found that only 0.7% of NPs successfully reached the tumor site, indicating that although passive targeting often results in low delivery efficacy, AuNPs have an improved ability to target tumor tissue. In addition, AuNPs can be attached to a variety of ligands, including drugs, peptides, antibodies and oligonucleotides, to enhance their targeted delivery properties (9799). AuNP carriers can protect peptides, antibodies and oligonucleotides from enzymatic degradation, thereby improving their effectiveness in transporting drugs into solid tumors (100). Table III presents some examples of targeted AuNPs and their properties.

Table III.

Properties of selected AuNPs targeting OC.

Table III.

Properties of selected AuNPs targeting OC.

First author, yearNamePropertiesPassive/active targetingDrug/targeting agentReceptorDrug release system(Refs.)
Lee et al, 2022HA-AuDEN-DoxMultivalent terminals of dendrimers can be functionalized with targeted ligands for active targeting, inhibiting ovary tumor growthActiveHA, DoxCD44pH and GSH stimuli-responsiveness(49)
Lee et al, 2020Dox-DNA-AuNPExcellent anticancer activity for OC cellsActiveDox-pH-dependent(48)
Kotcherlakota et al, 2017Au-TR-DX-siNon-toxic, target-specific uptake and significant OC tumor suppressionActiveDox, TR, erbB2 siRNAHER2-(50)
Kotcherlakota et al, 2019Au-C225-p53DNADelivers p53 DNA and C225 specifically to OC cells that overexpress EGFRActiveC225, p53 DNAEGFR-(51)
Piktel et al, 2021Peanut-shaped AuNPsReduce the viability and proliferation of OC cells by triggering ROS-mediated apoptosis and autophagyPassive---(6)
Piktel et al, 2021AuP@CSA-131Improved anticancer compared with CSA-131, enabling the effective dose to be reducedActiveCSA-131--(52)
Jabir et al, 2020LG/LGCSignificant antiproliferative effect on SK-OV-3 cellsActiveLinalool--(53)
Asl et al, 2023AuNPsPotent anticancer activity against CP-resistant OC cellsPassive---(54)
Xiong et al, 2014AuNPsSensitize OC cells to CP by depleting stem cell pools and inhibiting key molecular pathwaysActiveCP--(55)
Patra et al, 2010Au-PSH-CP-FAEnhanced cytotoxic effect on OC cells and protective against cytotoxic damage in normal cellsActiveCP, FAFolate receptorpH and GSH stimuli-responsiveness(57)
Wang et al, 201415P-PPy-NPsTarget SK-OV-3 cells in vitroActive15PVEGFR3-(59)
Shen et al, 2022RHMH18@AuDNPsGood biocompatibility and active chemotherapeutic photothermal synergistic effect on human ovarian tumorsActiveDTX-pH-dependent(60)
Van de Broek et al, 2011Branched AuNPsActively target HER2-expressing SK-OV-3 cellsActiveAnti-HER2HER2-(61)
Geng et al, 2011Glu-AuNPsEnhance the effectiveness of radiotherapy on OC cellsActiveGlu--(62)

[i] AuNP, gold nanoparticle; OC, ovarian cancer; HA, hyaluronic acid; AuDEN, dendrimer encapsulated AuNPs; Dox/DX, doxorubicin; GSH, glutathione; TR, bifunctional recombinant fusion protein TRAF(C); si/siRNA, small interfering RNA; C225, cetuximab; EGFR, epidermal growth factor receptor; ROS, reactive oxygen species; CSA-131, cationic steroid antibiotic 131; LG, linalool-AuNP; LGC, linalool-AuNP-CALNN peptide; CP, cisplatin; PSH, mercapto-PEG of molecular weight 2,000; FA, folic acid; 15P, SHSWHWLPNLRHYAS protein; PPy, polypyrrole; NPs, nanoparticles; VEGFR3, vascular endothelial growth factor receptor 3; AuDNPs, DTX-loaded AuNPs; DTX, docetaxel; Glu, 1-thio-β-glucose.

Drugs

Drug-conjugated AuNPs are considered to be highly promising and efficient nanoprodrugs. Such a conjugate may be constructed, for example, by the attachment of multiple thiol-terminated polyethylene glycol (PEG)-drug conjugates onto the surface of AuNPs via thiol-Au covalent bonds (63). The attachment of drugs to the surfaces of AuNPs offers several advantages while minimizing the risk of severe systemic toxicity (101). For instance, due to their small size, they can efficiently travel through capillaries to reach target cells. Chemotherapeutic agents can be loaded or attached to the AuNPs and can be passively or actively targeted to the tumor site (82). In addition, the incorporation of modifiers that are responsive to external stimuli, including pH or enzymes, into the linking molecules facilitates drug release (37).

The utilization of NP-based carriers for the delivery of anticancer agents is a promising strategy for reducing the dosages of antineoplastic compounds, as it minimizes their systemic toxicity while simultaneously enhancing their therapeutic efficacy (102). Piktel et al (52) used nanotechnology to manufacture a new nanosystem composed of AuNPs functionalized with a shell comprising cationic steroid antibiotic-131. This nanosystem exhibited marked activity against OC cells in vitro and prevented the development of ovarian tumors in animals with minimal toxicity. In another study, Dox-DNA-AuNPs exhibited an excellent anticancer effect in an in vitro propagation test, and efficacy in the prevention of tumor development in a xenograft mouse model over a 16-day treatment period. Compared with free Dox, Dox-DNA-AuNPs exhibited an ~2.5-fold greater inhibition of tumor development, demonstrating their strong ability to inhibit cancer development (48).

Cisplatin (CP) is a first-line chemotherapeutic drug for OC. Although CP is very useful as a cancer treatment, it has numerous side effects (103,104). Patra et al (57). described the manufacture and functional characterization of an AuNP-based drug delivery system for the potential treatment of OC. The system was fabricated by the reaction of AuNPs with folic acid (FA), mercapto-PEG of molecular weight 2,000 (PSH) and CP, to form an Au-PSH-CP-FA-based drug delivery system. In vitro proliferation assays revealed that the CP retained its cytotoxicity in this system, while normal cells were protected against cytotoxicity. Asl et al (54) successfully synthesized AuNPs using an extract derived from Satureja rechingeri Jamzad. The obtained spherical AuNPs displayed potent anticancer activity against CP-resistant OC cells, and low cytotoxicity to normal cells, indicating their biocompatibility. These findings indicate that AuNPs have strong potential for the treatment of OC.

By focusing on cancer cell markers that are more highly expressed in tumor tissues than in normal cells and tissues, active targeting agents can improve the precision of tumor tissue targeting (105107). Lee et al (49) created a targeted drug delivery system for the treatment of OC that was responsive to changes in pH and glutathione (GSH) levels. This was created by the attachment of hyaluronic acid molecules to the surface of dendrimer-encapsulated AuNPs via 1-ethyl-3-[3-(dimethylamino)propyl]carbodiimide and N-hydroxysuccinimide chemistry, and then loading Dox onto the Au surface. This nanodrug demonstrated high biocompatibility, excellent stability and effective targeting through the CD44 receptor. In addition, it effectively penetrated cancer cells, where the release of Dox was induced in response to the acidic pH and high GSH levels of the tumor microenvironment. This inhibited tumor growth while causing fewer toxic side effects in mice. Certain AuNPs have the ability to undergo photothermal transformation, which generates heat, thereby promoting in situ drug release and tumor ablation (108,109). For example, in one study, ultrasmall NPs were incorporated into RHMH8 fusion protein via biomimetic mineralization to form RHMH18@Au complexes which were further loaded with docetaxel (DTX). The resulting RHMH18@AuDTX NPs contained AuNPs clustered in the human serum albumin (HSA) portion of the fusion protein and histidine-encapsulated DTX. These RHMH18@AuD NPs formed a uniform dispersion in saline and exhibited chemo-photothermal therapeutic effects in ovarian tumor tissue. In addition, in vitro experiments demonstrated that under the influence of MMP-2, the RHMH18@AuDTX NPs decomposed into arginine-glycine-aspartic acid (RGD)-HSA@Au and His@DTX NPs. It is likely that these two components function in different areas of the tumor tissue, with RGD-HSA@Au playing a photothermal role in the extracellular matrix, and His@DTX NPs entering tumor cells due to their nanoscale size and charge interactions with the cell surface. This dual-targeting approach was demonstrated have good biocompatibility and a favorable anti-OC effect in vivo, and presents a promising novel strategy for tumor treatment (60).

Peptides

Various peptides have been employed for the specific delivery of therapeutic AuNPs (110).

A previous study demonstrated that SK-OV-3 cells are efficiently targeted by 15P (sequence, SHSWHWLPNLRHYAS) conjugated to AuNPs via polypyrrole (PPy) linkers. These conjugates demonstrated hyperthermic effects on the SK-OV-3 human OC cell line when exposed to near-infrared laser irradiation, with high tumor specificity. The hyperthermic effect of the PPy-conjugated AuNPs or 15P conjugates on tumor cells in vivo was investigated in nude mice bearing subcutaneous SK-OV-3 tumors. Significant inhibition of tumor growth was observed following near-infrared laser-mediated treatment with both types of conjugates. These findings indicate that 15P-PPy-AuNPs have excellent biocompatibility, and the ability to effectively induce the photothermal ablation of tumor cells in a tumor-targeted manner. The study also found that while 15P-PPy-NPs effectively bind to and ablate SK-OV-3 cells, they have no effect on HL-7702 or HepG2 cells (59).

Linalool is a monoterpene compound that is active against numerous cancer cell lines, but limited in its application by its high toxicity. A novel peptide conjugate of AuNPs and linalool was synthesized and characterized by Jabir et al (53), with the aim of reducing the general toxicity of linalool and improving its targeting ability. Linalool was loaded onto AuNPs by reaction with GSH and linalool, followed by the loading of CALNN peptide onto the surface of the linalool-loaded AuNPs via a chemical reaction. The peptide conjugate demonstrated strong antiproliferative effects on SK-OV-3 OC cells.

Antibodies

Drug-loaded AuNPs are able to actively target tumors by strategies using antibody-modified ligands (110112). Immunoglobulins and antibody fragments are the most frequently employed molecules for antibody targeting (104). HER2-positive OC is recognized as being aggressive in nature, resistant to chemotherapy and being associated with a high mortality rate (113). Therefore, targeting HER2 receptors is considered as a potential approach for improving the effectiveness of treatment and survival rates in patients with OC. Van de Broek et al (61) linked anti-HER2 nanobodies to branched AuNPs and demonstrated their specific effect on HER2-positive SK-OV-3 cells. These authors reported that the anti-HER2 conjugated AuNPs specifically bound to the cells, indicating that the nanobodies retained their specificity following conjugation to the AuNPs. In addition, targeted photothermal damage of the tumor cells was achieved in vitro by near-infrared laser irradiation of the branched AuNPs, while exposure of the cells to either near-infrared light or AuNPs alone did not affect cell viability; notably, when the two components were combined, cell death was limited to the area of laser/NP cotreatment. By contrast, AuNPs conjugated with anti-PSA nanobodies did not induce cell death upon laser exposure, underscoring the high specificity of these anti-HER2 AuNPs.

Genes

Kotcherlakota et al (51) developed stable AuNPs, designated Au-C225-p53DNA, for the specific delivery of p53 DNA to OC cells with upregulated epidermal growth factor receptor (EGFR) expression. The authors demonstrated that the targeted delivery of the wild-type p53 gene using these NPs effectively inhibited the growth of ovarian tumors in mice with SK-OV-3 ×enografts by the restoration of gene function. The C225 component of these NPs, also known as the EGFR-targeting antibody cetuximab, served as a targeted delivery system for the efficient administration of the p53 gene and the treatment of OC.

In another study, Kotcherlakota et al (50) combined AuNPs with the engineered bifunctional recombinant fusion protein TRAF(C) to fabricate a drug delivery system. This system facilitated the target-specific delivery of Dox and an erbB2-targeting small-interfering RNA into SK-OV-3 cells, which have upregulated expression levels of the HER2 receptor. These findings collectively suggest that AuNP-mediated gene therapy is a promising therapeutic approach for OC.

Others

In one study, thioglucose was used to modify the surface of AuNPs. The rationale behind this approach was that cancer cells have a greater metabolic rate and, therefore, a much higher glucose uptake rate than normal cells. The selective glucose uptake by cancer cells facilitated the specific internalization of the thioglucose-coated AuNPs (Glu-AuNPs) (114). In vivo data demonstrated that the accumulation of the Glu-GNPs in cancerous tissue was 10-fold greater than that in normal ovarian and uterine tissues. In another study, Geng et al demonstrated the potential of thioglucose-bound AuNPs as a sensitizer for the radiotherapy of OC. When SK-OV-3 cells were treated with the AuNPs alone, irradiation alone or the AuNPs in conjunction with irradiation, the intracellular accumulation of AuNPs resulted in greater antiproliferative activity compared with irradiation alone. The interaction between X-ray radiation and AuNPs was shown to lead to an increase in the production of ROS (62).

Combination therapies have garnered attention as a strategy to overcome the limitations associated with traditional cancer treatments. There has been an increasing interest in the use of ultrasound (US) to increase the intracellular concentration of chemotherapeutic agents, particularly in preclinical research. In addition, research has shown that NPs can enhance the efficacy of US therapy (115117). Kip et al (56) exploited the US-active property of nanocone-shaped AuNPs in a combined US and CP treatment strategy. Triple-combination therapy comprising US, AuNPs and a low dose of CP was found to effectively overcome drug resistance in OC cells in vitro, indicating its potential for the reduction of chemotherapy-induced side effects.

An economical, facile and eco-friendly method has been devised for the fabrication of anisotropic AuNPs utilizing whey proteins (Wh@AuNPs) (58). These Wh@AuNPs were found to exhibit potent catalytic activity and the ability to emit strong red fluorescence upon complexation by trypan blue, indicating their potential use in optical sensors and live/dead cell imaging. In addition, the Wh@AuNPs exhibited cytotoxic activity against breast cancer and OC cells but no toxicity toward normal cells, indicating that Wh@AuNPs may be novel theranostic agents that do not harm normal cells. However, further research is necessary to confirm the theranostic effectiveness of these Wh@AuNPs in vivo.

Therapeutic potential of AuNPs in OC

The poor biodegradability of AuNPs in vivo poses a significant challenge for clinical applications. Experiments in mice revealed that only 9% of 40-nm AuNPs administered by intravenous injection were excreted from the liver over 6 months (118). Other preclinical experiments demonstrated that a year postinjection, there was no detectible reduction in the quantity of 155-nm AuNPs retained in vivo (119). Higbee-Dempsey et al (120) synthesized biodegradable AuNPs modified with thiolated dextran, and introduced hydrophobic acetal groups onto the surface by the covalent modification of dextran. The acetal groups cleaved when exposed to an acidic environment, rendering the AuNPs highly soluble and susceptible to degradation. This carrier system was shown to facilitate the clearance of >85% of the AuNPs from the livers of mice within a span of 3 months. Therefore, this study resolves a key issue hindering the clinical translation of AuNPs and their use as nanocarrier systems.

Conjugated AuNPs have garnered widespread application as biomarkers and biodelivery vehicles within the medical sphere, with potential utility in early and advanced cancer diagnostics and therapeutics. This approach has demonstrated promise in the early identification of cancer stem cells within salivary gland tumors, as evidenced by a clinical trial using a nanocomposite of AuNPs conjugated to CD24 (NCT04907422) (121). Therefore, it is anticipated that clinical trials utilizing AuNP therapy for OC are likely to be underway in the future (Fig. 5).

Conclusions and prospects

NPs are widely used for targeted drug delivery, therapeutic purposes, catalysis, imaging and hyperthermia. AuNPs are used for various medical purposes, for example, as targeted therapeutic agents or drug delivery carriers, as well as in electronics and sensing applications. In the treatment of OC, AuNPs have been shown inhibit tumor growth, overcome drug resistance, reduce the toxicity of anticancer drugs, and prevent cancer cell invasion and migration. They can be combined with other therapies, including chemotherapy and radiotherapy, to provide an improved therapeutic effect. Despite these advances, certain constraints are associated with the development of AuNPs. First, AuNPs may exhibit toxicity at certain concentrations, particularly with long-term exposure (122). Therefore, further research is necessary to understand their toxicity and ensure biocompatibility. Second, the methods for synthesizing AuNPs are varied and often require strict experimental conditions and technical expertise. Thus, the development of simpler and more efficient preparation methods would be advantageous. Third, AuNPs often exhibit nonuniform particle size distributions, which affects their properties and application effectiveness (123). Therefore, improvements in their preparation methods are required to achieve more uniform particle size distributions. Fourth, AuNPs can aggregate and lose their activity during storage and use, which impacts their stability and long-term storage capability (124). Further research to develop more stable AuNP materials is essential. Finally, the cost of preparing AuNPs is high, limiting their potential for large-scale manufacture and use. Therefore, it is important to focus on reducing synthesis costs and improving scalability to enable the commercialization of AuNPs.

In summary, the development of AuNPs faces challenges and limitations that require further research and improvement. By addressing these issues, the application prospects of AuNPs can be further expanded.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

WH was responsible for writing the original draft of the manuscript, and for visualization. FY reviewed and edited the manuscript. QZ contributed to conceptualization of the study, supervision, editing and manuscript revision. KC conceived the idea of the study. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yang Y, Zheng X, Chen L, Gong X, Yang H, Duan X and Zhu Y: Multifunctional gold nanoparticles in cancer diagnosis and treatment. Int J Nanomedicine. 17:2041–2067. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Schoutrop E, Moyano-Galceran L, Lheureux S, Mattsson J, Lehti K, Dahlstrand H and Magalhaes I: Molecular, cellular and systemic aspects of epithelial ovarian cancer and its tumor microenvironment. Semin Cancer Biol. 86:207–223. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Zhang R, Siu MKY, Ngan HYS and Chan KKL: Molecular biomarkers for the early detection of ovarian cancer. Int J Mol Sci. 23:120412022. View Article : Google Scholar : PubMed/NCBI

4 

Bhardwaj BK, Thankachan S, Magesh P, Venkatesh T, Tsutsumi R and Suresh PS: Current update on nanotechnology-based approaches in ovarian cancer therapy. Reprod Sci. 30:335–349. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Jelovac D and Armstrong DK: Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J Clin. 61:183–203. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Piktel E, Ościłowska I, Suprewicz Ł, Depciuch J, Marcińczyk N, Chabielska E, Wolak P, Wollny T, Janion M, Parlinska-Wojtan M and Bucki R: ROS-Mediated apoptosis and autophagy in ovarian cancer cells treated with peanut-shaped gold nanoparticles. Int J Nanomedicine. 16:1993–2011. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Sperling RA and Parak WJ: Surface modification, functionalization and bioconjugation of colloidal inorganic nanoparticles. Philos Trans A Math Phys Eng Sci. 368:1333–1383. 2010.PubMed/NCBI

8 

Kamal A, Saba M, Ullah K, Almutairi SM, AlMunqedhi BM and Ragab abdelGawwad M: Mycosynthesis, characterization of zinc oxide nanoparticles, and its assessment in various biological activities. Crystals. 13:1712023. View Article : Google Scholar

9 

Huang CC, Yang Z, Lee KH and Chang HT: Synthesis of highly fluorescent gold nanoparticles for sensing mercury(II). Angew Chem Int Ed Engl. 46:6824–6828. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Huynh KH, Pham XH, Kim J, Lee SH, Chang H, Rho WY and Jun BH: Synthesis, properties, and biological applications of metallic alloy nanoparticles. Int J Mol Sci. 21:51742020. View Article : Google Scholar : PubMed/NCBI

11 

Yaqoob AA, Ahmad H, Parveen T, Ahmad A, Oves M, Ismail IMI, Qari HA, Umar K and Mohamad Ibrahim MN: Recent advances in metal decorated nanomaterials and their various biological applications: A review. Front Chem. 8:3412020. View Article : Google Scholar : PubMed/NCBI

12 

Vargas-Molinero HY, Serrano-Medina A, Palomino-Vizcaino K, López-Maldonado EA, Villarreal-Gómez LJ, Pérez-González GL and Cornejo-Bravo JM: Hybrid systems of nanofibers and polymeric nanoparticles for biological application and delivery systems. Micromachines (Basel). 14:2082023. View Article : Google Scholar : PubMed/NCBI

13 

Guo R, Song Y, Wang G and Murray RW: Does core size matter in the kinetics of ligand exchanges of monolayer-protected Au clusters? J Am Chem Soc. 127:2752–2757. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Daniel MC and Astruc D: Gold nanoparticles: Assembly, supramolecular chemistry, quantum-size-related properties, and applications toward biology, catalysis, and nanotechnology. Chem Rev. 104:293–346. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Jiang S, Gnanasammandhan MK and Zhang Y: Optical imaging-guided cancer therapy with fluorescent nanoparticles. J R Soc Interface. 7:3–18. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Aziz F, Ihsan A, Nazir A, Ahmad I, Bajwa SZ, Rehman A, Diallo A and Khan WS: Novel route synthesis of porous and solid gold nanoparticles for investigating their comparative performance as contrast agent in computed tomography scan and effect on liver and kidney function. Int J Nanomedicine. 12:1555–1563. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Spivak MY, Bubnov RV, Yemets IM, Lazarenko LM, Tymoshok NO and Ulberg ZR: Development and testing of gold nanoparticles for drug delivery and treatment of heart failure: a theranostic potential for PPP cardiology. EPMA J. 4:202013. View Article : Google Scholar : PubMed/NCBI

18 

Khan JA, Pillai B, Das TK, Singh Y and Maiti S: Molecular effects of uptake of gold nanoparticles in HeLa cells. Chembiochem. 8:1237–1240. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Scaletti F, Hardie J, Lee YW, Luther DC, Ray M and Rotello VM: Protein delivery into cells using inorganic nanoparticle-protein supramolecular assemblies. Chem Soc Rev. 47:3421–3432. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Rosi NL, Giljohann DA, Thaxton CS, Lytton-Jean AK, Han MS and Mirkin CA: Oligonucleotide-modified gold nanoparticles for intracellular gene regulation. Science. 312:1027–1030. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Dykman LA and Khlebtsov NG: Immunological properties of gold nanoparticles. Chem Sci. 8:1719–1735. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Chen YS, Hung YC, Lin WH and Huang GS: Assessment of gold nanoparticles as a size-dependent vaccine carrier for enhancing the antibody response against synthetic foot-and-mouth disease virus peptide. Nanotechnology. 21:1951012010. View Article : Google Scholar : PubMed/NCBI

23 

Connor EE, Mwamuka J, Gole A, Murphy CJ and Wyatt MD: Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity. Small. 1:325–327. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Ielo I, Rando G, Giacobello F, Sfameni S, Castellano A, Galletta M, Drommi D, Rosace G and Plutino MR: Synthesis, chemical-physical characterization, and biomedical applications of functional gold nanoparticles: A review. Molecules. 26:58232021. View Article : Google Scholar : PubMed/NCBI

25 

Xiong D, Chen M and Li H: Synthesis of para-sulfonatocalix[4]arene-modified silver nanoparticles as colorimetric histidine probes. Chem Commun (Camb). 880–882. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Xiao W, Xiong J, Zhang S, Xiong Y, Zhang H and Gao H: Influence of ligands property and particle size of gold nanoparticles on the protein adsorption and corresponding targeting ability. Int J Pharm. 538:105–111. 2018. View Article : Google Scholar : PubMed/NCBI

27 

De Luca G, Bonaccorsi P, Trovato V, Mancuso A, Papalia T, Pistone A, Casaletto MP, Mezzi A, Brunetti B, Minuti L, et al: Tripodal tris-disulfides as capping agents for a controlled mixed functionalization of gold nanoparticles. New J Chem. 42:16436–16440. 2018. View Article : Google Scholar

28 

Boyer C, Whittaker MR, Chuah K, Liu J and Davis TP: Modulation of the surface charge on polymer-stabilized gold nanoparticles by the application of an external stimulus. Langmuir. 26:2721–2730. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Presnova GV, Rubtsova MY, Presnov DE, Grigorenko VG, Yaminsky IV and Egorov AM: Conjugates of Streptavidin conjugates with gold nanoparticles for the visualization of DNA single interactions on the silicon surface. Biomed Khim. 60:538–542. 2014.(In Russian). View Article : Google Scholar : PubMed/NCBI

30 

Das RK, Pachapur VL, Lonappan L, Naghdi M, Pulicharla R, Maiti S, Cledon M, Dalila LMA, Sarma SJ and Brar SK: Biological synthesis of metallic nanoparticles: Plants, animals and microbial aspects. Nanotechnol. Environ. Eng. 2:182017.

31 

Thakkar KN, Mhatre SS and Parikh RY: Biological synthesis of metallic nanoparticles. Nanomedicine. 6:257–262. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Shnoudeh AJ, Hamad I, Abdo RW, et al: Synthesis, Characterization, and Applications of Metal Nanoparticles. Biomaterials and Bionanotechnology. pp527–612. 2019. View Article : Google Scholar

33 

Kharissova OV, Kharisov BI, Oliva González CM, Méndez YP and López I: Greener synthesis of chemical compounds and materials. R Soc Open Sci. 6:1913782019. View Article : Google Scholar : PubMed/NCBI

34 

Medici S, Peana M, Nurchi VM, Lachowicz JI, Crisponi G and Zoroddu MA: Noble metals in medicine: Latest advances. Coord Chem Rev. 284:329–350. 2015. View Article : Google Scholar

35 

Salem SS and Fouda A: Green synthesis of metallic nanoparticles and their prospective biotechnological applications: An overview. Biol Trace Elem Res. 199:344–370. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Gu X, Xu Z, Gu L, Xu H, Han F, Chen B and Pan X: Preparation and antibacterial properties of gold nanoparticles: A review. Environ Chem Lett. 19:167–187. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Amina SJ and Guo B: A review on the synthesis and functionalization of gold nanoparticles as a drug delivery vehicle. Int J Nanomedicine. 15:9823–9857. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Omran BA, Whitehead KA and Baek KH: One-pot bioinspired synthesis of fluorescent metal chalcogenide and carbon quantum dots: Applications and potential biotoxicity. Colloids Surf B Biointerfaces. 200:1115782021. View Article : Google Scholar : PubMed/NCBI

39 

Xu F, Li Y, Zhao X, Liu G, Pang B, Liao N, Li H and Shi J: Diversity of fungus-mediated synthesis of gold nanoparticles: Properties, mechanisms, challenges, and solving methods. Crit Rev Biotechnol. 44:924–940. 2024. View Article : Google Scholar : PubMed/NCBI

40 

Borse VB, Konwar AN, Jayant RD and Patil PO: Perspectives of characterization and bioconjugation of gold nanoparticles and their application in lateral flow immunosensing. Drug Deliv Transl Res. 10:878–902. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Murphy CJ: Materials science. Nanocubes and nanoboxes. Science. 298:2139–2141. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Das SK and Marsili E: A green chemical approach for the synthesis of gold nanoparticles: Characterization and mechanistic aspect. Rev Environ Sci Biotechnol. 9:199–204. 2010. View Article : Google Scholar

43 

Hamelian M, Varmira K and Veisi H: Green synthesis and characterizations of gold nanoparticles using Thyme and survey cytotoxic effect, antibacterial and antioxidant potential. J Photochem Photobiol B. 184:71–79. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Patil MP and Kim GD: Eco-friendly approach for nanoparticles synthesis and mechanism behind antibacterial activity of silver and anticancer activity of gold nanoparticles. Appl Microbiol Biotechnol. 101:79–92. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Patil MP and Kim GD: Marine microorganisms for synthesis of metallic nanoparticles and their biomedical applications. Colloids Surf B Biointerfaces. 172:487–495. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Shedbalkar U, Singh R, Wadhwani S, Gaidhani S and Chopade BA: Microbial synthesis of gold nanoparticles: Current status and future prospects. Adv Colloid Interface Sci. 209:40–48. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Beveridge TJ and Murray RG: Sites of metal deposition in the cell wall of Bacillus subtilis. J Bacteriol. 141:876–887. 1980. View Article : Google Scholar : PubMed/NCBI

48 

Lee CS, Kim TW, Oh DE, Bae SO, Ryu J, Kong H, Jeon H, Seo HK, Jeon S and Kim TH: In vivo and in vitro anticancer activity of doxorubicin-loaded DNA-AuNP nanocarrier for the ovarian cancer treatment. Cancers (Basel). 12:6342020. View Article : Google Scholar : PubMed/NCBI

49 

Lee CS, Kim TW, Kang Y, Ju Y, Ryu J, Kong H, Jang YS, Oh DE, Jang SJ, Cho H, et al: Targeted drug delivery nanocarriers based on hyaluronic acid-decorated dendrimer encapsulating gold nanoparticles for ovarian cancer therapy. Mater Today Chem. 26:1010832022. View Article : Google Scholar

50 

Kotcherlakota R, Srinivasan DJ, Mukherjee S, Haroon MM, Dar GH, Venkatraman U, Patra CR and Gopal V: Engineered fusion protein-loaded gold nanocarriers for targeted co-delivery of doxorubicin and erbB2-siRNA in human epidermal growth factor receptor-2+ ovarian cancer. J Mater Chem B. 5:7082–7098. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Kotcherlakota R, Vydiam K, Jeyalakshmi Srinivasan D, Mukherjee S, Roy A, Kuncha M, Rao TN, Sistla R, Gopal V and Patra CR: Restoration of p53 function in ovarian cancer mediated by gold nanoparticle-based EGFR targeted gene delivery system. ACS Biomater Sci Eng. 5:3631–3644. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Piktel E, Oscilowska I, Suprewicz Ł, Depciuch J, Marcińczyk N, Chabielska E, Wolak P, Głuszek K, Klimek J, Zieliński PM, et al: Peanut-Shaped gold nanoparticles with shells of ceragenin CSA-131 display the ability to inhibit ovarian cancer growth in vitro and in a tumor xenograft model. Cancers (Basel). 13:54242021. View Article : Google Scholar : PubMed/NCBI

53 

Jabir M, Sahib UI, Taqi Z, Taha A, Sulaiman G, Albukhaty S, Al-Shammari A, Alwahibi M, Soliman D, Dewir YH and Rizwana H: Linalool-Loaded glutathione-modified gold nanoparticles conjugated with CALNN peptide as apoptosis inducer and NF-κB translocation inhibitor in SKOV-3 cell line. Int J Nanomedicine. 15:9025–9047. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Asl SS, Tafvizi F and Noorbazargan H: Biogenic synthesis of gold nanoparticles using Satureja rechingeri Jamzad: A potential anticancer agent against cisplatin-resistant A2780CP ovarian cancer cells. Environ Sci Pollut Res Int. 30:20168–20184. 2023. View Article : Google Scholar : PubMed/NCBI

55 

Xiong X, Arvizo RR, Saha S, Robertson DJ, McMeekin S, Bhattacharya R and Mukherjee P: Sensitization of ovarian cancer cells to cisplatin by gold nanoparticles. Oncotarget. 5:6453–6465. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Kip B, Tunc CU and Aydin O: Triple-combination therapy assisted with ultrasound-active gold nanoparticles and ultrasound therapy against 3D cisplatin-resistant ovarian cancer model. Ultrason Sonochem. 82:1059032022. View Article : Google Scholar : PubMed/NCBI

57 

Patra CR, Bhattacharya R and Mukherjee P: Fabrication and functional characterization of goldnanoconjugates for potential application in ovarian cancer. J Mater Chem. 20:547–554. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Borghei YS and Hosseinkhani S: Bio-synthesis of a functionalized whey proteins theranostic nanoprobe with cancer-specific cytotoxicity and as a live/dead cell imaging probe. Journal of Photochemistry and Photobiology A: Chemistry. 431:1140252022. View Article : Google Scholar : PubMed/NCBI

59 

Wang L, Wang L, Xu T, Guo C, Liu C, Zhang H, Li J and Liang Z: Synthesis of 15P-conjugated PPy-modified gold nanoparticles and their application to photothermal therapy of ovarian cancer. Chem Res Chin Univ. 30:959–964. 2014. View Article : Google Scholar

60 

Shen Y, Wang M, Wang H, Zhou J and Chen J: Multifunctional human serum albumin fusion protein as a docetaxel nanocarrier for chemo-photothermal synergetic therapy of ovarian cancer. ACS Appl Mater Interfaces. 14:19907–19917. 2022. View Article : Google Scholar : PubMed/NCBI

61 

Van de Broek B, Devoogdt N, D'Hollander A, Gijs HL, Jans K, Lagae L, Muyldermans S, Maes G and Borghs G: Specific cell targeting with nanobody conjugated branched gold nanoparticles for photothermal therapy. ACS Nano. 5:4319–4328. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Geng F, Song K, Xing JZ, Yuan C, Yan S, Yang Q, Chen J and Kong B: Thio-glucose bound gold nanoparticles enhance radio-cytotoxic targeting of ovarian cancer. Nanotechnology. 22:2851012011. View Article : Google Scholar : PubMed/NCBI

63 

Cui T, Liang JJ, Chen H, Geng DD, Jiao L, Yang JY, Qian H, Zhang C and Ding Y: Performance of doxorubicin-conjugated gold nanoparticles: Regulation of drug location. ACS Appl Mater Interfaces. 9:8569–8580. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Wani WA, Baig U, Shreaz S, Shiekh RA, Iqbal PF, Jameel E, Ahmad A, Mohd-Setapar SH, Mushtaque M and Hun LT: Recent advances in iron complexes as potential anticancer agents. New J Chem. 40:1063–1090. 2016. View Article : Google Scholar

65 

Baetke SC, Lammers T and Kiessling F: Applications of nanoparticles for diagnosis and therapy of cancer. Br J Radiol. 88:201502072015. View Article : Google Scholar : PubMed/NCBI

66 

Fan M, Han Y, Gao S, Yan H, Cao L, Li Z, Liang XJ and Zhang J: Ultrasmall gold nanoparticles in cancer diagnosis and therapy. Theranostics. 10:4944–4957. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Wang J, Potocny AM, Rosenthal J and Day ES: Gold nanoshell-linear tetrapyrrole conjugates for near infrared-activated dual photodynamic and photothermal therapies. ACS Omega. 5:926–940. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Zhao X, Campbell S, Wallace GQ, Claing A, Bazuin CG and Masson JF: Branched Au nanoparticles on nanofibers for surface-enhanced raman scattering sensing of intracellular pH and extracellular pH gradients. ACS Sens. 5:2155–2167. 2020. View Article : Google Scholar : PubMed/NCBI

69 

Skrabalak SE, Chen J, Sun Y, Lu X, Au L, Cobley CM and Xia Y: Gold nanocages: Synthesis, properties, and applications. Acc Chem Res. 41:1587–1595. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Yang M, Wang W, Qiu J, Bai MY and Xia Y: Direct visualization and semi-quantitative analysis of payload loading in the case of gold nanocages. Angew Chem Int Ed Engl. 58:17671–17674. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Murthy SK: Nanoparticles in modern medicine: State of the art and future challenges. Int J Nanomedicine. 2:129–141. 2007.PubMed/NCBI

72 

Ghosh P, Yang X, Arvizo R, Zhu ZJ, Agasti SS, Mo Z and Rotello VM: Intracellular delivery of a membrane-impermeable enzyme in active form using functionalized gold nanoparticles. J Am Chem Soc. 132:2642–2645. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Zhou T, Du Y and Wei T: Transcriptomic analysis of human breast cancer cells reveals differentially expressed genes and related cellular functions and pathways in response to gold nanorods. Biophys Rep. 1:106–114. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Hu X, Zhang Y, Ding T, Liu J and Zhao H: Multifunctional gold nanoparticles: A novel nanomaterial for various medical applications and biological activities. Front Bioeng Biotechnol. 8:9902020. View Article : Google Scholar : PubMed/NCBI

75 

Zhang J, Mou L and Jiang X: Surface chemistry of gold nanoparticles for health-related applications. Chem Sci. 11:923–936. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Shukla R, Bansal V, Chaudhary M, Basu A, Bhonde RR and Sastry M: Biocompatibility of gold nanoparticles and their endocytotic fate inside the cellular compartment: A microscopic overview. Langmuir. 21:10644–10654. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Ben Haddada M, Gerometta E, Chawech R, Sorres J, Bialecki A, Pesnel S, Spadavecchia J and Morel AL: Assessment of antioxidant and dermoprotective activities of gold nanoparticles as safe cosmetic ingredient. Colloids Surf B Biointerfaces. 189:1108552020. View Article : Google Scholar : PubMed/NCBI

78 

Taratummarat S, Sangphech N, Vu CTB, Palaga T, Ondee T, Surawut S, Sereemaspun A, Ritprajak P and Leelahavanichkul A: Gold nanoparticles attenuates bacterial sepsis in cecal ligation and puncture mouse model through the induction of M2 macrophage polarization. BMC Microbiol. 18:852018. View Article : Google Scholar : PubMed/NCBI

79 

Feng ZV, Gunsolus IL, Qiu TA, Hurley KR, Nyberg LH, Frew H, Johnson KP, Vartanian AM, Jacob LM, Lohse SE, et al: Impacts of gold nanoparticle charge and ligand type on surface binding and toxicity to Gram-negative and Gram-positive bacteria. Chem Sci. 6:5186–5196. 2015. View Article : Google Scholar : PubMed/NCBI

80 

Cho TJ, MacCuspie RI, Gigault J, Gorham JM, Elliott JT and Hackley VA: Highly stable positively charged dendron-encapsulated gold nanoparticles. Langmuir. 30:3883–3893. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Schaeublin NM, Braydich-Stolle LK, Schrand AM, Miller JM, Hutchison J, Schlager JJ and Hussain SM: Surface charge of gold nanoparticles mediates mechanism of toxicity. Nanoscale. 3:410–420. 2011. View Article : Google Scholar : PubMed/NCBI

82 

Singh P, Pandit S, Mokkapati VRSS, Garg A, Ravikumar V and Mijakovic I: Gold nanoparticles in diagnostics and therapeutics for human cancer. Int J Mol Sci. 19:19792018. View Article : Google Scholar : PubMed/NCBI

83 

Ghosh C, Priegue P, Leelayuwapan H, Fuchsberger FF, Rademacher C and Seeberger PH: Synthetic Glyconanoparticles Modulate Innate Immunity but Not the Complement System. ACS Appl Bio Mater. 5:2185–2192. 2022. View Article : Google Scholar : PubMed/NCBI

84 

Nath D and Banerjee P: Green nanotechnology-a new hope for medical biology. Environ Toxicol Pharmacol. 36:997–1014. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Pan Y, Leifert A, Ruau D, Neuss S, Bornemann J, Schmid G, Brandau W, Simon U and Jahnen-Dechent W: Gold nanoparticles of diameter 1.4 nm trigger necrosis by oxidative stress and mitochondrial damage. Small. 5:2067–2076. 2009. View Article : Google Scholar : PubMed/NCBI

86 

Yasinska IM, Calzolai L, Raap U, Hussain R, Siligardi G, Sumbayev VV and Gibbs BF: Targeting of basophil and mast cell pro-allergic reactivity using functionalised gold nanoparticles. Front Pharmacol. 10:3332019. View Article : Google Scholar : PubMed/NCBI

87 

Weaver JL, Tobin GA, Ingle T, Bancos S, Stevens D, Rouse R, Howard KE, Goodwin D, Knapton A, Li X, et al: Evaluating the potential of gold, silver, and silica nanoparticles to saturate mononuclear phagocytic system tissues under repeat dosing conditions. Part Fibre Toxicol. 14:252017. View Article : Google Scholar : PubMed/NCBI

88 

Golchin K, Golchin J, Ghaderi S, Alidadiani N, Eslamkhah S, Eslamkhah M, Davaran S and Akbarzadeh A: Gold nanoparticles applications: From artificial enzyme till drug delivery. Artif Cells Nanomed Biotechnol. 46:250–254. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Parveen S, Misra R and Sahoo SK: Nanoparticles: A boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine. 8:147–166. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Byrne JD, Betancourt T and Brannon-Peppas L: Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev. 60:1615–1626. 2008. View Article : Google Scholar : PubMed/NCBI

91 

Attia MF, Anton N, Wallyn J, Omran Z and Vandamme TF: An overview of active and passive targeting strategies to improve the nanocarriers efficiency to tumour sites. J Pharm Pharmacol. 71:1185–1198. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Blanco MD, Teijon C, Olmo RM and Teijo JM: Targeted Nanoparticles for Cancer Therapy. In: Recent Advances in Novel Drug Carrier Systems. InTech; 2012

93 

Melancon M, Lu W and Li C: Gold-Based magneto/optical nanostructures: Challenges for in vivo applications in cancer diagnostics and therapy. Mater Res Bull. 34:415–421. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Wu J: The enhanced permeability and retention (EPR) Effect: The significance of the concept and methods to enhance its application. J Pers Med. 11:7712021. View Article : Google Scholar : PubMed/NCBI

95 

Matsumura Y and Maeda H: A new concept for macromolecular therapeutics in cancer chemotherapy: Mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 46((12 Pt 1)): 6387–6392. 1986.PubMed/NCBI

96 

Wilhelm S, Tavares AJ, Dai Q, Ohta S, Audet J, Dvorak HF and Chan WCW: Analysis of nanoparticle delivery to tumours. Nat Rev Mater. 1:160142016. View Article : Google Scholar

97 

Daraee H, Eatemadi A, Abbasi E, Fekri Aval S, Kouhi M and Akbarzadeh A: Application of gold nanoparticles in biomedical and drug delivery. Artif Cells Nanomed Biotechnol. 44:410–422. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Bai X, Wang Y, Song Z, Feng Y, Chen Y, Zhang D and Feng L: The basic properties of gold nanoparticles and their applications in tumor diagnosis and treatment. Int J Mol Sci. 21:24802020. View Article : Google Scholar : PubMed/NCBI

99 

Wang X and Guo Z: Targeting and delivery of platinum-based anticancer drugs. Chem Soc Rev. 42:202–224. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Ruan S, Xiao W, Hu C, Zhang H, Rao J, Wang S, Wang X, He Q and Gao H: Ligand-Mediated and enzyme-directed precise targeting and retention for the enhanced treatment of glioblastoma. ACS Appl Mater Interfaces. 9:20348–20360. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Bao QY, Geng DD, Xue JW, Zhou G, Gu SY, Ding Y and Zhang C: Glutathione-mediated drug release from Tiopronin-conjugated gold nanoparticles for acute liver injury therapy. Int J Pharm. 446:112–118. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Piktel E, Niemirowicz K, Wątek M, Wollny T, Deptuła P and Bucki R: Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J Nanobiotechnology. 14:392016. View Article : Google Scholar : PubMed/NCBI

103 

Hartmann JT, Kollmannsberger C, Kanz L and Bokemeyer C: Platinum organ toxicity and possible prevention in patients with testicular cancer. Int J Cancer. 83:866–869. 1999. View Article : Google Scholar : PubMed/NCBI

104 

Thompson SW, Davis LE, Kornfeld M, Hilgers RD and Standefer JC: Cisplatin neuropathy. Clinical, electrophysiologic, morphologic, and toxicologic studies. Cancer. 54:1269–1275. 1984. View Article : Google Scholar : PubMed/NCBI

105 

Yoo J, Park C, Yi G, Lee D and Koo H: Active targeting strategies using biological ligands for nanoparticle drug delivery systems. Cancers. 11:6402019. View Article : Google Scholar : PubMed/NCBI

106 

Wang Z, Qiao R, Tang N, Lu Z, Wang H, Zhang Z, Xue X, Huang Z, Zhang S, Zhang G and Li Y: Active targeting theranostic iron oxide nanoparticles for MRI and magnetic resonance-guided focused ultrasound ablation of lung cancer. Biomaterials. 127:25–35. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Suh MS, Shen J, Kuhn LT and Burgess DJ: Layer-by-layer nanoparticle platform for cancer active targeting. Int J Pharm. 517:58–66. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Slovak R, Ludwig JM, Gettinger SN, Herbst RS and Kim HS: Immuno-thermal ablations-boosting the anticancer immune response. J Immunother Cancer. 5:782017. View Article : Google Scholar : PubMed/NCBI

109 

Kroemer G, Galassi C, Zitvogel L and Galluzzi L: Immunogenic cell stress and death. Nat Immunol. 23:487–500. 2022. View Article : Google Scholar : PubMed/NCBI

110 

Goddard ZR, Marín MJ, Russell DA and Searcey M: Active targeting of gold nanoparticles as cancer therapeutics. Chem Soc Rev. 49:8774–8789. 2020. View Article : Google Scholar : PubMed/NCBI

111 

Emami F, Banstola A, Vatanara A, Lee S, Kim JO, Jeong JH and Yook S: Doxorubicin and Anti-PD-L1 antibody conjugated gold nanoparticles for colorectal cancer photochemotherapy. Mol Pharm. 16:1184–1199. 2019. View Article : Google Scholar : PubMed/NCBI

112 

Huang N, Liu Y, Fang Y, Zheng S, Wu J, Wang M, Zhong W, Shi M, Xing M and Liao W: Gold nanoparticles induce tumor vessel normalization and impair metastasis by inhibiting endothelial smad2/3 signaling. ACS Nano. 14:7940–7958. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Berchuck A, Kamel A, Whitaker R, Kerns B, Olt G, Kinney R, Soper JT, Dodge R, Clarke-Pearson DL, Marks P, et al: Overexpression of HER-2/neu is associated with poor survival in advanced epithelial ovarian cancer. Cancer Res. 50:4087–4091. 1990.PubMed/NCBI

114 

Kong T, Zeng J, Wang X, Yang X, Yang J, McQuarrie S, McEwan A, Roa W, Chen J and Xing JZ: Enhancement of radiation cytotoxicity in breast-cancer cells by localized attachment of gold nanoparticles. Small. 4:1537–1543. 2008. View Article : Google Scholar : PubMed/NCBI

115 

Zhang Y, Yu J, Bomba HN, Zhu Y and Gu Z: Mechanical force-triggered drug delivery. Chem Rev. 116:12536–12563. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Wood AK and Sehgal CM: A review of low-intensity ultrasound for cancer therapy. Ultrasound Med Biol. 41:905–928. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Zhang Y, Wan Y, Chen Y, Blum NT, Lin J and Huang P: Ultrasound-Enhanced chemo-photodynamic combination therapy by using albumin ‘Nanoglue’-Based Nanotheranostics. ACS Nano. 14:5560–5569. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Sadauskas E, Danscher G, Stoltenberg M, Vogel U, Larsen A and Wallin H: Protracted elimination of gold nanoparticles from mouse liver. Nanomedicine. 5:162–169. 2009. View Article : Google Scholar : PubMed/NCBI

119 

Gad SC, Sharp KL, Montgomery C, Payne JD and Goodrich GP: Evaluation of the toxicity of intravenous delivery of auroshell particles (gold-silica nanoshells). Int J Toxicol. 31:584–594. 2012. View Article : Google Scholar : PubMed/NCBI

120 

Higbee-Dempsey EM, Amirshaghaghi A, Case MJ, Bouché M, Kim J, Cormode DP and Tsourkas A: Biodegradable Gold nanoclusters with improved excretion due to pH-Triggered hydrophobic-to-hydrophilic transition. J Am Chem Soc. 142:7783–7794. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Kesharwani P, Ma R, Sang L, Fatima M, Sheikh A, Abourehab MAS, Gupta N, Chen ZS and Zhou Y: Gold nanoparticles and gold nanorods in the landscape of cancer therapy. Mol Cancer. 22:982023. View Article : Google Scholar : PubMed/NCBI

122 

Mironava T, Hadjiargyrou M, Simon M, Jurukovski V and Rafailovich MH: Gold nanoparticles cellular toxicity and recovery: Effect of size, concentration and exposure time. Nanotoxicology. 4:120–137. 2010. View Article : Google Scholar : PubMed/NCBI

123 

Mikhailova EO: Gold nanoparticles: Biosynthesis and potential of biomedical application. J Funct Biomater. 12:2021. View Article : Google Scholar : PubMed/NCBI

124 

Epanchintseva AV, Poletaeva JE, Pyshnyi DV, Ryabchikova EI and Pyshnaya IA: Long-term stability and scale-up of noncovalently bound gold nanoparticle-siRNA suspensions. Beilstein J Nanotechnol. 10:2568–2578. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 28 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He W, Yang F, Chen K and Zeng Q: Targeted gold nanoparticles for ovarian cancer (Review). Oncol Lett 28: 589, 2024.
APA
He, W., Yang, F., Chen, K., & Zeng, Q. (2024). Targeted gold nanoparticles for ovarian cancer (Review). Oncology Letters, 28, 589. https://doi.org/10.3892/ol.2024.14723
MLA
He, W., Yang, F., Chen, K., Zeng, Q."Targeted gold nanoparticles for ovarian cancer (Review)". Oncology Letters 28.6 (2024): 589.
Chicago
He, W., Yang, F., Chen, K., Zeng, Q."Targeted gold nanoparticles for ovarian cancer (Review)". Oncology Letters 28, no. 6 (2024): 589. https://doi.org/10.3892/ol.2024.14723