Next Article in Journal
Repurposed Drugs That Activate Autophagy in Filarial Worms Act as Effective Macrofilaricides
Previous Article in Journal
Photodynamic Antibacterial Therapy of Gallic Acid-Derived Carbon-Based Nanoparticles (GACNPs): Synthesis, Characterization, and Hydrogel Formulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review of Advances in Coating and Functionalization of Gold Nanoparticles: From Theory to Biomedical Application

by
Wilmmer Alexander Arcos Rosero
,
Angelica Bueno Barbezan
,
Carla Daruich de Souza
and
Maria Elisa Chuery Martins Rostelato
*
Instituto de Pesquisas Energéticas e Nucleares, São Paulo 05508-000, Brazil
*
Author to whom correspondence should be addressed.
Pharmaceutics 2024, 16(2), 255; https://doi.org/10.3390/pharmaceutics16020255
Submission received: 27 October 2023 / Revised: 27 November 2023 / Accepted: 2 January 2024 / Published: 9 February 2024
(This article belongs to the Section Nanomedicine and Nanotechnology)

Abstract

:
Nanoparticles, especially gold nanoparticles (Au NPs) have gained increasing interest in biomedical applications. Used for disease prevention, diagnosis and therapies, its significant advantages in therapeutic efficacy and safety have been the main target of interest. Its application in immune system prevention, stability in physiological environments and cell membranes, low toxicity and optimal bioperformances are critical to the success of engineered nanomaterials. Its unique optical properties are great attractors. Recently, several physical and chemical methods for coating these NPs have been widely used. Biomolecules such as DNA, RNA, peptides, antibodies, proteins, carbohydrates and biopolymers, among others, have been widely used in coatings of Au NPs for various biomedical applications, thus increasing their biocompatibility while maintaining their biological functions. This review mainly presents a general and representative view of the different types of coatings and Au NP functionalization using various biomolecules, strategies and functionalization mechanisms.

1. Introduction

Nanotechnology, resulting from technological advances in microscopy, involves the manipulation of matter at a nanometric scale, very close to quantum limits. Although the analysis of archaeological objects has revealed the presence of nanoparticles in decorative items, the boom in nanotechnology research occurred at the end of the past century, addressing questions about obtaining nanomaterials. In databases, there are thousands of articles on nanotechnology, covering synthesis, characterization and applications in various fields from medicine to geology. On a nanometric scale, matter exhibits characteristics distinct from those on a macroscopic scale, forming the basis for significant technological advances [1,2].
Nanoparticles are typically presented as colloids, consisting of a solid phase and a liquid phase, usually water or an organic solvent, due to the methods of obtaining them. Two main routes for obtaining nanoparticles are top–down and bottom–up. The former is primarily of a physical nature, such as laser ablation, while the latter is mainly a chemical route, starting with molecules that interact to produce stable nanometric systems with specific characteristics [3].
Colloids are of great interest in various research areas due to their intrinsic properties, including a high surface-to-volume ratio, facilitating interactions and increasing reactivity and functionalization possibilities of nanomaterials.
The optical properties resulting from surface plasmon resonance in metallic nanomaterials have found applications in a wide spectrum of technological and medical applications. When illuminated by light, metal particles undergo a coherent collective oscillation of free electrons in response to the oscillating electromagnetic field of the light. This electronic oscillation around the particle surface induces a charge separation with the ionic lattice, resulting in a dipole oscillation aligned with the electric field of light (Figure 1A), which can favor electrostatic interactions of the dipole–dipole type. This phenomenon, known as surface plasmon resonance (SPR), manifests as a maximal amplitude oscillation at a specific frequency. Notably, SPR in plasmon nanoparticles, particularly those composed of noble metals like Au and Ag, exhibits a significantly stronger absorption of incident light compared to other metals. Measurement of SPR can be conducted using a UV-Vis absorption spectrometer. The intensity and wavelength of the SPR band are influenced by factors affecting electron charge density on the particle surface, including metal type, particle size, shape, structure, composition and the dielectric constant of the surrounding medium, as elucidated by Mie theory [4].
Metallic nanoparticles in colloidal form, particularly iron oxide, gold and silver, have attracted attention due to their versatility, ease of synthesis and stability [5]. Au NPs, in particular, are extensively studied for medical applications, as they can be coupled with various functionalizing moieties, including ligands, medicinal agents, DNA, amino acids, proteins, peptides and oligonucleotides, through different types of interactions (Figure 1B). Numerous studies have demonstrated that Au NPs enter organelles and blood vessels to reach the tumor site, reflecting the extensive research in this area [1].
Innovative nanotechnologies are transforming clinical practices, particularly in diagnosing, treating and managing human diseases [6]. These technologies focus on the nanoscale drug delivery system, which ensures targeted and efficient drug delivery. Active targeting is achieved by integrating a ligand specific to the disease’s receptor or epitope [7].
Key to these nanotechnologies is their biocompatibility and biodegradability. This ensures that, once the drug is delivered, the nanocarrier breaks down into harmless components, facilitating its safe removal from the body [6].
Upon administration, nanoparticles undergo biodistribution, crossing epithelial barriers and circulating through the vascular system. Smaller particles (<20–30 nm) are typically excreted renally, while larger ones may be absorbed by the mononuclear phagocytic system (MPS) in organs like the liver and spleen [8].
The clearance rates of these particles are influenced by factors such as endothelial fenestral pore sizes, age, sex and genetics. This variability presents challenges in assessing nanomedicine efficacy and toxicity across different individuals [6].
Finally, the uptake of nanocarriers by macrophages is regulated through opsonization, a process mediated by the innate immune system [9].
Currently, nanoparticle-based therapeutic systems, exemplified by Doxil, have gained traction in the pharmaceutical market. Doxil, a widely used chemotherapeutic drug, employs a polyethylene glycol (PEG) liposome delivery system. While liposomes dominate the landscape of FDA-approved nanotechnology drugs, recent attention has shifted toward inorganic materials characterized by a high atomic number (Z). In the realm of radiotherapy, these materials aim to enhance the local radiation dosage while minimizing damage to healthy tissues [2,10].
Research on lymphatic administration of liposomal formulations dates to the 1940s, with J.Y. Johnson pioneering the development of artificial phospholipid vesicles, known as liposomes, for pharmaceutical industry applications [11].
In the following years, various researchers proposed similar methods for liposome creation, particularly emphasizing medical applications. This period marked significant advances, especially when compared to gold nanoparticles, an area that has recently seen increased research activity.
Between 2019 and 2021, there was a notable reduction in publications on gold nanoparticles in biomedical applications, an impact attributed to the COVID-19 pandemic. However, in 2022, with the easing of restrictions and the search for alternatives to combat the virus, a resurgence in research involving AuNPs was observed.
Many of the recent studies utilizing AuNPs as a vehicle are discussed in this review. Despite the increased focus on gold nanoparticles, their biomedical applications continue to show promising prospects.
Comparing methods for obtaining liposomes with gold nanoparticles (AuNPs), we find that liposomes spontaneously form through the amphiphilic properties of phospholipids, creating bilayers [12]. Although obtaining AuNPs is not inherently complex, it requires specific conditions. A significant advantage of AuNPs is the multitude of published synthesis methods, many with ecological considerations [13,14].
Liposomes and AuNPs offer numerous advantages, each showcasing diverse applications and commercialized products, reflecting variations in research timelines. Notably, a critical limitation of AuNPs is their cytotoxicity, which depends on particle size, morphology, environmental conditions and production methods [15]. In contrast, liposomes present versatile targeting options, employing both active and passive techniques to focus on specific tissues.
The clearance of liposomes by the mononuclear phagocytic system (MPS), due to their size range of 400–5000 nm, is a notable challenge. PEGylation helps mitigate opsonization, thereby reducing MPS clearance and extending their circulation half-life.
Gold nanoparticles (Au NPs) are recognized as the most stable metal nanoparticles, exhibiting unique properties not found in macroscopic materials. Their nanometric form allows for various advantageous features, especially in detection applications where intrinsic nanoparticle properties are utilized for intelligent sensors [16,17]. The synthesis of AuNPs is relatively straightforward, with versatility in altering size and shape through synthesis parameter modifications [16,17].
An additional noteworthy characteristic of AuNPs is their large surface-to-volume ratio. This facilitates the immobilization of substantial quantities of specific functional groups, resulting in swift responses and enhanced sensitivity to the target analyte. The plasmon resonance in AuNPs can be easily modified, allowing for variable optical properties tailored to specific needs. Moreover, nanoparticles exhibit excellent chemical and biological compatibility with active molecules, making them ideal for transport scaffolding with minimal impact on the functional activity of their active coating. This versatility extends to applications in colorimetric, fluorimetric, spectroscopic and electrochemical methods, positioning AuNPs as multifaceted detection platforms [2,16,18,19].
The diverse applications of AuNPs are dictated by their unique characteristics. The ability to couple with a wide array of functionalizing moieties, including ligands, therapeutic agents, DNA, amino acids, proteins, peptides and oligonucleotides, has fueled interest in their medical applications. Extensive research indicates that AuNPs not only traverse blood vessels to reach tumor locations but also penetrate organelles. The inefficient removal of AuNPs from tumors, attributed to poor lymphatic drainage in malignant tissues, enhances their passive accumulation. This underscores the potential of AuNPs as ‘magic bullets’, broadening their scope of applications [1,10,19,20,21].

2. Materials and Methods

This survey aims to bring together a research bank in a single location, with a prevailing reference to the scientific literature that emerged in the past 10 years. For this purpose, a systematic literature search was carried out in the databases of the following platforms: ScienceDirect, Google Scholar and Web of Science.
The keywords used were the following:
  • Coating of Gold Nanoparticles;
  • Encapsulating of Gold Nanoparticles;
  • Encapsulation of Gold Nanoparticles;
  • Capped of Gold Nanoparticles;
  • Cell membrane camouflaged of Gold Nanoparticles.
Our exhaustive investigation spanned from 2014 to 2023, yielding over 255,000 studies related to Au NPs. In an endeavor to enhance research quality, we conducted a subsequent search, employing article relevance as a parameter. This refined search yielded an additional 400 scientific studies.
This extensive undertaking stems from the imperative to elucidate the interplay between nanoparticles (the fundamental unit in nanotechnology) and cells (the basic unit in biology). This comprehension is crucial for advancing diagnostic and therapeutic techniques across various pathologies. A profound understanding of the interface between nanotechnology and biology is essential for achieving superior outcomes in medical applications (Scheme 1).

3. Literature Review on Gold Nanoparticles

3.1. Synthesis of Gold Nanoparticles

Two primary approaches exist for synthesizing gold nanoparticles (AuNPs), “Top-Down” and “Bottom-Up” [1,10], as illustrated in Figure 2. Similar to many other kinds of inorganic nanomaterials, top–down or bottom–up methods could be used to create AuNP. When using a top–down strategy, physical techniques like laser ablation, aerosol technology, UV and IR irradiation and ion sputtering are used to convert a bulk gold into AuNP. Contrarily, the bottom–up method of synthesizing AuNP begins at the atomic level (gold ions) and works its way up to nanoparticles of the required size and shape by using the right chemistries. The reduction of Au ions with the appropriate reducing agents in the presence of capping agents is the key step in the chemical process used to prepare spherical AuNP.
The “Top-Down” approach entails the synthesis of AuNPs from bulk materials, breaking them down into nanoparticles through various methods.
The “Bottom-Up” method, the focus of this review, initiates nanoparticle synthesis from the atomic level. It comprises two primary stages:
  • Reduction of a gold precursor, typically a salt in an aqueous solution. The choice of reducing agent, ranging from citrate to sodium borohydride, directly influences nanoparticle size [1,10];
  • Stabilization of the AuNPs, achieved through coatings forming covalent bonds or electrostatic stabilization. The selection of coating depends on the intended application and its interaction with the target [1,10,17,22].
In contrast, the “Top-Down” approach, although intuitive, may yield AuNPs with variations in size and shape, potentially being less cost-efficient. Conversely, the “Bottom-Up” approach provides precise control over AuNP properties and is more economical for large-scale production. However, its optimization can pose challenges due to synthesis variables. This review focuses on the “Bottom-Up” approach due to its relevance in biomedical applications, where nanoparticle specificity and functionalization are crucial [1,10].

3.2. Methods of Synthesis of Au NPs

Over the years, various methods have been employed, each with its unique features:
  • Turkevich Method;
  • Brust Method;
  • Seed-Mediated Growth;
  • Digestive Ripening;
  • Martin Method;
  • Radiation;
  • Biological Synthesis.
Currently, the pathways for synthesizing gold nanoparticles (AuNPs), as illustrated in Figure 2, are broadly understood, encompassing a detailed grasp of the variables influencing the outcome of the AuNPs and their corresponding mechanistic perspectives. Widely acknowledged chemical protocols for AuNP synthesis include the Frens/Turkevich method (yielding 10–100 nm hydrophilic spherical AuNPs), the Brust method (producing 1–3 nm hydrophobic spherical AuNPs), the Murphy/El-Sayed surfactant-assisted seed-mediated method (employed for gold nanorods) and the polyolgalvanic method (used in preparing gold hollow polyhedral nanoparticles). Additionally, modified methods rooted in green chemistry principles are also documented in the literature.
Collectively, AuNPs derive advantage from the existence of well-optimized, repeatable and tunable synthetic methodologies, enabling the generation of a diverse library of AuNP with varying sizes and shapes through accessible chemical processes. Notably, the current landscape facilitates the acquisition of AuNP from diverse industrial providers, offering a spectrum of options in terms of size, shape and surface chemistry [1,2,3,5,6].
  • Turkevich Method (1951)
This widely used method creates spherical AuNPs by reducing gold ions (Au3+) to gold atoms (Au0) using various reducing agents, like citrate or UV radiation. A variant involves sodium borohydride (NaBH4) for a simplified approach without heating [1,10,22].
  • Brust Method (1994)
Utilizing a two-phase reaction with organic solvents, this method yields AuNPs ranging from 1.5 to 5.2 nm. It employs sodium borohydride and alkanethiol, with Tetraoctylammonium bromide facilitating the phase transfer [1,10,22].
  • Seed-Mediated Growth
Predominantly used for creating nanometric gold rods, this method leverages pre-synthesized particles, which subsequently grow with weak reducing agents [1,10].
  • Digestive Ripening
Optimal for producing monodisperse AuNPs, this method relies on temperature modulation, heating colloidal suspensions up to 138 °C and then cooling them [1,10].
  • Martin Method
Emphasizing stoichiometry control, this approach adjusts the ratio of reactants for optimal nanoparticle synthesis. This method allows AuNPs to combine with hydrophilic molecules for various applications [23,24].
  • Radiation
Employing radiation influences the synthesis of AuNPs. The radiation dose rate determines the size of the nanoparticles, with higher rates leading to smaller sizes [5].
  • Biological Synthesis
An eco-friendly approach uses living organisms, from bacteria to plants and fungi, for AuNP synthesis. This method stands out for its environmental benefits and cost-effectiveness [1,10,22].
  • Other Methods
Various other techniques, like the use of bacteria, fungi, plants, algae and biomolecules, also contribute to the broad landscape of gold nanoparticle synthesis. Each method, whether using bacterial cell walls, fungal secretions, plant components or algae biomass, offers unique pathways for producing AuNPs with distinct properties [1,10,22].

4. Coating and Functioning

Due to the often overlapping and sometimes ambiguous terminology associated with nanoparticle studies, it is necessary to clarify two central terms: ‘coating’ and ‘functionalization’. This section aims to dispel any doubts regarding these terms, ensuring a clear and distinct understanding of each within the context of this work. Gold nanoparticles are extensively researched for their unique properties and potential applications in therapy, diagnostics and technology. Modifying these intrinsic properties can be achieved through two main processes, coating and functionalization, each with its nuances and objectives.
Coating involves encapsulating or covering the nanoparticle’s surface with a specific material. The primary goal is to provide stability, protect against degradation and optimize dispersion in specific media. Materials such as polymers (e.g., PEG), surfactants and organic entities serve as protective barriers, promoting a more harmonious integration of nanoparticles with their introduced environment.
In contrast, functionalization refers to the intentional modification of the nanoparticle’s surface to confer a desired functionality. This process is typically achieved through the covalent bonding of specific molecules or functional groups to the nanoparticle’s surface. Such modifications enable nanoparticles to respond to specific stimuli, bind to defined targets (such as cells or proteins) or serve as carriers for therapeutic agents. An illustrative example is the attachment of antibodies to nanoparticles, directing them selectively to tumor cells.
In practice, many applications initially require a coating to ensure the stability and biocompatibility of nanoparticles. Subsequently, additional functionalization is carried out to instill specific functionalities. Together, these techniques enable the customization of gold nanoparticles, optimizing their therapeutic efficacy and diagnostic capability. This study will delve into both processes in detail, shedding light on their intricacies and implications [25].

4.1. Applications and Functionalization of Coated AuNPs

The use of coated gold nanoparticles (AuNPs) has rapidly expanded in recent years, primarily due to their ability to bind to a variety of molecules, offering diverse applications in biomedicine. Furthermore, functionalization plays a pivotal role in nanoparticle design. By attaching a specific target molecule to the surface of the AuNPs, it is possible to direct the nanoparticle to a specific organ or tumor tissue. This approach has the potential to enhance treatment efficacy by reducing side effects, as the drug is delivered directly to the target site.

4.1.1. Drug Delivery

Chemotherapeutic agents, as drugs, inhibit cell replication and induce apoptosis, thereby limiting cellular function. This elimination capacity extends to healthy cells, restricting the use of certain drugs due to their impact on healthy tissues. Consequently, extensive research focuses on developing selective drugs to minimize collateral damage [22,26,27].
The expanding body of research has enhanced our comprehension of various physicochemical properties, including size, surface charge and their modifications, influencing the cellular absorption and fate of Au NPs. Notably, larger nanoparticles tend to accumulate near the vasculature, while smaller ones rapidly diffuse throughout the tumor matrix. The optimal size range for Au NPs in tumor treatment falls between 25 and 50 nm, depending on the tumor type. Regarding charge, it plays a crucial role; Au NPs with a positive charge exhibit 5 to 10 times greater absorption than their negative or neutral counterparts. This is attributed to their high affinity with negatively charged cell surfaces, facilitating enhanced adhesion to the cell membrane through the generation of transient holes [20,28].
As drug carriers, as depicted in Figure 3, gold nanoparticles (Au NPs) transport therapeutic agents through either covalent bonding or non-covalent attachment. Covalent bonding requires the chemical modification of drugs and external triggers for drug release, while non-covalent adsorption onto Au NPs provides an alternative method for efficient drug transport and release [28].
Due to their potential applications as delivery vehicles, diagnostic tools and therapeutic agents, gold nanoparticles are among the most extensively studied metallic nanomaterials. The extensive surface area of GNPs makes them well suited for functionalization with biomolecules via physical absorption or with reactive groups via ionic or covalent bonding for ligand or antibody modification. In general, GNPs are biocompatible and have low toxicity, and they have good biocompatibility [29].
Considering the goals of cancer therapy, which include achieving greater absorption, drug permeability, site specificity and control over the drug release rate, the role of Au NPs as drug chaperones is pivotal. Numerous studies suggest that, depending on the objective, Au NPs can incorporate specific functional groups to aid in orientation, exhibit fluorescence or possess imaging capabilities to trace their path [2,10,30].
The Retention and Permeability Effect (RPE) refers to the improper formation of vasculature, resulting in reduced pore size. This facilitates passive directional transport to tumor vasculature, enabling Au NPs to bypass natural body barriers. Consequently, Au NPs evade degradation or drug metabolism, allowing accumulation in tumor regions. The limited lymphatic drainage in these areas further prevents nanoparticle recirculation.
Numerous advantages of nanoparticles in transporting chemotherapeutic drugs have been elucidated, including a significant increase in drug circulation, inhibiting rapid elimination by the liver and kidneys [2,10,30].
Another strategy for Au NPs in drug delivery involves leveraging active targeting mechanisms. This entails binding specific ligands, such as peptides, aptamers or antibodies, to the Au NPs’ surface. The inclusion of these ligands ensures preferential binding to target cell receptors, serving as recognition moieties. This highly specific targeting minimizes effects on healthy tissue, thereby enhancing treatment efficacy [27,30,31].

4.1.2. Au NPs in Immunotherapy

Oncological immunotherapy relies on harnessing the patient’s immune system to combat neoplasms [32], recognized as a highly effective approach in cancer treatment. Interest is growing in delivering immunotherapeutic agents through nanoparticles, with Au NPs emerging as potent vectors for this purpose [32]. Due to their advanced surface chemistry, Au NPs can carry various antigens and adjuvants, enhancing the immune response against tumor cells and inducing cytotoxic T lymphocytes. These nanoparticles can mimic antigen-presenting cells by being functionalized with co-stimulatory molecules and proteins containing antigenic peptides, enabling interaction with dendritic cells and enhancing cytokine production, positioning them as promising adjuvants [32,33,34].
In oncological vaccinology, the current trend focuses on activated dendritic cells [35], where Au NPs serve as effective tools for delivering therapeutic vaccines [36,37]. Studies indicate that Au NPs coated with tumor antigens promote dendritic cell maturation and subsequent lymphocyte proliferation [38]. The induced modifications in dendritic cells, including phagocytosis, activation, migration and T-cell co-stimulation, are fundamental for the success of vaccines based on these cells [32,33,34,35,36,37,39].
Experimental data highlight the relevance of Au NPs in immunological interactions, showcasing their antigenic, adjuvant and pro-inflammatory properties. Thus, the future of combined oncological therapy, integrating phototherapy, chemotherapy and immunotherapy, may be shaped by the strategic development of multifunctional nanoparticles, with a particular emphasis on those based on Au NPs [32].

4.1.3. Au NPs as Radiosensitizers

Gold nanoparticles (AuNPs), with their elevated atomic number, present an enlarged X-ray absorption cross-section, rendering them optimal candidates as radiosensitizers to enhance radiotherapy efficacy. Additionally, the photothermal conversion capability of AuNPs can generate localized heat within the tumor region [2,10,20].
The versatile biomedical applications of AuNPs encompass the detection of cancer-associated biomarkers. Yang et al. devised a sandwich-structured electrochemiluminescence (ECL) sensor for detecting phosphatidylserine-positive exosomes, crucial in an early ovarian cancer (OC) diagnosis. This ECL sensor featured glassy carbon electrodes coated with gold “nanoflowers” (AuNFls) and a signal nanoprobe comprising graphitic carbon nitride sheets loaded with luminol-coated gold nanoparticles (Lum-AuNPs@g-C3N4). The high specific surface area of AuNFls and g-C3N4 facilitated the initial modification of abundant specific binding peptides (FNFRLKAGAKIRFRGC) of phosphatidylserine onto these materials [40].
Another application involves a rapid colorimetric method using gold nano-probes designed for detecting OC-associated miRNAs. In this method, miRNAs formed pairs with complementary AuNPs-miRNA probes, resulting in AuNP aggregation and a shift in the absorption spectrum.
Moreover, fluorescence resonance energy transfer (FRET) probes, utilizing carbon quantum dots as energy donors and AuNPs as energy acceptors, were developed for highly sensitive CA125 detection, a clinical marker for OC [40].
The localized intracellular presence of AuNPs heightens the likelihood of ionization events, increasing the accumulation of local energy and causing more harm to tumor cells. The distinction in mass energy absorption coefficients between gold and soft tissues, enabling dose escalation, underlies the physical mechanism of AuNP radiosensitization within the initial nanoseconds of exposure. Electrons emitted from AuNPs exhibit a limited range of a few micrometers, resulting in very localized ionizing events. Therefore, precise delivery to and absorption by tumor cells during radiotherapy are imperative to realize any benefits [2].
The chemical mechanism of AuNP radiosensitization involves radiochemical sensitization of DNA, mediating an increase in catalytic surface activity and augmenting free radical generation on AuNPs’ surface. Contrary to previous belief, the electronically active surface of AuNPs has been demonstrated to catalyze chemical reactions, primarily through interaction with molecular oxygen, producing free radicals. These catalytic effects, in combination with radiation, appear to enhance [2].
Radiobiological mechanisms contributing to cellular response to AuNP irradiation include the generation of reactive oxygen species (ROS), oxidative stress, induction of DNA damage, potential bystander effects and impacts on the cell cycle. Oxidative stress can result in the oxidation of lipids, proteins and DNA, leading to necrotic, apoptotic or other forms of cell death. Elevated ROS levels also affect mitochondrial activity, depleting cellular energy and causing necrotic death [2].

5. Stabilization of AuNPs

5.1. Electrostatic Stabilization

van der Waals forces, leading to system instability and precipitation, induce the aggregation of nanoparticles in colloidal systems. Numerous techniques have been employed since the inception of nanomaterial research to stabilize nanoparticles and ensure electrostatic stability. As per the Derjaguin–Landau–Verwey–Overbeek hypothesis, the total potential energy governing the interaction between two colloidal particles comprises both attractive (van der Waals) and repulsive forces arising from the electrical double layer of charges [1,41,42,43].
A particle is deemed stable if its total potential energy surpasses the kinetic energy. However, defining nanoparticle stability solely in terms of electrostatic stabilization is insufficient; surface energy must also be considered. Metallic surfaces, in conjunction with attractive dipole–dipole interactions, typically exhibit surface energies ranging from 1000 to 2000 mJm−2, markedly higher than those observed in other organic and inorganic materials, contributing to the instability of these nanomaterials [5,10].
A charged layer can form on the colloidal nanoparticle surface when ionic groups from the liquid dispersion medium adhere to it. Consequently, the colloidal nanoparticles are enveloped by an equal number of ions with opposing charges, forming electroneutral double layers. Figure 4 illustrates this stabilization through an electrical double layer generated by conflicting pressures, inhibiting nanoparticle aggregation in the solution phase through electrostatic repulsions. Electrostatic stabilization can be controlled by adjusting key factors, including pH, concentration and temperature [1].
An alternative method of stabilization involves combining steric and electrostatic approaches to maintain the stability of metallic nanoparticles in the solution phase. Polyelectrolytes, when used as polymeric surfactants, integrate steric and electrostatic stability effects into a single molecule. An ionic surfactant with extended end chains and a polar head group creates a two-fold electric layer around the nanoparticle, providing steric repulsion within the particles. This configuration prevents agglomeration, resulting in a mutual stabilization system. Figure 4 elucidates how steric and electrostatic interactions combine to stabilize AuNPs [1].

5.2. Steric Stabilization

Steric stabilization (Figure 5B) is employed to uphold the stability of nanoparticles through the attraction or repulsion of adsorbed ions or molecules amidst adjacent particles. Generally, enhanced steric stabilization is achieved with larger adsorbed molecules. A variety of functional groups, such as hydroxyls, surfactants, oligomers and polymers, are utilized to accomplish this type of stabilization. The spatial conformation of molecules plays a crucial role, and it has been demonstrated that elongated or conical conformations contribute to greater stability. Notably, if the size of the nanoparticle is smaller than that of the stabilizer, typically observed with long polymer chains, an encapsulation process can occur, resulting in the passivation of the particle [1,45].

6. Functionalization of Au NPs

When selecting Au NPs for biological applications, careful consideration must be given to the interaction medium. The complexity of a biological medium, as evidenced by in vitro tests, is heightened when introduced into a living organism.
Within the body, prior to reaching the target, Au NPs undergo compositional changes. When injected into the bloodstream, they interact with serum proteins, forming a coating known as the corona protein. At this stage, the corona dictates the biological functions of Au NPs, including internalization, biodistribution, elimination and potential toxicity. Various pathways, such as endocytosis mediated by specific or nonspecific receptors or the enhanced effect of permeability and retention, enable Au NPs to enter cells [10,18].
Functionalizing Au NPs for drug delivery or diverse applications often involves non-covalent interactions for drug loading. Alterations in native physical forces are sufficient for release, without necessitating specific bond cleavage. Hydrophobic drug release can be achieved by inducing changes in local hydrophobicity. Covalent ligation with the drug, forming cleavable bonds through internal or external incentives, presents another alternative.
To enhance binding to biological molecules and optimize Au NPs as drug carriers with increased specificity, certain functionalization techniques employ synthetic pathways containing functional residues. Utilizing one or more functional groups, such as oligo or polyethylene glycol (PEG), bovine serum albumin (BSA), amino acids, polypeptides, oligonucleotides, antibodies, receptors and similar particles, is common. A widely adopted method for attaching fractions, such as chemotherapeutic drugs, involves employing biomolecules with thiol groups [2,45].
Research on self-assembled monolayers (SAMs) has deepened the understanding of a widely used coating agent for gold nanoparticles. Chemically, sulfur is softer and less electronegative than oxygen, exhibiting a greater number of oxidation states and a stronger affinity for metals on the right in the d block. Gold, in particular, displays a significant affinity for sulfur [45,46].
Studies on the crystal structure of thiolate-coated gold clusters have uncovered stable SR(-Au-SR) motifs, seemingly crucial for the stability of Au-S bonds in thiol-stabilized Au NPs. In these systems, gold atoms are intercalated between sulfur atoms of thiolates, presenting several stable motifs, as illustrated in Figure 5, where it demonstrates that sulfur atoms in stable structures act as stereogenic centers, attached to four different substituents: two gold atoms, the organic R part of the thiolate, and a lone pair of electrons. Initially, the organic part R of the thiolate can assume two positions, with easy interconversion due to a low barrier [45,46].
With an increasing number of studies, additional stable motifs in gold clusters protected by thiolates have been discovered. Structural determinations reveal trimeric SR(-Au-SR) motifs, thiolate bridges, cyclic structures or even bare sulfur atoms. Structural studies indicate that the Au-S interface is dynamic, involving processes like thiolate migration, thiolate desorption and re-adsorption, contributing to flexibility [45,46].
Two primary techniques exist for functionalizing Au NPs with specific molecules. An efficient approach entails synthesizing Au NPs directly with the molecule of interest linked to a thiol group, as previously discussed. This method offers the mentioned advantages. Alternatively, a second approach involves post functionalization of Au NPs. Click chemistry and the succinimidyl ester reaction have both been employed to covalently bind molecules of interest to the ligand stabilizing Au NPs. Alternatively, ligand exchange may be utilized if the molecule of interest possesses a thiol group [47].

6.1. Protein Corona Formation

Nonspecific targeting and immune system activation are notable challenges in Au NP drug delivery. Coating affected cells with PEG, as proposed in some studies, resolves these issues by concealing the cell surface, inhibiting surface protein attachment, and diminishing the likelihood of immune system activation. However, a flawlessly coated surface hinders nanoparticle adhesion to specific receptors, rendering them “invisible” to the immune system. To address this, gold nanoconjugates undergo further modification with specific ligands to navigate in vivo challenges, despite potential side effects. Incorporating functional moieties on the particle surface facilitates the attraction of specific “stealth” proteins, forming a protein corona that shields the nanoparticles from phagocytic cells [48].
Understanding the intricacies of the immune system is crucial to overcoming challenges like those mentioned above. While certain Au NPs demonstrate promising in vitro results, the biological complexity of in vivo systems necessitates a comprehensive understanding and anticipation of the formation of the so-called crown protein for the successful application of Au NPs in biological contexts.
In the body, proteins adsorb onto the surface of Au NPs, leading to the formation of a dynamic protein crown, a process vividly illustrated in Figure 6. This protein coating is subject to ongoing competitive interactions until an equilibrium state is reached. Initially, a rapid formation of proteins creates the ‘soft crown’. Over time, proteins with a higher affinity for the surface gradually replace the initial layer, culminating in the formation of a more stable ‘hard crown’. As a result, both the composition and the thickness of this protein crown are subject to dynamic changes over time.
The composition of the protein–nanoparticle complex undergoes continuous changes during the nanoparticle’s interaction with the body. Initially, it is highly probable that proteins characterized by elevated plasma concentrations and robust association rates will occupy the nanoparticle’s surface. Over time, these proteins may disassociate, making room for others with lower concentrations, slower exchange rates or stronger affinities. This phenomenon, encompassing the competitive adherence of proteins to a finite surface based on abundance, affinities and incubation time, is commonly referred to as the “Vroman Effect” [52,53]. This effect holds considerable significance concerning the distribution of particles within the body. The evolution of the protein corona, occurring as the nanoparticle traverses different body compartments during its circulation, may be influenced by fluctuations in protein levels and binding affinities [54,55].
This holds particular relevance for nanoparticles due to the heightened importance of surface effects at this scale. Numerous studies have demonstrated that biological responses to nanoparticles exhibit a stronger correlation with surface area than with mass. Nanoscale materials possess significantly higher surface-to-volume ratios compared to larger particles, as surface areas decrease more gradually than volumes with diminishing size. Consequently, a nanoparticle with a higher surface-to-volume ratio is expected to bind a greater quantity of proteins in comparison to its mass, as supported by existing research [54,55].
The impact of varying protein binding on the biological response to a specific nanoparticle remains uncertain, as does the process of protein binding (e.g., uptake by phagocytic cells of the reticuloendothelial system and clearance). However, it is evident that the type and quantity of proteins present on a particle’s surface influence the biodistribution of nanoparticles. A comprehensive understanding of the kinetics, affinities and stoichiometries of protein attachment and dissociation from the nanoparticle, as well as the specific proteins associated with the particle, is crucial for a thorough comprehension of the protein corona [56].
The predominant focus of research investigating the impact of protein binding on absorption has involved preincubating particles with bulk serum/plasma, preincubating particles with specific proteins or attaching individual proteins to the particle’s surface, followed by the analysis of uptake by macrophages. This research consistently reveals a direct correlation between surface charge and protein binding, with neutrally charged particles opsonizing at a substantially slower pace than charged particles [9,57].

6.2. PEGylation of Au NPs

Poly (ethylene glycol) (PEG) contributes to a widely accepted and clinically approved coating strategy, renowned for its efficacy in prolonging blood circulation half-life to optimize pharmacokinetics. This stealth capability arises from the rapid chain motion and large excluded volume of PEG, inducing steric repulsion upon binding to foreign substances. PEG’s low interfacial free energy in water facilitates the easy synthesis and stabilization of PEG-coated nanoparticles (NPs) in aqueous media. Previous studies have underscored the significance of PEG coating density and chain length as key determinants influencing the stealth quality of PEG coatings [58].
Despite being generally considered non-toxic, PEG functions as an anchored hydrophilic polymer when used to functionalize Au NPs. Thiol-terminated PEG, commonly possessing biotin, nitriloacetic acid, -COOH, OH or -OCH3 end groups, allows the conjugation of various biomolecules, such as proteins [59,60].
While PEG imparts greater stability to Au NPs in vivo and mitigates undesirable immune responses, its usage diminishes cellular uptake compared to citrate stabilized Au NPs. This drawback can be overcome by co-functionalizing the Au NP surface with moieties like receptor-mediated endocytosis (RME) peptides, enhancing cellular uptake while preserving stability and immune response characteristics achieved through PEG modification [61,62].
The widespread application of PEG in the functionalization of Au NPs, primarily due to its easy adaptability with various molecules, is clearly demonstrated in Figure 7. Recognized for its clinical approval for intravenous use, the amphiphilic nature of PEG contributes to the stabilization of nanoparticles in biological environments. It is important to note that Au NPs are often not exclusively bound to PEG. As shown in Figure 7B, they can be linked to PEG either before or after binding to other fragments such as peptides, oligonucleotides or different molecules tailored to specific applications [1,61].
Consequently, PEG-Au NPs find utility not only in cellular functionalization but also in intracellular internalization. Studies indicate that PEG inhibits agglomeration and cytotoxicity of Au NPs in high-ion-concentration environments, supporting prolonged circulation and increased utility of Au NPs. The hydrophilic corona formed by PEG on the Au NP surface significantly reduces nonspecific protein adsorption, evading opsonization and complement activation in the blood. Notably, PEG coatings may, however, negatively impact drug delivery efficacy, potentially reducing cellular uptake and endosomal release of therapeutics loaded in the NPs, and diminishing NP tumor targeting efficacy [1,63].
To achieve complete surface saturation with peptides or other biologically active groups, a hetero-bifunctional PEG linker is proposed, potentially enhancing direct cell/peptide contact with the target cell. However, concerns have been raised regarding the stability compromise and anti-PEG effects associated with PEG linkers [64].
The degree of PEGylation alters basic physiochemical properties of Au NPs, influencing uptake by immune cells. Particle size and PEG density-dependent adsorption of serum proteins by PEGylated Au NPs impact the efficiency of uptake by macrophages [60].
Data on critical coagulation concentrations (CCCs) indicate that PEG length, nanoparticle diameter and the PEG molecule/nanoparticle ratio influence nanoparticle stability in high-ionic-strength media. Longer PEGs, smaller nanoparticles and higher PEG/nanoparticle ratios favor more stable sols, aligning with DLVO theory and Flory–Krigbaum theory predictions [65].
Considering the crucial role of diameter in biomolecule–nanoparticle conjugates, especially for cell nucleus access, PEGs of different sizes (900, 1500 and 5000) increase the hydrodynamic radius of 20 nm gold nanoparticles by 3.5, 5 and 9 nm, respectively. PEG 5000, while enhancing stability, may compromise nuclear translocation effectiveness due to its additional bulk [65].
Studies have demonstrated that PEGylated nanoparticles resist removal by the reticuloendothelial system, with extracted nanoparticles from the systemic circulation accumulating in the liver after payload release [66].
PEGylation encompasses a broad range of sizes and classes within the PEG molecule. Typically, another molecule is covalently linked, with its purpose depending on the desired application. As presented in Table 1, diverse molecular systems supported on PEG have been reported for functionalizing Au NPs.

6.3. Antibody and Protein Functionalized Au NPs

The active targeting of nanoparticles to known oncogenes prompted the immediate consideration of antibodies as the optimal choice for this purpose. Antibodies exhibit a robust affinity for specific receptors, rendering them highly selective. Examples of gold nanoparticles (Au NPs) frequently employed for enhanced radiotherapy in targeting malignancies include cetuximab and trastuzumab. Receptor-mediated endocytosis facilitates the uptake of antibodies into cells; however, the conjugation of antibodies is known to alter their pharmacokinetics, leading to distinct subcellular localizations between the antibody and the Au NPs. This process provides a target-specific internalization mechanism for antibodies-Au NPs. Post-conjugation, alterations in the antibody’s kinetics and absorption are anticipated. While documented examples suggest favorable changes for improved absorption, further research is warranted in this area [17,91].
To cross-link Au NPs with carboxylic acids on antibody surfaces, a hydrazine-terminated PEG linker can be employed (Figure 8B). However, the literature predominantly utilizes some variant of amide bond formation involving lysines on antibodies, given the higher abundance of carboxylic acids than amines. This chemistry tends to be relatively random, generating more variants [17].
Random amide bond formation may result in bonds forming at or near the antibody’s active site or orienting the active site toward the gold nucleus, diminishing conjugate attachment. To optimize activity, the orientation of antibodies on an Au NP surface can be adjusted using protein G [17].
While antibodies exhibit indisputable affinity for receptors, their size may impede tumor penetration. An alternative approach involves using antibody fragments. Functional antibody fragments are produced by reducing disulfide links between an antibody’s heavy chains, exposing free thiols that can bind to the gold nucleus. This method positions the antibody’s active site away from the nucleus, enhancing accessibility [17,92].
The physical interaction between antibodies and gold nanoparticles is determined based on three phenomena: (a) ionic attraction between positively charged antibodies and negatively charged gold; (b) hydrophobic attraction between antibodies and the gold surface; and (c) dative binding between gold-conducting electrons and amino acid sulfur atoms of antibodies. Various methods facilitate chemical interactions, including (i) chemisorption using thiol derivatives, (ii) the use of bifunctional linkers and (iii) the use of adapter molecules such as streptavidin and biotin [93].
While spontaneous protein-Au NP adsorption is recognized, limited information is available on the structural and molecular details of this interaction (Figure 8A). Recent studies indicate reduced activity of enzymes like lysozyme and chymotrypsin upon adsorption to Au NPs, with a similar effect observed for fibrinogen. Pepsin remains active when attached to Au NPs, and adsorption enhances the stability of bovine catalase at high temperatures. CD tests reveal that although enzyme activity may diminish on the nanoparticle surface, proteins seem to retain their secondary structure. Therefore, the effect of enzyme activity retention after adsorption appears to be protein-specific [94,95].
In comparison to other targeted therapies, the utilization of native proteins as targeted agents remains largely unexplored. Natural receptor ligands or lectins, proteins binding to carbohydrates often extracted from fruits and vegetables, serve as targeting moieties. However, the abundance of competing ligands in the human body lacking Au NPs may reduce selection effectiveness. This underscores the need for further exploration in this area [17,96,97,98].
Figure 8. Antibody and protein functionalized Au NPs. (A) Coating of gold nanoparticles with proteins by means of electrostatic interactions. Protein binding association constants on AuNPs increase progressively with AuNP diameter between 5 and 60 nm. The binding association constant, on the other hand, was discovered to be slowly varying for some AuNPs with diameters greater than about 80 nm [99]. (B) Functionalization of antibodies by means of a PEG bridge. Every year, more antibodies are used to functionalize Au NPs. Most of these systems are stabilized with thiolated linkers like PEG chains, which are typically terminated in carboxylic acids or succinimidyl esters that are then activated for the addition of antibodies by forming random amide bonds with free amine residues on the surface of the antibodies.
Figure 8. Antibody and protein functionalized Au NPs. (A) Coating of gold nanoparticles with proteins by means of electrostatic interactions. Protein binding association constants on AuNPs increase progressively with AuNP diameter between 5 and 60 nm. The binding association constant, on the other hand, was discovered to be slowly varying for some AuNPs with diameters greater than about 80 nm [99]. (B) Functionalization of antibodies by means of a PEG bridge. Every year, more antibodies are used to functionalize Au NPs. Most of these systems are stabilized with thiolated linkers like PEG chains, which are typically terminated in carboxylic acids or succinimidyl esters that are then activated for the addition of antibodies by forming random amide bonds with free amine residues on the surface of the antibodies.
Pharmaceutics 16 00255 g008
The modification of Au NPs or the promotion of self-assembly can be achieved using thiols and protein scaffolds. An alternative approach involves the direct adsorption of proteins onto the Au NP surface, exemplified by the “crown protein” (Au NP CP), which garners considerable attention. This entity, governed by biological dynamics, binding affinity and protein adsorption rate, crucially influences the exchange time and lifetime of interfacial interactions. Investigations reveal that the corona protein presence enhances surface free energy, and proteins can be conjugated with Au NPs through glutamic acid. This involves bonding with amino groups and carboxyl groups extending outward, forming connections with protein amino groups [1,98].
Interaction between Au NPs and proteins (Figure 8A) can induce intrinsic characteristic changes in both. Notably, physiological modifications such as alterations in bound protein structure, complement activation, blood clotting and protein aggregation may arise [100,101,102,103]. The hydrophilicity or hydrophobicity of Au NPs plays a regulatory role in the amount and composition of protein adsorption. For instance, hydrophilic polyethylene glycol (PEG) modification results in strong resistance to plasma proteins, such as complement subunit adsorption, enabling extended circulation time due to resistance to the reticuloendothelial system (RES) [104].
Colloidal gold nanoparticles capped with citrate attract immunoglobulins and other proteins through non-covalent interactions. These interactions involve hydrophobic attractions to the metal surface, electrostatic interactions between positively charged amino acids, the protein’s N-terminus and the negatively charged surface of citrate-capped particles [93,105]. Gold–sulfhydryl group bonds, particularly with cysteine side chains of proteins, contribute to tight, practically irreversible protein binding on the AuNP surface, especially at a pH just above their pI values [95].
Tighter protein binding may impact function and structure. Immunoglobulins, despite their large size (150 kDa), maintain antigen-binding activity when combined with AuNPs. However, smaller proteins may face inactivation risks, although some enzymes may retain functionality. Conjugation may occasionally enhance enzyme stability and activity, while protein immobilization on NP surfaces can increase tolerance to thermal and pH gradients and reduce susceptibility to denaturation and degradation [95].
To enhance Au NP dispersibility, surface treatment with electrolytes like citrate, CTAB, PSS, PDDAC, etc., is common. This alteration imparts net charges on Au NPs, leading to electrostatic attraction to functional groups in proteins with opposing charges. Fine-tuning the conjugation pH to approximately 0.5 units above the protein’s isoelectric point is essential to prevent protein aggregation due to electrostatic attraction. This adjustment maintains hydrophobic interactions, facilitating protein–gold conjugation. System optimization for specific antibodies to each Au NP preparation involves determining the optimal pH value for conjugation [105].
While recombinant proteins have been studied, many investigations employ native receptor proteins, benefiting from the advantageous properties of recombinant proteins derived from mutations and variants of wild-type proteins. Table 2 and Table 3 present antibodies and proteins used as coatings on Au NPs. Copper-pro- moted alkyne-azide cycloaddition reaction CuAAC can efficiently react the azo group on gold nanoparticles with the alkyne group on the protein surface, and a protein labeled with an alkyne-containing photoaffinity probe in solution can also be click-captured, enriched on a clickable reagent, and easily released by thiol exchange reaction [97].

6.4. Peptides and Amino Acid Functionalized Au NPs

A methodology to enhance biocompatibility involves surface engineering or custom coating of metal nanoparticles with peptides or amino acids, as depicted in Figure 9 [138].
Peptides, shorter polymers of amino acids, are chemically synthesizable and fully characterizable to specifications. Despite a lower affinity toward receptors compared to proteins, peptides have garnered significant attention due to their simplicity and rapid absorption kinetics. Their excellent biocompatibility, non-toxicity and environmental friendliness make peptides with specific sequences ideal conjugates for Au NPs. Figure 9 showcases different types of amino acid interactions with bacteria (Figure 9A) and the ease of functionalizing nanoparticles with peptides for gene delivery, including several types of interactions between peptide fragments (Figure 9B). The versatility of peptide-based Au NP systems, coupled with gentler manufacturing conditions, unique self-assembly methods and specific substrate-binding properties, enables their utilization in biosensors, drug delivery and cellular uptake [1,96].
Peptides, spanning 0.4 to 1 nm and comprising 2 to 50 amino acids, facilitate cell-specific targeting when bound to target molecules on specifically designed carriers, leveraging the nanoparticles’ increased reactive surface area. Their notable attributes include high stability (long shelf life), substantial carrier capacity (incorporating multiple drug molecules in the particle matrix), the ability to carry both hydrophilic and lipophilic molecules and facilitating hassle-free drug administration through various routes [139,140].
Amine groups in amino acids and peptides, negatively charged, bind to negatively charged Au NPs, stabilizing them as negatively charged carboxylic groups expand outward. Some amino acids, such as lysine or glycine, effectively conjugate to DNA without inducing cytotoxicity. Modifying the peptide sequence allows for influencing the assembly and disassembly of Au NPs [1]. Amino acid conjugated nanoparticles show different interactions with bacteria (Figure 9A). The amino acid conjugated to the nanoparticle can interact through hydrogen bonding between the (N) of amine in amino acid and (O) of the cell surface carboxyl group. Hydrophobic interaction occurs between carbon chains of amino acid and a methyl group on the bacterial cell wall. These interactions can lead to free radical generation, which can cause membrane perturbations (pores in the membrane). Due to perturbations, there is oozing out of the bacterial cytoplasm, leading the cell to death due to loss of proteins, nucleic acids and cell structural rigidity [141,142].
Figure 9. Peptides and amino acid functionalized Au NPs. (A) Different types of amino acid interactions with bacteria. (B) Nanoparticles allow easy functionalization of peptides for gene delivery, with several types of interactions between the peptide fragments being possible; stability increases [141,142].
Figure 9. Peptides and amino acid functionalized Au NPs. (A) Different types of amino acid interactions with bacteria. (B) Nanoparticles allow easy functionalization of peptides for gene delivery, with several types of interactions between the peptide fragments being possible; stability increases [141,142].
Pharmaceutics 16 00255 g009
Organic inhibitors, featuring hetero atoms, aromatic rings and conjugated systems, stand out as highly effective components in industrial applications [14,143,144]. These inhibitors serve as additives for the modification of sol–gel coatings. Notably, amino acids, characterized by their environmentally friendly nature, affordability and purity exceeding 99%, represent a compelling inhibitor class. The corrosion-inhibiting ability of amino acids stems from their propensity to establish hydrogen bonds with oxide or hydroxide groups on metal surfaces [14,143,144].
Amino acids exhibit intrinsic biological properties, such as antibacterial effects and enhanced viability for eukaryotic cells. The particle size, nanoparticle core and specific amino acid employed in surface functionalization influence biodistribution and host immunity [142]. Single amino acids provide both amino and carboxy-terminal functionalities for biomolecule conjugation. Biocompatible amino-acid-based coatings address challenges related to size, biodistribution, immune cell interaction and inflammation induction, and offer heightened biocompatibility [142].
The implementation of peptide coatings has enabled precise control over the aggregation of Au NPs. The thiol–gold interaction facilitates the anchoring of positively charged peptides, containing cysteine residues, neutralizing negative charges that might otherwise induce aggregation. Additionally, peptides disperse negatively charged Au NPs [96,97]. This innovative approach yields more stable Au NPs, simplifying the conventional citrate-mediated synthesis. The synthesis involves mixing dilute acid and an aqueous solution of multidomain peptides (MDPs) at room temperature, with 3,4-dihydroxy-L-phenylalanine (Dopa) residues ensuring excellent biological reducibility. The method offers advantages such as low toxicity, short reaction time, high engineering efficiency and precise size control [96,97].
Peptides, being smaller than proteins or antibodies, excel as targeting agents. Their relative stability on antibodies and proteins allows for linkage after binding to Au NPs or before ligand exchange [17]. Peptide coatings enhance the physiological stability of nanoparticles, enabling programming of surface chemistry and immunomodulatory activity [145]. Cell penetration peptides (CPPs) are commonly utilized for Au NP conjugation. CPPs, known for membrane translocation capability, facilitate the delivery of therapeutic molecules, proteins, liposomes, nanoparticles, antisense molecules, siRNA and nucleic acids. Categorization of CPPs includes cationic, amphipathic and hydrophobic peptides, with cationic CPPs, rich in arginine and lysine, being extensively employed for various cellular payloads [146,147].
The physical–chemical properties of CPPs allow classification into three groups: cationic, amphipathic and hydrophobic peptides. The majority of known CPPs fall under the cationic category, primarily rich in arginine and lysine. These peptides have been widely utilized for delivering diverse payloads into cells, including proteins, nucleic acids, small molecule medicines and NPs. Classification systems for CPPs are often based on physico-chemical characteristics, such as amphipathicity or hydrophobicity [148,149].
Amino acids, featuring amine and carboxylic groups, can function as both reducing and capping agents in the creation of Au NPs. All amino acids, except cysteine, have demonstrated the capability to reduce HAuCl4 to gold NPs. Studies indicate that gold NPs with amphiphilic amino acid surfaces exhibit superior biocompatibility [150]. Table 4 shows a series of peptides and amino acids reported as coating Au NPs.

6.5. Microorganism, DNA, RNA and Aptameros Functionalized Au NPs

DNA serves as a functionalization tool for Au NPs and is increasingly embraced in the scientific community due to its precisely defined structures and functions, coupled with its programmable assembly process based on DNA sequence, length and structure [1,17]. Since the advent of chemical DNA synthesis, DNA oligonucleotides have evolved into indispensable biopolymers in materials science. Nearly any position in DNA is modifiable during synthesis, utilizing various functional groups. The persistence length of a duplex DNA, around 50 nm or 150 base pairs, is easily achievable through synthesis. By extending the chain in increments of 0.34 nm with the addition of a base pair, DNA manipulation with sub-nanometer precision is feasible. The high programmability of DNA has led to the creation of numerous remarkable nanostructures [164].
In the realm of AuNPs, conjugation predominantly employs terminal thiol-modified DNA. This modification imparts strong electrostatic and steric stabilization from the phosphate backbone, rendering the DNA highly negatively charged (Figure 10B,C). AuNPs adorned with a dense DNA layer exhibit excellent colloidal stability. Thiolated DNA, owing to its substantial Au-S affinity (exceeding 200 kJ/mol), has long been employed for Au surface functionalization. However, a noteworthy observation is the DNA desorption noticed after prolonged storage. Intriguingly, research into the nature of the Au-S bond persists [165,166].
Controlling the separation distance between nanoparticle assemblies during synthesis allows for the creation of nanoparticle crystals, gummers or large oligomerizations. One approach involves coating the surface of Au NPs with thiol-capped single-stranded DNA molecules [1,17,96]. DNA hydrolyzing enzymes enhance the stability of DNA-Au NP complexes, with current research exploring factors influencing stabilization and techniques for achieving it [167]. Sequence specificity of DNA on Au NPs, along with the immobilization of DNA on gold improving thermal stability, has been reported [167].
Cationic-glycopolymer-stabilized Au NPs, when complexed with DNA, present a potential solution for endosomal withdrawal and nuclear uptake challenges in gene delivery. Transfection efficiencies of cationic glyconanoparticles exhibit size dependency as gene delivery aspects [168]. The stability of the double helix with Au NPs is attributed to chemical and electrostatic interactions, with low-density DNA strands potentially adopting a bent conformation for effective stabilization [169].
In the context of anticancer medications targeting DNA, understanding drug interactions with DNA is crucial for developing effective medication delivery systems. The surface density of DNA-AuNP-based sensors significantly influences their performance. At low surface densities, nonspecific binding restricts hybridization, while high densities may render capture probes ineffective, posing challenges for stability and reproducibility [170].
Storage issues, including nonspecific probe DNA contact and contamination, affect the stability of DNA-AuNPs over time. Common techniques involving gradual thiolated DNA attachment to AuNPs over 12 h require careful storage to maintain high-quality DNA-AuNPs [171]. Ionic strength, pH and local environment influence interactions between DNA and citrate-AuNPs, necessitating the addition of salt for adsorption [172].
The pH of the solution is critical for DNA-AuNP stability, with destabilization occurring below pH 5 or above pH 9. Citrate-capped AuNPs remain stable between pH 5 and pH 9, although low pH can partially protonate citrate [173,174]. Kinetic studies reveal DNA strands spreading over Au NP surfaces over time, and functionalizing gold nanoparticles with oligonucleotides for aptamer hybridization is a superior method [1,17,96].
RNA molecules offer a low-risk alternative to DNA-based treatments, enabling selective gene expression modulation. Recombinant proteins, while useful, require repeated injections and lengthy purification procedures. RNA therapies, including siRNA- and mRNA-based modulations, present a highly effective and secure alternative [175,176,177].
RNA interference (RNAi) holds potential for silencing disease-causing genes. Delivery remains a fundamental barrier, with siRNAs facing challenges in pharmacokinetics and metabolic stability. Melanomas, with constitutively activated STAT3, are targeted effectively with siRNA [178]. To extend siRNA half-life, molecular modification or carrier substances are employed [179].
Therapeutic siRNA release over time requires a biomaterial supporting storage and controlled release without negatively impacting cell viability. RNA molecules are prone to degradation and immune recognition, necessitating modification strategies for in vivo applications [180]. Aptamers, whether DNA or RNA oligonucleotides, offer advantages in cancer biomarker recognition and anticancer drug delivery [181,182].
Aptamers, being non-immunogenic and stable in various conditions, act as both targeting moieties and drug vehicles [181,183]. They demonstrate success in targeting multiple cancer cell receptors, enhancing treatment selectivity [1,96]. Aptamers address therapeutic voids similar to monoclonal antibodies, providing low immunogenicity, minimal batch variation, ease of production, prolonged shelf life, stability and targeting potential [181,184]. Aptamers also facilitate gene delivery by preventing DNase activity from reaching the target site [185]. Table 5 presents various DNA, RNA and aptamers used as coatings for Au NPs.

6.6. Carbohydrate Directed Au NPs

Carbohydrates play a central role in cell growth and infection events, being implicated in major infections such as HIV, tuberculosis and malaria. Glycan recognition results in the formation of lectins that selectively bind to carbohydrates. Although lectin–carbohydrate interactions exhibit weaker binding compared to specific enzyme–substrate or antibody–antigen motifs, cells overcome this limitation through multivalency. By presenting multiple glycan moieties in proximity, cells enable cooperative binding to receptors, leading to a greater increase in affinity than simple monovalent interactions. This multivalency is effectively mimicked by Au NPs, serving as excellent cell mimics with specific attributes, such as shape, size and a high specific surface area, facilitating the presentation of carbohydrates with varying spacing, ligand density and spatial orientation [198].
Significant biological macromolecules, such as alginate, carrageenan, porphyran, fucoidan, ulvan, agarose and chitosan, are widely found in marine algae and animals, a variety of which are illustrated in Figure 11. These macromolecules possess desirable properties such as biocompatibility, biodegradability and lack of irritation, making them extensively useful in biomedicine and nanomaterials. Specifically, carbohydrate functionalized AuNPs, employing a greener preparation method, demonstrate enhanced biocompatibility and targeting capabilities. This is exemplified in Figure 11, where the diverse applications of these marine-derived macromolecules in AuNP functionalization are depicted [199].
In the realm of metal nanomaterials, carbohydrates, especially in AuNPs, play a crucial role. For example, the chitosan/alginate complex enhances the biocompatibility and anticancer activity of AuNPs [200]. The selective modification of carbohydrate surfaces, such as sulfation and acetylation, holds promise for improving their use in nanomaterials [199,201].
To broaden the applications of carbohydrate-based AuNP nanosystems, AuNPs can be modified using specific ligands like functional peptides and targeting receptors [202].
Non-covalent forces, including hydrogen bonds, hydrophobic interactions and van der Waals forces, facilitate the binding of carbohydrates to AuNPs. While covalent bonds are weaker, non-covalent bonds, commonly referred to as sorption or physisorption, can effectively bind carbohydrates to NPs through multiple interactions, collectively resulting in strong binding interactions [198].
They exhibit a diverse array of functional groups, such as hydroxyl, amino, carboxylate, sulfate and ester groups, amenable to functionalization through hydrogen bonding, electrostatic attraction or chemical modification [199,203].
Carbohydrates, when encapsulated, and gold nanoparticles (Au NPs), when appropriately stabilized, demonstrate prolonged blood circulation, enhancing their biomedical efficacy. These have found extensive application in drug delivery systems, mitigating adverse effects on healthy cells through electrostatic interaction, hydrogen bonding or non-covalent interaction [118,204].
Carbohydrate ligands on glyco-Au NPs play multifaceted roles in formulations, contributing to (1) increased circulation times by minimizing clearance, (2) reduced nanoparticle cytotoxicity and (3) targeting glycan-responsive receptors. Exploiting the Warburg effect, where cancer cells exhibit a preference for glucose metabolism over oxidative phosphorylation, carbohydrates can actively target cancer cells, capitalizing on the overexpression of sugar transporters in these cells. Furthermore, glycosylation of magnetic nanoparticles (MNPs) has demonstrated enhanced in vivo circulation times for NPs [198].
The distinction between cancer cells and healthy cells lies in the differential expression of lectins on their surfaces. Considering this, investigations into the affinity of carbohydrates for these lectins have been conducted to target cancer cells. Notably, molecules such as hyaluronic acid (HA), a natural polysaccharide with multiple free carboxylic acids in its polymeric structure, find widespread use in drug functionalization [17]. Table 6 presents the most frequently employed carbohydrates for coating Au NPs in biological applications.

6.7. Drug Small, Medium and Big Molecule Directed Au NPs

The economic viability of synthesizing small molecules presents significant advantages, demonstrating increased stability compared to previously discussed coating types. Small molecules, owing to their size, facilitate efficient delivery of payloads into tumors. Some molecules, due to their reduced size, necessitate conjugation through linkers such as PEG or polyethyleneimine via amide bonds to attach to Au NPs; examples of functionalization through electrostatic interactions have also been documented [1,17].
The field of nanotechnology has proven invaluable for various applications, including drug delivery, diagnostics and therapeutics, thanks to its special optical and imaging properties, as well as its highly tunable nature [222]. Numerous agents for leveling and stabilizing the surface functionalization of Au NPs have been reported, encompassing thiol/thiolated groups, disulfides, carboxylate groups, amines, hydroxyl compounds, surfactants and phosphate-based chelating agents. Thiol-protected Au NPs, due to their desirable stability and ease of control, characterization and functionalization, exhibit robust Au-S bonds. Concurrently, heteroatoms such as nitrogen (N), oxygen (O) and phosphorus (P) in carboxylate, amine and hydroxyl compounds, and phosphate-based capping agents and stabilizers, engage in electrostatic interactions and surface functionalization with chelator agents, and GDPs [223,224].
Au NPs can be directly linked to small molecules, such as certain drugs or antibiotics, through physical absorption or covalent or ionic bonding. Methotrexate (MTX), an analog of folic acid, stands out as a widely used molecule with demonstrated enhanced cytotoxic activity when conjugated with Au NPs compared to its free state [1].
Attributing antibacterial properties to Au NPs can be achieved through Au-S binding. In 2010, Au-DAPT, functionalized Au NPs with 4,6-diamino-2-pyrimidinethiol (DAPT), demonstrated antibacterial activity against multidrug-resistant Gram-negative bacteria, highlighting the synergistic effect of the components [97]. Au NPs exhibit broad-spectrum antibacterial activities, even against superbugs resistant to most antibiotics, achieved through surface functionalization with an N-heterocyclic molecule [97]. Physical adsorption serves as an additional surface modification method for Au NPs, with pharmaceutical intermediates such as 7-aminocephalosporanic acid (7-ACA), 6-aminopenicillanic acid (6-APA) and 7-aminodeacetoxycephalosporanic acid (7-ADCA) adsorbing onto the surface through physical adsorption between amino and gold groups [97].
Surface functionalization of Au NPs with diverse molecules holds promise for the development of novel antimicrobial materials. Meanwhile, further investigation is essential to understand the effects of different sizes, shapes and surface properties of Au NPs on their antimicrobial properties. Table 7 presents all types of drugs, large and small molecules, used to coat AuNPs for biological applications.

6.8. Polymer and Bio-Functionalized Au NP

The synthesis of water-soluble Au particles stabilized with starch dates back to Helcher’s treatise in 1718, marking the inception of the study of polymer-stabilized Au nanoparticles (P-Au NPs) [283]. P-Au NPs, a subject of ongoing nanotechnological exploration, hold potential for applications in drug delivery. Commonly employed synthetic routes for P-Au NPs include “direct synthesis”, “grafting to” and “grafting from” methods [284].
In the majority of cases, end-functional polymers or selective block copolymers are either physically or chemically adsorbed onto the AuNP surface. This process results in the formation of densely packed surface-tethered polymer chain assemblies, commonly referred to as polymer brushes. Notably, the polymer coating of these AuNPs typically consists of a single type of polymer [285].
P-Au NPs are frequently coated with diverse polymers, such as polyethyleneimine [106,286,287], chitosan [2,187,202], PVA [5] and Pluronic hydrogel [20], among others. In the realm of cancer chemotherapy, polymeric ligands offer several advantages, including prolonged stability of Au NPs, modified solubility, enhanced outer surface hydrophilicity, adjusted shell surface density, reduced immunogenicity and improved biocompatibility [288].
Biocompatible polymers, including poly (ethylene glycol), heparin, hyaluronic acid, chitosan, polystyrenesulfonate, polyethyleneimine and xanthan gum, find application in surface modification of Au NPs for various purposes. These modifications serve to increase the capacity of NPs and payloads, facilitating long systemic circulation followed by cellular uptake. This usage positions Au NPs as a promising system for drug/nucleic acid delivery in cancer therapy [288].
The “direct synthesis” method involves the reduction of tetrachloroauric acid (HAuCl4) in the presence of sulfur-terminated polymers, resulting in Au NPs in a single step (Figure 12). On the other hand, “grafting to” refers to the creation of P-AuNPs by attaching functionalized polymers to their surface, utilizing polymers with thiol or amine groups at the beginning, middle or end of the polymer to stabilize Au NPs. The Brust–Schiffrin method outlines this process, wherein the reaction proceeds through ligand substitution [289].
Gold nanoparticles functionalized with biopolymers, particularly those based on polysaccharides, exhibit outstanding biocompatibility and minimal toxicity [96]. The positive charge and hydrophobicity of biopolymers, such as chitosan, contribute to the effective stabilization of Au NPs, mitigating their pronounced agglomeration tendency and fostering electrostatic repulsion [96]. Moreover, polymers not only deter the aggregation of Au NPs but also possess the capability to modulate their morphology by controlling reaction conditions.
Chitosan, renowned for its abundance and cost-effectiveness, emerges as one of the most widely employed biopolymers, serving dual roles as a stabilizer and a reducing agent. Additionally, macrocyclic supramolecules like cyclodextrin, characterized by unique and size-adjustable cavity structures, find application in AuNP stabilization. The synthesis of P-Au NPs involves the “grafting to” method, wherein functionalized polymers are attached to the NP surface, typically employing polymers with thiol or amine groups [96]. Ligand substitution further propels the reaction. Another approach, known as reverse addition fragmentation chain transfer (RAFT) or “grafting from”, entails polymerization at the Au surface, initiated by a species or chain transfer agent, leading to subsequent polymer chain growth from the AuNP surface [96,288].
Beyond chitosan, various polysaccharides and carbohydrates, such as heparin, serve as effective reducing agents, enhancing biocompatibility, stability and anticoagulant activity of synthesized Au NPs [96]. In Table 8, we provide an overview of key biopolymers and biofunctionalizations extensively employed in biological applications of Au NPs.

6.9. Cell-Membrane-Coated Au NPs

Cell-membrane-coated gold nanoparticles (Au NPs) offer a versatile approach for treating various pathologies, leveraging inherent properties inherited from the cells of origin. This innovative technology has been applied to a diverse range of cell types, including red blood cells, platelets, tumor cells, immune cells and various bacteria (Table 9), playing a pivotal role in disease surveillance and prevention [303].
The fabrication of nanoparticles with cell membrane coatings involves several methodologies.
  • Firstly, membrane-coated nanoparticles can be prepared through membrane fusion using physical extrusion, a process akin to liposome synthesis. This technique employs porous membrane materials with uniform pore diameters, facilitating the creation of nanoparticles (Figure 13) [103].
  • Secondly, ultrasonic treatment also induces the spontaneous formation of nanoparticles from core–shell structures enveloped by cell membranes, exhibiting morphologies comparable to those obtained through physical extrusion [304].
  • An alternative and noteworthy approach involves the utilization of microfluidic systems. Reports indicate that combining rapid mixing and electroporation in microfluidic devices can synthesize cell-membrane-coated nanoparticles with enhanced structural integrity and functional reserve, ensuring more efficient production. This cutting-edge technology employs a microfluidic device comprising a Y-type channel, an S-shaped channel mixer and an electroporation area. Through this system, transient pores are generated in cellular membrane memory via electronic pulses, facilitating nanoparticle encapsulation by cell membranes. Notably, this method outperforms alternative strategies by preserving memory and membrane integrity while minimizing protein loss from the membrane surface.
In summary, the use of cell-membrane-coated nanoparticles presents a promising avenue for addressing various pathologies. The diverse synthesis methods, including physical extrusion, ultrasonic treatment and microfluidic systems with electroporation, provide researchers with versatile tools to tailor nanoparticles for specific applications, while also ensuring the preservation of cellular properties critical for therapeutic efficacy [103,303,306].
In view of the multifaceted nature of the challenges faced by cell membrane coating technology, recent studies emphasize the importance of a holistic approach to optimization. This approach involves a comprehensive examination of the manufacturing processes, with a keen focus on identifying and mitigating bottlenecks that impede yield. By strategically addressing these bottlenecks, researchers aim to not only enhance the efficiency of the manufacturing process but also pave the way for a more seamless translation of this technology into diverse biomedical applications [303].
Table 9. Cell membrane AuNP coatings. DD—Drug delivery, CT—Cancer treatment, DA—Diagnostic application, BA—Biomedical application.
Table 9. Cell membrane AuNP coatings. DD—Drug delivery, CT—Cancer treatment, DA—Diagnostic application, BA—Biomedical application.
CoatingApplication
EpCam-RPAuNs (Au nanocages (AuNs) encapsulated anti-tumor drug paclitaxel coated by RBC membranes modified by anti-EpCam antibodies)DD [307]
4t1 (breast tumor cell)CT [307]
MacrophagesCT [308]
ErythrocytesDD [309]
LeukocytesCT [310]
MonocytesCT [309]
Escherichia coliBA [176]
HUVEC (endothelial cells)CT [311]
OMVs (bacterial outer membrane vesicles)BA [311]
RBC (red blood cells)BA [312]
MPCM-AuNSs (macrophage-cell-membrane-camouflaged AuNS)BA [313]
THP-1 (human monocyte leukemia cell membrane)CT [310]
AuNP-pep@MemBA [314]
NSC (neural-stem-cell-mediated—intratumoral)DD [315]
BM-AuNPs (bacterial-membrane-coated gold nanoparticles)BA [316]
MDA MB-435 (breast tumor cell)CT [317]
HTC (rat hepatoma)CT [318]
MCF-7 (breast cancer cell)CT [318]
RAW264.7 (Macrophage Abelson Murine Leukemia Virus Transformed)BA [318]
Cell membraneCT [319]

7. Conclusions

The performance of gold nanoparticles (AuNPs) can be significantly adapted and optimized through the appropriate choice of coatings. Each coating introduces unique characteristics that can enhance stability, circulation time, target specificity and other crucial properties of the AuNPs. Selecting the right coating is, therefore, a critical step to achieve the desired functionality and ensure the therapeutic success of the nanoparticles.
  • Impact of Coatings on the Optimization of Gold Nanoparticle Performance
Each of the coatings for gold nanoparticles offers unique advantages. However, their application in practical scenarios can also come with challenges or obstacles. Let us delve into the potential obstacles for each coating:
1—PEG: While it offers biocompatibility, it may induce immune reactions after repeated administrations, a phenomenon known as the “accelerated blood clearance effect”.
2—Antibodies: They can be expensive to produce and stabilize. Furthermore, they might not have a long circulation if not properly modified.
3—Proteins and Peptides: Stability can vary and there is potential for degradation, especially in hostile environments like the human body.
4—Amino Acids: Although they are more accessible, they might not offer the same specificity or functionality as other more complex molecules.
5—Microorganisms (DNA, RNA, Aptamers): Stability can be an issue, and there are potential concerns about the immunogenicity of certain sequences.
6—Carbohydrates: Their functionalization might not be as straightforward or effective as other molecules, and there is potential for immune reactions.
7—Drugs (Medications): Controlled drug release can be a challenge, as well as ensuring the drug remains stable and active.
8—Polymers: Biocompatibility can vary, and stabilizing nanoparticles might be challenging depending on the polymer chemistry.
9—Cell Membranes: There is potential for rapid degradation in the body, and concerns about immunogenicity, depending on the origin of the membrane.
In the realm of gold nanoparticle (AuNP) coatings, PEG stands out, particularly in combination with other molecules. PEG, a non-toxic hydrophilic polymer commonly used in pharmaceutical formulations, enhances stability, and reduces nonspecific interactions, prolonging blood circulation. PEGylated AuNPs, such as PEG-SH, offer stability, biocompatibility, prolonged circulation and cytotoxicity. Despite advances in drug delivery systems like liposomes and PEG nanoparticles, their efficiency and yield remain limited.
Recent advancements enable pharmaceuticals to bind to various inorganic nanomaterials, facilitating a reliable drug delivery platform. Gold nanorods (GNRs) exhibit high conjugation efficiency due to their large surface area. While PEG’s effects on stability and circulation are extensively studied, its impact on payload release is not fully understood.
Aptamers, notably associated with chitosan nanoparticles, show promise for targeted drug delivery. However, challenges persist, including quick filtration in the plasma compartment and susceptibility to nucleases affecting aptamer affinity.
DNA duplexes on gold nanoparticles exhibit higher thermal stability than pure duplexes, impacting payload release. The complex interaction between DNA bases and AuNPs, influenced by keto and amino groups, presents challenges in understanding their binding affinity.
RNA therapeutics, including siRNAs, ASOs and mRNAs, have entered clinical trials, benefiting from advanced design and chemistry. Various approaches, such as AVV-based delivery and nanoparticle methods, are explored, with challenges in effective transfection and RNA molecule encapsulation.
Carbohydrates like carrageenan, fucoidan and chitosan, due to their biocompatibility and ease of modification, are frequently used in AuNP coatings for diverse applications. Amino acids, an underutilized option, can reduce toxicity and increase stability.
Protein-AuNP complexes, dynamic and complex entities, depend on factors like surface chemistry and curvature, influencing biodistribution. Peptides, with lower receptor affinity, offer site-specific conjugation advantages, but their short circulatory half-life poses challenges.
Small molecules, though with high tumor penetration, exhibit lower affinity and selectivity. Polymer ligands, with positive charge and hydrophobicity, enhance solution stability and enable additional functionality.
Membrane-coated nanoparticles, using cell membranes like those of red blood cells, offer versatility in immune response modulation and homologous targeting. Challenges include comprehending membrane-coated particle properties and ensuring stability.
AuNPs, while demonstrating potential in pre-clinical studies, face challenges in toxicity assessment, biodistribution and clinical translation. Advances in coatings and controlled release strategies show promise, but careful consideration is necessary for successful therapeutic application.
In conclusion, despite the substantial benefits and cellular versatility of AuNPs in medical applications, challenges persist in toxicity, stability and clinical translation. Ongoing innovations in nanomedicine offer potential, emphasizing the need for meticulous approaches to overcome existing hurdles and enhance therapeutic efficacy. The development of nanocarriers holds promise for future improvements in human health.

Author Contributions

W.A.A.R.: Conceptualization, methodology, software, validation, formal analysis, investigation, resources, data curation, writing-original draft preparation, writing review and editing, visualization, supervision, project administration, funding acquisition. A.B.B.: Conceptualization, methodology, validation, formal analysis, investigation, data curation, writing-original draft preparation, writing review and editing, visualization, supervision, project administration, funding acquisition. C.D.d.S.: validation, formal analysis, data curation, writing review and editing, visualization, supervision, project administration. M.E.C.M.R.: Conceptualization, methodology, formal analysis, investigation, resources, data curation, writing-original draft preparation, writing review and editing, visualization, supervision, project administration, funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by: FAPESP 2017/50332-0 and 2020/07065-4, CNPQ PCI 300407/2023-8, CNPQ INTERAS 406761/2022-1, IPEN 2018.05.IPEN.9, 2020.06.IPEN.37 and AIEA BRA-6062, for project funding and scholarship.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Amina, S.J.; Guo, B. A review on the synthesis and functionalization of gold nanoparticles as a drug delivery vehicle. Int. J. Nanomed. 2020, 15, 9823–9857. [Google Scholar] [CrossRef]
  2. Bromma, K.; Chithrani, D.B. Advances in gold nanoparticle-based combined cancer therapy. Nanomaterials 2020, 10, 1671. [Google Scholar] [CrossRef]
  3. Selmani, A.; Kovačević, D.; Bohinc, K. Nanoparticles: From synthesis to applications and beyond. Adv. Colloid Interface Sci. 2022, 303, 102640. [Google Scholar] [CrossRef] [PubMed]
  4. Huang, X.; El-sayed, M.A. Gold nanoparticles: Optical properties and implementations in cancer diagnosis and photothermal therapy. J. Adv. Res. 2010, 1, 13–28. [Google Scholar] [CrossRef]
  5. De Freitas, L.F.; Varca, G.H.C.; Batista, J.G.D.S.; Lugão, A.B. An overview of the synthesis of gold nanoparticles using radiation technologies. Nanomaterials 2018, 8, 939. [Google Scholar] [CrossRef] [PubMed]
  6. Zhang, L.; Webster, T.J. Nanotechnology and nanomaterials: Promises for improved tissue regeneration. Nanotoday 2009, 4, 66–80. [Google Scholar] [CrossRef]
  7. Yaghini, E.; Seifalian, M.A.; MacRobert, J.A. Quantum dots and their potential biomedical applications in photosensitization for photodynamic therapy R eview. Futur. Med. 2009, 4, 353–363. [Google Scholar]
  8. Gaumet, M.; Vargas, A.; Gurny, R.; Delie, F. Nanoparticles for drug delivery: The need for precision in reporting particle size parameters. Eur. J. Pharm. Biopharm. 2008, 69, 1–9. [Google Scholar] [CrossRef] [PubMed]
  9. Owens, D.E.; Peppas, N.A. Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm. 2006, 307, 93–102. [Google Scholar] [CrossRef] [PubMed]
  10. Kumari, Y.; Kaur, G.; Kumar, R.; Kumar, S.; Gowthamarajan, K.; Karri, V.V.S.N.R. Gold Nanoparticles: New Routes Across Old Boundaries. Adv. Colloid Interface Sci. 2019, 274, 102037. [Google Scholar] [CrossRef]
  11. Idoudi, S.; Ismail, R. The Golden Liposomes: Preparation and Biomedical Applications of Gold-Liposome Nanocomposites. Nanotheranostics 2023, 4, 201–227. [Google Scholar] [CrossRef]
  12. Giordani, S.; Marassi, V.; Zattoni, A.; Roda, B.; Reschiglian, P. Liposomes characterization for market approval as pharmaceutical products: Analytical methods, guidelines and standardized protocols. J. Pharm. Biomed. Anal. 2023, 236, 115751. [Google Scholar] [CrossRef] [PubMed]
  13. Firdhouse, M.J.; Lalitha, P. Biogenic green synthesis of gold nanoparticles and their applications—A review of promising properties. Inorg. Chem. Commun. 2022, 143, 109800. [Google Scholar] [CrossRef]
  14. Bhattarai, B.; Zaker, Y.; Bigioni, T.P. ScienceDirect Green synthesis of gold and silver nanoparticles: Challenges and opportunities. Curr. Opin. Green Sustain. Chem. 2018, 12, 91–100. [Google Scholar] [CrossRef]
  15. Cell, C.; Khan, M.Y.; Roy, M. Synthesis, Limitation and Application of Gold Nanoparticles in Treatment of Cancerous Cell. Int. J. Sci. Res. Multidiscip. Stud. 2019, 5, 9. [Google Scholar] [CrossRef]
  16. Chen, H.; Zhou, K.; Zhao, G. Gold nanoparticles: From synthesis, properties to their potential application as colorimetric sensors in food safety screening. Trends Food Sci. Technol. 2018, 78, 83–94. [Google Scholar] [CrossRef]
  17. Goddard, Z.R.; Marín, M.J.; Russell, D.A.; Searcey, M. Active targeting of gold nanoparticles as cancer therapeutics. Chem. Soc. Rev. 2020, 49, 8774–8789. [Google Scholar] [CrossRef]
  18. Matczuk, M.; Ruzik, L.; Aleksenko, S.S.; Keppler, B.K.; Jarosz, M.; Timerbaev, A.R. Analytical methodology for studying cellular uptake, processing and localization of gold nanoparticles. Anal. Chim. Acta 2019, 1052, 1–9. [Google Scholar] [CrossRef]
  19. Siddique, S.; Chow, J.C.L. Gold nanoparticles for drug delivery and cancer therapy. Appl. Sci. 2020, 10, 3824. [Google Scholar] [CrossRef]
  20. Beik, J.; Khateri, M.; Khosravi, Z.; Kamrava, S.K.; Kooranifar, S.; Ghaznavi, H.; Shakeri-zadeh, A. Gold nanoparticles in combinatorial cancer therapy strategies. Coord. Chem. Rev. 2019, 387, 299–324. [Google Scholar] [CrossRef]
  21. Zhao, Y.; Sultan, D.; Liu, Y. Biodistribution, Excretion, and Toxicity of Nanoparticles; Elsevier Inc.: Amsterdam, The Netherlands, 2019. [Google Scholar] [CrossRef]
  22. Akturk, O.; Erdemli, O.; Tunali, B.C. Chapter 15. Gold nanocomposites for biomedical applications. In Materials for Biomedical Engineering: Bioactive Materials, Properties, and Applications; Elsevier Inc.: Amsterdam, The Netherlands, 2019; pp. 485–526. [Google Scholar] [CrossRef]
  23. Chandran, P.R.; Thomas, R.T. Chapter 14. Gold Nanoparticles in Cancer Drug Delivery. In Nanotechnology Applications for Tissue Engineering; Elsevier Inc.: Amsterdam, The Netherlands, 2015; pp. 221–238. [Google Scholar] [CrossRef]
  24. Hammami, I.; Alabdallah, N.M.; Al, A. Gold nanoparticles: Synthesis properties and applications. J. King Saud Univ.-Sci. 2021, 33, 101560. [Google Scholar] [CrossRef]
  25. Pinelli, F.; Perale, G.; Rossi, F. Coating and Functionalization Strategies for Nanogels and Nanoparticles for Selective Drug Delivery. Gels 2020, 6, 6. [Google Scholar] [CrossRef]
  26. Ramzy, L.; Nasr, M.; Metwally, A.A.; Awad, G.A.S. Cancer nanotheranostics: A review of the role of conjugated ligands for overexpressed receptors. Eur. J. Pharm. Sci. 2017, 104, 273–292. [Google Scholar] [CrossRef]
  27. Mohd-Zahid, M.H.; Mohamud, R.; Che Abdullah, C.A.; Lim, J.; Alem, H.; Wan Hanaffi, W.N.; Iskandar, Z.A. Colorectal cancer stem cells: A review of targeted drug delivery by gold nanoparticles. RSC Adv. 2019, 10, 973–985. [Google Scholar] [CrossRef]
  28. Connor, D.M.; Broome, A. Gold Nanoparticles for the Delivery of Cancer Therapeutics, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2018. [Google Scholar] [CrossRef]
  29. Chen, Y.; Feng, X. Gold nanoparticles for skin drug delivery. Int. J. Pharm. 2022, 625, 122122. [Google Scholar] [CrossRef]
  30. Dianzani, C.; Zara, G.P.; Maina, G.; Pettazzoni, P.; Pizzimenti, S.; Rossi, F.; Gigliotti, C.L.; Ciamporcero, E.S.; Daga, M.; Barrera, G. Drug delivery nanoparticles in skin cancers. Biomed Res. Int. 2014, 2014, 895986. [Google Scholar] [CrossRef]
  31. Sharma, R.; Agrawal, U.; Mody, N.; Dubey, S.; Vyas, S.P. Engineered Nanoparticles as a Precise Delivery System in Cancer Therapeutics; Elsevier Inc.: Amsterdam, The Netherlands, 2016. [Google Scholar] [CrossRef]
  32. Ykman, L.A.D.; Hlebtsov, N.G.K. Gold nanoparticles in chemo-, immuno-, and combined therapy. Biomed. Opt. Express 2019, 10, 3152–3182. [Google Scholar] [CrossRef]
  33. Lin, A.Y.; Lunsford, J.; Bear, A.S.; Young, J.K.; Eckels, P.; Luo, L.; Foster, A.E.; Drezek, R.A. High-density sub-100-nm peptide-gold nanoparticle complexes improve vaccine presentation by dendritic cells in vitro. Nanoscale Res. Lett. 2013, 8, 72. [Google Scholar] [CrossRef] [PubMed]
  34. Paulo, J.; Almeida, M.; Lin, A.Y.; Figueroa, E.R.; Edward, A.; Drezek, R.A. In vivo gold nanoparticle delivery of peptide vaccine induces anti-tumor immune response in prophylactic and therapeutic tumor models. HHS Public Access 2016, 11, 1453–1459. [Google Scholar]
  35. Cohn, L.; Delamarre, L. Dendritic cell-targeted vaccines. Front. Immunol. 2014, 5, 255. [Google Scholar] [CrossRef] [PubMed]
  36. Vang, K.B.; Safina, I.; Darrigues, E.; Nedosekin, D.; Nima, Z.A.; Watanabe, F.; Kannarpady, G.; Kore, R.A.; Casciano, D.; Zharov, V.P.; et al. Modifying Dendritic Cell Activation with Plasmonic Nano Vectors. Sci. Rep. 2017, 7, 5513. [Google Scholar] [CrossRef]
  37. Zang, X.; Zhao, X.; Hu, H.; Qiao, M.; Deng, Y.; Chen, D. Nanoparticles for tumor immunotherapy. Eur. J. Pharm. Biopharm. 2017, 115, 243–256. [Google Scholar] [CrossRef]
  38. Fogli, S.; Paccosi, S.; Michelucci, E.; Berti, D.; Parenti, A. Inorganic nanoparticles as potential regulators of immune response in dendritic cells. Nanomedicine 2017, 12, 1647–1660. [Google Scholar] [CrossRef]
  39. Ahmad, S.; Zamry, A.A.; Tan, H.T.T.; Wong, K.K.; Lim, J.K.; Mohamud, R. Targeting dendritic cells through gold nanoparticles: A review on the cellular uptake and subsequent immunological properties. Mol. Immunol. 2017, 91, 123–133. [Google Scholar] [CrossRef] [PubMed]
  40. Yang, Y.; Huang, Q.; Xiao, Z.; Liu, M.; Zhu, Y.; Chen, Q. Nanomaterial-based biosensor developing as a route toward in vitro diagnosis of early ovarian cancer. Mater. Today Bio 2022, 13, 100218. [Google Scholar] [CrossRef] [PubMed]
  41. Spicer, C.D.; Jumeaux, C.; Gupta, B.; Stevens, M.M. Peptide and protein nanoparticle conjugates: Versatile platforms for biomedical applications. Chem. Soc. Rev. 2018, 47, 3574–3620. [Google Scholar] [CrossRef]
  42. Ali, H.S.; El-haj, B.M.; Saifullah, S.; Kawish, M. Gold nanoparticles in cancer diagnosis and therapy. In Metal Nanoparticles for Drug Delivery and Diagnostic Applications; Elsevier Inc.: Amsterdam, The Netherlands, 2020; pp. 43–58. [Google Scholar] [CrossRef]
  43. Tsirikis, P.; Xiang, S.D.; Selomulya, C.; Plebanski, M. Design of nanoparticle structures for cancer immunotherapy. In Nanostructures for Cancer Therapy; Elsevier Inc.: Melbourne, VIC, Australia, 2017; pp. 307–328. [Google Scholar] [CrossRef]
  44. Carone, A.; Emilsson, S.; Mariani, P.; Anthony, D. Nanoscale Advances Gold nanoparticle shape dependence of colloidal stability domains. Nanoscale Adv. 2023, 5, 2017–2026. [Google Scholar] [CrossRef] [PubMed]
  45. Bürgi, T. Nanoscale from self-assembled monolayers to clusters. Nanoscale 2015, 7, 15553–15567. [Google Scholar] [CrossRef]
  46. Knoppe, S.; Bu, T. Chirality in Thiolate-Protected Gold Clusters. Acc. Chem. Res. 2014, 47, 1318–1326. [Google Scholar] [CrossRef]
  47. Porret, E.; Le Guevel, X.; Coll, J.-L. Gold Nanoclusters for Biomedical Applications: Toward In Vivo Studies. J. Mater. Chem. B 2020, 8, 2216–2232. [Google Scholar] [CrossRef]
  48. Müller, J.; Bauer, K.N.; Prozeller, D.; Simon, J.; Mailänder, V.; Wurm, F.R.; Winzen, S.; Landfester, K. Coating nanoparticles with tunable surfactants facilitates control over the protein corona. Biomaterials 2017, 115, 1–8. [Google Scholar] [CrossRef]
  49. Walkey, C.D.; Chan, W.C.W. Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. Chem. Soc. Rev. 2012, 41, 2780–2799. [Google Scholar] [CrossRef]
  50. Del Pino, P.; Pelaz, B.; Zhang, Q.; Maffre, P.; Nienhaus, G.U.; Parak, W.J. Protein corona formation around nanoparticles-From the past to the future. Mater. Horizons 2014, 1, 301–313. [Google Scholar] [CrossRef]
  51. Vinluan, R.D., III; Zheng, J. Serum protein adsorption and excretion pathways of metal nanoparticles. Nanomedicine 2015, 10, 2781–2794. [Google Scholar] [CrossRef]
  52. Sen, S.; Thaker, A.; Sirajudeen, L.; Williams, D.; Nannenga, B.L. Protein–Nanoparticle Complex Structure Determination by Cryo-Electron Microscopy. Appl. Bio Mater. 2022, 5, 4696–4700. [Google Scholar] [CrossRef]
  53. Ucuzian, A.A.; Greisler, H. Chapter 7.29 Vascular Grafts; Elsevier Ltd.: Amsterdam, The Netherlands, 2017. [Google Scholar]
  54. Haranath, C.; Vamsi, K.S.; Qarmout, Y.A.M.; Ahad, H.A.; Chandana, S. Impact of Vroman’ s Effect on Pharmacodynamics and Pharmacokinetics on Nanoparticulate Drug Delivery Systems. J Young Pharm 2022, 14, 355–358. [Google Scholar] [CrossRef]
  55. Angioletti-uberti, S.; Ballauff, M.; Dzubiella, J.; Angioletti-uberti, S.; Ballauff, M. Competitive adsorption of multiple proteins to nanoparticles: The Vroman effect revisited revisited. Mol. Phys. 2018, 116, 3154–3163. [Google Scholar] [CrossRef]
  56. Cedervall, T.; Lynch, I.; Lindman, S.; Berggård, T.; Thulin, E.; Nilsson, H.; Dawson, K.A.; Linse, S. Understanding the nanoparticle–protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles. Proc. Natl. Acad. Sci. USA 2007, 104, 2050–2055. [Google Scholar] [CrossRef]
  57. Aggarwal, P.; Hall, J.B.; Mcleland, C.B.; Dobrovolskaia, M.A.; Mcneil, S.E. Nanoparticle interaction with plasma proteins as it relates to particle biodistribution, biocompatibility and therapeutic efficacy. Adv. Drug Deliv. Rev. 2009, 61, 428–437. [Google Scholar] [CrossRef] [PubMed]
  58. Lin, J.; Zhang, H.; Dargazany, R. PEGylation on Mixed Monolayer Gold Nanoparticles: Effect of Grafting Density, Chain Length, and Surface Curvature. J. Colloid Interface Sci. 2017, 504, 325–333. [Google Scholar] [CrossRef]
  59. Tlotleng, N.; Vetten, M.A.; Keter, F.K.; Skepu, A.; Tshikhudo, R.; Gulumian, M. Cytotoxicity, intracellular localization and exocytosis of citrate capped and PEG functionalized gold nanoparticles in human hepatocyte and kidney cells. Cell Biol. Toxicol. 2016, 32, 305–321. [Google Scholar] [CrossRef]
  60. Chen, T.; Chen, M.; Liu, S.; Lin, J.; Yang, Y.; Huang, H.; Chen, J.; Yang, C.; Lin, K.M. Assessment of Polyethylene Glycol-Coated Gold Nanoparticle Toxicity and Inflammation In Vivo Using NF-κ B Reporter Mice. Int. J. Mol. Sci. 2020, 21, 8158. [Google Scholar] [CrossRef] [PubMed]
  61. Harrison, E.; Nicol, J.R.; Macias, M.; Burke, G.A.; Coulter, J.A.; Meenan, B.J.; Dixon, D. A comparison of gold nanoparticle surface co-functionalization approaches using Polyethylene Glycol (PEG) and the effect on stability, non-speci fi c protein adsorption and internalization. Mater. Sci. Eng. C 2016, 62, 710–718. [Google Scholar] [CrossRef]
  62. Zhou, J.; Cao, Z.; Panwar, N.; Hu, R.; Wang, X.; Qu, J.; Chuan, S.; Xu, G.; Yong, K. Functionalized gold nanorods for nanomedicine: Past, present and future. Coord. Chem. Rev. 2017, 352, 15–66. [Google Scholar] [CrossRef]
  63. Cheng, X.; Li, H.; Ge, X.; Chen, L.; Liu, Y.; Mao, W. Tumor-Microenvironment-Responsive Size-Shrinkable Drug-Delivery Nanosystems for Deepened Penetration Into Tumors. Front. Mol. Biosci. 2020, 7, 576420. [Google Scholar] [CrossRef] [PubMed]
  64. Nanocrystals, G.; Huang, X.; Peng, X.; Wang, Y.; Wang, Y.; Shin, D.M.; El-, M.A.; Nie, S. A Reexamination of Active and Passive Tumor Targeting by Using Rod-Shaped. ACS Nano 2010, 4, 5887–5896. [Google Scholar]
  65. Liu, Y.; Shipton, M.K.; Ryan, J.; Kaufman, E.D.; Franzen, S.; Feldheim, D.L. Synthesis, Stability, and Cellular Internalization of Gold Nanoparticles Containing Mixed Peptide-Poly (ethylene glycol) Monolayers. Anal. Chem. 2007, 79, 2221–2229. [Google Scholar] [CrossRef]
  66. Kumar, D.; Meenan, J.B.; Dixon, D. Glutathione-mediated release of Bodipy ® from PEG cofunctionalized gold nanoparticles. Int. J. Nanomed. 2012, 7, 4007–4022. [Google Scholar] [CrossRef]
  67. Dunlop, I.E.; Ryan, M.P.; Goode, A.E.; Schuster, C.; Terrill, N.J.; Weaver, J.V.M. Direct synthesis of PEG-encapsulated gold nanoparticles using branched copolymer. RSC Adv. 2014, 4, 27702–27707. [Google Scholar] [CrossRef]
  68. Scaletti, F.; Feis, A.; Centi, S.; Pini, R.; Rotello, V.M.; Messori, L. Tuning the interactions of PEG-coated gold nanorods with BSA and model proteins through insertion of amino or carboxylate groups. J. Inorg. Biochem. 2015, 10, 2781–2794. [Google Scholar] [CrossRef]
  69. Hinkley, G.K.; Carpinone, P.; Munson, J.W.; Powers, K.W.; Roberts, S.M. Oral absorption of PEG-coated versus uncoated gold nanospheres: Does agglomeration matter? Oral absorption of PEG-coated versus uncoated gold nanospheres: Does agglomeration matter? Part. Fibre Toxicol. 2015, 12, 9. [Google Scholar] [CrossRef] [PubMed]
  70. Lazarus, G.G.; Singh, M. In vitro cytotoxic activity and transfection efficiency of polyethyleneimine functionalized gold nanoparticles. Colloids Surf. B Biointerfaces 2016, 145, 906–911. [Google Scholar] [CrossRef] [PubMed]
  71. Lai, P.; Letchman, E.; Mashouf, S.; Pignol, J.; Really, R.M. Depot system for controlled release of gold nanoparticles with precise intratumoral placement by permanent brachytherapy seed implantation (PSI) techniques. Int. J. Pharm. 2016, 515, 729–739. [Google Scholar] [CrossRef] [PubMed]
  72. Moyano, D.F.; Ayaz, F.; Osborne, B.A.; Rotello, V.M.; Moyano, D.F.; Liu, Y.; Ayaz, F.; Hou, S.; Puangploy, P. Immunomodulatory Effects of Coated Gold Nanoparticles in LPS-Stimulated In Vitro and In Vivo Murine Model Systems. Chem 2016, 1, 320–327. [Google Scholar] [CrossRef] [PubMed]
  73. Pino, P.; Yang, F.; Pelaz, B.; Zhang, Q.; Kantner, K.; Hartmann, R.; De Baroja, N.M.; Gallego, M.; Mçller, M.; Manshian, B.B.; et al. Basic Physicochemical Properties of Polyethylene Glycol Coated Gold Nanoparticles that Determine Their Interaction with Cells. Angew. Chemie-Int. Ed. 2016, 128, 5573–5577. [Google Scholar] [CrossRef]
  74. Alhussan, A.; Palmerley, N.; Smazynski, J.; Karasinska, J.; Renouf, D.J.; Schaeffer, D.F.; Beckham, W.; Alexander, A.S.; Chithrani, D.B. Potential of Gold Nanoparticles in Current Radiotherapy Using a Co-Culture Model of Cancer Cells and Cancer Associated Fibroblasts. Cancers 2022, 14, 3586. [Google Scholar] [CrossRef]
  75. Liu, X.; Liu, Y.; Zhang, P.; Jin, X.; Zheng, X.; Ye, F.; Chen, W.; Li, Q. The synergistic radiosensitizing effect of tirapazamine-conjugated gold nanoparticles on human hepatoma HepG2 cells under X-ray irradiation. Int. J. Nanomed. 2016, 11, 3517–3531. [Google Scholar] [CrossRef]
  76. Leopold, L.F.; Tódor, I.S.; Andrei, Ş.; Leopold, N.; Coman, C. Assessment of PEG and BSA-PEG gold nanoparticles cellular interaction. Colloids Surf. A 2017, 532, 70–76. [Google Scholar] [CrossRef]
  77. He, B.; Zhang, Y.; Zhang, Q.; He, B.; Dai, W.; Wang, X.; Yang, D.; Qin, M.; Zhang, H.; Yin, C.; et al. Increased cellular uptake of peptide-modified PEGylated gold nanoparticles. Biochem. Biophys. Res. Commun. 2017, 494, 339–345. [Google Scholar] [CrossRef]
  78. Hoshikawa, A.; Tagami, T.; Morimura, C.; Fukushige, K.; Ozeki, T. Ranibizumab biosimilar/polyethyleneglycol-conjugated gold nanoparticles as a novel drug delivery platform for age-related macular degeneration. J. Drug Deliv. Sci. Technol. 2017, 38, 45–50. [Google Scholar] [CrossRef]
  79. Ben, M.; Aouidat, F.; Monteil, M.; Lecouvey, M.; Lamy, M.; Chapelle, D.; Spadavecchia, J. A simple assay for direct colorimetric detection of prostatic acid phosphatase (PAP) at fg levels using biphosphonated loaded PEGylated gold nanoparticles. Front. Lab. Med. 2017, 1, 186–191. [Google Scholar] [CrossRef]
  80. Ghorbani, M.; Hamishehkar, H. Decoration of gold nanoparticles with thiolated pH-responsive polymeric (PEG-b-p(2-dimethylamio ethyl methacrylat-co-Itaconic acid) shell: A novel platform for targeting of anticancer agent. Mater. Sci. Eng. C 2017, 81, 561–570. [Google Scholar] [CrossRef]
  81. Kumar, B.; Gupta, B.; Ramasamy, T.; Kumar, R.; Pathak, S.; Taek, K.; Jeong, J.; Choi, H.; Soon, C. Colloids and Surfaces B: Biointerfaces PEGylated thermosensitive lipid-coated hollow gold nanoshells for effective combinational chemo-photothermal therapy of pancreatic cancer. Colloids Surf. B Biointerfaces 2017, 160, 73–83. [Google Scholar] [CrossRef]
  82. Pulagam, K.R.; Gona, K.B.; Gomez-Vallejo, V.; Meijer, J.; Zilberfain, C.; Estrela-lopis, I.; Baz, Z.; Cossio, U.; Llop, J. Gold Nanoparticles as Boron Carriers for Boron and In Vivo Evaluation. Molecules 2019, 24, 3609. [Google Scholar] [CrossRef]
  83. Elbialy, N.S.; Fathy, M.M.; Khalil, W.M. Doxorubicin loaded magnetic gold nanoparticles for in vivo targeted drug delivery. Int. J. Pharm. 2015, 490, 190–199. [Google Scholar] [CrossRef] [PubMed]
  84. Wolfe, T.; Chatterjee, D.; Lee, J.; Grant, J.D.; Bhattarai, S.; Tailor, R.; Goodrich, G.; Nicolucci, P.; Krishnan, S. Targeted gold nanoparticles enhance sensitization of prostate tumors to megavoltage radiation therapy in vivo. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1277–1283. [Google Scholar] [CrossRef] [PubMed]
  85. Rosman, C.; Pierrat, S.; Tarantola, M.; Schneider, D.; Sunnick, E.; Janshoff, A.; Sönnichsen, C. Mammalian cell growth on gold nanoparticle-decorated substrates is influenced by the nanoparticle coating. Beilstein J. Nanotechnol. 2014, 5, 2479–2488. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, F.; Zhang, W.; Shen, Y.; Huang, Q.; Zhou, D.; Guo, S. Efficient RNA delivery by integrin-targeted glutathione responsive polyethyleneimine capped gold nanorods. Acta Biomater. 2015, 23, 136–146. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, N.; Chen, H.; Liu, A.Y.; Shen, J.J.; Shah, V.; Zhang, C.; Hong, J.; Ding, Y. Gold conjugate-based liposomes with hybrid cluster bomb structure for liver cancer therapy. Biomaterials 2016, 74, 280–291. [Google Scholar] [CrossRef] [PubMed]
  88. Wai, L.; Ho, C.; Ka, C.; Chan, W.; Han, R.; Fong, Y.; Lau, Y.; Li, H.; Ho, Y.; Zhuang, X.; et al. Mammalian Cells Exocytose Alkylated Gold Nanoparticles via Extracellular Vesicles. ACS Nano 2022, 16, 2032–2045. [Google Scholar] [CrossRef]
  89. Shameer, A.B.; Baijal, G.; Somashekar, R.; Iyer, S.; Nayak, V. Comparative study of one pot synthesis of PEGylated gold and silver nanoparticles for imaging and radiosensitization of oral cancers. Radiat. Phys. Chem. 2022, 194, 109990. [Google Scholar] [CrossRef]
  90. David, S.; Patel, D.Y.; Cardona, S.M.; Kirby, N. Cellular uptake and cytotoxicity of PEGylated gold nanoparticles in C33A cervical cancer cells Cellular uptake and cytotoxicity of PEGylated gold nanoparticles in C33A cervical cancer cells. Nano Express 2022, 3, 025006. [Google Scholar] [CrossRef]
  91. Kim, H.; Park, J.H.; Choi, Y.J.; Oh, J.; Park, J. Hyaluronic acid-coated gold nanoparticles as a controlled drug delivery system for poorly water-soluble drugs. RSC Adv. 2023, 13, 5529–5537. [Google Scholar] [CrossRef]
  92. Venditti, I.; Hassanein, T.F.; Fratoddi, I.; Fontana, L.; Battocchio, C.; Rinaldi, F.; Carafa, M.; Marianecci, C.; Diociaiuti, M.; Agostinelli, E.; et al. Bioconjugation of gold-polymer core-shell nanoparticles with bovine serum amine oxidase for biomedical applications. Colloids Surf. B Biointerfaces 2015, 134, 314–321. [Google Scholar] [CrossRef]
  93. Hadi, M.; Amani, H.; Akbar, A.; Pazoki-toroudi, H.; Sedighimoghaddam, B. Various methods of gold nanoparticles (GNPs) conjugation to antibodies. SBSR 2016, 9, 17–22. [Google Scholar] [CrossRef]
  94. Wang, A.; Vangala, K.; Vo, T.; Zhang, D.; Fitzkee, N.C. A Three-Step Model for Protein–Gold Nanoparticle Adsorption. J. Phys. Chem. 2014, 118, 8134–8142. [Google Scholar] [CrossRef]
  95. Bailes, J.; Gazi, S.; Ivanova, R.; Soloviev, M. Chapter 7 Effect: Effect of Gold Nanoparticle Conjugation on the Activity and Stability of Functional Proteins. In Nanoparticles in Biology and Medicine: Methods and Protocols, Methods in Molecular Biology; Springer: Berlin/Heidelberg, Germany, 2012; pp. 89–99. [Google Scholar] [CrossRef]
  96. Zhicong, M.; Gao, Z.; Chen, R.; Angeles, L. Surface-bioengineered Gold Nanoparticles for Biomedical Applications Surface-bioengineered Gold Nanoparticles for Biomedical Applications. Curr. Med. Chem. 2018, 25, 1920–1944. [Google Scholar] [CrossRef]
  97. Zhang, J.; Jiang, X. Surface chemistry of gold nanoparticles for health-related. Chem. Sci. 2020, 11, 923–936. [Google Scholar] [CrossRef] [PubMed]
  98. Luther, D.C.; Huang, R.; Jeon, T.; Zhang, X.; Lee, W.; Nagaraj, H.; Rotello, V.M. Delivery of Drugs, Proteins, and Nucleic Acids using Inorganic Nanoparticles David. Adv. Drug Deliv. Rev. 2020, 156, 188–213. [Google Scholar] [CrossRef] [PubMed]
  99. Lacerda, S.H.D.P.; Park, J.J.; Meuse, C.; Pristinski, D.; Becker, M.L.; Karim, A.; Douglas, J.F. Interaction of Gold Nanoparticles with Common Human Blood Proteins. ACS Nano 2010, 4, 365–379. [Google Scholar] [CrossRef] [PubMed]
  100. Deng, Z.J.; Liang, M.; Monteiro, M.; Toth, I.; Minchin, R.F. Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation. Nat. Nanotechnol. 2011, 6, 4–9. [Google Scholar] [CrossRef]
  101. Deng, Z.J.; Liang, M.; Toth, I.; Monteiro, M.J.; Minchin, R.F. Molecular Interaction of Poly (acrylic acid) Gold Nanoparticles with Human Fibrinogen. ACS Nano 2012, 6, 8962–8969. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, P.; Wang, X.; Wang, L.; Hou, X. Interaction of gold nanoparticles with proteins and cells. Sci. Technol. Adv. Mater. 2015, 16, 034610. [Google Scholar] [CrossRef]
  103. Hu, C.M.J.; Fang, R.H.; Wang, K.C.; Luk, B.T.; Thamphiwatana, S.; Dehaini, D.; Nguyen, P.; Angsantikul, P.; Wen, C.H.; Kroll, A.V.; et al. Nanoparticle biointerfacing by platelet membrane cloaking. Nature 2015, 526, 118–121. [Google Scholar] [CrossRef] [PubMed]
  104. Fischer, H.C.; Chan, W.C.W. Nanotoxicity: The growing need for in vivo study. Pharm. Biotechnol. 2007, 18, 565–571. [Google Scholar] [CrossRef]
  105. Di Nardo, F.; Baggiani, C.; Giovannoli, C.; Spano, G.; Anfossi, L. Multicolor immunochromatographic strip test based on gold nanoparticles for the determination of aflatoxin B1 and fumonisins. Microchim Acta 2017, 184, 1295–1304. [Google Scholar] [CrossRef]
  106. Taghizadeh, S.; Alimardani, V.; Roudbali, P.L.; Ghasemi, Y. Gold nanoparticles application in liver cancer. Photodiagnosis Photodyn. Ther. 2019, 25, 389–400. [Google Scholar] [CrossRef]
  107. Aioub, M.; Austin, L.A.; El-sayed, M.A. Chapter 2. Gold nanoparticles for cancer diagnostics, spectroscopic imaging, drug delivery, and plasmonic photothermal therapy. In Inorganic Frameworks as Smart Nanomedicines; Elsevier Inc.: Amsterdam, The Netherlands, 2018; pp. 41–92. [Google Scholar]
  108. Haume, K.; Rosa, S.; Grellet, S.; Śmiałek, M.A.; Butterworth, K.T.; Solov’yov, A.V.; Prise, K.M.; Golding, J.; Mason, N.J. Gold nanoparticles for cancer radiotherapy: A review. Cancer Nanotechnol. 2016, 7, 8. [Google Scholar] [CrossRef]
  109. Norouzi, M. Gold Nanoparticles in Glioma Theranostics. Pharmacol. Res. 2020, 156, 104753. [Google Scholar] [CrossRef]
  110. Ngernpimai, S.; Srijampa, S.; Thongmee, P.; Teerasong, S.; Puangmali, T.; Maleewong, W.; Chompoosor, A.; Tippayawat, P. Insight into the Covalently Oriented Immobilization of Antibodies on Gold Nanoparticle Probes to Improve Sensitivity in the Colorimetric Detection of Listeria monicytogenes. Bioconjug. Chem. 2022, 33, 2103–2112. [Google Scholar] [CrossRef]
  111. Mostafa, O.; Mohammad, H.; Salaheldin, T.A.; Ibrahim, A.K.; Elfeky, S.A. Fluorescein/gold nanoparticles conjugated EGFR antibody for imaging and P53 upregulation in hamster mucosal cells carcinoma. J. Drug Deliv. Sci. Technol. 2022, 76, 103293. [Google Scholar] [CrossRef]
  112. Martinez-liu, C.; Machain-williams, C.; Martinez-acuña, N.; Lozano-sepulveda, S.; Galan-huerta, K.; Arellanos-soto, D.; Mel, M.; Diana, Á.; Katya, P.; Galindo-rodr, S.; et al. Development of a Rapid Gold Nanoparticle-Based Lateral Flow Immunoassay for the Detection of Dengue Virus. Biosensors 2022, 12, 495. [Google Scholar] [CrossRef]
  113. Momenbeitollahi, N.; Van Der Zalm, J.; Chen, A.; Li, H. Entrapping gold nanoparticles in membranes for simple-to-use enhanced fl uorescence detection of proteins. Anal. Chim. Acta 2022, 1195, 339443. [Google Scholar] [CrossRef]
  114. Pyne, A.; Nandi, S.; Ghosh, M.; Roy, T.; Dhara, S.; Sarkar, N. Denaturant-Mediated Modulation of the Formation and Drug Encapsulation Responses of Gold Nanoparticles. Langmuir 2020, 36, 7634–7647. [Google Scholar] [CrossRef] [PubMed]
  115. Seo, J.M.; Kim, E.B.; Hyun, M.S.; Kim, B.B.; Park, T.J. Self-assembly of biogenic gold nanoparticles and their use to enhance drug delivery into cells. Colloids Surf. B Biointerfaces 2015, 135, 27–34. [Google Scholar] [CrossRef] [PubMed]
  116. Talib, A.; Khan, M.S.; Gedda, G.R.; Wu, H.F. Stabilization of gold nanoparticles using natural plant gel: A greener step towards biological applications. J. Mol. Liq. 2016, 220, 463–467. [Google Scholar] [CrossRef]
  117. Haynes, B.; Zhang, Y.; Liu, F.; Li, J.; Petit, S.; Kothayer, H.; Bao, X.; Westwell, A.D.; Mao, G.; Shekhar, M.P.V. Gold nanoparticle conjugated Rad6 inhibitor induces cell death in triple negative breast cancer cells by inducing mitochondrial dysfunction and PARP-1 hyperactivation: Synthesis and characterization. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 745–757. [Google Scholar] [CrossRef]
  118. Khutale, G.V.; Casey, A. Synthesis and characterization of a multifunctional gold-doxorubicin nanoparticle system for pH triggered intracellular anticancer drug release. Eur. J. Pharm. Biopharm. 2017, 119, 372–380. [Google Scholar] [CrossRef]
  119. Cho, J.H.; Kim, A.R.; Kim, S.H.; Lee, S.J.; Chung, H.; Yoon, M.Y. Development of a novel imaging agent using peptide-coated gold nanoparticles toward brain glioma stem cell marker CD133. Acta Biomater. 2017, 47, 182–192. [Google Scholar] [CrossRef]
  120. Colin, J.A.; Pech-Pech, I.E.; Oviedo, M.; Águila, S.A.; Romo-Herrera, J.M.; Contreras, O.E. Gold nanoparticles synthesis assisted by marine algae extract: Biomolecules shells from a green chemistry approach. Chem. Phys. Lett. 2018, 708, 210–215. [Google Scholar] [CrossRef]
  121. Seo, B.; Lim, K.; Kim, S.S.; Oh, K.T.; Lee, E.S.; Choi, H.G.; Shin, B.S.; Youn, Y.S. Small gold nanorods-loaded hybrid albumin nanoparticles with high photothermal efficacy for tumor ablation. Colloids Surf. B Biointerfaces 2019, 179, 340–351. [Google Scholar] [CrossRef] [PubMed]
  122. Nosrati, H.; Salehiabar, M.; Charmi, J.; Yaray, K.; Ghaffarlou, M.; Balcioglu, E.; Ertas, Y.N. Enhanced In Vivo Radiotherapy of Breast Cancer Using Gadolinium Oxide and Gold Hybrid Nanoparticles. Appl. Bio Mater. 2023, 6, 784–792. [Google Scholar] [CrossRef] [PubMed]
  123. González-López, M.A.; Gutiérrez-Cárdenas, E.M.; Sánchez-Cruz, C.; Hernández-Paz, J.F.; Pérez, I.; Olivares-Trejo, J.J.; Hernández-González, O. Reducing the effective dose of cisplatin using gold nanoparticles as carriers. Cancer Nanotechnol. 2020, 11, 4. [Google Scholar] [CrossRef]
  124. Lu, B.; Lu, F.; Ran, L.; Yu, K.; Xiao, Y.; Li, Z.; Dai, F.; Wu, D.; Lan, G. Self-assembly of natural protein and imidazole molecules on gold nanoparticles: Applications in wound healing against multi-drug resistant bacteria. Int. J. Biol. Macromol. 2018, 119, 505–516. [Google Scholar] [CrossRef] [PubMed]
  125. Yoon, H.Y.; Selvan, S.T.; Yang, Y.; Kim, M.J.; Yi, D.K.; Kwon, I.C.; Kim, K. Engineering nanoparticle strategies for effective cancer immunotherapy. Biomaterials 2018, 178, 597–607. [Google Scholar] [CrossRef] [PubMed]
  126. Jang, J.W.; Kim, H.; Kim, I.; Lee, S.W.; Jung, H.G.; Hwang, K.S.; Lee, J.H.; Lee, G.; Lee, D.; Yoon, D.S. Surface Functionalization of Enzyme-Coronated Gold Nanoparticles with an Erythrocyte Membrane for Highly Selective Glucose Assays. Anal. Chem. 2022, 94, 6473–6481. [Google Scholar] [CrossRef] [PubMed]
  127. Jafarzadeh, S.; Bargahi, N.; Bagherpour, S.; Soleymani, J. Concanavalin A-conjugated gold nanoparticle/silica quantum dot (AuNPs/SiQDs-Con A)-based platform as a fluorescent nanoprobe for the bioimaging of glycan-positive cancer cells. RSC Adv. 2022, 12, 8492–8501. [Google Scholar] [CrossRef] [PubMed]
  128. Poonaki, E.; Nickel, A.; Ardestani, M.S.; Rademacher, L.; Kaul, M.; Apartsin, E.; Meuth, S.G.; Gorji, A.; Janiak, C.; Kahlert, U.D. CD133-Functionalized Gold Nanoparticles as a Carrier Platform for Telaglenastat (CB-839) against Tumor Stem Cells. Int. J. Mol. Sci. 2022, 23, 5479. [Google Scholar] [CrossRef]
  129. Das, G.; Seo, S.; Yang, I.; Thi, L.; Nguyen, H.; Shin, H.; Patra, J.K.; Das, G.; Seo, S. Synthesis of Biogenic Gold Nanoparticles by Using Sericin Protein from Bombyx mori Silk Cocoon and Investigation of Its Wound Healing, Antioxidant, and Antibacterial Potentials. Int. J. Nanomed. 2023, 18, 17–34. [Google Scholar] [CrossRef]
  130. Aniu, S.; Allwin, Y.; Vinitha, T.; Balamurugan, K.; Dharuman, V. Streptavidin Fe2O3-gold nanoparticles functionalized theranostic liposome for antibiotic resistant bacteria and biotin sensing. Biosens. Bioelectron. 2023, 219, 114849. [Google Scholar] [CrossRef]
  131. Gupta, N.; Kumar, R.; Maitra, A.; Shrivastava, A. In-vitro and in-vivo efficacy of hollow gold nanoparticles encapsulating horseradish peroxidase: Oxidative stress-mediated tumor cell killing. J. Drug Deliv. Sci. Technol. 2023, 79, 103979. [Google Scholar] [CrossRef]
  132. Lach, M.; Strelow, C.; Meyer, A.; Mews, A.; Beck, T. Encapsulation of Gold Nanoparticles into Redesigned Ferritin Nanocages for the Assembly of Binary Superlattices Composed of Fluorophores and Gold Nanoparticles. Appl. Bio Mater. 2022, 14, 10656–10668. [Google Scholar] [CrossRef] [PubMed]
  133. Villalonga, A.; Sánchez, A.; Vilela, D.; Mayol, B.; Martínez, P. Electrochemical aptasensor based on anisotropically modified (Janus-type) gold nanoparticles for determination of C-reactive protein. Microchim. Acta 2022, 189, 309. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, Y.; Huang, X.; Zhu, H.; Lu, Y.; Song, H.; Niu, J.; Chen, H. Cancer cell membrane functionalized gold nanoparticles: Natural receptor Tenascin-C as biomimetic probe for sensitive detection of circulating exosomes. Sens. Actuators B. Chem. 2022, 372, 132673. [Google Scholar] [CrossRef]
  135. Memdi, I.; Nasrin, F.; Dutta, A.; Park, E.Y. Advancement of dengue virus NS1 protein detection by 3D-nanoassembly complex gold nanoparticles utilizing competitive sandwich aptamer on disposable electrode. Anal. Chim. Acta 2022, 1207, 339817. [Google Scholar] [CrossRef]
  136. Kim, J.; Yu, A.M.; Kubelick, K.P.; Emelianov, S.Y. Gold nanoparticles conjugated with DNA aptamer for photoacoustic detection of human matrix metalloproteinase-9. Photoacoustics 2022, 25, 100307. [Google Scholar] [CrossRef] [PubMed]
  137. Pal, S.; Koneru, J.K.; Andreou, C.; Rakshit, T.; Rajasekhar, V.K.; Wlodarczyk, M.; Healey, J.H.; Kircher, M.F.; Mondal, J. DNA-Functionalized Gold Nanorods for Perioperative Optical Imaging and Photothermal Therapy of Triple-Negative Breast Cancer. Appl. Nanomater. 2022, 5, 9159–9169. [Google Scholar] [CrossRef]
  138. Avila-Alejo, J.O.; González-Palomo, A.K.; Plascencia-Villa, G.; José-Yacamán, M.; Navarro-Contreras, H.R.; Pérez-Maldonado, I.N. Low cytotoxicity of anisotropic gold nanoparticles coated with lysine on peripheral blood mononuclear cells “in vitro”. Environ. Toxicol. Pharmacol. 2017, 56, 210–218. [Google Scholar] [CrossRef]
  139. Sachdeva, V.; Monga, A.; Vashisht, R.; Singh, D.; Singh, A. Iron Oxide Nanoparticles: The precise strategy for targeted delivery of genes, oligonucleotides and peptides in cancer therapy. J. Drug Deliv. Sci. Technol. 2022, 74, 103585. [Google Scholar] [CrossRef]
  140. Mudshinge, S.R.; Deore, A.B.; Patil, S.; Bhalgat, C.M. Nanoparticles: Emerging carriers for drug delivery. Saudi Pharm. J. 2011, 19, 129–141. [Google Scholar] [CrossRef]
  141. Xu, X.; Liu, Y.; Yang, Y.; Wu, J.; Cao, M.; Sun, L. One-pot synthesis of functional peptide-modified gold nanoparticles for gene delivery. Colloids Surf. A Physicochem. Eng. Asp. 2022, 640, 128491. [Google Scholar] [CrossRef]
  142. Chakraborty, A.; Boer, J.C.; Selomulya, C.; Plebanski, M. Amino Acid Functionalized Inorganic Nanoparticles as Cutting-Edge Therapeutic and Diagnostic Agents. Bioconjug. Chem. 2018, 29, 657–671. [Google Scholar] [CrossRef]
  143. Rodriguez, A.F.R.; dos Santos, C.C.; Lüdtke-Buzug, K.; Bakenecker, A.C.; Chaves, Y.O.; Mariúba, L.A.M.; Brandt, J.V.; Amantea, B.E.; de Santana, R.C.; Marques, R.F.C.; et al. Evaluation of antiplasmodial activity and cytotoxicity assays of amino acids functionalized magnetite nanoparticles: Hyperthermia and flow cytometry applications. Mater. Sci. Eng. C 2021, 125, 112097. [Google Scholar] [CrossRef]
  144. Alvarez, P.E.; Fiori-Bimbi, M.V.; Neske, A.; Brandán, S.A.; Gervasi, C.A. Rollinia occidentalis extract as green corrosion inhibitor for carbon steel in HCl solution. J. Ind. Eng. Chem. 2018, 58, 92–99. [Google Scholar] [CrossRef]
  145. Yang, H.; Kozicky, L.; Saferali, A.; Fung, S.Y.; Afacan, N.; Cai, B.; Falsafi, R.; Gill, E.; Liu, M.; Kollmann, T.R.; et al. Endosomal pH modulation by peptide-gold nanoparticle hybrids enables potent anti-inflammatory activity in phagocytic immune cells. Biomaterials 2016, 111, 90–102. [Google Scholar] [CrossRef]
  146. Gessner, I.; Neundorf, I. Nanoparticles modified with cell-penetrating peptides: Conjugation mechanisms, physicochemical properties, and application in cancer diagnosis and therapy. Int. J. Mol. Sci. 2020, 21, 2536. [Google Scholar] [CrossRef]
  147. Li, Y.X.; Pang, H.B. Macropinocytosis as a cell entry route for peptide-functionalized and bystander nanoparticles. J. Control. Release 2021, 329, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  148. Copolovici, D.M.; Langel, K.; Eriste, E. Cell-Penetrating Peptides: Design, Synthesis, and Applications. ACS Nano 2014, 8, 1972–1994. [Google Scholar] [CrossRef] [PubMed]
  149. Guidotti, G.; Brambilla, L.; Rossi, D. Cell-Penetrating Peptides: From Basic Research to Clinics. Trends Pharmacol. Sci. 2017, 38, 406–424. [Google Scholar] [CrossRef] [PubMed]
  150. Cai, H.; Yao, P. Gold nanoparticles with different amino acid surfaces: Serum albumin adsorption, intracellular uptake and cytotoxicity. Colloids Surf. B Biointerfaces 2014, 123, 900–906. [Google Scholar] [CrossRef] [PubMed]
  151. Tiwari, P.M.; Eroglu, E.; Bawage, S.S.; Vig, K.; Miller, M.E.; Pillai, S.; Dennis, V.A.; Singh, S.R. Enhanced intracellular translocation and biodistribution of gold nanoparticles functionalized with a cell-penetrating peptide (VG-21) from vesicular stomatitis virus. Biomaterials 2014, 35, 9484–9494. [Google Scholar] [CrossRef]
  152. Brann, T.; Patel, D.; Chauhan, R.; James, K.T.; Bates, P.J.; Malik, M.T.; Keynton, R.S.; O’Toole, M.G. Gold Nanoplates as Cancer-Targeted Photothermal Actuators for Drug Delivery and Triggered Release. J. Nanomater. 2016, 2016, 2036029. [Google Scholar] [CrossRef]
  153. Pérez-Ortiz, M.; Zapata-Urzúa, C.; Acosta, G.A.; Álvarez-Lueje, A.; Albericio, F.; Kogan, M.J. Gold nanoparticles as an efficient drug delivery system for GLP-1 peptides. Colloids Surf. B Biointerfaces 2017, 158, 25–32. [Google Scholar] [CrossRef]
  154. Tan, H.; Huang, Y.; Xu, J.; Chen, B.; Zhang, P.; Ye, Z.; Liang, S.; Xiao, L.; Liu, Z. Spider toxin peptide lycosin-I functionalized gold nanoparticles for in vivo tumor targeting and therapy. Theranostics 2017, 7, 3168–3178. [Google Scholar] [CrossRef] [PubMed]
  155. de Alteriis, E.; Falanga, A.; Galdiero, S.; Guida, M.; Maselli, V.; Galdiero, E. Genotoxicity of gold nanoparticles functionalized with indolicidin towards Saccharomyces cerevisiae. J. Environ. Sci. 2018, 66, 138–145. [Google Scholar] [CrossRef] [PubMed]
  156. Wu, L.; Lin, B.; Yang, H.; Chen, J.; Mao, Z.; Wang, W.; Gao, C. Enzyme-responsive multifunctional peptide coating of gold nanorods improves tumor targeting and photothermal therapy efficacy. Acta Biomater. 2019, 86, 363–372. [Google Scholar] [CrossRef] [PubMed]
  157. Naapuri, M.J.; Losada, G.N.; Deska, J.; Palomo, M.J. Synthesis of silver and gold nanoparticles–enzyme–polymer conjugate hybrids as dual-activity catalysts for chemoenzymatic cascade reactions. Nanoscale 2022, 14, 5701–5715. [Google Scholar] [CrossRef] [PubMed]
  158. Wei, B.; Zhu, W.; Li, K.; Liu, Q.; Zhang, J.; Kou, H.; Xu, C. Natural collagen peptides-encapsulated gold nanoclusters for the simultaneous detection of multiple antibiotics in milk and molecular logic operations. LWT 2022, 153, 112416. [Google Scholar] [CrossRef]
  159. Liu, M.; Lai, W.; Chen, M.; Wang, P.; Liu, J.; Fang, X.; Yang, Y.; Wang, C. Prominent enhancement of peptide-mediated targeting efficiency for human hepatocellular carcinomas with composition-engineered protein corona on gold nanoparticles. Colloids Surf. A Physicochem. Eng. Asp. 2023, 662, 131016. [Google Scholar] [CrossRef]
  160. Hou, J.; Qian, X.; Xu, Y.; Guo, Z.; Thierry, B.; Yang, C.; Zhou, X.; Mao, C. Rapid and reliable ultrasensitive detection of pathogenic H9N2 viruses through virus-binding phage nanofibers decorated with gold nanoparticles. Biosens. Bioelectron. 2023, 237, 115423. [Google Scholar] [CrossRef]
  161. Shelar, S.B.; Barick, K.C.; Dutta, B.; Basu, M.; Hassan, P.A. Selective targeting of gold nanoparticles for radiosensitization of somatostatin 2 receptor-expressing cancer cells. J. Drug Deliv. Sci. Technol. 2023, 82, 104381. [Google Scholar] [CrossRef]
  162. Griveau, A.; Arib, C.; Spadavecchia, J.; Eyer, J. Biological activity of gold nanoparticles combined with the NFL-TBS. 40-63 peptide, or with other cell penetrating peptides, on rat glioblastoma cells. Int. J. Pharm. X 2022, 4, 100129. [Google Scholar] [CrossRef] [PubMed]
  163. Dizman, H.M.; Arsu, N. Rapid and sensitive colorimetric determination of dopamine and serotonin in solution and polymer matrix with photochemically prepared and N -acetyl-L-cysteine functionalized gold nanoparticles. Mater. Today Commun. 2023, 35, 105599. [Google Scholar] [CrossRef]
  164. Liu, B.; Liu, J. Interface-Driven Hybrid Materials Based on DNA-Functionalized Gold Nanoparticles. Matter 2019, 1, 825–847. [Google Scholar] [CrossRef]
  165. Chuong, T.T.; Pallaoro, A.; Chaves, C.A.; Li, Z.; Lee, J.; Eisenstein, M.; Stucky, G.D. Dual-reporter SERS-based biomolecular assay with reduced false-positive signals. Proc. Natl. Acad. Sci. USA 2017, 114, 9056–9061. [Google Scholar] [CrossRef] [PubMed]
  166. Inkpen, M.S.; Liu, Z.F.; Li, H.; Campos, L.M.; Neaton, J.B.; Venkataraman, L. Non-chemisorbed gold–sulfur binding prevails in self-assembled monolayers. Nat. Chem. 2019, 11, 351–358. [Google Scholar] [CrossRef]
  167. Delong, R.K.; Reynolds, C.M.; Malcolm, Y.; Schaeffer, A.; Severs, T.; Wanekaya, A. Functionalized gold nanoparticles for the binding, stabilization, and delivery of therapeutic DNA, RNA, and other biological macromolecules. Nanotechnol. Sci. Appl. 2010, 3, 53–63. [Google Scholar] [CrossRef]
  168. Ahmed, M.; Deng, Z.; Narain, R. Study of Transfection Efficiencies of Cationic Glyconanoparticles of Different Sizes in human cell line. ACS Appl. Mater. Interfaces 2009, 1, 1980–1987. [Google Scholar] [CrossRef]
  169. Akamatsu, K.; Kimura, M.; Shibata, Y.; Nakano, S. A DNA Duplex with Extremely Enhanced Thermal Stability Based on Controlled Immobilization on Gold Nanoparticles. NANO Lett. 2006, 6, 491–495. [Google Scholar] [CrossRef]
  170. Giljohann, D.A.; Seferos, D.S.; Patel, P.C.; Millstone, J.E.; Rosi, N.L.; Mirkin, C.A. Oligonucleotide Loading Determines Cellular Uptake of DNA-Modified Gold Nanoparticles. NANO Lett. 2007, 7, 3818–3821. [Google Scholar] [CrossRef]
  171. Brown, K.A.; Park, S.; Hamad-schifferli, K. Nucleotide-Surface Interactions in DNA-Modified Au-Nanoparticle Conjugates: Sequence Effects on Reactivity and Hybridization. J. Phys. Chem. Lett. 2008, 112, 7517–7521. [Google Scholar] [CrossRef]
  172. Zeng, Z.; Zhang, B.; Li, C.; Peng, X.; Liu, X.; Meng, S.; Feng, Y. A key point of porphyrin structure affect DSSCs performance based on porphyrin sensitizers. Dye. Pigment. 2014, 100, 278–285. [Google Scholar] [CrossRef]
  173. Debnath, M.; Fatma, K.; Dash, J. Chemical Regulation of DNA i-Motifs for Nanobiotechnology and Therapeutics Angewandte. Angew. Chemie-Int. Ed. 2019, 58, 2942–2957. [Google Scholar] [CrossRef] [PubMed]
  174. Chandrasekaran, A.R.; Rusling, D.A. Triplex-forming oligonucleotides: A third strand for DNA nanotechnology. Nucleic Acids Res. 2018, 46, 1021–1037. [Google Scholar] [CrossRef]
  175. Granot, Y.; Peer, D. Delivering the right message: Challenges and opportunities in lipid nanoparticles-mediated modi fi ed mRNA therapeutics—An innate immune system standpoint. Semin. Immunol. 2017, 34, 68–77. [Google Scholar] [CrossRef]
  176. Li, B.; Zhang, X.; Dong, Y. Nanoscale platforms for messenger RNA delivery. Nanomed. Nanobiotechnol. 2019, 11, e1530. [Google Scholar] [CrossRef]
  177. Kaczmarek, J.C.; Kowalski, P.S.; Anderson, D.G. Advances in the delivery of RNA therapeutics: From concept to clinical reality. Genome Med. 2017, 9, 60. [Google Scholar] [CrossRef] [PubMed]
  178. Labala, S.; Jose, A.; Chawla, S.R.; Khan, M.S.; Bhatnagar, S.; Kulkarni, O.P.; Venuganti, V.V.K. Effective melanoma cancer suppression by iontophoretic co-delivery of STAT3 siRNA and imatinib using gold nanoparticles. Int. J. Pharm. 2017, 525, 407–417. [Google Scholar] [CrossRef] [PubMed]
  179. Koenig, O.; Nothdurft, D.; Perle, N.; Neumann, B.; Behring, A.; Degenkolbe, I.; Walker, T.; Schlensak, C.; Wendel, H.P.; Nolte, A. An Atelocollagen Coating for Efficient Local Gene Silencing by Using Small Interfering RNA. Mol. Ther. Nucleic Acid 2017, 6, 290–301. [Google Scholar] [CrossRef]
  180. Dalpke, A.H.; Helm, M.; Dalpke, A.H.; Helm, M. Health and disease RNA mediated toll-like receptor stimulation in health and disease. RNA Biol. 2012, 6286, 828–842. [Google Scholar] [CrossRef]
  181. Sheikh, A.; Alhakamy, N.A.; Kesharwani, P. Recent development of aptamer conjugated chitosan nanoparticles as cancer therapeutics. Int. J. Pharm. 2022, 620, 121751. [Google Scholar] [CrossRef] [PubMed]
  182. Cadinoiu, A.N.; Rata, D.M.; Atanase, L.I.; Daraba, O.M.; Gherghel, D.; Vochita, G.; Popa, M. Aptamer-Functionalized Liposomes as a Potential Treatment for Basal Cell Carcinoma. Polymers 2019, 11, 1515. [Google Scholar] [CrossRef] [PubMed]
  183. Gao, G.; Liu, C.; Jain, S.; Li, D.; Wang, H.; Zhao, Y.; Liu, J. Potential Use of Aptamers for Diagnosis and Treatment of Pancreatic Cancer. J. Drug Target. 2018, 2330, 1564924. [Google Scholar] [CrossRef] [PubMed]
  184. Varshney, A.; Bala, J.; Santosh, B.; Bhaskar, A. Identification of an RNA aptamer binding hTERT-derived peptide and inhibiting telomerase activity in MCF7 cells. Mol. Cell. Biochem. 2017, 427, 157–167. [Google Scholar] [CrossRef]
  185. Dehghani, S.; Alibolandi, M.; Amiri, Z.; Kazemi, R.; Nosrati, R.; Soltani, F.; Ramezani, M. Self-assembly of an aptamer-decorated chimeric peptide nanocarrier for targeted cancer gene delivery. Colloids Surf. B Biointerfaces 2021, 208, 112047. [Google Scholar] [CrossRef] [PubMed]
  186. Labala, S.; Jose, A.; Venuganti, V.V.K. Transcutaneous iontophoretic delivery of STAT3 siRNA using layer-by-layer chitosan coated gold nanoparticles to treat melanoma. Colloids Surf. B Biointerfaces 2016, 146, 188–197. [Google Scholar] [CrossRef]
  187. Mu, B.; Wang, R.; Gao, J.; Li, Z.; Li, X. Nano gold incorporated into Aerva javanica chitosan hydrogels disrupting agents against infections of burn wound. Mater. Technol. 2021, 36, 783–792. [Google Scholar] [CrossRef]
  188. Thomas, R.K.; Sukumaran, S.; Prasanth, S.; Sudarsanakumar, C. Revealing the interaction strategy of Diosmin functionalized gold nanoparticles with ctDNA: Multi-spectroscopic, calorimetric and thermodynamic approach. J. Lumin. 2019, 205, 265–276. [Google Scholar] [CrossRef]
  189. Ruttala, H.B.; Ramasamy, T.; Poudel, B.K.; Ruttala, R.R.T.; Jin, S.G.; Choi, H.G.; Ku, S.K.; Yong, C.S.; Kim, J.O. Multi-responsive albumin-lonidamine conjugated hybridized gold nanoparticle as a combined photothermal-chemotherapy for synergistic tumor ablation. Acta Biomater. 2020, 101, 531–543. [Google Scholar] [CrossRef]
  190. Dester, E.; Kao, K.; Alocilja, E.C. Detection of Unamplified E. coli O157 DNA Extracted from Large Food Samples Using a Gold Nanoparticle Colorimetric Biosensor. Biosensors 2022, 12, 274. [Google Scholar] [CrossRef] [PubMed]
  191. Chaudhari, R.; Nasra, S.; Meghani, N.; Kumar, A. MiR-206 conjugated gold nanoparticle based targeted therapy in breast cancer cells. Sci. Rep. 2022, 12, 4713. [Google Scholar] [CrossRef]
  192. Xu, R.; Cheng, Y.; Qi, X.; Li, X.; Zhang, Z.; Chen, L.; Sun, T.; Gao, Z.; Zhu, M. Target-induced gold nanoparticles colorimetric sensing coupled with aptamer for rapid and high-sensitivity detecting kanamycin. Anal. Chim. Acta 2022, 1230, 340377. [Google Scholar] [CrossRef]
  193. Liu, C.; Gou, S.; Bi, Y.; Gao, Q.; Sun, J.; Hu, S.; Guo, W. Smart DNA-gold nanoparticle hybrid hydrogel film based portable, cost-effective and storable biosensing system for the colorimetric detection of lead (II) and uranyl ions. Biosens. Bioelectron. 2022, 210, 114290. [Google Scholar] [CrossRef]
  194. Li, X.; Xu, R.; Shi, L.; Ni, T. Design of flavonol-loaded cationic gold nanoparticles with enhanced antioxidant and antibacterial activities and their interaction with proteins. Int. J. Biol. Macromol. 2023, 253, 127074. [Google Scholar] [CrossRef] [PubMed]
  195. Khorshid, M.; Varshosaz, J.; Rostami, M.; Haghiralsadat, F. Anti HER-2 aptamer functionalized gold nanoparticles of dasatinib for targeted chemo-radiotherapy in breast cancer cells. Biomater. Adv. 2023, 154, 213591. [Google Scholar] [CrossRef] [PubMed]
  196. Kumawat, M.; Madhyastha, H.; Umapathi, A.; Singh, M.; Revaprasadu, N.; Daima, H.K. Surface Engineered Peroxidase-Mimicking Gold Nanoparticles to Subside Cell Inflammation. Langmuir 2022, 38, 1877–1887. [Google Scholar] [CrossRef]
  197. Yu, N.; Ma, G.; Chen, Y.; Huang, S.; Gong, Y.; Li, S.; Gu, H.; You, H.; Miao, P. MnO 2 nanosheets and gold nanoparticles supported electrochemical detection of circulating tumor cells. Colloids Surf. B Biointerfaces 2023, 229, 113482. [Google Scholar] [CrossRef] [PubMed]
  198. Ndugire, W.; Liyanage, S.H.; Yan, M. Carbohydrate-Presenting Metal Nanoparticles: Synthesis, Characterization and Applications, 2nd ed.; Elsevier B.V.: Amsterdam, The Netherlands, 2021. [Google Scholar] [CrossRef]
  199. Chen, X.; Zhao, X.; Wang, G. Review on marine carbohydrate-based gold nanoparticles represented by alginate and chitosan for biomedical application. Carbohydr. Polym. 2020, 244, 116311. [Google Scholar] [CrossRef] [PubMed]
  200. Martins, A.F.; Facchi, S.P.; Monteiro, J.P.; Nocchi, S.R.; Silva, C.T.P.; Nakamura, C.V.; Girotto, E.M.; Rubira, A.F.; Muniz, E.C. Preparation and cytotoxicity of N, N, N -trimethyl chitosan/alginate beads containing gold nanoparticles. Int. J. Biol. Macromol. 2015, 72, 466–471. [Google Scholar] [CrossRef]
  201. Zahir-Jouzdani, F.; Mahbod, M.; Soleimani, M.; Vakhshiteh, F. Chitosan and thiolated chitosan: Novel therapeutic approach for preventing corneal haze after chemical injuries. Carbohydr. Polym. J. 2018, 179, 42–49. [Google Scholar] [CrossRef] [PubMed]
  202. Yadav, P.; Prakash, S.; Kumar, A.; Shanavas, A.; Srivastava, R. Gold laced bio-macromolecules for theranostic application. Int. J. Biol. Macromol. 2018, 110, 39–53. [Google Scholar] [CrossRef]
  203. Santos-Fidencio, G.C.; Gonçalves, A.G.; Noseda, M.D.; Eugênia, M.; Duarte, R.; Ducatti, D.R.B.; De Bioquímica, D.; Federal, U.; Politécnico, C.; Box, P.O. Effects of carboxyl group on the anticoagulant activity of oxidized carrageenans. Carbohydr. Polym. 2019, 214, 286–293. [Google Scholar] [CrossRef]
  204. Manivasagan, P.; Oh, J. Marine polysaccharide-based nanomaterials as a novel source of nanobiotechnological applications. Int. J. Biol. Macromol. 2016, 82, 315–327. [Google Scholar] [CrossRef]
  205. Qiu, S.; Granet, R.; Mbakidi, J.P.; Brégier, F.; Pouget, C.; Micallef, L.; Sothea-Ouk, T.; Leger, D.Y.; Liagre, B.; Chaleix, V.; et al. Delivery of tanshinone IIA and α-mangostin from gold/PEI/cyclodextrin nanoparticle platform designed for prostate cancer chemotherapy. Bioorg. Med. Chem. Lett. 2016, 26, 2503–2506. [Google Scholar] [CrossRef] [PubMed]
  206. Dichello, G.A.; Fukuda, T.; Maekawa, T.; Whitby, R.L.D.; Mikhalovsky, S.V.; Alavijeh, M.; Pannala, A.S.; Sarker, D.K. Preparation of liposomes containing small gold nanoparticles using electrostatic interactions. Eur. J. Pharm. Sci. 2017, 105, 55–63. [Google Scholar] [CrossRef] [PubMed]
  207. Van Deun, J.; Roux, Q.; Deville, S.; Van Acker, T.; Rappu, P.; Miinalainen, I.; Heino, J.; Vanhaecke, F.; De Geest, B.G.; De Wever, O.; et al. Feasibility of Mechanical Extrusion to Coat Nanoparticles with Extracellular Vesicle Membranes. Cells 2020, 9, 1797. [Google Scholar] [CrossRef] [PubMed]
  208. Charest, G.; Tippayamontri, T.; Shi, M.; Wehbe, M.; Anantha, M.; Bally, M.; Sanche, L. Concomitant chemoradiation therapy with gold nanoparticles and platinum drugs co-encapsulated in liposomes. Int. J. Mol. Sci. 2020, 21, 4848. [Google Scholar] [CrossRef]
  209. Borran, A.A.; Aghanejad, A.; Farajollahi, A.; Barar, J.; Omidi, Y. Gold nanoparticles for radiosensitizing and imaging of cancer cells. Radiat. Phys. Chem. 2018, 152, 137–144. [Google Scholar] [CrossRef]
  210. Gomez-Caballero, L.F.; Pichardo-Molina, J.L.; Basurto-Islas, G. Cellulose dialysis membrane tubing doped with gold nanoparticles as SERS substrate. Mater. Lett. 2022, 313, 131718. [Google Scholar] [CrossRef]
  211. Cardellini, J.; Caselli, L.; Lavagna, E.; Salassi, S.; Amenitsch, H.; Calamai, M.; Montis, C.; Rossi, G.; Berti, D. Membrane Phase Drives the Assembly of Gold Nanoparticles on Biomimetic Lipid Bilayers. J. Phys. Chem. 2022, 126, 4483–4494. [Google Scholar] [CrossRef] [PubMed]
  212. Lv, R.; Qian, Z.; Zhao, X.; Xiong, F.; Xu, Y.; Fan, W.; Yao, X.; Huang, W. Self-assembly of polysarcosine amphiphilic polymers-tethered gold nanoparticles for precise photo-controlled syneristic therapy. Nano Res. 2023, 16, 5685–5694. [Google Scholar] [CrossRef]
  213. Sarker, S.R.; Polash, S.A.; Karim, N.; Saha, T.; Dekiwadia, C.; Bansal, V.; Sabri, Y.; Kandjani, A.E.; Bhargava, S.K. Functionalized Concave Cube Gold Nanoparticles as Potent Antimicrobial Agents against Pathogenic Bacteria. Appl. Bio Mater. 2022, 5, 492–503. [Google Scholar] [CrossRef]
  214. Huo, Y.; Song, X.; Zhang, W.; Zhou, Z.; Lv, Q.; Cui, H. Thermosensitive Biomimetic Hybrid Membrane Camouflaged Hollow Gold Nanoparticles for NIR-Responsive Mild-Hyperthermia Chemo-/Photothermal Combined Tumor Therapy. Appl. Bio Mater. 2022, 5, 5113–5125. [Google Scholar] [CrossRef]
  215. Asadullah, S.; Caliskan-aydogan, O.; Alocilja, E. Carbohydrate-coated magnetic and gold nanoparticles for point-of-use food contamination testing. Biosens. Bioelectron. X 2023, 13, 100322. [Google Scholar] [CrossRef]
  216. Gellini, C.; Feis, A. Thioglucose functionalized gold nanoparticles as active substrates for surface enhanced Raman spectroscopy of lectins. Vib. Spectrosc. 2022, 123, 103468. [Google Scholar] [CrossRef]
  217. Ayyanaar, S.; Palsamy, M. One-pot biogenic synthesis of gold nanoparticles @ saponins niosomes: Sustainable nanomedicine for antibacterial, anti-inflammatory and anticancer therapeutics. Colloids Surf. A Physicochem. Eng. Asp. 2023, 676, 132229. [Google Scholar] [CrossRef]
  218. Doghish, A.S.; Hashem, A.H.; Shehabeldine, A.M.; Sallam, A.M.; El-sayyad, G.S.; Salem, S.S. Nanocomposite based on gold nanoparticles and carboxymethyl cellulose: Synthesis, characterization, antimicrobial, and anticancer activities. J. Drug Deliv. Sci. Technol. 2022, 77, 103874. [Google Scholar] [CrossRef]
  219. Karimzadeh, Z.; Jouyban, A.; Khoubnasabjafari, M.; Gharakhani, A. Utilizing Sucrose-Functionalized Gold Nanoparticles for Daclatasvir: Chemometric Optimization and Determination. Plasmonics 2022, 17, 1999–2008. [Google Scholar] [CrossRef]
  220. Huang, W.; Wang, L.; Zou, Y.; Ding, X.; Li, J.; Zhao, H.; Qi, R.; Li, S.; Geng, X. Preparation of gastrodin-modified dendrimer-entrapped gold nanoparticles as a drug delivery system for cerebral ischemia–reperfusion injury. Brain Behav. 2022, 12, e2810. [Google Scholar] [CrossRef] [PubMed]
  221. Pinilla-Torres, A.M.; Sanchez-Dominguez, C.N.; Basilio-Bernabe, K.; Carrion-Garcia, P.Y.; Roacho-Perez, J.A.; Garza-Treviño, E.N.; Gallardo-Blanco, H.; Sanchez-Dominguez, M. Green Synthesis of Mesquite-Gum-Stabilized Gold Nanoparticles for Biomedical Applications: Physicochemical Properties and Biocompatibility Assessment. Polymers 2023, 15, 3533. [Google Scholar] [CrossRef] [PubMed]
  222. Zhou, S.; Li, X.; Zhu, M.; Yu, H.; Chu, R.; Chen, W.; Wang, B.; Wang, M.; Zheng, L.; Chai, Z.; et al. Hepatic impacts of gold nanoparticles with different surface coatings as revealed by assessing the hepatic drug-metabolizing enzyme and lipid homeostasis in mice. NanoImpact 2020, 20, 100259. [Google Scholar] [CrossRef]
  223. Priyadarshini, E.; Pradhan, N. Chemical Gold nanoparticles as efficient sensors in colorimetric detection of toxic metal ions: A review. Sens. Actuators B. Chem. 2017, 238, 888–902. [Google Scholar] [CrossRef]
  224. Wang, X.; Wu, J.; Liang, F.; Yang, Y. In Situ Gold Nanoparticle Synthesis Mediated by a Water-Soluble Leaning Pillar[6]arene for Self-Assembly, Detection, and Catalysis. Org. Lett. 2019, 21, 9–12. [Google Scholar] [CrossRef]
  225. Oni, Y.; Hao, K.; Dozie-Nwachukwu, S.; Obayemi, J.D.; Odusanya, O.S.; Anuku, N.; Soboyejo, W.O. Gold nanoparticles for cancer detection and treatment: The role of adhesion. J. Appl. Phys. 2014, 115, 084305. [Google Scholar] [CrossRef]
  226. Marcelo, G.; Kaplan, E.; Tarazona, M.P.; Mendicuti, F. Interaction of gold nanoparticles with Doxorubicin mediated by supramolecular chemistry. Colloids Surf. B Biointerfaces 2015, 128, 237–244. [Google Scholar] [CrossRef] [PubMed]
  227. Licciardi, M.; Li Volsi, A.; Mauro, N.; Scialabba, C.; Cavallaro, G.; Giammona, G. Preparation and Characterization of Inulin Coated Gold Nanoparticles for Selective Delivery of Doxorubicin to Breast Cancer Cells. J. Nanomater. 2016, 2016, 2078315. [Google Scholar] [CrossRef]
  228. Fan, J.; Chen, S.; Gao, Y. Coating gold nanoparticles with peptide molecules v ia a peptide elongation approach. Colloids Surf. B 2003, 28, 199–207. [Google Scholar] [CrossRef]
  229. Hosseinzadeh, H.; Atyabi, F.; Varnamkhasti, B.S.; Hosseinzadeh, R.; Ostad, S.N.; Ghahremani, M.H.; Dinarvand, R. SN38 conjugated hyaluronic acid gold nanoparticles as a novel system against metastatic colon cancer cells. Int. J. Pharm. 2017, 526, 339–352. [Google Scholar] [CrossRef]
  230. Stolarczyk, E.U.; Stolarczyk, K.; Łaszcz, M.; Kubiszewski, M.; Maruszak, W.; Olejarz, W.; Bryk, D. Synthesis and characterization of genistein conjugated with gold nanoparticles and the study of their cytotoxic properties. Eur. J. Pharm. Sci. 2017, 96, 176–185. [Google Scholar] [CrossRef]
  231. Bettini, S.; Ottolini, M.; Valli, D.; Pagano, R.; Ingrosso, C.; Roeffaers, M.; Hofkens, J.; Valli, L.; Giancane, G. Synthesis and Characterization of Gold Chiral Nanoparticles Functionalized by a Chiral Drug. Nanomaterials 2023, 13, 1526. [Google Scholar] [CrossRef]
  232. Bishop, C.J.; Tzeng, S.Y.; Green, J.J. Degradable Polymer-Coated Gold Nanoparticles for Co-Delivery of DNA and siRNA. Acta Biomater. 2014, 11, 393–403. [Google Scholar] [CrossRef]
  233. Mahalakshmi, M.; Kumar, P. Phloroglucinol-conjugated gold nanoparticles targeting mitochondrial membrane potential of human cervical (HeLa) cancer cell lines. Spectrochim. Acta-Part A Mol. Biomol. Spectrosc. 2019, 219, 450–456. [Google Scholar] [CrossRef] [PubMed]
  234. Kamalabadi, M.A.; Neshastehriz, A.; Ghaznavi, H.; Mohammad, S. Folate functionalized gold-coated magnetic nanoparticles effect in combined electroporation and radiation treatment of HPV-positive oropharyngeal cancer. Med. Oncol. 2022, 39, 196. [Google Scholar] [CrossRef]
  235. Wang, Y.; Wang, H.; Khan, M.S.; Husain, F.M.; Ahmad, S.; Bian, L. Bioconjugation of gold nanoparticles with aminoguanidine as a potential inhibitor of non-enzymatic glycation reaction. J. Biomol. Struct. Dyn. 2021, 39, 2014–2020. [Google Scholar] [CrossRef]
  236. Pędziwiatr-Werbicka, E.; Gorzkiewicz, M.; Michlewska, S.; Ionov, M.; Shcharbin, D.; Klajnert-Maculewicz, B.; Peña-González, C.E.; Sánchez-Nieves, J.; Gómez, R.; de la Mata, F.J.; et al. Evaluation of dendronized gold nanoparticles as siRNAs carriers into cancer cells. J. Mol. Liq. 2021, 324, 114726. [Google Scholar] [CrossRef]
  237. Diyana, N.; Ibrahim, N.; Yuziana, N.; Yusof, M.; Ta, C.; Ling, L. Optical reflectometric measurement of SARS-CoV-2 (COVID-19) RNA based on cationic cysteamine-capped gold nanoparticles. Opt. Laser Technol. 2023, 157, 108763. [Google Scholar] [CrossRef] [PubMed]
  238. Paraiso, W.K.D.; Tanaka, H.; Sato, Y.; Shirane, D.; Suzuki, N.; Ogra, Y.; Tange, K.; Nakai, Y.; Yoshioka, H.; Harashima, H.; et al. Preparation of envelope-type lipid nanoparticles containing gold nanorods for photothermal cancer therapy. Colloids Surf. B Biointerfaces 2017, 160, 715–723. [Google Scholar] [CrossRef]
  239. Alex, S.A.; Chandrasekaran, N.; Mukherjee, A. Impact of gold nanorod functionalization on biocorona formation and their biological implication. J. Mol. Liq. 2017, 248, 703–712. [Google Scholar] [CrossRef]
  240. Luchini, A.; D’Errico, G.; Leone, S.; Vaezi, Z.; Bortolotti, A.; Stella, L.; Vitiello, G.; Paduano, L. Structural organization of lipid-functionalized-Au nanoparticles. Colloids Surf. B Biointerfaces 2018, 168, 2–9. [Google Scholar] [CrossRef]
  241. Li, X.; Wang, H.; Zou, X.; Su, H.; Li, C. Methotrexate-loaded folic acid of solid-phase synthesis conjugated gold nanoparticles targeted treatment for rheumatoid arthritis. Eur. J. Pharm. Sci. 2022, 170, 106101. [Google Scholar] [CrossRef]
  242. Her, S.; Jaffray, D.A.; Allen, C. Gold nanoparticles for applications in cancer radiotherapy: Mechanisms and recent advancements. Adv. Drug Deliv. Rev. 2017, 109, 84–101. [Google Scholar] [CrossRef]
  243. Tonthat, L.; Kimura, M.; Ogawa, T.; Kitamura, N.; Kobayashi, Y.; Gonda, K.; Yabukami, S. Development of gold-coated magnetic nanoparticles as a theranostic agent for magnetic hyperthermia and CT imaging applications. AIP Adv. 2023, 13, 025239. [Google Scholar] [CrossRef]
  244. Falahati, M.; Attar, F.; Shari, M.; Akbar, A.; Salihi, A.; Mohammad, F.; Kostova, I.; Burda, C.; Priecel, P.; Lopez-sanchez, J.A.; et al. Gold nanomaterials as key suppliers in biological and chemical sensing, catalysis, and medicine. BBA-Gen. Subj. J. 2019, 1864, 129435. [Google Scholar] [CrossRef] [PubMed]
  245. Ghorani-azam, A.; Mottaghipisheh, J.; Amiri, M.S.; Mashreghi, M.; Hashemzadeh, A.; Haddad-mashadrizeh, A.; Nourbakhsh, F.; Nadaf, M.; Qayoomian, M.; Ehsan, M.; et al. Resveratrol-Mediated Gold-Nanoceria Synthesis as Green Nanomedicine for Phytotherapy of Hepatocellular Carcinoma. Front. Biosci.-Landmark 2022, 27, 227. [Google Scholar] [CrossRef] [PubMed]
  246. Liang, Z.; Cao, C.; Gao, J.; Cai, W.; Li, J.; Wu, D.; Kong, Y. Gold Nanorods@Mesoporous SiO2@Hyaluronic Acid Core–Shell Nanoparticles for Controlled Drug Delivery. Appl. Bio Mater. 2022, 5, 7440–7448. [Google Scholar] [CrossRef]
  247. Raj, V.; Alex, S. Phosphotungstic Acid Capped Gold Nanoparticles for Spectrophotometric Determination of Uric Acid in Diagnostics of Gout. J. Anal. Chem. 2022, 77, 1267–1273. [Google Scholar] [CrossRef]
  248. Hwan, J.; Choe, P.H.; Im, S.K.G.; Ho, S.; Kim, U.J.; Ho, S. Silica-Capped and Gold-Decorated Silica Nanoparticles for Enhancing Effect of Gold Nanoparticle-Based Photothermal Therapy. Tissue Eng. Regen. Med. 2022, 19, 1161–1168. [Google Scholar] [CrossRef]
  249. Jaiswal, V.D.; Pangam, D.S.; Dongre, P.M. Biophysical study of cisplatin loaded albumin-gold nanoparticle and its interaction with glycans of gp60 receptor. Int. J. Biol. Macromol. 2023, 231, 123368. [Google Scholar] [CrossRef] [PubMed]
  250. Lorenzoni, S.; Cerra, S.; Angulo-elizari, E.; Salamone, T.A.; Battocchio, C.; Marsotto, M.; Scaramuzzo, F.A.; Sanmartín, C.; Plano, D.; Fratoddi, I. Organoselenium compounds as functionalizing agents for gold nanoparticles in cancer therapy. Colloids Surf. B Biointerfaces 2022, 219, 112828. [Google Scholar] [CrossRef]
  251. Nwude, F.; Chika, R.; Ovatlarnporn, C. Insights into the remarkable attenuation of hen egg white lysozyme amyloid fibril formation mediated by biogenic gold nanoparticles stabilized by quercetin-functionalized tara gum. Int. J. Biol. Macromol. 2023, 232, 123044. [Google Scholar] [CrossRef]
  252. Ding, Y.; Huang, P.J.; Zandieh, M.; Wang, J.; Liu, J. Gold Nanoparticles Synthesized Using Various Reducing Agents and the Effect of Aging for DNA Sensing. Langmuir 2023, 39, 256–264. [Google Scholar] [CrossRef]
  253. Bacteria, M.; Zhao, X.; Tang, H.; Jiang, X. Deploying Gold Nanomaterials in Combating Multi-Drug-Resistant Bacteria. ACS Nano 2022, 16, 10066–10087. [Google Scholar] [CrossRef]
  254. Kariuki, R.; Penman, R.; Bryant, S.J.; Orrell-trigg, R.; Meftahi, N.; Crawford, R.J.; Mcconville, C.F.; Bryant, G.; Vo, K.; Conn, C.E.; et al. Behavior of Citrate-Capped Ultrasmall Gold Nanoparticles on a Supported Lipid Bilayer Interface at Atomic Resolution. ACS Nano 2022, 16, 17179–17196. [Google Scholar] [CrossRef]
  255. Yadav, T.C.; Gupta, P.; Saini, S.; Mohiyuddin, S.; Pruthi, V.; Prasad, R. Plausible Mechanistic Insights in Bio fi lm Eradication Potential against Candida spp. Using In Situ-Synthesized Tyrosol-Functionalized Chitosan Gold Nanoparticles as a Versatile Antifouling Coating on Implant Surfaces. ACS Omega 2022, 7, 8350–8363. [Google Scholar] [CrossRef]
  256. Journal, A.I.; Khan, A.; Jabeen, H.; Ahmad, T.; Rehman, N.U.; Khan, S.; Shareef, H.; Sarwar, R.; Yahya, S.; Uddin, J.; et al. Comparative efficacy of cephradine-loaded silver and gold nanoparticles against resistant human pathogens. Artif. Cells Nanomed. Biotechnol. 2022, 50, 312–321. [Google Scholar] [CrossRef]
  257. Abbasgholi, B.; Asbaghi, N.; Alsadig, A.; Casalis, L.; Parisse, P.; Niemela, J.; Bellucci, S.; Cabrera, H. An electrophoresis approach with online thermal lens detection to monitoring DNA surface coatings on gold nanoparticles. Microchem. J. 2022, 173, 106961. [Google Scholar] [CrossRef]
  258. Blanck, C.; Schmutz, M. Materials Advances electro-cross-linking of tannic acid capped gold nanoparticles and enzymes. Mater. Adv. 2022, 3, 2222–2233. [Google Scholar] [CrossRef]
  259. Imanparast, A.; Attaran, N.; Eshghi, H.; Sazgarnia, A. Surface modification of gold nanoparticles with 6-mercapto-1-hexanol to facilitate dual conjugation of protoporphyrin IX and folic acid for improving the targeted photochemical internalization. Iran. J. Basic Med. Sci. 2022, 25, 970–979. [Google Scholar] [CrossRef] [PubMed]
  260. Mayuri, S.; Sweta, N.; Kumar, S. Curcumin-capped gold nanorods as optical sensing platform for sequence specific detection of DNA based on their self-assembly. Int. J. Biol. Macromol. 2023, 253, 126829. [Google Scholar] [CrossRef] [PubMed]
  261. Selim, A.; Lila, A.; Huwaimel, B.; Alobaida, A.; Hussain, T.; Rafi, Z.; Mehmood, K.; Abdallah, M.H.; Hagbani, T.A.; Mohd, S.; et al. Delafloxacin-Capped Gold Nanoparticles (DFX-AuNPs): An Effective Antibacterial Nano-Formulation of Fluoroquinolone Antibiotic. Materials 2022, 15, 5709. [Google Scholar] [CrossRef]
  262. Mohd, S.; Rizvi, D.; Hussain, T.; Alshammari, F.; Sonbol, H.; Ahmad, N. Nano-Conversion of Ineffective Cephalosporins into Potent One against Resistant Clinical Uro-Pathogens via Gold Nanoparticles. Nanomaterials 2023, 13, 475. [Google Scholar] [CrossRef]
  263. Ncobeni, N.; De Torre, B.G.; Albericio, F. Active targeting of CD4+ T lymphocytes by PEI-capped, peptide-functionalized gold nanoparticles. Nanotechnology 2022, 22, 405101. [Google Scholar] [CrossRef]
  264. Sharifiaghdam, Z.; Mohammad, S.; Dalouchi, F. Apigenin-coated gold nanoparticles as a cardioprotective strategy against doxorubicin-induced cardiotoxicity in male rats via reducing apoptosis. Heliyon 2023, 9, e14024. [Google Scholar] [CrossRef]
  265. Shabani, L.; Reza, S.; Shreeshivadasan, K.; Milad, C.; Hossein, A. An investigation into green synthesis of Ru template gold nanoparticles and the in vitro photothermal effect on the MCF-7 human breast cancer cell line. Appl. Phys. A 2023, 129, 564. [Google Scholar] [CrossRef]
  266. Chen, P.; Bian, L.; Hu, X. Synergic Fabrication of Gold Nanoparticles Embedded Dextran/Silk Sericin Nanomaterials for the Treatment and Care of Wound Healing. J. Clust. Sci. 2022, 33, 2147–2156. [Google Scholar] [CrossRef]
  267. Safdar, M.; Ozaslan, M. Enhanced Catalytic, Antibacterial and Anti-cancer Activities of Erythromycin Capped Gold Nanoparticles. J. Inorg. Organomet. Polym. Mater. 2022, 32, 1819–1827. [Google Scholar] [CrossRef]
  268. Faid, A.H.; Shouman, S.A.; Badr, Y.A.; Sharaky, M.; Mostafa, E.M. Gold nanoparticles loaded chitosan encapsulate 6-mercaptopurine as a novel nanocomposite for chemo-photothermal therapy on breast cancer. BMC Chem. 2022, 16, 94. [Google Scholar] [CrossRef] [PubMed]
  269. Mal, S.; Saha, T.; Halder, A.; Kumar, S.; Das, S.; Tin, W.; Chatterji, U.; Roy, P. EGF-conjugated bio-safe luteolin gold nanoparticles induce cellular toxicity and cell death mediated by site-specific rapid uptake in human triple negative breast cancer cells. J. Drug Deliv. Sci. Technol. 2023, 80, 104148. [Google Scholar] [CrossRef]
  270. Li, X.; Ni, T.; Xu, R. Study on the preparation of novel flavonol-gold nanoparticles antioxidants and the mechanism of protein corona formation. J. Mol. Struct. 2023, 1294, 136413. [Google Scholar] [CrossRef]
  271. Lalaoui, N.; Gentil, K.; Ghandari, I.; Cosnier, S.; Giroud, F. Nitrob enzoic acid-functionalize d gold nanoparticles: DET promoter of multicopper oxidases and electrocatalyst for NAD-dependent glucose dehydrogenase. Electrochim. Acta 2022, 408, 139894. [Google Scholar] [CrossRef]
  272. Salamone, T.A.; Rutigliano, L.; Pennacchi, B.; Cerra, S.; Matassa, R.; Nottola, S.; Sciubba, F.; Battocchio, C.; Marsotto, M.; Del, A.; et al. Thiol functionalised gold nanoparticles loaded with methotrexate for cancer treatment: From synthesis to in vitro studies on neuroblastoma cell lines. J. Colloid Interface Sci. 2023, 649, 264–278. [Google Scholar] [CrossRef]
  273. Wang, M.; Wang, X.; Liu, B.; Lang, C.; Wang, W.; Liu, Y.; Wang, X. Synthesis of Cipro fl oxacin-capped Gold Nanoparticles Conjugates with Enhanced Sonodynamic Antimicrobial Activity in vitro. J. Pharm. Sci. 2023, 112, 336–343. [Google Scholar] [CrossRef] [PubMed]
  274. Akturk, O. The anticancer activity of doxorubicin-loaded levan-functionalized gold nanoparticles synthesized by laser ablation. Int. J. Biol. Macromol. 2022, 196, 72–85. [Google Scholar] [CrossRef] [PubMed]
  275. Emik, S.; Gürses, C.; Kaya, M. Blood-brain barrier targeted delivery of lacosamide-conjugated gold nanoparticles: Improving outcomes in absence seizures. Epilepsy Reseach 2022, 184, 106939. [Google Scholar] [CrossRef]
  276. Peranic, N.; Barbir, R.; Hall, C.R.; Smith, T.A.; Sani, M.A. Spectroscopic study of L-DOPA and dopamine binding on novel gold nanoparticles towards more efficient drug-delivery system for Parkinson ’ s disease. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 268, 120707. [Google Scholar] [CrossRef]
  277. Nguyen, C.; Banh, K.; Nguyen, T.; An, M.; Vu-quang, H.; Nguyen, T. Cyclodextrin modified with different groups to enhance the drug delivery efficiency of gold nanoparticles to treat cancer. Arab. J. Chem. 2023, 16, 105079. [Google Scholar] [CrossRef]
  278. Zamaswazi, Z.; Leigh, J.; Precious, N.; Ngema, P.; Nwabisa, A.; Iheanyichukwu, E.; Mulaudzi, T.; Fiona, S.; Fanelwa, R. Thioglycolic acid-capped gold nanoparticle/cytochrome P450-2E1 electrochemical biosensor for isoniazid. Sens. Bio-Sens. Res. 2023, 41, 100583. [Google Scholar] [CrossRef]
  279. Kumar, P.; Sivapunniyam, A.; Ramalingam, P. Fabrication of a citrus flavonoid hesperetin-capped gold nanoparticles-reduced graphene oxide nanocomposites (Hes-Au/rGONCs) as a potential therapeutic agent for triple negative breast cancer and bacterial infections. Surf. Interfaces 2023, 42, 103347. [Google Scholar] [CrossRef]
  280. Arya, S.S.; Rookes, J.E.; Cahill, D.M.; Lenka, S.K. Reduced Genotoxicity of Gold Nanoparticles with Protein Corona in Allium cepa. Front. Bioeng. Biotechnol. 2022, 10, 849464. [Google Scholar] [CrossRef]
  281. Shivashankarappa, A.; Sanjay, K.R.; Shah, D.; Tagat, A. Decalepis hamiltonii derived gold nanoparticles and photodynamic cytotoxic evaluation on skin melanoma (B16F10) cells as an effective drug delivery vehicle. J. Drug Deliv. Sci. Technol. 2022, 76, 103766. [Google Scholar] [CrossRef]
  282. Xiong, Y.; Chen, T.; Chen, L.; Cai, R. Gold Nanoparticles Coated with SH-PEG-NH2 and Loaded with Ziyuglycoside I for Promoting Autophagy in Hematopoietic Stem Cells. Int. J. Nanomed. 2023, 18, 1347–1362. [Google Scholar] [CrossRef]
  283. Daniel, M.; Astruc, D. Gold Nanoparticles: Assembly, Supramolecular Chemistry, Quantum-Size-Related Properties, and Applications toward Biology, Catalysis, and Nanotechnology. Chem. Rev. 2004, 104, 293–346. [Google Scholar] [CrossRef]
  284. Li, D.; He, Q.; Li, J. Smart core/shell nanocomposites: Intelligent polymers modi fi ed gold nanoparticles. Adv. Colloid Interface Sci. 2009, 149, 28–38. [Google Scholar] [CrossRef]
  285. Chen, L.; Klok, H.A. “Multifaceted” Polymer Coated, Gold Nanoparticles. Soft Matter 2013, 9, 10678–10688. [Google Scholar] [CrossRef]
  286. Kumar, B.; Jajodia, K.; Kumar, P.; Gautam, H.K. AC Stability. J. Drug Deliv. Sci. Technol. 2017, 41, 260–268. [Google Scholar] [CrossRef]
  287. Chaudhary, A.; Garg, S. siRNA delivery using polyelectrolyte-gold nanoassemblies in neuronal cells for BACE1 gene silencing. Mater. Sci. Eng. C 2017, 80, 18–28. [Google Scholar] [CrossRef] [PubMed]
  288. Muddineti, O.S.; Ghosh, B.; Biswas, S. Current trends in using polymer coated gold nanoparticles for cancer therapy. Int. J. Pharm. 2015, 484, 252–267. [Google Scholar] [CrossRef]
  289. Ehlert, S.; Taheri, S.M.; Pirner, D.; Drechsler, M.; Schmidt, H. Polymer Ligand Exchange to Control Stabilization and Compatibilization of Nanocrystals. ACS Nano 2014, 8, 6114–6122. [Google Scholar] [CrossRef] [PubMed]
  290. Khurana, S.; Kukreti, S.; Kaushik, M. Prospecting the cancer therapeutic edge of chitosan-based gold nanoparticles through conformation selective binding to the parallel G-quadruplex formed by short telomeric DNA sequence: A multi-spectroscopic approach. Int. J. Biol. Macromol. 2023, 253, 126835. [Google Scholar] [CrossRef] [PubMed]
  291. Kumar, K.; Vulugundam, G.; Jaiswal, P.K.; Shyamlal, B.R.K.; Chaudhary, S. Efficacious cellular codelivery of doxorubicin and EGFP siRNA mediated by the composition of PLGA and PEI protected gold nanoparticles. Bioorganic Med. Chem. Lett. 2017, 27, 4288–4293. [Google Scholar] [CrossRef]
  292. Engstrom, A.M.; Faase, R.A.; Marquart, G.W.; Baio, J.E.; Mackiewicz, M.R.; Harper, S.L. Size-dependent interactions of lipid-coated gold nanoparticles: Developing a better mechanistic understanding through model cell membranes and in vivo toxicity. Int. J. Nanomed. 2020, 15, 4091–4104. [Google Scholar] [CrossRef]
  293. Abdelmahmoud, M.; Maki, A.; Teng, M.S.; Tan, K.F.; Kumar, P.V. Polyamidoamine-stabilized and hyaluronic acid-functionalized gold nanoparticles for cancer therapy. OpenNano 2023, 13, 100182. [Google Scholar] [CrossRef]
  294. Chen, Y.; Xu, Z.; Zhu, D.; Tao, X.; Gao, Y.; Zhu, H.; Mao, Z.; Ling, J. Gold nanoparticles coated with polysarcosine brushes to enhance their colloidal stability and circulation time in vivo. J. Colloid Interface Sci. 2016, 483, 201–210. [Google Scholar] [CrossRef]
  295. Wang, C.; Tang, F.; Wang, X.; Li, L. Synthesis and application of biocompatible gold core—Poly-(L-Lysine) shell nanoparticles. Colloids Surf. A Physicochem. Eng. Asp. 2016, 506, 425–430. [Google Scholar] [CrossRef]
  296. Feng, Y.; Li, X.; Ji, D.; Tian, J.; Peng, Q.; Shen, Y. Functionalised penetrating peptide-chondroitin sulphate-gold nanoparticles: Synthesis, characterization, and applications as an anti-Alzheimer’ s disease drug. Int. J. Biol. Macromol. 2023, 230, 123125. [Google Scholar] [CrossRef] [PubMed]
  297. Baghani, L.; Heris, N.N.; Khonsari, F.; Dinarvand, S.; Dinarvand, M.; Atyabi, F. Trimethyl-Chitosan Coated Gold Nanoparticles Enhance Delivery, Cellular Uptake and Gene Silencing Effect of EGFR-siRNA in Breast Cancer Cells. Front. Mol. Biosci. 2022, 9, 871541. [Google Scholar] [CrossRef] [PubMed]
  298. Tunali, B.C.; Akturk, O.; Sahingoz, D.; Turk, M.; Keskin, A.C. Synthesis and characterization of polyethyleneimine/silk fibroin/gold nanoparticle nanocomposites: Potential application as a gene carrier in breast cancer cell lines. Eur. Polym. J. 2023, 191, 112042. [Google Scholar] [CrossRef]
  299. Beltran-vargas, N.E.; Peña-mercado, E. Sodium Alginate/Chitosan Scaffolds for Cardiac Tissue Engineering: The Influence of Its Three-Dimensional Material Preparation and the Use of Gold Nanoparticles. Polymers 2022, 14, 3233. [Google Scholar] [CrossRef] [PubMed]
  300. Deb, M.; Hunter, R.; Taha, M.; Abdelbary, H.; Anis, H. Rapid detection of bacteria using gold nanoparticles in SERS with three different capping agents: Thioglucose, polyvinylpyrrolidone, and citrate. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 280, 121533. [Google Scholar] [CrossRef]
  301. Clasky, A.J.; Chen, P.Z.; Brooks, W.; Mccabe, K.; Gu, F.X. Polymer Co-Coating of Gold Nanoparticles Enables Their Integration Into Contact Lenses for Stable, Selective Ocular Light Filters. Adv. Mater. Interfaces 2022, 9, 2201549. [Google Scholar] [CrossRef]
  302. Srivastava, I.; Xue, R.; Jones, J.; Rhee, H.; Flatt, K.; Gruev, V.; Nie, S. Biomimetic Surface-Enhanced Raman Scattering Nanoparticles with Improved Dispersibility, Signal Brightness, and Tumor Targeting Functions. ACS Nano 2022, 16, 8051–8063. [Google Scholar] [CrossRef]
  303. Liu, H.; Miao, Z.; Zha, Z. Cell membrane-coated nanoparticles for immunotherapy. Chinese Chem. Lett. 2022, 33, 1673–1680. [Google Scholar] [CrossRef]
  304. Hu, C.M.J.; Fang, R.H.; Luk, B.T.; Zhang, L. Nanoparticle-detained toxins for safe and effective vaccination. Nat. Nanotechnol. 2013, 8, 933–938. [Google Scholar] [CrossRef] [PubMed]
  305. Hu, C.J.; Zhang, L.; Aryal, S.; Cheung, C.; Fang, R.H.; Zhang, L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl. Acad. Sci. USA 2011, 108, 10980–10985. [Google Scholar] [CrossRef]
  306. Ma, Q.; Fan, Q.; Xu, J.; Bai, J.; Han, X.; Dong, Z.; Zhou, X.; Liu, Z.; Gu, Z.; Wang, C. Calming Cytokine Storm in Pneumonia by Targeted Delivery of TPCA-1 Using Platelet-Derived Extracellular Vesicles. Matter 2020, 3, 287–301. [Google Scholar] [CrossRef]
  307. Le, W.; Mei, T.; Wang, Y.; Chen, B. Cell membrane camou fl aged nanoparticles: A new biomimetic platform for cancer photothermal therapy. Int. J. Nanomed. 2019, 14, 4431–4448. [Google Scholar]
  308. Jiménez-Jiménez, C.; Manzano, M.; Vallet-Regí, M. Nanoparticles coated with cell membranes for biomedical applications. Biology 2020, 9, 406. [Google Scholar] [CrossRef] [PubMed]
  309. Narain, A.; Asawa, S.; Chhabria, V.; Patil-sen, Y. Cell membrane coated nanoparticles: Next-generation therapeutics. Nanomedicine 2017, 12, 2677–2692. [Google Scholar] [CrossRef] [PubMed]
  310. Vijayan, V.; Uthaman, S.; Park, I. Cell Membrane-Camouflaged Nanoparticles: A Promising Biomimetic Strategy for Cancer Theragnostics. Polymers 2018, 10, 983. [Google Scholar] [CrossRef]
  311. Fang, R.H.; Kroll, A.V.; Gao, W.; Zhang, L. Cell Membrane Coating Nanotechnology. Adv. Mater. 2018, 30, 1706759. [Google Scholar] [CrossRef]
  312. Zhai, Y.; Su, J.; Ran, W.; Zhang, P.; Yin, Q.; Zhang, Z.; Yu, H. Preparation and Application of Cell Membrane-Camouflaged Nanoparticles for Cancer Therapy. Theranostics 2023, 7, 2575–2592. [Google Scholar] [CrossRef] [PubMed]
  313. Li, R.; He, Y.; Zhang, S.; Qin, J.; Wang, J. Cell membrane-based nanoparticles: A new biomimetic platform for tumor diagnosis and treatment. Acta Pharm. Sin. B 2018, 8, 14–22. [Google Scholar] [CrossRef] [PubMed]
  314. Hu, X.; Li, H.; Huang, X.; Zhu, Z.; Zhu, H.; Gao, Y.; Zhu, Z. Cell membrane-coated gold nanoparticles for apoptosis imaging in living cells based on fluorescent determination. Microchim Acta 2020, 187, 175. [Google Scholar] [CrossRef]
  315. Tan, S.; Wu, T.; Zhang, D.; Zhang, Z. Cell or Cell Membrane-Based Drug Delivery Systems. Theranostics 2015, 5, 16–21. [Google Scholar] [CrossRef]
  316. Gao, W.; Zhang, L.; Gao, W.; Zhang, L. Coating nanoparticles with cell membranes for targeted drug delivery. J. Drug Target. 2015, 23, 619–626. [Google Scholar] [CrossRef]
  317. Choi, B.; Park, W.; Park, S.; Rhim, W.; Han, D.K. Recent trends in cell membrane-cloaked nanoparticles for therapeutic applications. Methods 2023, 177, 2–14. [Google Scholar] [CrossRef] [PubMed]
  318. Pincela Lins, P.M. Cell-Derived Nanoplatforms for Cancer Therapy. Ph.D. Thesis, Universidade de São Paulo, São Paulo, Brazil, 2021. [Google Scholar]
  319. Lian, M.; Shao, S.; Liu, M.; Shi, Y.; Zhang, H.; Chen, D. Cell membrane-coated nanoparticles as peroxidase mimetics for cancer cell targeted detection and therapy. Talanta 2023, 238, 123071. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) Surface plasmon resonance (SPR) phenomenon involving what happens on the surface of gold nanoparticles when interacting with light. (B) Some types of coatings used for gold nanoparticles, ligands, medicinal agents, DNA, antibody, etc. These interact through covalent-type ligations such as S-Au or electrostatic, and at the same time molecules linked to gold atoms can form interactions such as H-H hydrogen bridges.
Figure 1. (A) Surface plasmon resonance (SPR) phenomenon involving what happens on the surface of gold nanoparticles when interacting with light. (B) Some types of coatings used for gold nanoparticles, ligands, medicinal agents, DNA, antibody, etc. These interact through covalent-type ligations such as S-Au or electrostatic, and at the same time molecules linked to gold atoms can form interactions such as H-H hydrogen bridges.
Pharmaceutics 16 00255 g001
Scheme 1. Flowchart for Inclusion of Studies Performed.
Scheme 1. Flowchart for Inclusion of Studies Performed.
Pharmaceutics 16 00255 sch001
Figure 2. Exploring diverse routes in bottom–up nanoparticle synthesis: from chemical techniques to eco-friendly biological and radiative methods. In the bottom–up approach, there is a wide variety of routes, completely chemical methods such as the Turkevich method and variants, biological methods where microorganisms such as fungi are used, often called eco-friendly, and methods of obtaining the use of gamma radiation.
Figure 2. Exploring diverse routes in bottom–up nanoparticle synthesis: from chemical techniques to eco-friendly biological and radiative methods. In the bottom–up approach, there is a wide variety of routes, completely chemical methods such as the Turkevich method and variants, biological methods where microorganisms such as fungi are used, often called eco-friendly, and methods of obtaining the use of gamma radiation.
Pharmaceutics 16 00255 g002
Figure 3. Schematic illustration of doxorubicin (DOX)-tethered responsive gold nanoparticles.
Figure 3. Schematic illustration of doxorubicin (DOX)-tethered responsive gold nanoparticles.
Pharmaceutics 16 00255 g003
Figure 4. Electrostatic and steric repulsion model. (A) Electrostatic stabilization of AuNPs is based on Coulomb repulsion between particles with the same surface charge sign, preventing the particles from coming into close contact where the attractive short-distance van der Waals forces dominate. (B) For steric stabilization, the contact is avoided by adsorbing bulky molecules on the surface, which act as a spacer to prevent particle aggregation [1,44].
Figure 4. Electrostatic and steric repulsion model. (A) Electrostatic stabilization of AuNPs is based on Coulomb repulsion between particles with the same surface charge sign, preventing the particles from coming into close contact where the attractive short-distance van der Waals forces dominate. (B) For steric stabilization, the contact is avoided by adsorbing bulky molecules on the surface, which act as a spacer to prevent particle aggregation [1,44].
Pharmaceutics 16 00255 g004
Figure 5. Stereochemistry in protecting units on the surface of gold clusters. (A) Schematic drawing of a monomeric Au–SR–Au staple motif. (B) Stereochemistry in protecting units on the surface of gold clusters. While the gold atoms stabilized between two sulfur atoms are formal Au, the gold atoms at the bottom of the structures are Au0 atoms at the surface of the cluster core (I). The stereodescriptors are indicated in red, and lowercase letters indicate pseudochirality. Priority for the determination of absolute configuration is Au0 > Au1 > R > electron lone pair (schematic and image taken from “Chirality in Thiolate-Protected Gold Clusters”) [46].
Figure 5. Stereochemistry in protecting units on the surface of gold clusters. (A) Schematic drawing of a monomeric Au–SR–Au staple motif. (B) Stereochemistry in protecting units on the surface of gold clusters. While the gold atoms stabilized between two sulfur atoms are formal Au, the gold atoms at the bottom of the structures are Au0 atoms at the surface of the cluster core (I). The stereodescriptors are indicated in red, and lowercase letters indicate pseudochirality. Priority for the determination of absolute configuration is Au0 > Au1 > R > electron lone pair (schematic and image taken from “Chirality in Thiolate-Protected Gold Clusters”) [46].
Pharmaceutics 16 00255 g005
Figure 6. Au NP protein corona complex. The affinity and exchange rate of the protein crown depend on the size, charge, composition, geometry, incubation conditions (temperature, concentration, time), the type of proteins and their stability. The formation of the crown defines the cellular internalization, biodistribution and toxicity of the particle [47,49,50,51].
Figure 6. Au NP protein corona complex. The affinity and exchange rate of the protein crown depend on the size, charge, composition, geometry, incubation conditions (temperature, concentration, time), the type of proteins and their stability. The formation of the crown defines the cellular internalization, biodistribution and toxicity of the particle [47,49,50,51].
Pharmaceutics 16 00255 g006
Figure 7. Co-functionalization of AuNPs typically employs two techniques. The first method entails producing a mixed monolayer, for instance, by directly adhering PEG and RME to the surface of AuNP using thiol chemistries (A). The second method makes use of a hetero-bifunctional PEG linker, such as thiol-PEGsuccinimidyl glutaramide (SH-PEG-SGA), which has an NHS ester group at one end for attachment to the AuNP surface and a thiol at the other end for direct binding to the RME peptide via the N-terminus group (B). Therefore, the latter configuration has the ability to fully saturate the AuNP surface with PEG before saturating the free end of PEG with a physiologically active group.
Figure 7. Co-functionalization of AuNPs typically employs two techniques. The first method entails producing a mixed monolayer, for instance, by directly adhering PEG and RME to the surface of AuNP using thiol chemistries (A). The second method makes use of a hetero-bifunctional PEG linker, such as thiol-PEGsuccinimidyl glutaramide (SH-PEG-SGA), which has an NHS ester group at one end for attachment to the AuNP surface and a thiol at the other end for direct binding to the RME peptide via the N-terminus group (B). Therefore, the latter configuration has the ability to fully saturate the AuNP surface with PEG before saturating the free end of PEG with a physiologically active group.
Pharmaceutics 16 00255 g007
Figure 10. Methods for conjugating DNA on AuNPs. (A) The typical salt-aging process involving initial DNA adsorption, and gradual increase in NaCl concentration. (B) Attaching DNA in the presence of stabilizing ligands (e.g., surfactants) to improve the colloidal stability of AuNPs. (C) Low-pH loading requiring the DNA containing a poly(A) block to assemble into a parallel duplex. (D) The freezing method does not require additional reagents. (E) Anchoring poly(A) containing nonthiolated DNA on AuNPs with controlled density; a longer poly(A) block yields a lower DNA density (adaptation from Biwu et al.) [164].
Figure 10. Methods for conjugating DNA on AuNPs. (A) The typical salt-aging process involving initial DNA adsorption, and gradual increase in NaCl concentration. (B) Attaching DNA in the presence of stabilizing ligands (e.g., surfactants) to improve the colloidal stability of AuNPs. (C) Low-pH loading requiring the DNA containing a poly(A) block to assemble into a parallel duplex. (D) The freezing method does not require additional reagents. (E) Anchoring poly(A) containing nonthiolated DNA on AuNPs with controlled density; a longer poly(A) block yields a lower DNA density (adaptation from Biwu et al.) [164].
Pharmaceutics 16 00255 g010
Figure 11. Structure and biological activity of carbohydrates that have been functionalized with Au NPs (figure is modified from Xiangyan et al.) [199].
Figure 11. Structure and biological activity of carbohydrates that have been functionalized with Au NPs (figure is modified from Xiangyan et al.) [199].
Pharmaceutics 16 00255 g011
Figure 12. Methods for synthesis of polymeric AuNPs. P-AuNPs have the potential to be used in medicine delivery thanks to advancements in nanotechnology. The “direct synthesis”, “grafting to” and “grafting from” techniques are the main synthetic pathways for P-AuNPs (figure is modified from Muddineti et al.) [288].
Figure 12. Methods for synthesis of polymeric AuNPs. P-AuNPs have the potential to be used in medicine delivery thanks to advancements in nanotechnology. The “direct synthesis”, “grafting to” and “grafting from” techniques are the main synthetic pathways for P-AuNPs (figure is modified from Muddineti et al.) [288].
Pharmaceutics 16 00255 g012
Figure 13. Schematic depicting the fabrication of red blood cell (RBC)-membrane-coated Au NPs (adapted from Hu et al.) [305].
Figure 13. Schematic depicting the fabrication of red blood cell (RBC)-membrane-coated Au NPs (adapted from Hu et al.) [305].
Pharmaceutics 16 00255 g013
Table 1. PEG-derived Au NP coatings. BA—Biomedical application, DD—Drug delivery, BC—Brachytherapy, CT—Cancer treatment, RT—Radiation therapy.
Table 1. PEG-derived Au NP coatings. BA—Biomedical application, DD—Drug delivery, BC—Brachytherapy, CT—Cancer treatment, RT—Radiation therapy.
CoatingApplication
PEGMA5–DEAMA95–EGDMA15–TG20BA [67]
O-(3-carboxypropyl)-O″-[2-(3-mercaptopropionylamino) ethyl]-polyethylene glycol (MW 5000, HS-PEG-COOH)DD [68]
(MW 5000, HS-PEG-NH2)BA [69]
Thiol-terminated polyethylene glycol (5 kDa)BA [70]
Polyethyleneimine (branched, 25 kDa)BC [71]
Mercaptopolyethylene glycol monomethyl ether (MeO-PEG-SH) (2 kDa)BA [72]
Zwitterionic and non-ionic tetraethylene glycol (TEG)BA [73]
PEGCT [74]
2-Hydroxypropane-1,2,3-tricarboxylic acidRT [75]
HS-PEG-CarboxylBA [76]
Tirapazamine ModifiedBA [77]
Bovine serum albumin (BSA-PEG)BA [78]
CPP fragments (CALNNPFVYLI, CALRRRRRRRR)BA [79]
Ranibizumab biosimilar (Mab)/polyethyleneglycol (PEG)-conjugated, PEG chain (5 kDa and 10 kDa)DD [80]
Dicarboxylic PolyEthylene Glycol (PEG)-600 (PEG)-Bisphosphonate (Bis-PO)BA [81]
(Methoxy-poly(ethylene glycol)-b-poly((2-dimethylamino) ethyl methacrylate-co-itaconic acid) (mPEG-b-p(DMAEMA-co-IA)BA [82]
DSPE-PEG2000DD [83]
Polyethylene glycol (PEG) and functionalized with the boron-rich anion cobalt bis(dicarbollide)CT [84]
Thiolated polyethylene glycol(PEG-SH, MW5000)BA [85]
Thiol terminated methoxy polyethylene glycolDD [86]
Poly(ethylene glycol) thiols (X–PEG–SH, where X = COOH, NH2, CH3O; MW = 5000 Da)CT [87]
PEGylated, RGD peptide modified and disulfide cross-linked short polyethylenimines (DSPEIs)DD [86]
Paclitaxel-PEG 400DD [68]
Thiolated methoxy polyethylene glycol (HS-mPEG)BA [1]
PEGylated-poly(DL-lactic-co-glycolic acid)BA [22]
Dodecyl-PEGBA [88]
PEG 600CT [89]
PEG 7.5 kCT [90]
Table 2. Antibody Au NP coatings. CT—Cancer treatment, BA—Biomedical application, DD—Drug delivery.
Table 2. Antibody Au NP coatings. CT—Cancer treatment, BA—Biomedical application, DD—Drug delivery.
Coating Application
TrastuzumabCT [17]
CetuximabCT [17]
EGFRCT [17]
Anti-transferrinBA [17]
CD30 CT [17]
MUC-1 CT [17]
hERG1CT [17]
DR5 CT [17]
TROP-2CT [17]
CD44CT [17]
Anti-human IgGBA [1]
Anti-CD4DD [19]
Anti-HER2/c erb-2antibodyCT [10]
Anti-EGFRBA [10]
F19 monoclonal antibodiesCT [10]
Anti-CA15-3-HRP antibodyCT [10]
Her-2/neu antigenCT [10]
ScFv (single-chain variable fragment)CT [10]
SM5-1CT [106]
Rabbit antihuman IgGCT [107]
Mouse antihuman f-PSA BA [107]
Anti-HER2CT [107]
Cetuximab antibodyCT [108]
Anti-interleukin-13 receptor alpha 2 (anti-IL13Ra2)CT [109]
Listeria monocytogene antibody (mAb-Lis)BA [110]
Anti-EGFRCT [111]
4G2BA [112]
Goat anti-rabbit IgGBA [113]
Table 3. Protein Au NP coatings. DD—Drug delivery, BA—Biomedical application, CT—Cancer treatment, TA—Theranostic application, DA—Diagnostic application.
Table 3. Protein Au NP coatings. DD—Drug delivery, BA—Biomedical application, CT—Cancer treatment, TA—Theranostic application, DA—Diagnostic application.
Coating (Proteins)Application
Human serum albumin (alb-AuNP)DD [114]
Luteinizing hormone releasing hormone (LHRH)BA [47]
Heavy metal binding proteins (HMBPs) in recombinant Escherichia coliDD [115]
Gel from an Aloe vera plantBA [116]
Inhibitor SMI#9CT [117]
Polyami-doamine (PAMAM) G4 dendrimerCT [118]
CBP4CT [119]
Extract from the alga Egregia sp. intrinsicallyCT [120]
Hybrid albuminCT [121]
Bovine serum albumin (BSA)CT [122]
BromelainCT [123]
Chicken egg white (CEW) BA [124]
FGF1 CT [17]
Lectin Jacalin CT [17]
T antigen CT [17]
Apolipoprotein E DD [114]
Ovalbumin (OVA)DD [125]
CollagenTA [22]
Silk fibroinTA [22]
Wheat-protein-isolate-based polyacrylamide hydrogelBA [22]
CYT-6091 DA [20]
Tumor necrosis factor-α (TNF-α)TA [20]
TransferrinCT [108]
α- and β-integrinCT [17]
Neuropilin-1 CT [17]
β-galactosidase DD [1]
Phospho-tyrosineBA [1]
Glucose-oxidaseBA [126]
Concanavalin ACT [127]
CD133/AC133CT [128]
SericinBA [129]
StreptavidinBA [130]
Horseradish peroxidaseCT [131]
FerritinBA [132]
Horseradish peroxidaseBA [133]
Tenascin-CBA [134]
Streptavidin–biotinBA [135]
Matrix metalloproteinase-9BA [136]
5′ thiol-ended DNACT [137]
Table 4. Peptides and Amino Acid Au NP coatings. BA—Biomedical application, DD—Drug delivery, CT—Cancer treatment.
Table 4. Peptides and Amino Acid Au NP coatings. BA—Biomedical application, DD—Drug delivery, CT—Cancer treatment.
CoatingApplication
VG-21BA [151]
Peptides: CLPSSD, CLPTTD, CLPAAD, CLPVVD, CLPLLD, CLPIID, CLPYYD, CLPWWD, CLPYYDBA [145]
HairpinDD [152]
Aspartate (Asp), glycine (Gly), leucine (Leu), lysine (Lys), serine (Ser), arginine (Arg)BA [138]
Glucagon-like peptide-1 (GLP-1) DD [153]
L-aspartateCT [106]
Lycosin-ICT [154]
IndolicidinBA [155]
Plectin-1 (KTLLPTP)CT [17]
KTLLPTPYC CT [17]
c(RGD) CT [17]
CRGDK CT [17]
GE11 (YHWYGYTPQNVI) CT [17]
Aspartic acid DD [1]
Glutamic acidDD [1]
Phenylalanine DD [1]
Tryptophan DD [1]
L-cysteine DD [1]
Crotamine DD [19]
Cyclic RGD (cRGD) DD [47]
Peptide p12 (TSFAEYWNLLSP)CT [17]
PDC: peptide–drug conjugate containing chlorambucil, melphalan or bendamustineCT [2]
RGD: arginyl-glycyl-aspartic acid tripeptideCT [2]
PKC-peptideCT [10]
Peptide (CCVVGRKKRRQRRRPQGGPLGVEKEKEKEK) and non-responsive peptide (CCVVGRKKRRQRRRPQGGGLPVEKEKEKEK) CT [156]
RGD-125IPt-PDA-AuNRsCT [20]
RGD-177Lu-AuNPsCT [20]
RGD-GdTc-AuNPsCT [20]
RGDCT [74]
Candida antarctica lipaseBA [157]
Tilapia-scale collagen peptidesBA [158]
RRGTIAFDNWVDTGTRVYDPPPPCCCT [159]
Primer: 5′-
CCGATGCTGTCTTTCGCTGTATCAGGTTCATCACCTGACTCA-3′; reverse primer: 5′-GGCCGCTTTTGCGGGATCTGAGTCAGGTGATGAACCTG
ATAC-3′
BA [160]
LanreotideCT [161]
NFL-TBS.40–63 peptide (NH2-YSSYSAPVSSSLSVRRSYSSSSGS-CONH2), BIOT-NFL-peptideBA [162]
TAT.48–60 peptide (BIOT-TAT-peptide; BIOTGRKKRRQRRRPPQ-CONH2)BA [162]
Vim-TBS.58–81 peptide (BIOT-Vim-peptide; BIOT-GGAYVTRSSAVRLRSSVPGVRLLQCONH2)BA [162]
N-acetyl-L-cysteineBA [163]
Table 5. Microorganisms, DNA, RNA and aptamers. CT—Cancer treatment, BA—Biomedical application, DD—Drug delivery, DA—Diagnostic application.
Table 5. Microorganisms, DNA, RNA and aptamers. CT—Cancer treatment, BA—Biomedical application, DD—Drug delivery, DA—Diagnostic application.
CoatingApplication
siRNACT [186]
anti-STAT3 siRNACT [187]
Diosmin with calf thymus DNA (ctDNA)BA [188]
AS1411 (AS-LAGN)CT [189]
AS1411—a 26-base guanine (Apt)DD [17]
sgc8c (Apt)CT [17]
Nucleolin (apt)CT [17]
PSMA (apt)CT [17]
PTK7 (apt)CT [17]
HER2 aptamer HAptCT [17]
Antisense oligonucleotides (DNA)DD [1]
Mimics of tumor suppressive miRNA-miR-205 (DNA)BA [1]
NU-0129 CT [2]
26-base G-rich DNA oligonucleotideDA [47]
miR-375CT [106]
MycobacteriumDD [19]
DNABA [190]
ssDNA aptamerBA [133]
miR-206CT [191]
Kana aptamer (5′-TGGGGGTTGAGGCTAAGCCG-3′)BA [192]
Thiol-modified DNABA [193]
L-arginineBA [194]
L-lysineBA [194]
L-histidineBA [194]
Anti-HER-2BA [195]
TyrosineBA [196]
5′-Methylene blue (MB)-TCTAACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGATTTTTTT-(CH2)6-SH-3′.DA [197]
Table 6. Carbohydrate Au NP coatings. DD—Drug delivery, BA—Biomedical application, TA—Theranostic application, DA—Diagnostic application, CT—Cancer treatment.
Table 6. Carbohydrate Au NP coatings. DD—Drug delivery, BA—Biomedical application, TA—Theranostic application, DA—Diagnostic application, CT—Cancer treatment.
CoatingApplication
Β-cyclodextrinDD [205]
Lyophilized lipids: 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC); 2-dipalmitoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (DPPG); 1,2-dipalmitoyl-3-trimethylammonium-propane (DPTAP); 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-5000] (DPPE-PEG5000)BA [206]
Gellan gumTA [22]
Extracellular vesicle membranesD [207]
LyposomesCT [208]
Glucose CT [2]
Lactose CT [17]
Galectin-1 CT [17]
Guar gumBA [22]
Gum karayaBA [22]
Lactobionic acidDA [106]
1-Thio-D-glucoseCT [209]
Fluoro-6-deoxythio-1-D-glucoseCT [209]
CarrageenanBA [202]
AlginateBA [202]
ChitinBA [202]
CelluloseBA [210]
AgaroseBA [202]
1,2-Dioleoyl-sn-glycero-3-phosphocholine (DOPC)BA [211]
1-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC)BA [211]
Lipoic acidBA [212]
Tripeptide glutathioneBA [213]
Thermosensitive lipidCT [214]
DextrinBA [215]
ThioglucoseBA [216]
Saponin niosomesCT [217]
Carboxymethyl celluloseCT [218]
SucroseBA [219]
G5 PAMAM dendrimersBA [220]
GastrodinBA [220]
Mesquite-gumBA [221]
GelatinTA [22]
Table 7. Drug Au NP coatings. BA—Biomedical application, CT—Cancer treatment, DD—Drug delivery, DA—Diagnostic Application, TA—Theranostic application, RT—Radiation therapy.
Table 7. Drug Au NP coatings. BA—Biomedical application, CT—Cancer treatment, DD—Drug delivery, DA—Diagnostic Application, TA—Theranostic application, RT—Radiation therapy.
CoatingApplication
PaclitaxelBA [225]
CatecholDD [226]
Doxorubicin hydrochlorideCT [123]
Inulin-folate derivative (INU-FA)DD [227]
1-Dodecanethiol, 1-hexanethioBA [228]
SN38CT [229]
GE-genistein,BA [230]
Cetyltrimethylammonium bromide (CTAB)DD [231]
11-Mercaptoundecanoic acid (11-MUA)DD [232]
Gosereline CT [84]
Imatinib mesylate (IM)CT [187]
2-Mercapto-1-methylimidazole (MMT) BA [124]
PhloroglucinolCT [233]
Lonidamine (LND) CT [189]
Folic acid CT [234]
AminoguanidineBA [235]
Cationic carbosilane dendronsDD [236]
Oxaliplatin CT [107]
Anisamide CT [17]
α- and β-bicalutamideCT [17]
Sigma receptor CT [17]
GPRC6A/MAR CT [17]
3-Mercaptopropionic acid DD [1]
4-Mercaptophenol DD [1]
DocetaxelCT [17]
Glutathione CT [2]
AMD3100 DA [47]
Cysteamine–folic acidDA [237]
XAV939DD [107]
GSHCT [108]
Hyaluronic acid (HA)CT [91]
ssPalmM/DOPE/cholesterol CT [238]
DMG-mPEG2000CT [238]
PSSDD [239]
LysophosphocholineDD [240]
Triethylenetetramine CT [1]
2-Chlorotrityl resin CT [1]
CALNN BA [1]
CALNNR8 BA [1]
6-Mercaptopurine-9-b-D-ribofuranosideCT [10]
N-Succinimidyl 3-(2-puridyldithio) propionate DD [19]
MethotrexateDD [241]
StreptomycinBA [22]
AmpicillinBA [22]
KanamycinBA [22]
BleomycinCT [2]
Cationic tetraalkyl ammonium DD [1]
5-FluorouracilCT [27]
CisplatinCT [123]
MitoxantroneCT [27]
AmoxicillinBA [22]
CefaclorBA [22]
CiprofloxacinBA [22]
GentamycinBA [22]
RifampicinBA [22]
VancomycinBA [22]
Methylene blueBA [22]
HerceptinCT [10]
PenicillamineDA [106]
TioproninCT [242]
N-acetyl cysteineCT [242]
TamoxifenCT [107]
Methyl thioglycolate CT [1]
Biotina CT [2]
Cetyltrimethyl ammonium bromide CT [2]
Triphenyl monosulfonate CT [2]
EGCGBA [5]
Phthalocyanine CT [1]
EthanediamineBA [47]
Ethanedioic acid BA [47]
Ce6 carbon dots TA [47]
2-MercaptoethylamineBA [22]
Pyridinium amphiphiles DD [19]
Amphiphilic triethyleneglycol mercaptanDD [19]
Fe3O4 BA [243]
Thiol hydroxyethylmethacrylate (HEMA)BA [22]
Diatrizoic acidBA [244]
P-mercaptobenzoic acidBA [10]
Diethylenetriamine-N,N,N′,N″,N″-pentaacetic acid (DTPA)BA [106]
[3-Butoxy-2-hydroxypropyl 2-(2,4-dihydroxyphenyl) acetate]CT [106]
CyNAMLA-381BA [107]
11-MercaptoundecanesulfonateRT [242]
OctanethiolRT [242]
PH2PC6H4SO3HRT [108]
TPPTSRT [108]
PlatinumBA [20]
4,6-Diamino-2-pyrimidinethiol (DAPT)BA [97]
GemcitabineCT [17]
CeO2CT [245]
Gadolinium oxideRT [122]
Tetraethyl orthosilicate (TEOS)DD [246]
Phosphotungstic acidBA [247]
3-Aminopropyltriethoxysilane (APTES)DD [246]
SilicaCT [248]
CisplatinBA [249]
4-SelenocyanatoanilineCT [250]
4,4′-DiselanediyldianilineCT [250]
N-(4-selenocyanatophenyl)cinnamamide CT [250]
N-(3-selenocyanatopropyl)cinnamamideCT [250]
IsoniazidBA [196]
Quercetin (Qrc, antioxidant)BA [251]
Sodium citrateBA [252]
4,6-Diamino- 2-pyrimidinethiolBA [253]
1,2-Di(9Z-octadecenoyl)-sn-glycero-3-phosphocholineBA [254]
1,2-Dipalmitoyl-sn-glycero-3-phosphocholineBA [254]
TyrosolBA [255]
CephradineBA [256]
cF9-SHBA [257]
Tannic acidBA [258]
Protoporphyrin IXCT [259]
CurcuminCT [260]
DelafloxacinBA [261]
CefotetanBA [262]
CefiximeBA [262]
N-succinimidyl 3-(2-pyridyldithio) propionateBA [263]
ApigeninBA [264]
RutinCT [265]
PAMAM G-4BA [133]
Dextran/silk sericinBA [266]
ErythromycinCT [267]
6-MercaptopurineBA [268]
LuteolineCT [269]
Galangin (3,5,7-trihydroxyflavone)BA [270]
Kaempferol (3,5,7,4′-tetrahydroxyflavone)BA [270]
Quercetin (3,5,7,3′,4′-pentahydroxyflavone)BA [270]
Myricetin (3,5,7,3′,4′,5′-hexahydroxyflavone)BA [270]
Nitrobenzoic acidBA [271]
3-MercaptopropanesulfonateCT [272]
2-(Diethylamino) ethanethiol hydrochlorideCT [272]
CiprofloxacinBA [273]
LevanCT [274]
(11-Mercaptoundecyl)-N,N,N-trimethylammonium bromideBA [194]
LacosamideBA [275]
L-dopaBA [276]
DopamineBA [276]
β-cyclodextrinCT [277]
Thioglycolic acidBA [278]
HesperetinCT [279]
VanillinBA [280]
Hexadecyltrimethylammonium bromideCT [137]
5-Aminolevulinic acidCT [281]
ZiyuglycosideBA [282]
Table 8. Polymer and biofunctionalized Au NP coatings. DD—Drug delivery, BA—Biomedical Application, CT—Cancer treatment, TA—Theranostic application, DA—Diagnostic application.
Table 8. Polymer and biofunctionalized Au NP coatings. DD—Drug delivery, BA—Biomedical Application, CT—Cancer treatment, TA—Theranostic application, DA—Diagnostic application.
CoatingApplication
Polyethyleneimine, citraconic anhydride modified poly (allylamine) hydrochloride and poly L-arginineDD [287]
ChitosanBA [290]
Poly (lactic-co-glycolic acid) (PLGA)DD [291]
L-α-phosphatidylcholine-hydrogenated soy (HPC), 1.2-dipalmitoyl-d62-sn-glycero-3-phosphocholine (dDPPC)DD [292]
Polyvinyl-alcohol (PVA)TA [5]
PolyamidoamineBA [293]
Amphipathic polymer (hydrophobic PCL/poly(2-hydroxyethyl methacrylate) and hydrophilic poly[2-(2-methoxyethoxy) ethyl methacrylate])TA [22]
PolypyrroleTA [10]
Pluronic® F127 (Au-DOX-Gel)CT [106]
Poly(ethylene glycol)-b-poly(ԑ-caprolactone)TA [242]
Pluronic hydrogelCT [20]
PEI AuNPs/51 (polyethyleneimine) CT [20]
GO-Au-PANI (Polyaniline) core–shellCT [20]
Au-IPN-pNIPAAm (poly(N-isopropylacrylamide))CT [20]
PLGA-AuNR-MnO2 (Poly lactic-co-glycolic acid)CT [20]
Poly (lactic-co-glycolic acid) (PLGA) polymerDD [291]
PCLBA [22]
Graphene oxide (GO)-polymer-AuDA [244]
Hydrophilic polymer, poly(3-dimethylammonium-1-propyne hydrochloride) (PDMPAHCl)BA [92]
Poly-lysineBA [1]
PolysarcosineBA [294]
LiposomesCT [87]
Poly-(l-lysine)BA [295]
Poly(diallydimethyl ammonium chloride; PDADMAC)DD [1]
Poly(sodium-4-stryrenesulfonate; PSS) DD [1]
Poly(allylamine hydrochloride; PAH) DD [1]
Hydrogel poly (Nisopropylacryamide) (PNIPAM)CT [1]
AuroLase CT [2]
Hydroxylated polyethylene terephthalate (PET)BA [22]
Polycaprolactone (PCL) gelatinBA [22]
PVA-BSABA [22]
StarchBA [202]
Chondroitin sulfateBA [296]
FucoidaBA [202]
Trimethyl-chitosanCT [297]
PolyethyleneimineCT [298]
Pluronic F-127BA [299]
Polysarcosine–bpolycaprolactoneBA [212]
PolyvinylpyrrolidoneBA [300]
Poly(allylamine–hydrochloride)BA [210]
Methacrylate monomerBA [301]
Red blood cellCT [302]
MCF-7 cancer cellCT [214]
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

Arcos Rosero, W.A.; Bueno Barbezan, A.; Daruich de Souza, C.; Chuery Martins Rostelato, M.E. Review of Advances in Coating and Functionalization of Gold Nanoparticles: From Theory to Biomedical Application. Pharmaceutics 2024, 16, 255. https://doi.org/10.3390/pharmaceutics16020255

AMA Style

Arcos Rosero WA, Bueno Barbezan A, Daruich de Souza C, Chuery Martins Rostelato ME. Review of Advances in Coating and Functionalization of Gold Nanoparticles: From Theory to Biomedical Application. Pharmaceutics. 2024; 16(2):255. https://doi.org/10.3390/pharmaceutics16020255

Chicago/Turabian Style

Arcos Rosero, Wilmmer Alexander, Angelica Bueno Barbezan, Carla Daruich de Souza, and Maria Elisa Chuery Martins Rostelato. 2024. "Review of Advances in Coating and Functionalization of Gold Nanoparticles: From Theory to Biomedical Application" Pharmaceutics 16, no. 2: 255. https://doi.org/10.3390/pharmaceutics16020255

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

Article Metrics

Back to TopTop