Next Article in Journal
Computational Design and Biological Evaluation of Analogs of Lupin Peptide P5 Endowed with Dual PCSK9/HMG-CoAR Inhibiting Activity
Next Article in Special Issue
Functionalized Silver and Gold Nanomaterials with Diagnostic and Therapeutic Applications
Previous Article in Journal
3,4-Dihydroxyphenylethanol (DPE or Hydroxytyrosol) Counteracts ERK1/2 and mTOR Activation, Pro-Inflammatory Cytokine Release, Autophagy and Mitophagy Reduction Mediated by Benzo[a]pyrene in Primary Human Colonic Epithelial Cells
Previous Article in Special Issue
Iron–Gold Nanoflowers: A Promising Tool for Multimodal Imaging and Hyperthermia Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gold Nanorods for Drug and Gene Delivery: An Overview of Recent Advancements

by
Atieh Jahangiri-Manesh
1,†,
Marziyeh Mousazadeh
1,†,
Shirinsadat Taji
1,†,
Abbas Bahmani
2,
Atefeh Zarepour
3,
Ali Zarrabi
3,*,
Esmaeel Sharifi
4,* and
Mostafa Azimzadeh
5,6,*
1
Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-154, Iran
2
Institute for Nanoscience & Nanotechnology (INST), Sharif University of Technology, Tehran P.O. Box 14588-89694, Iran
3
Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
4
Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan P.O. Box 65178-38736, Iran
5
Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd P.O. Box 887-89165, Iran
6
Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd P.O. Box 887-89165, Iran
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2022, 14(3), 664; https://doi.org/10.3390/pharmaceutics14030664
Submission received: 22 February 2022 / Revised: 14 March 2022 / Accepted: 15 March 2022 / Published: 17 March 2022
(This article belongs to the Special Issue Recent Advances in Metallic Nanoparticles as Theranostics Platforms)

Abstract

:
Over the past few decades, gold nanomaterials have shown great promise in the field of nanotechnology, especially in medical and biological applications. They have become the most used nanomaterials in those fields due to their several advantageous. However, rod-shaped gold nanoparticles, or gold nanorods (GNRs), have some more unique physical, optical, and chemical properties, making them proper candidates for biomedical applications including drug/gene delivery, photothermal/photodynamic therapy, and theranostics. Most of their therapeutic applications are based on their ability for tunable heat generation upon exposure to near-infrared (NIR) radiation, which is helpful in both NIR-responsive cargo delivery and photothermal/photodynamic therapies. In this review, a comprehensive insight into the properties, synthesis methods and toxicity of gold nanorods are overviewed first. For the main body of the review, the therapeutic applications of GNRs are provided in four main sections: (i) drug delivery, (ii) gene delivery, (iii) photothermal/photodynamic therapy, and (iv) theranostics applications. Finally, the challenges and future perspectives of their therapeutic application are discussed.

1. Introduction

The applications of nanotechnology in our lives are constantly increasing by way of innovations in design, synthesis, and functionalization of nanomaterials and nanostructures [1]. Those advancements have been applied in many fields to overcome past limitations and create new applications. The nano dimension brings new properties mainly because of the restriction in the electron motions of the particles and quantum confinement, which is also dependent on the material’s shape [2]. Some nanoscale-specific physiochemical properties include high surface area, outstanding electronic and optical properties such as localized surface plasmon resonance (LSPR) for noble metal nanoparticles, high mechanical strength, ease of functionalization, enhanced thermal conductivity and magnetic properties [3]. These traits make nanoparticles a great choice for various applications [3]. Nanoparticles can be applied in many different aspects including electrochemical applications (as electrolytes in batteries) [4,5], catalytic applications [5], solar cells [6], biosensors [7,8], imaging [9,10], therapeutic purposes [11,12], biomolecules tracking [13,14], tissue engineering [15,16] and gene or drug delivery [17,18].
As one of the most important applications of nanotechnology, nano-drug delivery systems (nDDSs) have attracted remarkable attention. nDDSs have unique features such as easier cell uptake, controlled drug release, prolonged stability of drugs inside the cells, long-lasting circulation time, reduction in drug side effects, enhanced bioavailability and biocompatibility, the potency of targeted delivery, lower administration dose [19,20], controllable pharmacokinetics, and traceability of delivery system [21,22]. These systems could be designed in the form of liposome [23], micelles, polymers [24], polysaccharides [25], self-assembled peptides [26], dendrimers, silica-based nanomaterials, bioactive glasses [27], hydrogels, carbon-based nanomaterials [28], metal nanoparticles, recently exosomes [29] and several other forms which could be used for delivering both drugs and oligonucleotides (DNAs or RNAs) [30,31,32,33,34,35,36]. Some examples of commercially available drugs containing nDDSs are Doxil® (Doxorubicin, polyethylene glycol (PEG)ylated liposome) [19,37], Abraxane® (Paclitaxel, Polymer nanoparticle) [38,39], and Vivagel® (Dendrimer nanoparticle) [32,40]. There are also some nanoparticles used for delivery systems in clinical trials such as polymeric micelles (Paclitaxel, Phase II Trial) [41], dendrimers (Docetaxel, Phase II Trial) [42] and vaccination platforms for antigen delivery in cancer treatments [43].
nDDSs are considered systems in which therapeutic agents are introduced to the body with improved properties [44,45]. For example, P-glycoproteins which are expressed in cancer cells, pump antitumor drugs out of the cells. It has been evidenced that nDDSs can transfer anticancer drugs to the cell without being pumped out [22,46,47]. In addition, the blood–brain barrier (BBB), which does not allow drugs to pass through, can also be challenged by nDDSs [31,48]. Gulyaev et al. showed that doxorubicin with polysorbate 80-coated nanoparticles could be transported to BBB [49] via apo-lipoprotein E [50].
To understand the main mechanism in which nanomaterials can lead to good and efficient drug delivery, the interaction between nanomaterial and biological environment should be studied [22]. Therapeutic elements can be absorbed or attached to the surface or encapsulated/trapped inside the carrier. Covalent bonding, the strongest type of binding, is a strategy to control the administered dosage of the drug component [51]. Then, drugs can be released from nDDSs in response to the changes in the physiological environment such as temperature, pH, reducing nature, enzymatic activity or even concentration [52]. There are two possible mechanisms for enhancing drug availability at the targeted sites: (1) active delivery, in which some binding/recognition elements such as antibodies or aptamers are functionalized on the surface of carriers to develop a targeted DDS; (2) passive delivery, which only has the role of carrying drug particles through the reticuloendothelial system. Nano-drug delivery systems may be designed for both mechanisms [31,53]. The mechanism through which nanoparticles are passively delivered to the tumor site is called the enhanced permeability and retention (EPR) effect [54]. Due to the dysfunctionalities in vascular nature of solid tumors, the high molecular weight material such as nanoparticles will accumulate in the solid tumors and increase its permeability to drugs. This effect increases the nanodrugs entrance to the tumor sites [55]. This phenomenon, which was firstly discovered by Maeda in the mid-1980s, has different affecting factors such as the location and type of the tumor. For example, EPR effect in the brain tumors is very low due to the BBB presence [56]. Furthermore, high tumor interstitial fluid pressure (IFP), poor blood flow inside tumors, and abnormal vascular distribution compromise the EPR effect [57,58].
Among different conventional used nanomaterials, gold nanostructures have attracted great interest, making them one of the most important platforms in nanobiotechnology and nanomedicine [59,60]. The significance of gold nanomaterials in medicine originates from their optical properties, which strongly depends on their size, aspect ratio and shape [61,62]. Due to the unique optical properties of gold nanomaterials, their ease of functionalization, and their high biocompatibility, gold nanomaterials have been increasingly used in medical applications, with potential use as carriers [62,63] for delivery of antigens for vaccination [64], gene delivery [65], other therapeutic targets [66,67]. Rod-shaped gold nanoparticles (gold nanorods or GNRs) have unique properties useful in medical applications. In this review, the synthesis methods and the properties of the GNRs are introduced and their therapeutic applications (drug/gene delivery, photothermal/photodynamic therapy, and theranostics) are focused and the challenges and future perspectives of their therapeutic application are discussed. Scheme 1 is representing general applications and properties of the GNRs discussed in this review.

2. Properties and Synthesis of GNRs

Gold nanorods are tiny rod shape structures with unique physical, optical and chemical features that make them favorable candidates in medical research [68]. The optical property of these tiny nanostructures returns to LSPR [69,70], which is an extraordinary optical property of metal nanoparticles induced by an incident electromagnetic wave in a particular wavelength; hence, it leads to collective oscillation of electrons [71,72]. Anisotropic structure of GNRs exhibit two surface plasmon bands in the UV–Vis spectrum, which are related to surface electron oscillation toward the transversal side of the particle and longitudinal side of GNRs [73]. Their ability to bind amine and thiol groups of other biomolecules facilitates their functionalization and bioconjugation processes and makes them ideal for use as nanocarrier systems [70,74].

2.1. GNR Synthesis Methods

One of the other advantages of GNRs is related to their accessibility and synthesizing of particles. Herein, there are quite number of synthesis methods which are divided into three group methods including chemical, physical and green synthesis method [62]. Despite of recent interests to replace more eco-friendly methods with less deposit problems and less toxicity, some limitations due to size controllability and synthesis yield led to those particularly chemical methods, are mostly preferred for GNRs synthesis among other types of chemical such as photochemical or electrochemical methods [62]. Electrochemical, seed-mediated, and synthesis with AgNO3 are the most common GNR synthesis methods. Electrochemical method has a high-quality yield; seed-mediated is the most favorable method with high yield, monodispersity, and ease of fabrication; AgNO3 method has up to 50% yield and is affected by many variable factors [75]. Unknown mechanism for some of the synthesis methods and their dependency on the environmental factors could negatively affect the reproducibility and controllable high yield production of GNRs [75] for industrial purposes as a main market limitation. The method called “seed-mediated growth” has been the most common method of producing colloidal GNRs (in 20–100 nm size range) since the 1920s [62]. To explain, GNRs are sequentially synthesized in a way in which Au salts are first reduced by reductants such as Citrate, forming spherical gold particles (seeds). Then, in a second step, adding reducing agents will induce assembling the gold spherical particles to produce gold nanorods as well [76]. Advances in synthesis could lead to the introduction of a new seed-mediated synthesis protocol that was proposed by Murphy et al. for the first time, by adding a kind of surfactant, so-called cetyltrimethylammonium bromide (CTAB), that clearly induces the zipping mechanism, aiming to achieve quite good rod morphology and better aspect ratio of GNRs. In this regard, the structural modification of GNRs is due to the fact that the CTAB works as a template for one-dimensional growth of these nanoparticles. Other surfactants will also affect the structural modifications [77]. Since then, different versions of optimizations have been published, such as the optimized protocol by Tohidi Moghadam et al. in Iran [78].

2.2. Photothermal Effect

GNRs have unique physicochemical properties, but undoubtedly, the most distinctive characteristics are due to their photothermal therapy effect, which can be defined as converting NIR radiation (650–900 nm) to heat energy [72]. Therefore, local heat generation leads to provoking cell death mainly via apoptosis and necrosis, even in deeper sides of soft tissues, especially for cancer therapy. Indeed, in recent years GNRs have been widely implemented in multiplexed cancer studies and could be useful in combating barriers of conventional cancer medications [72,79,80]. Recent studies have reported that some parameters such as aspect ratio of nanoparticles (length/width of a particle), radiation wavelength (Selected wavelength of NIR radiation raging 700–2000 nm), and coating (Covering agent of particle) can directly control the depth of penetration and efficacy of irradiation. For example, several experimental investigations have emphasized that 808 nm wavelength is an optimal point of laser radiation [81,82].

2.3. Toxicity Related to GNRs’ Surface Modifications

Nanoparticles such as GNRs are novel advanced materials, so their toxicity and safety issues should be under consideration by researchers. Accumulation, uptake, pharmacokinetics, and clearance are effective factors in toxicity analysis. Since these factors are different between nanomaterials and biomolecules, the biocompatibility and toxicity should be studied [83]. In terms of accumulation, nanoparticles are opsonized by resident macrophages in the tissues and then highly present in some healthy organs such as liver and spleen. This nonspecific presence and distribution may have toxicity and side effects on these healthy organs [84]. Uptake is basically the regulated cellular uptake (nanoparticles entrance to the cells) by mechanisms such as receptor-mediated endocytosis [85]. The pharmacokinetics feature represents the profile of nanoparticles’ bio distribution and clearance among time and the volume of utilized nanoparticles [86]. The final term, clearance, is the deletion of nanoparticles from the systematic circulating blood vessels [87]. Studies have shown that the surface modification of GNRs has the most impactful effect on their cytotoxicity. In research conducted by Alkilany et al., the toxicity of GNRs coated with polyelectrolytes (PE-GNRs) and polyethylene glycol (PEG-GNRs) in the therapeutic concentration of 1 nM were investigated on the vascular endothelial and smooth muscle cells, which were isolated from rat aortic rings. The results showed that PE-GNRs were toxic for vascular endothelial cells but not for the smooth muscle cells, while PEG coated nanorods did not have a toxic effect on vascular endothelial and smooth muscle cells. PE-GNRs cannot harm smooth muscle cells because the endothelium layer and sub-endothelial connective tissue are separated from direct contact with the GNRs. The uptake of PE-GNRs in vascular endothelium rather than PEG-GNRs is one the main reasons make it toxic for these cells. PE-GNRs induce significant changes in the vasorelaxation response which was measured by the tension changes in the aortic ring. Furthermore, the nitric oxide increase (as an inflammation indicator), and microscopic studied of the cells have shown toxic effects on vascular cells [88].
In the case of direct injection of GNRs into the blood vessels, their toxicity should be considered cautiously. Additionally, blood vessels are a route most nanoparticles come through during their transport in the body. Due to the small size of nanoparticles, their wide bio distribution in the body negatively affects the hemocompatibility in two ways: affecting the coagulations pathways and changing the blood cellular fates; disrupting the hemostatic balance, disseminated intravascular coagulopathy (DIC), and deep vein thrombosis (DVT) are hazardous effects of nanoparticles on hemocompatibility [89]. Mitochondria is one of the cell components which can be highly affected by the nanoparticles and changes the fate of the cells. In a study conducted by Nunes et al., the effect of CTAB-GNRs on mitochondrial integrity was evaluated. CTAB-GNRs decrease the respiratory ratio and phosphorylation pathways in the mitochondria and disrupt the electron chain transport. These results validate the toxicity of CTAB-GNRs by affecting the mitochondrial integrity [90].
On the other hand, the more a nanoparticle is used, the more environmental contamination it will have. So, in various research, the effect of GNRs on aquatic organisms was studied. Fish are the most used animal models in the environmental contamination investigations. In a study conducted by Souza et al., after the fish were subjected to sub-lethal exposure to the CTAB-GNRs (LC50 = 0.03 mg/L), the fish model showed a decrease in the neonate and an increase in the reactive oxygen species (ROS) level, which shows the increased toxicity levels in the fish [91]. In another similar study, conducted by Mesquita, after fish were exposed to CTAB-GNRs, no DNA damage was seen in the fish model, while malformation in the body development was observed even in the low doses (LC50 = 110.2 μg/L) [92].
To compare the toxicity effect of shape and coating, GNRs and gold nanoparticles (GNPs) with two different coatings, (polyethylene imine (PEI) and bovine serum albumin (BSA)), were used. Here, four different types of nanomaterial (PEI-GNRs, BSA-GNRs, PEI-GNPs, and BSA-GNPs) were synthesized, and their effects in different biological liquids such as serum, buffer, and cell culture media were investigated. It was showed that GNPs are less toxic and more biocompatible than GNRs. Since the surface functionalization of GNRs is critical in their cytotoxicity, this research proved that PEI-GNRs are nontoxic and safe and could easily penetrate normal and cancerous cells without damaging them [93].

2.4. The GNRs–Proteins Interaction

Since active biological material has a strong structure–function relationship, an appropriate carrier for drug delivery systems should also have no effect on the cargo’s structure. Since protein-based drugs could be one of the GNR’s cargos in nDDSs, investigating the effects of nanoparticle morphology on the protein’s structure is also important. GNRs and GNPs were both decorated with BSA, which showed that BSA’s biological activity was lost when it was joined with GNRs, while it did not change with the GNPs [94]. Thus, it seems that GNRs could unfold the protein cargo, while GNPs do not have such an effect, and therefore, GNPs could be better options for protein cargos.

3. GNRs for Therapeutic Applications

GNRs can be functionalized and simultaneously loaded by drugs or genes, acting as promising nanocarriers for targeted delivery of cargo. The load in such systems can be released in the designated location through the heat generation following NIR radiation. Thus, GNRs can be applied in enhancing the efficiency of targeted treatments. They are also considered as promising photothermal therapy agents. In this section, we have reviewed recent applications of GNRs for therapeutics purposes in three categories: drug delivery, gene delivery, and hyperthermia/photodynamic therapy.

3.1. GNRs for Drug Delivery Systems

There are quite many numbers of research investigating GNRs application in drug delivery systems for various purposes. Zhang et al. designed a nanocarrier for targeted drug delivery, responsive to pH and NIR, by conjugating a thiolated pH-responsive DNA to GNRs. A thiolated poly (ethylene glycol)-biotin was used to passivate the nanocarrier to provide a particular binding to cancer cells that showed over-expression of biotin receptors. Doxorubicin (DOX) was then intercalated in the double-stranded pH-responsive DNA (Figure 1A). Lower pH environment (pH~5) and/or 808 nm radiation resulted in the effective release of DOX. It was reported that the designed nanomedicine improved cell uptake remarkably and reduced drug efflux by the multidrug-resistant (MDR) breast cancer cell lines [95].
GNRs can also be used for improving skin permeability of proteins in the case of transdermal protein delivery, which can be an interesting method for therapeutic applications or for vaccination purposes. Haine et al. developed a method to enhance transdermal protein delivery by conjugating a polysaccharide-based hydrogel containing fluorescein isothiocyanate-modified ovalbumin (FITC-OVA) with GNRs on its surface. Using GNRs’ photothermal property generated heat due to a continuous-wave laser irradiation that led to an increase in the skin temperature and thus facilitated the protein’s permeation [96].
Figure 1. (A) Schematic drawing of the pH and NIR-responsive nDDS. Cancerous cells with overexpressed biotin receptors will uptake the Biotin-PEG-GNR-DNA/DOX system and enter its endosomes. Following the maturation of the endosomes, the pH will decrease, and thus DOX will be released. The system can alternatively be stimulated by NIR radiation for a local release. Reprinted from [95] with permission from Elsevier, Amsterdam, Netherlands. (B) The schematic representation of tri-sensitive nDDS for simultaneous photothermal and photodynamic therapy and fluorescence tracing for HER2/CD44 positive breast cancer cells. Reprinted from [97] with permission from Elsevier, Amsterdam, Netherlands.
Figure 1. (A) Schematic drawing of the pH and NIR-responsive nDDS. Cancerous cells with overexpressed biotin receptors will uptake the Biotin-PEG-GNR-DNA/DOX system and enter its endosomes. Following the maturation of the endosomes, the pH will decrease, and thus DOX will be released. The system can alternatively be stimulated by NIR radiation for a local release. Reprinted from [95] with permission from Elsevier, Amsterdam, Netherlands. (B) The schematic representation of tri-sensitive nDDS for simultaneous photothermal and photodynamic therapy and fluorescence tracing for HER2/CD44 positive breast cancer cells. Reprinted from [97] with permission from Elsevier, Amsterdam, Netherlands.
Pharmaceutics 14 00664 g001
Using the GNRs’ photothermal effect, nanosystems can be produced for photothermal therapy or drug delivery systems. Niidome et al. designed an NIR-responsive system by modifying GNRs with poly (N-isopropyl acrylamide) gel. When the system was irradiated, the photothermal effect of GNRs led to a phase transition effect that converted the polymer layer to be hydrophobe, so the whole system shrank and accumulated in the irradiated tissue [98].
Innovative targeted and sustained drug releasing systems have also been integrated with GNRs relying on their electrical properties to achieve a natural heart-similar electrical conductivity. Since these particles could improve the conductivity of the hydrogels, Chen et al. designed an injectable bioactive hydrogel with high drug loading efficiency for myocardial infarction treatment [99]. They incorporated Astragaloside IV nanoparticles and GNRs as drug and conductivity enhancers, respectively, in a hydrogel formed from polyethylene glycol diacrylate/4-vinyl phenylboronic acid (PEGDA-PBA) and thiol hyaluronic acid (HA-SH). The system showed effectiveness in improvement of cardiac dysfunction and cardiac remodeling [99].
GNRs could also be coupled with fluorescent particles to design traceable drug delivery systems. These nanoparticles were used to design a system with photothermal and photodynamic therapeutic properties and traceability through fluorescent imaging. Photodynamic therapy is a two-step treatment in which a photosensitizer is activated by light stimulation, produces ROS that causes cell death. In their designed system, gold nanorods functionalized with hyaluronic acid, 5-aminolevulinic acid (ALA) and Cy 7.5 were used for photothermal treatment, photodynamic treatment, and fluorescent imaging. After entering the target cells, this system is destroyed in response to hyaluronidase, acidic pH and glutathione within the cell, and as a result of the separation of its components, the mentioned properties appear (Figure 1B) [97].
Herein, a comparative summary of the GNR-based nDDS is provided in Table 1.
An important property for materials to be used in drug delivery systems is non-(or low) toxicity. As described before, there are also efforts to reduce GNR’s toxicity and make them more appropriate for medical applications. PEG is widely used in this field, since it both improves GNRs’ stability and prevents the immuno-uptake of these nanomaterials [133]. In research by Niidome et al., mPEG-SH was added to surfactant-stabilized GNRs (hexadecyl trimethyl ammonium bromide or CTAB surfactant) to reduce its cytotoxicity. Intravenous injection of the PEG-GNRs to the mice showed that 54% of the injected material was found in the blood rather than accumulation in the liver, unlike the original CTAB-GNRs [134]. However, their method resulted in stability issues of some GNRs, which were later addressed by additional steps in the ligand exchange process, such as using Tris-HCl buffer [135] and ethanol-water mixture [136].
According to Table 1, the photothermal effect of GNRs to convert NIR to heat, is the most applied GNRs’ feature in drug delivery or therapeutic systems. Considering the systems’ functional mechanism, this property is mostly used for tissue hyperthermia in photothermal therapies or a local drug release [95,112]. This feature is also effective in increasing the cytotoxicity of drugs such as DOX [107].
Coupling GNRs with materials such as mesoporous silica would keep GNRs from aggregation and enhance the system’s drug loading capacity [137]. In many designed systems, the Au-core provides stability, and the monolayer shell tunes the desired surface features such as charge or hydrophobicity. The disadvantages of GNRs, such as high aggregation rate and short blood circulation lifetime, could also be improved by modifying them with polymeric materials [108]. Additionally, GNRs are promising candidates for theranostic applications, due to their imaging and thermal-induced therapy features. [117].
In general, to come up with the best choices for a gold nanoparticle-based drug delivery system design, investigation of the cell penetration rate of the carrier and the factors affecting it (e.g., surface chemistry, functionalization, geometry, morphology, and size), the assessment of the carrier’s effects on the cargo, and the induced biological responses is highly crucial. The complex behavior of GNRs and GNPs was studied by Pyshnaya et al. The study revealed that cell penetration of both positively charged GNRs and GNPs was better than negatively charged ones. It was also indicated that the initial surface charge was even more important than their final charge in the biological fluids. Therefore, it is assumed that GNRs and GNPs have almost similar cell uptake rates (natural and cancerous cells) based on their functionalization [93]. However, the surface charge does not seem to be the only effective factor. GNRs and GNPs were also compared for penetration in antigen-presenting cells (APCs) and humoral response in a study by Niikura et al. Despite the higher antibody titer against GNPs, GNRs had more cell uptake due to the higher cell-entrance to cell-exit ratio. In addition, other evidence such as different cytokine measurements indicated that GNRs had the lysosomal escaping ability and activated the inflammasome pathways, while GNPs readily exit the cell after the entrance. In other words, the morphology of gold nanostructures (GNPs/GNRs) could affect immune responses via activating nano inflammasome dependent manner [138].

3.2. GNRs for Gene Delivery Systems

GNRs could also be applied for targeted oligonucleotide delivery [139]. For example, DNA and RNA strands are stabilized on the surface of GNRs in different nanosystem designs for brain drug delivery. GNRs have been applied in a nanosystem designed for glioblastoma therapy by combining thermal therapy and inhibiting a protein expression, which is expected to make the glioblastoma more sensitive to chemotherapy [131]. The system consists of a liposome with a cationic-charged surface, modified by angiopep-2 peptide and loaded internally with DOX (a chemotherapeutic agent) and externally with GNRs (photothermal therapeutic agents) and yes-associated protein siRNA (YAP-siRNA), responsible for inhibiting the expression of YAP protein (Figure 2A). The system showed good tumor growth inhibition effects and could pass the blood–brain barrier [131].
GNRs have also been found useful in successful small interfering RNA (siRNA) delivery in cancer treatments. The application of siRNAs as important cancer treatment agents due to the ability to suppress protein expressions is challenging, since they have poor cellular uptake and rapidly degrade when exposed to RNase and serums or endosomes inside the cells. A nano siRNA-delivery system has been designed by assembling chitosan on GNRs (Figure 2B). This system had sufficient cargo protection, successful cell and tissue delivery and good endosomal escape, thus effective in cancer suppression. The photothermal property of GNRs was also helpful in enhancing the treatment efficacy [140]. Having a remarkable photothermal property, GNRs are now considered promising candidates for designing controlled-release systems that can be activated by near-infrared radiation. Shuji Yamashita et al. developed such a system by modifying dsDNA on GNRs. Irradiating GNRs by NIR resulted in the release of ssDNA due to the photothermal effect, during which the amount of release could be controlled by the power and time of radiation [141].
Combinational systems for gene delivery, PTT and thermal imaging are also achievable using GNR. In a recent study GNRs were grafted by poly(amidoamine) dendrimers and modified by GX1 peptide to design a system for simultaneous gene delivery (FAM172A gene), photothermal therapy and thermal imaging for tumor treatment [142]. A summary of the research in gene delivery applications of GNRs is provided in Table 2.
According to the above table (Table 2), the most important and the only reason for using GNRs in gene delivery applications is their photothermal effect. In addition, siRNAs and shRNAs are the most used nucleic acid-based cargoes in GNRs delivery systems. This may be due to the silencing ability of these cargoes for disease treatment, which has fewer unknown side effects than permanent DNA therapies [143]. One of the widely used nanoparticles with similar morphology to GNRs, is carbon nanotubes (CNTs). GNRs and CNTs are both one-dimensional nanoparticles with a high ability to bind to nucleic acids. They have high surface-to-volume ratios, ease of functionalization, and optical and conductive abilities. Additionally, they both represent a good photothermal effect. Their differences are that CNTs are hollow cylinder-shaped carbon-based nanoparticles while GNRs are filled and solid cylinder-shaped metal nanoparticles. Nucleic acids are mostly absorbed physically and electrostatically to the CNTs, while in GNRs delivery systems, covalently bonded nucleic acids are used with the same amounts as the electrostatically bonded ones. One important issue that limits the clinical use of CNTs in comparison with GNRs is their nanotoxicity or environmental effects [144]. Fabricating hybrid structures based on GNRs and CNTs could be an intriguing and novel delivery platform in future gene delivery systems [145].
Table 2. GNRs applications in gene delivery systems (research-based studies).
Table 2. GNRs applications in gene delivery systems (research-based studies).
PurposeSystem’s StructureMechanism of FunctionGNR’s Applied FeatureRef.
RnDDendrimer coated GNRsDelivery and enhanced expression of the brcaa1-shRNA to the targeted cells upon NIR irradiationPT[146]
DnDDisulfide cross-linked polyethyleneimine-conjugated GNRs grafted by PEG and RGD peptideGene release in response to high glutathione concentration in target cells and NIR irradiationPT[147]
RnDLayer-by-layer assembled chitosan-GNRsDelivery of siRNAs, accumulation in tumor tissue, NIR-mediated photothermal ablationPT[140]
DnDCationic-charged surfactant and DNA modified GNRsDNA release as a result of Photothermal hyperthermiaPT[148]
Drug +siRNA co-deliveryDOX, YAP-siRNA and GNRs loaded cationic liposomeTargeted chemo, PT and gene combination therapy using NIR irradiationPT[131]
DnDGNRs grafted with Poly(amidoamine) dendrimers and modified by GX1 peptide, FAM172A geneDNA release and PTT as a result of Photothermal hyperthermiaPT[142]
Abbreviations: DnD: DNA delivery, RnD: RNA delivery.

3.3. GNRs for Photothermal/Photodynamic Therapy

Therapeutic applications of GNRs are mainly based on their photothermal effect. The photothermal effect of GNRs can deteriorate tumors and cancerous cells using local irradiated heating, called photothermal therapy (PTT). Moreover, it can occur due to the singlet oxygen generation, called photodynamic therapy (PDT) [11,72]. In the last few decades, numerous studies have been carried out on this topic to improve fabrication and assemble optimal photothermal systems. Conventional methods of hyperthermia affect both tumors and healthy cells, so they have lower specificity. In addition, a temperature gradient forms on the way to the tissues, which provides a more severe issue for deep tumors. Nanoparticle-based hyperthermia can facilitate these limitations by targeting the tumor tissue and then generating local hyperthermia. Among the most essential nanostructures capable of generating hyperthermia, such as gold nanoparticles, magnetic nanoparticles (MNPs), CNTs, and ZnCuO [149], gold nanorods have remarkable capabilities to make them ideal for thermal therapy [150].
A comparison of gold nanorods with gold spherical nanoparticles shows that although gold spherical nanoparticles (GNPs) have the photothermal effect, they have several limitations. Indeed, GNPs not only have relatively low heating efficiency, but also the wavelength at which their resonance occurs, located in the mid-visible region that is outside of the “tissue window”. Therefore, the use of gold nanorods is a significant advantage because they are more efficient photo converters to heat and can coordinate the maximum heat wavelength within the tissue window [151]. However, several studies have been conducted on the PTT applications by means of GNPs [152,153].
In this case, MNPs have also the great potential of heat generation. However, the effectiveness of MNP hyperthermia treatment greatly depends on some magnetic parameters, such as alternating magnetic field (AMF), intensity of the AMF, penetration depth. For example, exposure to a high frequency alternating magnetic field is needed to convert electromagnetic energy into heat energy. Moreover, bio functionalization in addition to the targeted delivery, plays an essential role in generating the thermal stability of the particles. The targeted heating of MNPs is lower than GNRs, due to the fact that biomolecules are often physically bonded to MNPs. In contrast, GNRs could covalently and electrostatically be functionalized with a variety of biomolecules. Thus, bio functionalization is mostly less stable, and the heating damage of healthy tissue or overheating is possible. Recent studies in this area have focused on proposing next generations of MNPs such as superparamagnetic iron oxide nanoparticles (SPIONs), due to their unique optical and magnetic properties that help to decrease the overheating risks and enhance the heating efficacy. In addition, according to the highlighted optical properties of CNTs, they also express great potential of tumor ablation in both targeted and non-targeted manner. Despite the advantages of CNTs, the safety aspects of using them in vivo, including toxicity and different antitumor mechanisms of action, are still under investigation [154].
Several studies in the last two decades showed that photothermal therapy can be implemented for different sort of cells as well. In this case, Pissuan et al. could empirically assemble the system using functionalized GNRs with anti-CD11b (Tachyzoite surface antigens) to targeted delivery of a nanosystem. Thus, increasing the temperature showed promising results in killing the active protozoans, hampering the parasite life cycle (Figure 3A) [155].
In another study, Lee et al. have designed a new kind of virus-simulated system, using necotin receptor peptides (RVG) as coverage of PEGylated silica-coated gold nanorods (SiO2@GNRs-PEG) nano assembly, not only to transport the system through the blood–brain barrier but also to inhibit the growth of glioma tumor using hyperthermia. Results showed that implementing the system could lead to locally increasing the temperature above 50 centigrade just after 5 min of NIR exposure [156].
It is noteworthy that despite the number of studies focusing on GNR-mediated PTT, the research has not yet reached the end. In the most recent study in 2021, on hospitalized coronavirus patients, scientists tried to use a more external and less invasive medication procedure for patients with severe respiratory complications. So, functionalized GNRs with angiotensin-converting enzyme 2 (ACE2) have been chosen at the first line of treatment to degenerate targeted cells, inducing apoptosis or necrosis [157]. Additionally, in another study, by Meng et al., persistent luminescent nanoparticles (PLNPs)-conjugated GNRs have been used, forming a nanocomposite platform (PLNP-GNRs) which were encapsulated by biocompatible PW12 molecules (phosphotungstate derivatives). The results revealed that GNRs can effectively act as a promising photothermal conversion agent, using single 635 nm wavelength, for targeted photothermal therapy of deep-tissue tumors, particularly in skin and breast cancer cases (Figure 3B) [158].
Table 3 summarizes some of the photothermal therapeutic systems in which GNRs show photothermal effects.
Figure 3. (A) Schematic figure of GNRs attachment to targeted tachyzoit cells. Reprinted from [155] with permission from ACS Publications, Washington, DC, USA. (B) Scheme of PLNP-GNRs biocompatible nanocomposite platform for NIR-induced photothermal tumor therapy. Reprinted from [158] with permission from Elsevier, Amsterdam, Netherlands.
Figure 3. (A) Schematic figure of GNRs attachment to targeted tachyzoit cells. Reprinted from [155] with permission from ACS Publications, Washington, DC, USA. (B) Scheme of PLNP-GNRs biocompatible nanocomposite platform for NIR-induced photothermal tumor therapy. Reprinted from [158] with permission from Elsevier, Amsterdam, Netherlands.
Pharmaceutics 14 00664 g003

4. GNRs for Theranostics; Combination of Diagnostic and Therapy

The integration of diagnostic imaging and therapy, called theranostics, has emerged as an advancing field for high-performance cancer treatment. Hence, the majority of current studies have been reported on developing potential cancer theranostic agents. According to the imaging techniques, bio-imaging is universally preferred, as it is a non-invasive procedure for in situ visualization and offers real-time tracking of biological processes. So, GNRs are an excellent candidate for bio-imaging and photothermal therapy, due to the capability of local heat generation, triggered by laser beam [165]. GNRs can be applied solely or simultaneously by other imaging techniques such as PA imaging and positron emission tomography (PET) to improve both the imaging’s resolution and image-guided therapy [166,167,168,169,170,171]. Herein, Zhang and his colleagues designed a nanosystem as a theranostic platform by using chlorin e6 (fluorescent tag)-conjugated gold nanorods (Ce6-PEG-GNRs), resulted in enhancing the efficiency of therapy, because of dual PTT/PDT, and precisely-tumor tracking of nanosystem by pH-responsive fluorescent and IR imaging (Figure 4A) [72]. The combination of multimodal therapeutic method (photothermal, photodynamic and chemotherapy) and diagnosis was also shown in another study in which two different wavelength laser radiation (660 nm, 808 nm) were used to elevate the effect of treatment (Figure 4B) [172]. Drug delivery potency of GNRs can also be integrated with biomarkers to track a drug’s local release. Guo et al. designed such a system for targeted delivery of DOX and detection of intracellular ATP. The system was composed of GNRs, modified by three types of DNA (DNA1: complementary DNA sequence for ATP aptamer, DNA2: Ramos cell aptamer, DNA3: ATP aptamer) and loaded by DOX. The immobilization of ATP aptamers and Ramos cell aptamers made GNRs capable of specific recognition of tumor sites. The presence of ATP caused the release of DOX from the nDDS and increased its fluorescence intensity related to the ATP concentration (Figure 4C) [173].
The application of gold nanorods in some of the theranostic systems are mentioned in Table 4.

5. Conclusions

This study focused on the properties, synthesis, and applications of gold nanorods (GNRs), as promising candidates for utilization in nano-drug delivery systems (nDDS), photothermal therapy, rapid diagnostic and imaging, and theranostics applications. Among these applications, the capacity of GNRs in converting irradiation into heat has been considered the most distinctive feature in photothermal therapeutic strategies and localized drug release. This feature is also shown to be effective in drug diffusion and increase the cytotoxicity of drugs such as doxorubicin. Moreover, studies have shown the possibility of using an efficient hybrid system of GNRs with a wide range of materials such as SiO2, nucleic acids, polyvinyl alcohol, chitosan, magnetic ionic liquids, hyaluronic acid, graphene oxide, polymers, peptides and potential drugs.
Nevertheless, the interaction between GNRs and different chemical/biological elements and the possibility of unfavorable, irreversible changes in their physicochemical characteristics is an important issue that needs deep research and ex/in vivo experiments for nDDS and other potential applications, i.e., nanobiosensing and regenerative medicine. Manageable surface chemistry, high stability (less agglomeration and corrosion), low cytotoxicity and high biocompatibility, simple synthesis protocols with reproducibility in scaling up, strong signals for bioimaging, sustainability in delivery, and cost-effectiveness are also other concerns that need to be addressed in designing such hybrid systems.
Today, nDDS has shown to be a promising strategy for diverse applications. However, there are serious concerns in a biological medium such as immune response elicitation, the toxicity of nanoparticles, enhancement of the capacity and efficiency of drug loadings, unpredictable degradation patterns of nanoparticles inside the body, and possible environmental contaminations. Therefore, apart from optimizing synthesis protocols and stabilizing functional nanocarrier systems, understanding their biological effects/interactions in the host system would be important.
The anisotropic morphology of GNRs provides them a good potential for biomedical usage. Besides their applications in the field of drug delivery, developing new electrochemical and optical sensors are promising areas in the future of GNRs. Photothermal and photodynamic therapy based on the GNRs are also important especially due to their usage in the tumor ablation applications. Furthermore, current positive results in clinical trials based on plasmonic photothermal therapy of atherosclerosis with GNPs show that market of GNRs will be a bright one in the biomedical field and overtake the rivals, because of the highlighted GNRs privileges among other plasmonic nanoparticles such as silver or even gold nanoparticles. Despite the presence of two drug formulations based on the gold nanospheres, currently in clinical trial processes, there are no FDA-approved therapeutic formulations based on the GNR’s delivery system in the clinical trials until 2021. Aurolase is a core–shell silica gold nanoparticle (GNPs) coated with PEG and used for tumor ablation based on the photothermal effect of AuNPs (GNPs). Lung, head and neck, and prostate cancers are the diseases being investigated to treat solid tumors with this drug. NU-0129 is another drug consisting of spherical gold nanoparticles (GNPs) functionalized with nucleic acids, which targets the Bcl2L12 gene. This gene hampers the apoptosis in the glioblastoma cells, so blocking this gene through NU-0129 is a treatment for patients with recurrent glioblastoma or gliosarcoma. These cases show that GNRs could have good potential for future applications in gene delivery and photothermal applications.

Author Contributions

Conceptualization, investigation, writing—original draft preparation, writing—review and editing, A.J.-M., M.M. and S.T.; writing—review and editing, A.B. and A.Z. (Atefeh Zarepour), writing—review and editing, supervision, A.Z. (Ali Zarrabi), E.S. and M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

GNRGold nanorods
NIRNear-infrared
LSPRLocalized Surface Plasmon Resonance
nDDSsnano-Drug delivery systems
BBBBlood–brain barrier
CTABCetyltrimethylammonium bromide
PEPolyelectrolytes
PEGPolyethylene glycol
DICDisseminated intravascular coagulopathy
DVTDeep vein thrombosis
ROSReactive oxygen species
GNPsGold nanoparticles
PEIPolyethylene imine
BSABovine serum albumin
EPRPermeability and retention
IFPInterstitial fluid pressure
DOXDoxorubicin
MDRMultidrug-resistant
ALA5-Aminolevulinic acid
PTPhotothermal
DDDrug delivery
PDprotein delivery
PTTphotothermal therapy
PAPhotoacoustic
Coconductivity
CaCarrier
StStability
PDTphotodynamic therapy
ImImaging
GDGene delivery
ThTheranostic
siRNAsmall interfering RNA
YAPYes-associated protein
PLGAPoly(lactic-co-glycolic acid)
CNTsCarbon nanotubes
DnDDNA delivery
RnDRNA delivery
MNPsMagnetic nanoparticles
AMFAlternating magnetic field
ACE2Angiotensin-converting enzyme 2
TDTargeted delivery
PETPositron emission tomography
TRKTracking
PLPhotoluminescence

References

  1. Ranzoni, A.; Cooper, M.A. Chapter One—The Growing Influence of Nanotechnology in Our Lives. In Micro and Nanotechnology in Vaccine Development; Skwarczynski, M., Toth, I., Eds.; William Andrew Publishing: Norwich, NY, USA, 2017; pp. 1–20. [Google Scholar] [CrossRef]
  2. Huang, X.; Neretina, S.; El-Sayed, M.A. Gold Nanorods: From Synthesis and Properties to Biological and Biomedical Applications. Adv. Mater. 2009, 21, 4880–4910. [Google Scholar] [CrossRef]
  3. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
  4. Kar, M.; Simons, T.J.; Forsyth, M.; MacFarlane, D.R. Ionic liquid electrolytes as a platform for rechargeable metal–air batteries: A perspective. Phys. Chem. Chem. Phys. 2014, 16, 18658–18674. [Google Scholar] [CrossRef] [PubMed]
  5. He, Z.; Alexandridis, P. Ionic liquid and nanoparticle hybrid systems: Emerging applications. Adv. Colloid Interface Sci. 2017, 244, 54–70. [Google Scholar] [CrossRef] [PubMed]
  6. Zhao, Y.; Bostrom, T. Application of Ionic Liquids in Solar Cells and Batteries: A Review. Curr. Org. Chem. 2015, 19, 556–566. [Google Scholar] [CrossRef]
  7. Mousazadeh, M.; Jahangiri-Manesh, A.; Nikkhah, M.; Abbasian, S.; Moshaii, A.; Masroor, M.J.; Norouzi, P. Detection of hexanal gas as a volatile organic compound cancer biomarker using a nanocomposite of gold nanoparticles and selective polymers. J. Electroanal. Chem. 2021, 905, 115962. [Google Scholar] [CrossRef]
  8. Jahangiri-Manesh, A.; Mousazadeh, M.; Nikkhah, M.; Abbasian, S.; Moshaii, A.; Masroor, M.J.; Norouzi, P. Molecularly imprinted polymer-based chemiresistive sensor for detection of nonanal as a cancer related biomarker. Microchem. J. 2021, 173, 106988. [Google Scholar] [CrossRef]
  9. Su, X.; Fu, B.; Yuan, J. Gold nanocluster-coated gold nanorods for simultaneously enhanced photothermal performance and stability. Mater. Lett. 2016, 188, 111–114. [Google Scholar] [CrossRef]
  10. Naz, G.; Othaman, Z.; Shamsuddin, M.; Ghoshal, S.K. Gold nanorod libraries enhancement for optical imaging. Mater. Lett. 2016, 166, 63–66. [Google Scholar] [CrossRef]
  11. Yan, J.; Sun, H.; Li, J.; Qi, W.; Wang, H. A theranostic plaster combining photothermal therapy and photodynamic therapy based on chlorin e6/gold nanorods (Ce6/Au nrs) composite. Colloids Surf. A Physicochem. Eng. Asp. 2018, 537, 460–466. [Google Scholar] [CrossRef]
  12. Freitas, L.; Hamblin, M.; Anzengruber, F.; Perussi, J.R.; Ribeiro, A.; Martins, V.; Plepis, A. Zinc phthalocyanines attached to gold nanorods for simultaneous hyperthermic and photodynamic therapies against melanoma in vitro. J. Photochem. Photobiol. B Biol. 2017, 173, 181–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhou, J.; Cao, Z.; Panwar, N.; Hu, R.; Wang, X.; Qu, J.; Tjin, S.C.; Xu, G.; Yong, K.-T. Functionalized gold nanorods for nanomedicine: Past, present and future. Coord. Chem. Rev. 2017, 352, 15–66. [Google Scholar] [CrossRef]
  14. Kilin, V.; Anton, H.; Anton, N.; Steed, E.; Vermot, J.; Vandamme, T.F.; Mely, Y.; Klymchenko, A.S. Counterion-enhanced cyanine dye loading into lipid nano-droplets for single-particle tracking in zebrafish. Biomaterials 2014, 35, 4950–4957. [Google Scholar] [CrossRef] [PubMed]
  15. Taheri, R.A.; Akhtari, Y.; Moghadam, T.T.; Ranjbar, B. Assembly of Gold Nanorods on HSA Amyloid Fibrils to Develop a Conductive Nanoscaffold for Potential Biomedical and Biosensing Applications. Sci. Rep. 2018, 8, 9333. [Google Scholar] [CrossRef]
  16. Bigham, A.; Rahimkhoei, V.; Abasian, P.; Delfi, M.; Naderi, J.; Ghomi, M.; Moghaddam, F.D.; Waqar, T.; Ertas, Y.N.; Sharifi, S.; et al. Advances in tannic acid-incorporated biomaterials: Infection treatment, regenerative medicine, cancer therapy, and biosensing. Chem. Eng. J. 2021, 432, 134146. [Google Scholar] [CrossRef]
  17. Kesharwani, P.; Gorain, B.; Low, S.Y.; Tan, S.A.; Ling, E.C.S.; Lim, Y.K.; Chin, C.M.; Lee, P.Y.; Lee, C.M.; Ooi, C.H.; et al. Nanotechnology based approaches for anti-diabetic drugs delivery. Diabetes Res. Clin. Pract. 2018, 136, 52–77. [Google Scholar] [CrossRef]
  18. Kumar, B.; Jalodia, K.; Kumar, P.; Gautam, H.K. Recent advances in nanoparticle-mediated drug delivery. J. Drug Deliv. Sci. Technol. 2017, 41, 260–268. [Google Scholar] [CrossRef]
  19. Amirmahani, N.; Mahmoodi, N.O.; Galangash, M.M.; Ghavidast, A. Advances in nanomicelles for sustained drug delivery. J. Ind. Eng. Chem. 2017, 55, 21–34. [Google Scholar] [CrossRef]
  20. Ould-Ouali, L.; Noppe, M.; Langlois, X.; Willems, B.; Riele, P.T.; Timmerman, P.; Brewster, M.E.; Ariën, A.; Préat, V. Self-assembling PEG-p(CL-co-TMC) copolymers for oral delivery of poorly water-soluble drugs: A case study with risperidone. J. Control. Release 2005, 102, 657–668. [Google Scholar] [CrossRef]
  21. Kipp, J.E. The role of solid nanoparticle technology in the parenteral delivery of poorly water-soluble drugs. Int. J. Pharm. 2004, 284, 109–122. [Google Scholar] [CrossRef]
  22. Benita, S. Microencapsulation: Methods and Industrial Applications; CRC Press: Boca Raton, FL, USA, 2005. [Google Scholar]
  23. Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal Formulations in Clinical Use: An Updated Review. Pharmaceutics 2017, 9, 12. [Google Scholar] [CrossRef] [PubMed]
  24. Ramasamy, T.; Ruttala, H.B.; Gupta, B.; Poudel, B.K.; Choi, H.-G.; Yong, C.S.; Kim, J.O. Smart chemistry-based nanosized drug delivery systems for systemic applications: A comprehensive review. J. Control. Release 2017, 258, 226–253. [Google Scholar] [CrossRef] [PubMed]
  25. Caro, C.; Pozo, D. Polysaccharide Colloids as Smart Vehicles in Cancer Therapy. Curr. Pharm. Des. 2015, 21, 4822–4836. [Google Scholar] [CrossRef] [PubMed]
  26. Delfi, M.; Sartorius, R.; Ashrafizadeh, M.; Sharifi, E.; Zhang, Y.; De Berardinis, P.; Zarrabi, A.; Varma, R.S.; Tay, F.R.; Smith, B.R.; et al. Self-assembled peptide and protein nanostructures for anti-cancer therapy: Targeted delivery, stimuli-responsive devices and immunotherapy. Nano Today 2021, 38, 101119. [Google Scholar] [CrossRef] [PubMed]
  27. Sharifi, E.; Bigham, A.; Yousefiasl, S.; Trovato, M.; Ghomi, M.; Esmaeili, Y.; Samadi, P.; Zarrabi, A.; Ashrafizadeh, M.; Sharifi, S. Mesoporous Bioactive Glasses in Cancer Diagnosis and Therapy: Stimuli-Responsive, Toxicity, Immunogenicity, and Clinical Translation. Adv. Sci. 2022, 9, 2102678. [Google Scholar] [CrossRef]
  28. Ashrafizadeh, M.; Saebfar, H.; Gholami, M.H.; Hushmandi, K.; Zabolian, A.; Bikarannejad, P.; Hashemi, M.; Daneshi, S.; Mirzaei, S.; Sharifi, E.; et al. Doxorubicin-loaded graphene oxide nanocomposites in cancer medicine: Stimuli-responsive carriers, co-delivery and suppressing resistance. Expert Opin. Drug Deliv. 2022. accepted. [Google Scholar] [CrossRef]
  29. Butreddy, A.; Kommineni, N.; Dudhipala, N. Exosomes as Naturally Occurring Vehicles for Delivery of Biopharmaceuticals: Insights from Drug Delivery to Clinical Perspectives. Nanomaterials 2021, 11, 1481. [Google Scholar] [CrossRef]
  30. Demetzos, C. Advanced Drug Delivery Nanosystems: Perspectives and Regulatory Issues. GeNeDis 2014, 822, 195–198. [Google Scholar] [CrossRef]
  31. Nazarov, G.V.; Galan, S.E.; Nazarova, E.V.; Karkishchenko, N.N.; Muradov, M.M.; Stepanov, V.A. Nanosized forms of drugs (A Review). Pharm. Chem. J. 2009, 43, 163–170. [Google Scholar] [CrossRef]
  32. Arias, J.L. Porous Silica Nanoparticles for Drug Delivery and Controlled Release. In Nanotechnology and Drug Delivery; CRC Press: Boca Raton, FL, USA, 2014; Volume 1, pp. 301–338. [Google Scholar]
  33. Mohammadinejad, R.; Dehshahri, A.; Madamsetty, V.S.; Zahmatkeshan, M.; Tavakol, S.; Makvandi, P.; Khorsandi, D.; Pardakhty, A.; Ashrafizadeh, M.; Afshar, E.G.; et al. In Vivo gene delivery mediated by non-viral vectors for cancer therapy. J. Control. Release 2020, 325, 249–275. [Google Scholar] [CrossRef]
  34. Delfi, M.; Ghomi, M.; Zarrabi, A.; Mohammadinejad, R.; Taraghdari, Z.; Ashrafizadeh, M.; Zare, E.; Agarwal, T.; Padil, V.; Mokhtari, B.; et al. Functionalization of Polymers and Nanomaterials for Biomedical Applications: Antimicrobial Platforms and Drug Carriers. Prosthesis 2020, 2, 117–139. [Google Scholar] [CrossRef]
  35. Assadi, Z.; Emtiazi, G.; Zarrabi, A. Hyperbranched polyglycerol coated on copper oxide nanoparticles as a novel core-shell nano-carrier hydrophilic drug delivery model. J. Mol. Liq. 2018, 250, 375–380. [Google Scholar] [CrossRef]
  36. Jahandar, M.; Zarrabi, A.; Shokrgozar, M.A.; Mousavi, H. Synthesis, characterization and application of polyglycerol coated Fe3O4nanoparticles as a nano-theranostics agent. Mater. Res. Express 2015, 2, 125002. [Google Scholar] [CrossRef]
  37. Mishra, A.K. Nanomedicine for Drug Delivery and Therapeutics; John Wiley & Sons: Hoboken, NJ, USA, 2013. [Google Scholar]
  38. Tacar, O.; Sriamornsak, P.; Dass, C.R. Doxorubicin: An update on anticancer molecular action, toxicity and novel drug delivery systems. J. Pharm. Pharmacol. 2013, 65, 157–170. [Google Scholar] [CrossRef] [PubMed]
  39. Chen, H.; Huang, X.; Wang, S.; Zheng, X.; Lin, J.; Li, P.; Lin, L. Nab-paclitaxel (abraxane)-based chemotherapy to treat elderly patients with advanced non-small-cell lung cancer: A single center, randomized and open-label clinical trial. Chin. J. Cancer Res. 2015, 27, 190–196. [Google Scholar] [CrossRef] [PubMed]
  40. Ma, P.; Mumper, R.J. Paclitaxel nano-delivery systems: A comprehensive review. J. Nanomed. Nanotechnol. 2013, 4, 1000164. [Google Scholar] [CrossRef] [Green Version]
  41. Lee, S.-W.; Kim, Y.-M.; Cho, C.H.; Kim, Y.T.; Kim, S.M.; Hur, S.Y.; Kim, J.-H.; Kim, B.-G.; Kim, S.-C.; Ryu, H.-S.; et al. An Open-Label, Randomized, Parallel, Phase II Trial to Evaluate the Efficacy and Safety of a Cremophor-Free Polymeric Micelle Formulation of Paclitaxel as First-Line Treatment for Ovarian Cancer: A Korean Gynecologic Oncology Group Study (KGOG-3021). Cancer Res. Treat. 2018, 50, 195–203. [Google Scholar] [CrossRef]
  42. Docetaxel (Taxotere®). Available online: https://starpharma.com/drug_delivery/dep_docetaxel (accessed on 12 March 2022).
  43. Grippin, A.J.; Sayour, E.J.; Mitchell, D.A. Translational nanoparticle engineering for cancer vaccines. OncoImmunology 2017, 6, e1290036. [Google Scholar] [CrossRef] [Green Version]
  44. Michaelis, K.; Hoffmann, M.M.; Dreis, S.; Herbert, E.; Alyautdin, R.N.; Michaelis, M.; Kreuter, J.; Langer, K. Covalent linkage of apolipoprotein e to albumin nanoparticles strongly enhances drug transport into the brain. J. Pharmacol. Exp. Ther. 2006, 317, 1246–1253. [Google Scholar] [CrossRef] [Green Version]
  45. Rowland, M.; Noe, C.R.; Smith, D.A.; Tucker, G.T.; Crommelin, D.J.; Peck, C.C.; Rocci, M.L., Jr.; Besançon, L.; Shah, V.P. Impact of the pharmaceutical sciences on health care: A reflection over the past 50 years. J. Pharm. Sci. 2012, 101, 4075–4099. [Google Scholar] [CrossRef]
  46. Suri, S.S.; Fenniri, H.; Singh, B. Nanotechnology-based drug delivery systems. J. Occup. Med. Toxicol. 2007, 2, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Koziara, J.M.; Whisman, T.R.; Tseng, M.T.; Mumper, R.J. In-Vivo efficacy of novel paclitaxel nanoparticles in paclitaxel-resistant human colorectal tumors. J. Control. Release 2006, 112, 312–319. [Google Scholar] [CrossRef] [PubMed]
  48. Koziara, J.M.; Lockman, P.; Allen, D.D.; Mumper, R.J. Paclitaxel nanoparticles for the potential treatment of brain tumors. J. Control. Release 2004, 99, 259–269. [Google Scholar] [CrossRef] [PubMed]
  49. Wilczewska, A.Z.; Niemirowicz, K.; Markiewicz, K.H.; Car, H. Nanoparticles as drug delivery systems. Pharmacol. Rep. 2012, 64, 1020–1037. [Google Scholar] [CrossRef]
  50. Gulyaev, A.E.; Gelperina, S.; Skidan, I.N.; Antropov, A.S.; Kivman, G.Y.; Kreuter, J. Significant Transport of Doxorubicin into the Brain with Polysorbate 80-Coated Nanoparticles. Pharm. Res. 1999, 16, 1564–1569. [Google Scholar] [CrossRef]
  51. Ljubimova, J.Y.; Sun, T.; Mashouf, L.; Ljubimov, A.V.; Israel, L.; Ljubimov, V.A.; Falahatian, V.; Holler, E. Covalent nano delivery systems for selective imaging and treatment of brain tumors. Adv. Drug Deliv. Rev. 2017, 113, 177–200. [Google Scholar] [CrossRef]
  52. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; del Pilar Rodriguez-Torres, M.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [Green Version]
  53. Rupp, R.; Rosenthal, S.L.; Stanberry, L.R. VivaGel™(SPL7013 Gel): A candidate dendrimer–microbicide for the prevention of HIV and HSV infection. Int. J. Nanomed. 2007, 2, 561. [Google Scholar]
  54. Wilhelm, S.; Tavares, A.J.; Dai, Q.; Ohta, S.; Audet, J.; Dvorak, H.F.; Chan, W.C.W. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 2016, 1, 16014. [Google Scholar] [CrossRef]
  55. Nichols, J.W.; Bae, Y.H. Odyssey of a cancer nanoparticle: From injection site to site of action. Nano Today 2012, 7, 606–618. [Google Scholar] [CrossRef] [Green Version]
  56. Caro, C.; Avasthi, A.; Paez-Muñoz, J.M.; Leal, M.P.; García-Martín, M.L. Passive targeting of high-grade gliomas via the EPR effect: A closed path for metallic nanoparticles? Biomater. Sci. 2021, 9, 7984–7995. [Google Scholar] [CrossRef] [PubMed]
  57. Nichols, J.W.; Bae, Y.H. EPR: Evidence and fallacy. J. Control. Release 2014, 190, 451–464. [Google Scholar] [CrossRef]
  58. Dai, Q.; Wilhelm, S.; Ding, D.; Syed, A.; Sindhwani, S.; Zhang, Y.; Chen, Y.Y.; MacMillan, P.; Chan, W.C.W. Quantifying the Ligand-Coated Nanoparticle Delivery to Cancer Cells in Solid Tumors. ACS Nano 2018, 12, 8423–8435. [Google Scholar] [CrossRef] [PubMed]
  59. Fan, H.-H.; Le, Q.; Lan, S.; Liang, J.-X.; Tie, S.-L.; Xu, J.-L. Modifying the mechanical properties of gold nanorods by copper doping and triggering their cytotoxicity with ultrasonic wave. Colloids Surf. B Biointerfaces 2018, 163, 47–54. [Google Scholar] [CrossRef] [PubMed]
  60. Kong, F.-Y.; Zhang, J.-W.; Li, R.-F.; Wang, Z.-X.; Wang, W.-J.; Wang, W. Unique Roles of Gold Nanoparticles in Drug Delivery, Targeting and Imaging Applications. Molecules 2017, 22, 1445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Haine, A.T.; Niidome, T. Gold Nanorods as Nanodevices for Bioimaging, Photothermal Therapeutics, and Drug Delivery. Chem. Pharm. Bull. 2017, 65, 625–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Pérez-Juste, J.; Pastoriza-Santos, I.; Liz-Marzán, L.M.; Mulvaney, P. Gold nanorods: Synthesis, characterization and applications. Coord. Chem. Rev. 2005, 249, 1870–1901. [Google Scholar] [CrossRef]
  63. Ghosh, P.; Han, G.; De, M.; Kim, C.K.; Rotello, V.M. Gold nanoparticles in delivery applications. Adv. Drug Deliv. Rev. 2008, 60, 1307–1315. [Google Scholar] [CrossRef]
  64. Pulliam, B.; Sung, J.C.; Edwards, D.A. Design of nanoparticle-based dry powder pulmonary vaccines. Expert Opin. Drug Deliv. 2007, 4, 651–663. [Google Scholar] [CrossRef]
  65. Ragusa, A.; García, I.; Penadés, S. Nanoparticles as nonviral gene delivery vectors. IEEE Trans. Nano Biosci. 2007, 6, 319–330. [Google Scholar] [CrossRef]
  66. Streicher, R.M.; Schmidt, M.; Fiorito, S. Nanosurfaces and nanostructures for artificial orthopedic implants. Nanomedicine 2007, 2, 861–874. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, P.C.; Mwakwari, S.C.; Oyelere, A.K. Gold nanoparticles: From nanomedicine to nanosensing. Nanotechnol. Sci. Appl. 2008, 1, 45. [Google Scholar] [PubMed] [Green Version]
  68. Naderi, M.S.; Moghadam, T.T.; Khajeh, K.; Ranjbar, B. Improving the stability of chondroitinase ABC I via interaction with gold nanorods. Int. J. Biol. Macromol. 2018, 107, 297–304. [Google Scholar] [CrossRef] [PubMed]
  69. Guo, J.; Ning, T.; Han, Y.; Sheng, Y.; Li, C.; Zhao, X.; Lu, Z.; Man, B.; Jiao, Y.; Jiang, S. Preparation, characterization, and nonlinear optical properties of hybridized graphene@ gold nanorods nanocomposites. Appl. Surf. Sci. 2018, 433, 45–50. [Google Scholar] [CrossRef]
  70. Stone, J.; Jackson, S.; Wright, D. Biological applications of gold nanorods. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2011, 3, 100–109. [Google Scholar] [CrossRef]
  71. Cao, J.; Sun, T.; Grattan, K.T.V. Gold nanorod-based localized surface plasmon resonance biosensors: A review. Sens. Actuators B Chem. 2014, 195, 332–351. [Google Scholar] [CrossRef]
  72. Zhang, C.; Cheng, X.; Chen, M.; Sheng, J.; Ren, J.; Jiang, Z.; Cai, J.; Hu, Y. Fluorescence guided photothermal/photodynamic ablation of tumours using pH-responsive chlorin e6-conjugated gold nanorods. Colloids Surf. B Biointerfaces 2017, 160, 345–354. [Google Scholar] [CrossRef]
  73. Wu, H.-Y.; Chu, H.-C.; Kuo, T.-J.; Kuo, C.-L.; Huang, M.H. Seed-Mediated Synthesis of High Aspect Ratio Gold Nanorods with Nitric Acid. Chem. Mater. 2005, 17, 6447–6451. [Google Scholar] [CrossRef]
  74. Gao, F.; Bai, L.; Liu, S.; Zhang, R.; Zhang, J.; Feng, X.; Zheng, Y.; Zhao, Y. Rationally encapsulated gold nanorods improving both linear and nonlinear photoacoustic imaging contrast in vivo. Nanoscale 2016, 9, 79–86. [Google Scholar] [CrossRef]
  75. Ishtiaq, S.; Shah, K.U.; Ur-Rehman, T.; Ud-Din, F. Gold nanorods: New generation drug delivery platform. In Metal Nanoparticles for Drug Delivery and Diagnostic Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 59–84. [Google Scholar]
  76. Brown, K.R.; Natan, M.J. Hydroxylamine Seeding of Colloidal Au Nanoparticles in Solution and on Surfaces. Langmuir 1998, 14, 726–728. [Google Scholar] [CrossRef]
  77. Gao, J.; Bender, A.C.M.; Murphy, C.J. Dependence of the Gold Nanorod Aspect Ratio on the Nature of the Directing Surfactant in Aqueous Solution. Langmuir 2003, 19, 9065–9070. [Google Scholar] [CrossRef]
  78. Moghadam, T.T.; Ranjbar, B.; Khajeh, K. Conformation and activity of lysozyme on binding to two types of gold nanorods: A comparative study. Int. J. Biol. Macromol. 2012, 51, 91–96. [Google Scholar] [CrossRef] [PubMed]
  79. Liz-Marzán, L. (Ed.) Colloidal Synthesis of Plasmonic Nanometals; CRC Press: Boca Raton, FL, USA, 2020. [Google Scholar] [CrossRef]
  80. Ali, M.R.K.; Rahman, M.A.; Wu, Y.; Han, T.; Peng, X.; Mackey, M.A.; Wang, D.; Shin, H.J.; Chen, Z.G.; Xiao, H.; et al. Efficacy, long-term toxicity, and mechanistic studies of gold nanorods photothermal therapy of cancer in xenograft mice. Proc. Natl. Acad. Sci. USA 2017, 114, E3110–E3118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Morales-Dalmau, J.; Vilches, C.; de Miguel, I.; Sanz, V.; Quidant, R. Optimum morphology of gold nanorods for light-induced hyperthermia. Nanoscale 2017, 10, 2632–2638. [Google Scholar] [CrossRef] [Green Version]
  82. Marasini, R.; Pitchaimani, A.; Nguyen, T.D.T.; Comer, J.; Aryal, S. The influence of polyethylene glycol passivation on the surface plasmon resonance induced photothermal properties of gold nanorods. Nanoscale 2018, 10, 13684–13693. [Google Scholar] [CrossRef]
  83. Singh, N.; Joshi, A.; Toor, A.P.; Verma, G. Drug delivery: Advancements and challenges. In Nanostructures for Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2017; pp. 865–886. [Google Scholar] [CrossRef]
  84. Blanco, E.; Shen, H.; Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 2015, 33, 941–951. [Google Scholar] [CrossRef]
  85. Donahue, N.D.; Acar, H.; Wilhelm, S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv. Drug Deliv. Rev. 2019, 143, 68–96. [Google Scholar] [CrossRef] [PubMed]
  86. Bailly, A.-L.; Correard, F.; Popov, A.; Tselikov, G.; Chaspoul, F.; Appay, R.; Al-Kattan, A.; Kabashin, A.V.; Braguer, D.; Esteve, M.-A. In Vivo evaluation of safety, biodistribution and pharmacokinetics of laser-synthesized gold nanoparticles. Sci. Rep. 2019, 9, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Alexis, F.; Pridgen, E.; Molnar, L.K.; Farokhzad, O.C. Factors Affecting the Clearance and Biodistribution of Polymeric Nanoparticles. Mol. Pharm. 2008, 5, 505–515. [Google Scholar] [CrossRef] [Green Version]
  88. Alkilany, A.; Shatanawi, A.; Kurtz, T.; Caldwell, R. Toxicity and Cellular Uptake of Gold Nanorods in Vascular Endothelium and Smooth Muscles of Isolated Rat Blood Vessel: Importance of Surface Modification. Small 2012, 8, 1270–1278. [Google Scholar] [CrossRef] [PubMed]
  89. De La Harpe, K.M.; Kondiah, P.P.; Choonara, Y.E.; Marimuthu, T.; Du Toit, L.C.; Pillay, V. The Hemocompatibility of Nanoparticles: A Review of Cell–Nanoparticle Interactions and Hemostasis. Cells 2019, 8, 1209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Nunes, Á.M.; da Silva, K.R.M.; Calado, C.M.S.; Saraiva, K.L.A.; Figueiredo, R.C.B.Q.; Leite, A.C.R.; Meneghetti, M.R. Evaluation of gold nanorods toxicity on isolated mitochondria. Toxicology 2019, 413, 24–32. [Google Scholar] [CrossRef] [PubMed]
  91. Souza, J.P.; Mansano, A.S.; Venturini, F.P.; Marangoni, V.S.; Lins, P.M.P.; Silva, B.P.C.; Dressler, B.; Zucolotto, V. Toxicity of gold nanorods on Ceriodaphnia dubia and Danio rerio after sub-lethal exposure and recovery. Environ. Sci. Pollut. Res. 2021, 28, 25316–25326. [Google Scholar] [CrossRef] [PubMed]
  92. Mesquita, B.S.M. Acute and Developmental Toxicity of Gold Nanorods on Zebrafish (Danio Rerio) Embryos; University of Porto: Porto, Portugal, 2016. [Google Scholar]
  93. Pyshnaya, I.A.; Razum, K.; Poletaeva, Y.; Pyshnyi, D.; Zenkova, M.A.; Ryabchikova, E.I. Comparison of Behaviour in Different Liquids and in Cells of Gold Nanorods and Spherical Nanoparticles Modified by Linear Polyethyleneimine and Bovine Serum Albumin. BioMed. Res. Int. 2014, 2014, 908175. [Google Scholar] [CrossRef]
  94. Chakraborty, S.; Joshi, P.; Shanker, V.; Ansari, Z.A.; Singh, S.P.; Chakrabarti, P. Contrasting Effect of Gold Nanoparticles and Nanorods with Different Surface Modifications on the Structure and Activity of Bovine Serum Albumin. Langmuir 2011, 27, 7722–7731. [Google Scholar] [CrossRef]
  95. Zhang, W.; Wang, F.; Wang, Y.; Wang, J.; Yu, Y.; Guo, S.; Chen, R.; Zhou, D. pH and near-infrared light dual-stimuli responsive drug delivery using DNA-conjugated gold nanorods for effective treatment of multidrug resistant cancer cells. J. Control. Release 2016, 232, 9–19. [Google Scholar] [CrossRef] [Green Version]
  96. Haine, A.T.; Koga, Y.; Hashimoto, Y.; Higashi, T.; Motoyama, K.; Arima, H.; Niidome, T. Enhancement of transdermal protein delivery by photothermal effect of gold nanorods coated on polysaccharide-based hydrogel. Eur. J. Pharm. Biopharm. 2017, 119, 91–95. [Google Scholar] [CrossRef]
  97. Xu, W.; Qian, J.; Hou, G.; Wang, Y.; Wang, J.; Sun, T.; Ji, L.; Suo, A.; Yao, Y. A dual-targeted hyaluronic acid-gold nanorod platform with triple-stimuli responsiveness for photodynamic/photothermal therapy of breast cancer. Acta Biomater. 2018, 83, 400–413. [Google Scholar] [CrossRef]
  98. Niidome, T.; Shiotani, A.; Mori, T.; Katayama, Y. Targeted delivery of gold nanorods modified with thermo-sensitive polymer. J. Control. Release 2010, 148, e65–e66. [Google Scholar] [CrossRef]
  99. Chen, J.; Han, X.; Deng, J.; Zhang, J.; Li, L.; Ni, J.; Huang, Y.; Xie, X.; Chen, S.; Ke, L.; et al. An injectable hydrogel based on phenylboronic acid hyperbranched macromer encapsulating gold nanorods and Astragaloside IV nanodrug for myocardial infarction. Chem. Eng. J. 2020, 413, 127423. [Google Scholar] [CrossRef]
  100. Shanmugam, V.; Chien, Y.-H.; Cheng, Y.-S.; Liu, T.-Y.; Huang, C.-C.; Su, C.-H.; Chen, Y.-S.; Kumar, U.; Hsu, H.-F.; Yeh, C.-S. Oligonucleotides—Assembled Au Nanorod-Assisted Cancer Photothermal Ablation and Combination Chemotherapy with Targeted Dual-Drug Delivery of Doxorubicin and Cisplatin Prodrug. ACS Appl. Mater. Interfaces 2014, 6, 4382–4393. [Google Scholar] [CrossRef] [PubMed]
  101. Liang, Y.; Gao, W.; Peng, X.; Deng, X.; Sun, C.; Wu, H.; He, B. Near infrared light responsive hybrid nanoparticles for synergistic therapy. Biomaterials 2016, 100, 76–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wang, Y.; Wang, L.; Yan, M.; Dong, S.; Hao, J. Near-Infrared-Light-Responsive Magnetic DNA Microgels for Photon- and Magneto-Manipulated Cancer Therapy. ACS Appl. Mater. Interfaces 2017, 9, 28185–28194. [Google Scholar] [CrossRef]
  103. Zhou, H.; Xu, H.; Li, X.; Lv, Y.; Ma, T.; Guo, S.; Huang, Z.; Wang, X.; Xu, P. Dual targeting hyaluronic acid—RGD mesoporous silica coated gold nanorods for chemo-photothermal cancer therapy. Mater. Sci. Eng. C 2017, 81, 261–270. [Google Scholar] [CrossRef] [PubMed]
  104. Khan, M.S.; Pandey, S.; Bhaisare, M.L.; Gedda, G.; Talib, A.; Wu, H.-F. Graphene oxide@gold nanorods for chemo-photothermal treatment and controlled release of doxorubicin in mice Tumor. Colloids Surf. B Biointerfaces 2017, 160, 543–552. [Google Scholar] [CrossRef] [PubMed]
  105. Parida, S.; Maiti, C.; Rajesh, Y.; Dey, K.K.; Pal, I.; Parekh, A.; Patra, R.; Dhara, D.; Dutta, P.K.; Mandal, M. Gold nanorod embedded reduction responsive block copolymer micelle-triggered drug delivery combined with photothermal ablation for targeted cancer therapy. Biochim. Biophys. Acta (BBA) Gen. Subj. 2017, 1861, 3039–3052. [Google Scholar] [CrossRef]
  106. Song, Z.; Liu, Y.; Shi, J.; Ma, T.; Zhang, Z.; Ma, H.; Cao, S. Hydroxyapatite/mesoporous silica coated gold nanorods with improved degradability as a multi-responsive drug delivery platform. Mater. Sci. Eng. C 2017, 83, 90–98. [Google Scholar] [CrossRef]
  107. Chen, J.; Li, X.; Zhaoa, X.; Wub, Q.Q.; Zhub, H.; Maoa, Z.; Gaoa, C. Doxorubicin-conjugated pH-responsive gold nanorods for combined photothermal therapy and chemotherapy of cancer. Bioact. Mater. 2018, 3, 347–354. [Google Scholar] [CrossRef]
  108. Ghamkhari, A.; Mahmoodzadeh, F.; Ghorbani, M.; Hamishehkar, H. A novel gold nanorods coated by stimuli-responsive ABC triblock copolymer for chemotherapy of solid tumors. Eur. Polym. J. 2019, 115, 313–324. [Google Scholar] [CrossRef]
  109. Hou, G.; Qian, J.; Xu, W.; Sun, T.; Wang, Y.; Wang, J.; Ji, L.; Suo, A. A novel pH-sensitive targeting polysaccharide-gold nanorod conjugate for combined photothermal-chemotherapy of breast cancer. Carbohydr. Polym. 2019, 212, 334–344. [Google Scholar] [CrossRef]
  110. Jiang, P.; Hu, Y.; Li, G. Biocompatible Au@Ag nanorod@ZIF-8 core-shell nanoparticles for surface-enhanced Raman scattering imaging and drug delivery. Talanta 2019, 200, 212–217. [Google Scholar] [CrossRef] [PubMed]
  111. Lee, C.; Lim, K.; Kim, S.S.; Thien, L.X.; Lee, E.S.; Oh, K.T.; Choi, H.-G.; Youn, Y.S. Chlorella-gold nanorods hydrogels generating photosynthesis-derived oxygen and mild heat for the treatment of hypoxic breast cancer. J. Control. Release 2019, 294, 77–90. [Google Scholar] [CrossRef]
  112. Alenezi, A.; Hulander, M.; Atefyekta, S.; Andersson, M. Development of a photon induced drug-delivery implant coating. Mater. Sci. Eng. C 2019, 98, 619–627. [Google Scholar] [CrossRef]
  113. Rodrigues, C.F.; Reis, C.A.; Moreira, A.F.; Ferreira, P.; Correia, I.J. Optimization of gold core-mesoporous silica shell functionalization with TPGS and PEI for cancer therapy. Microporous Mesoporous Mater. 2019, 285, 1–12. [Google Scholar] [CrossRef]
  114. Qi, Z.; Shi, J.; Zhang, Z.; Cao, Y.; Li, J.; Cao, S. PEGylated graphene oxide-capped gold nanorods/silica nanoparticles as multifunctional drug delivery platform with enhanced near-infrared responsiveness. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 104, 109889. [Google Scholar] [CrossRef] [PubMed]
  115. Mohebbi, S.; Moghadam, T.T.; Nikkhah, M.; Behmanesh, M. RGD-HK Peptide-Functionalized Gold Nanorods Emerge as Targeted Biocompatible Nanocarriers for Biomedical Applications. Nanoscale Res. Lett. 2019, 14, 13. [Google Scholar] [CrossRef] [PubMed]
  116. Zhu, F.; Tan, G.; Zhong, Y.; Jiang, Y.; Cai, L.; Yu, Z.; Liu, S.; Ren, F. Smart nanoplatform for sequential drug release and enhanced chemo-thermal effect of dual drug loaded gold nanorod vesicles for cancer therapy. J. Nanobiotechnol. 2019, 17, 44. [Google Scholar] [CrossRef]
  117. Jiang, Y.; Guo, Z.; Fang, J.; Wang, B.; Lin, Z.; Chen, Z.-S.; Chen, Y.; Zhang, N.; Yang, X.; Gao, W. A multi-functionalized nanocomposite constructed by gold nanorod core with triple-layer coating to combat multidrug resistant colorectal cancer. Mater. Sci. Eng. C 2020, 107, 110224. [Google Scholar] [CrossRef]
  118. Puleio, R.; Licciardi, M.; Varvarà, P.; Scialabba, C.; Cassata, G.; Cicero, L.; Cavallaro, G.; Giammona, G. Effect of actively targeted copolymer coating on solid tumors eradication by gold nanorods-induced hyperthermia. Int. J. Pharm. 2020, 587, 119641. [Google Scholar] [CrossRef]
  119. Du, Z.; Yan, K.; Cao, Y.; Li, Y.; Yao, Y.; Yang, G. Regenerated keratin-encapsulated gold nanorods for chemo-photothermal synergistic therapy. Mater. Sci. Eng. C 2020, 117, 111340. [Google Scholar] [CrossRef]
  120. Chen, R.; Shi, J.; Zhu, B.; Zhang, L.; Cao, S. Mesoporous hollow hydroxyapatite capped with smart polymer for multi-stimuli remotely controlled drug delivery. Microporous Mesoporous Mater. 2020, 306, 110447. [Google Scholar] [CrossRef]
  121. Cao, Y.; Shi, J.; Wu, Z.; Li, J.; Cao, S. Gold nanorods/polydopamine-capped hollow hydroxyapatite microcapsules as remotely controllable multifunctional drug delivery platform. Powder Technol. 2020, 372, 486–496. [Google Scholar] [CrossRef]
  122. Pourjavadi, A.; Bagherifard, M.; Doroudian, M. Synthesis of micelles based on chitosan functionalized with gold nanorods as a light sensitive drug delivery vehicle. Int. J. Biol. Macromol. 2020, 149, 809–818. [Google Scholar] [CrossRef]
  123. Matai, I.; Kaur, G.; Soni, S.; Sachdev, A.; Vikas; Mishra, S. Near-infrared stimulated hydrogel patch for photothermal therapeutics and thermoresponsive drug delivery. J. Photochem. Photobiol. B Biol. 2020, 210, 111960. [Google Scholar] [CrossRef] [PubMed]
  124. Sharma, V.; Vijay, J.; Ganesh, M.; Sundaramurthy, A. Multilayer capsules encapsulating nimbin and doxorubicin for cancer chemo-photothermal therapy. Int. J. Pharm. 2020, 582, 119350. [Google Scholar] [CrossRef] [PubMed]
  125. Xue, L.; Wang, D.; Zhang, X.; Xu, S.; Zhang, N. Targeted and triple therapy-based liposomes for enhanced treatment of rheumatoid arthritis. Int. J. Pharm. 2020, 586, 119642. [Google Scholar] [CrossRef]
  126. Guo, H.; Yi, S.; Feng, K.; Xia, Y.; Qu, X.; Wan, F.; Chen, L.; Zhang, C. In Situ formation of metal organic framework onto gold nanorods/mesoporous silica with functional integration for targeted theranostics. Chem. Eng. J. 2020, 403, 126432. [Google Scholar] [CrossRef]
  127. Guo, D.; Huang, Y.; Jin, X.; Zhang, C.; Zhu, X. A Redox-Responsive, In-Situ Polymerized Polyplatinum(IV)-Coated Gold Nanorod as An Amplifier of Tumor Accumulation for Enhanced Thermo-Chemotherapy. Biomaterials 2020, 266, 120400. [Google Scholar] [CrossRef]
  128. Darwish, W.M.; Abdoon, A.S.; Shata, M.S.; Elmansy, M. Vincristine-loaded polymeric corona around gold nanorods for combination (chemo-photothermal) therapy of oral squamous carcinoma. React. Funct. Polym. 2020, 151, 104575. [Google Scholar] [CrossRef]
  129. Cheng, D.; Ji, Y.; Wang, B.; Wang, Y.; Tang, Y.; Fu, Y.; Xu, Y.; Qian, X.; Zhu, W. Dual-responsive nanohybrid based on degradable silica-coated gold nanorods for triple-combination therapy for breast cancer. Acta Biomater. 2021, 128, 435–446. [Google Scholar] [CrossRef]
  130. Yin, M.; Qiao, Z.; Yan, D.; Yang, M.; Yang, L.; Wan, X.; Chen, H.; Luo, J.; Xiao, H. Ciprofloxacin conjugated gold nanorods with pH induced surface charge transformable activities to combat drug resistant bacteria and their biofilms. Mater. Sci. Eng. C 2021, 128, 112292. [Google Scholar] [CrossRef] [PubMed]
  131. Lihuang, L.; Qiuyan, G.; Yanxiu, L.; Mindan, L.; Jun, Y.; Yunlong, G.; Qiang, Z.; Benqiang, S.; Xiumin, W.; Liang-Cheng, L.; et al. Targeted combination therapy for glioblastoma by co-delivery of doxorubicin, YAP-siRNA and gold nanorods. J. Mater. Sci. Technol. 2020, 63, 81–90. [Google Scholar] [CrossRef]
  132. Ramasamy, T.; Ruttala, H.B.; Sundaramoorthy, P.; Poudel, B.K.; Youn, Y.S.; Ku, S.K.; Choi, H.-G.; Yong, C.S.; Kim, J.O. Multimodal selenium nanoshell-capped Au@ mSiO2 nanoplatform for NIR-responsive chemo-photothermal therapy against metastatic breast cancer. NPG Asia Mater. 2018, 10, 197–216. [Google Scholar] [CrossRef] [Green Version]
  133. Shi, X.; Perry, H.L.; Wilton-Ely, J.D.E.T. Strategies for the functionalisation of gold nanorods to reduce toxicity and aid clinical translation. Nanotheranostics 2021, 5, 155–165. [Google Scholar] [CrossRef] [PubMed]
  134. Niidome, T.; Yamagata, M.; Okamoto, Y.; Akiyama, Y.; Takahashi, H.; Kawano, T.; Katayama, Y.; Niidome, Y. PEG-modified gold nanorods with a stealth character for in vivo applications. J. Control. Release 2006, 114, 343–347. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, Z.; Lin, M. Fast loading of PEG–SH on CTAB-protected gold nanorods. RSC Adv. 2014, 4, 17760–17767. [Google Scholar] [CrossRef]
  136. Kinnear, C.; Dietsch, H.; Clift, M.J.; Endes, C.; Rothen-Rutishauser, B.; Petri-Fink, A. Gold nanorods: Controlling their surface chemistry and complete detoxification by a two-step place exchange. Angew. Chem. Int. Ed. 2013, 52, 1934–1938. [Google Scholar] [CrossRef] [Green Version]
  137. Zhao, X.; Ma, Z.; Sun, H. Simple preparation of photothermal nanomaterial GNR@ SiO2 with enhanced drug loading content. IET Nanobiotechnol. 2018, 13, 257–261. [Google Scholar] [CrossRef]
  138. Niikura, K.; Matsunaga, T.; Suzuki, T.; Kobayashi, S.; Yamaguchi, H.; Orba, Y.; Kawaguchi, A.; Hasegawa, H.; Kajino, K.; Ninomiya, T.; et al. Gold Nanoparticles as a Vaccine Platform: Influence of Size and Shape on Immunological Responses in Vitro and in Vivo. ACS Nano 2013, 7, 3926–3938. [Google Scholar] [CrossRef]
  139. Ramasamy, T.; Munusamy, S.; Ruttala, H.B.; Kim, J.O. Smart Nanocarriers for the Delivery of Nucleic Acid-Based Therapeutics: A Comprehensive Review. Biotechnol. J. 2020, 16, 1900408. [Google Scholar] [CrossRef]
  140. Yang, Z.; Liu, T.; Xie, Y.; Sun, Z.; Liu, H.; Lin, J.; Liu, C.; Mao, Z.-W.; Nie, S. Chitosan layered gold nanorods as synergistic therapeutics for photothermal ablation and gene silencing in triple-negative breast cancer. Acta Biomater. 2015, 25, 194–204. [Google Scholar] [CrossRef] [PubMed]
  141. Yamashita, S.; Fukushima, H.; Akiyama, Y.; Niidome, Y.; Mori, T.; Katayama, Y.; Niidome, T. Controlled-release system of single-stranded DNA triggered by the photothermal effect of gold nanorods and its in vivo application. Bioorganic Med. Chem. 2011, 19, 2130–2135. [Google Scholar] [CrossRef] [PubMed]
  142. Ye, L.; Chen, Y.; Mao, J.; Lei, X.; Yang, Q.; Cui, C. Dendrimer-modified gold nanorods as a platform for combinational gene therapy and photothermal therapy of tumors. J. Exp. Clin. Cancer Res. 2021, 40, 303. [Google Scholar] [CrossRef] [PubMed]
  143. Prabhune, M. RNAi vs. CRISPR: Guide to Selecting the Best Gene Silencing Method. Available online: https://www.synthego.com/blog/rnai-vs-crispr-guide#rn-ai-the-knockdown-pioneer (accessed on 30 September 2021).
  144. Zare, H.; Ahmadi, S.; Ghasemi, A.; Ghanbari, M.; Rabiee, N.; Bagherzadeh, M.; Karimi, M.; Webster, T.J.; Hamblin, M.R.; Mostafavi, E. Carbon Nanotubes: Smart Drug/Gene Delivery Carriers. Int. J. Nanomed. 2021, 16, 1681. [Google Scholar] [CrossRef]
  145. Ni, S.; Zhu, J.; Mezour, M.A.; Lennox, R.B. Single wall carbon nanotube (SWCNT)–gold nanorod (AuNR) conjugates via thermally-mild reaction conditions. New J. Chem. 2017, 41, 12392–12396. [Google Scholar] [CrossRef]
  146. Cui, D.; Huang, P.; Zhang, C.; Ozkan, C.S.; Pan, B.; Xu, P. Dendrimer-modified gold nanorods as efficient controlled gene delivery system under near-infrared light irradiation. J. Control. Release 2011, 152, e137–e139. [Google Scholar] [CrossRef]
  147. Wang, F.; Shen, Y.; Zhang, W.; Li, M.; Wang, Y.; Zhou, D.; Guo, S. Efficient, dual-stimuli responsive cytosolic gene delivery using a RGD modified disulfide-linked polyethylenimine functionalized gold nanorod. J. Control. Release 2014, 196, 37–51. [Google Scholar] [CrossRef]
  148. Petralia, S.; Forte, G.; Aiello, M.; Nocito, G.; Conoci, S. Photothermal-triggered system for oligonucleotides delivery from cationic gold nanorods surface: A molecular dynamic investigation. Colloids Surf. B Biointerfaces 2021, 201, 111654. [Google Scholar] [CrossRef]
  149. Ruttala, H.B.; Ramasamy, T.; Ruttala, R.R.T.; Tran, T.H.; Jeong, J.-H.; Choi, H.-G.; Ku, S.K.; Yong, C.S.; Kim, J.O. Mitochondria-targeting multi-metallic ZnCuO nanoparticles and IR780 for efficient photodynamic and photothermal cancer treatments. J. Mater. Sci. Technol. 2021, 86, 139–150. [Google Scholar] [CrossRef]
  150. Mortezaee, K.; Narmani, A.; Salehi, M.; Bagheri, H.; Farhood, B.; Haghi-Aminjan, H.; Najafi, M. Synergic effects of nanoparticles-mediated hyperthermia in radiotherapy/chemotherapy of cancer. Life Sci. 2021, 269, 119020. [Google Scholar] [CrossRef]
  151. Pissuwan, D.; Valenzuela, S.M.; Cortie, M.B. Prospects for Gold Nanorod Particles in Diagnostic and Therapeutic Applications. Biotechnol. Genet. Eng. Rev. 2008, 25, 93–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. 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. 2019, 101, 531–543. [Google Scholar] [CrossRef] [PubMed]
  153. Poudel, B.K.; Soe, Z.C.; Ruttala, H.B.; Gupta, B.; Ramasamy, T.; Thapa, R.K.; Gautam, M.; Ou, W.; Nguyen, H.T.; Jeong, J.-H.; et al. In situ fabrication of mesoporous silica-coated silver-gold hollow nanoshell for remotely controllable chemo-photothermal therapy via phase-change molecule as gatekeepers. Int. J. Pharm. 2018, 548, 92–103. [Google Scholar] [CrossRef]
  154. Lal, S.; Verma, J.; Van Noorden, C.J. Nanoparticles for hyperthermic therapy: Synthesis strategies and applications in glioblastoma. Int. J. Nanomed. 2014, 9, 2863–2877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Pissuwan, D.; Valenzuela, S.M.; Miller, C.M.; Cortie, M.B. A Golden Bullet? Selective Targeting of Toxoplasma gondii Tachyzoites Using Antibody-Functionalized Gold Nanorods. Nano Lett. 2007, 7, 3808–3812. [Google Scholar] [CrossRef] [PubMed]
  156. Lee, C.; Hwang, H.S.; Lee, S.; Kim, B.; Kim, J.O.; Oh, K.T.; Lee, E.S.; Choi, H.-G.; Youn, Y.S. Rabies Virus-Inspired Silica-Coated Gold Nanorods as a Photothermal Therapeutic Platform for Treating Brain Tumors. Adv. Mater. 2017, 29. [Google Scholar] [CrossRef] [PubMed]
  157. Labouta, H.I.; Hooshmand, N.; Upreti, T.; El-Sayed, M.A. Localized Plasmonic Photothermal Therapy as a Life-saving Treatment Paradigm for Hospitalized COVID-19 Patients. Plasmonics 2021, 16, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
  158. Meng, Y.; Yang, J.; Jiang, R.; Wang, S.; Zheng, L.; Wang, G.; Tian, X.; Zhu, H.; Yan, D.; Liu, C.; et al. Biocompatible PLNP-GNR composite nanoplatforms for monitoring deep-tissue photothermal therapy process. Appl. Surf. Sci. 2021, 562, 150189. [Google Scholar] [CrossRef]
  159. Du, Y.; Jiang, Q.; Beziere, N.; Song, L.; Zhang, Q.; Peng, D.; Chi, C.; Yang, X.; Guo, H.; Diot, G.; et al. DNA-Nanostructure-Gold-Nanorod Hybrids for Enhanced In Vivo Optoacoustic Imaging and Photothermal Therapy. Adv. Mater. 2016, 28, 10000–10007. [Google Scholar] [CrossRef]
  160. Bhana, S.; O’Connor, R.; Johnson, J.; Ziebarth, J.D.; Henderson, L.; Huang, X. Photosensitizer-loaded gold nanorods for near infrared photodynamic and photothermal cancer therapy. J. Colloid Interface Sci. 2016, 469, 8–16. [Google Scholar] [CrossRef] [Green Version]
  161. 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] [PubMed]
  162. Gonçalves, D.P.; Rodriguez, R.D.; Kurth, T.; Bray, L.J.; Binner, M.; Jungnickel, C.; Gür, F.N.; Poser, S.W.; Schmidt, T.L.; Zahn, D.R.; et al. Enhanced targeting of invasive glioblastoma cells by peptide-functionalized gold nanorods in hydrogel-based 3D cultures. Acta Biomater. 2017, 58, 12–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Jacinto, T.A.; Rodrigues, C.F.; Moreira, A.F.; Miguel, S.A.P.; Costa, E.C.; Ferreira, P.; Correia, I.J. Hyaluronic acid and vitamin E polyethylene glycol succinate functionalized gold-core silica shell nanorods for cancer targeted photothermal therapy. Colloids Surf. B Biointerfaces 2020, 188, 110778. [Google Scholar] [CrossRef] [PubMed]
  164. Lu, Q.; Lu, T.; Xu, M.; Yang, L.; Song, Y.; Li, N. SO2 prodrug doped nanorattles with extra-high drug payload for “collusion inside and outside” photothermal/pH triggered-gas therapy. Biomaterials 2020, 257, 120236. [Google Scholar] [CrossRef]
  165. Collini, E. Cooperative effects to enhance two-photon absorption efficiency: Intra-versus inter-molecular approach. Phys. Chem. Chem. Phys. 2012, 14, 3725–3736. [Google Scholar] [CrossRef]
  166. Liang, C.; Diao, S.; Wang, C.; Gong, H.; Liu, T.; Hong, G.; Shi, X.; Dai, H.; Liu, Z. Tumor Metastasis Inhibition by Imaging-Guided Photothermal Therapy with Single-Walled Carbon Nanotubes. Adv. Mater. 2014, 26, 5646–5652. [Google Scholar] [CrossRef]
  167. Jin, Y.; Li, Y.; Ma, X.; Zha, Z.; Shi, L.; Tian, J.; Dai, Z. Encapsulating tantalum oxide into polypyrrole nanoparticles for X-ray CT/photoacoustic bimodal imaging-guided photothermal ablation of cancer. Biomaterials 2014, 35, 5795–5804. [Google Scholar] [CrossRef]
  168. Wang, S.; Huang, P.; Nie, L.; Xing, R.; Liu, D.; Wang, Z.; Lin, J.; Chen, S.; Niu, G.; Lu, G.; et al. Single Continuous Wave Laser Induced Photodynamic/Plasmonic Photothermal Therapy Using Photosensitizer-Functionalized Gold Nanostars. Adv. Mater. 2013, 25, 3055–3061. [Google Scholar] [CrossRef] [Green Version]
  169. Zhou, M.; Li, J.; Liang, S.; Sood, A.K.; Liang, D.; Li, C. CuS Nanodots with Ultrahigh Efficient Renal Clearance for Positron Emission Tomography Imaging and Image-Guided Photothermal Therapy. ACS Nano 2015, 9, 7085–7096. [Google Scholar] [CrossRef] [Green Version]
  170. Liang, X.; Deng, Z.; Jing, L.; Li, X.; Dai, Z.; Li, C.; Huang, M. Prussian blue nanoparticles operate as a contrast agent for enhanced photoacoustic imaging. Chem. Commun. 2013, 49, 11029–11031. [Google Scholar] [CrossRef]
  171. Nima, Z.A.; Alwbari, A.M.; Dantuluri, V.; Hamzah, R.N.; Sra, N.; Motwani, P.; Arnaoutakis, K.; Levy, R.A.; Bohliqa, A.F.; Nedosekin, D.; et al. Targeting nano drug delivery to cancer cells using tunable, multi-layer, silver-decorated gold nanorods. J. Appl. Toxicol. 2017, 37, 1370–1378. [Google Scholar] [CrossRef]
  172. Zeng, J.-Y.; Zhang, M.-K.; Peng, M.-Y.; Gong, D.; Zhang, X.-Z. Porphyrinic Metal-Organic Frameworks Coated Gold Nanorods as a Versatile Nanoplatform for Combined Photodynamic/Photothermal/Chemotherapy of Tumor. Adv. Funct. Mater. 2017, 28. [Google Scholar] [CrossRef]
  173. Guo, Y.; Li, S.; Liu, J.; Yang, G.; Sun, Z.; Wan, J. Double functional aptamer switch probes based on gold nanorods for intracellular ATP detection and targeted drugs transportation. Sens. Actuators B Chem. 2016, 235, 655–662. [Google Scholar] [CrossRef]
  174. Newell, B.B.; Wang, Y.; Irudayaraj, J. Multifunctional gold nanorod theragnostics probed by multi-photon imaging. Eur. J. Med. Chem. 2012, 48, 330–337. [Google Scholar] [CrossRef]
  175. Jiang, Z.; Dong, B.; Chen, B.; Wang, J.; Xu, L.; Zhang, S.; Song, H. Multifunctional Au@ mSiO2/rhodamine B isothiocyanate nanocomposites: Cell imaging, photocontrolled drug release, and photothermal therapy for cancer cells. Small 2013, 9, 604–612. [Google Scholar] [CrossRef]
  176. Niidome, T.; Ohga, A.; Akiyama, Y.; Watanabe, K.; Niidome, Y.; Mori, T.; Katayama, Y. Controlled release of PEG chain from gold nanorods: Targeted delivery to tumor. Bioorganic Med. Chem. 2010, 18, 4453–4458. [Google Scholar] [CrossRef] [PubMed]
  177. Sun, Q.; You, Q.; Pang, X.; Tan, X.; Wang, J.; Liu, L.; Guo, F.; Tan, F.; Li, N. A photoresponsive and rod-shape nanocarrier: Single wavelength of light triggered photothermal and photodynamic therapy based on AuNRs-capped & Ce6-doped mesoporous silica nanorods. Biomaterials 2017, 122, 188–200. [Google Scholar] [PubMed]
  178. Choi, J.; Kim, S.Y. Photothermally enhanced photodynamic therapy based on glutathione-responsive pheophorbide a-conjugated gold nanorod formulations for cancer theranostic applications. J. Ind. Eng. Chem. 2020, 85, 66–74. [Google Scholar] [CrossRef]
  179. Chen, H.-Y.; Ghosh, S.; Ponnusamy, V.K.; Chattopadhyay, S. Graphene oxide as broadband hyperthermic agent and chemo-photothermal dissolution of kidney-stone mimicking calcium oxalate crystals. J. Photochem. Photobiol. A Chem. 2020, 405, 112917. [Google Scholar] [CrossRef]
  180. Gong, B.; Shen, Y.; Li, H.; Li, X.; Huan, X.; Zhou, J.; Chen, Y.; Wu, J.; Li, W. Thermo-responsive polymer encapsulated gold nanorods for single continuous wave laser-induced photodynamic/photothermal tumour therapy. J. Nanobiotechnol. 2021, 19, 41. [Google Scholar] [CrossRef]
  181. Luo, L.; Sun, W.; Feng, Y.; Qin, R.; Zhang, J.; Ding, D.; Shi, T.; Liu, X.; Chen, X.; Chen, H. Conjugation of a Scintillator Complex and Gold Nanorods for Dual-Modal Image-Guided Photothermal and X-ray-Induced Photodynamic Therapy of Tumors. ACS Appl. Mater. Interfaces 2020, 12, 12591–12599. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Overview of GNRs properties and medical applications.
Scheme 1. Overview of GNRs properties and medical applications.
Pharmaceutics 14 00664 sch001aPharmaceutics 14 00664 sch001b
Figure 2. (A) The designed nanosystem for a combination of chemo, thermal and gene delivery for glioblastoma. Reprinted from [131] with permission from Elsevier, Amsterdam, Netherlands. (B) Chitosan-GNR-based delivery system for siRNA-mediated cancer therapy. Reprinted from [140] with permission from Elsevier, Amsterdam, Netherlands.
Figure 2. (A) The designed nanosystem for a combination of chemo, thermal and gene delivery for glioblastoma. Reprinted from [131] with permission from Elsevier, Amsterdam, Netherlands. (B) Chitosan-GNR-based delivery system for siRNA-mediated cancer therapy. Reprinted from [140] with permission from Elsevier, Amsterdam, Netherlands.
Pharmaceutics 14 00664 g002
Figure 4. (A) Schematic figure of the theranostics designed nanosystem, using pH-induced chlorin e6@ gold nanorods (Ce6-PEG-AuNR). Reprinted from [72] with permission from Elsevier, Amsterdam, Netherlands. (B) Schematic illustration of AuNR@MOFs@CPT theranostics nanosystem for tumor’s multiplexed photodynamic/photothermal/chemotherapy. MOF represents as metal-organic frameworks. Reprinted from [172] with permission from Wiley Online Library. (C) a dual functional GNR-based system for targeted drug delivery and intracellular ATP detection. Reprinted from [173] with permission from Elsevier, Amsterdam, Netherlands.
Figure 4. (A) Schematic figure of the theranostics designed nanosystem, using pH-induced chlorin e6@ gold nanorods (Ce6-PEG-AuNR). Reprinted from [72] with permission from Elsevier, Amsterdam, Netherlands. (B) Schematic illustration of AuNR@MOFs@CPT theranostics nanosystem for tumor’s multiplexed photodynamic/photothermal/chemotherapy. MOF represents as metal-organic frameworks. Reprinted from [172] with permission from Wiley Online Library. (C) a dual functional GNR-based system for targeted drug delivery and intracellular ATP detection. Reprinted from [173] with permission from Elsevier, Amsterdam, Netherlands.
Pharmaceutics 14 00664 g004
Table 1. GNR applications in drug delivery or therapeutic systems (research-based studies).
Table 1. GNR applications in drug delivery or therapeutic systems (research-based studies).
PurposeSystem’s StructureMechanism of FunctionGNR’s Applied FeatureRef.
DDFolate targeted GNRs, cDNA and tether platinum [Pt(IV)]Photothermal hyperthermia and dual drug release with the NIR-laser stimulationPT[100]
DDNIR-responsive chromophore, b-cyclodextrins functionalized PEG, DOX-loaded GNRsNIR-stimulated drug releasePT[101]
DDBiotin targeted DNA-conjugated GNRs loaded by DOXNIR and pH-induced drug releasePT[95]
DDGNR core, magnetic ionic liquid, and DNA moieties shellPhotothermal hyperthermia, drug releasePT[102]
DD, PTTHyaluronic acid and RGD (Arg-Gly-Asp)-conjugated silica-coated GNRs, loaded by DOXPhotothermal hyperthermia, pH-enzyme sensitivity, and NIR-triggered drug releasePT[103]
PDGNRs coated polysaccharide-based hydrogel loaded by fluorescein isothiocyanate-modified ovalbumin (FITC-OVA)Photothermal hyperthermia and enhanced transdermal FITC-OVA deliveryPT[96]
DD, PTTGraphene oxide-conjugated GNRs loaded by DOXPhotothermal hyperthermia and NIR-stimulated drug releasePT[104]
DD, PTTGNRs with glutathione responsive diblock co-polymer micelles loaded by GW627368Xhigh glutathione concentration-responsive drug release, PTT-induced apoptosis, and necroptosisPT[105]
DDGNR core, mesoporous silica/hydroxyapatite shellNIR- and pH-induced drug releasePT[106]
DD, PTTDOX-loaded GNRs with a pH-sensitive linkagepH responsive DOX release, Photothermal hyperthermiaPT[107]
DD, PTT, PDT, ImHyaluronic acid (HA) functionalized GNRs, conjugating with 5-aminolevulinic acid (ALA), Cy7.5 and anti-HER2 antibodyHER2 and CD44 targeted system.
pH, glutathione, and hyaluronidase-responsive drug release.
NIR-stimulated PDT and PTT.
PT[97]
DDStimuli-responsive ABC triblock co-polymer coated GNRs loaded by DOXDOX release in response to pH and thermosensitive ABC triblock co-polymerSt[108]
DD, PTTConjugate of dihydroxy phenyl/hydrazide bifunctionalized hydroxyethyl chitosan-GNR, decorated with a pH-sensitive oxidized hyaluronic acid and loaded by DOXCD44 targeted, pH-responsive drug release, PTTPT[109]
DD, ImAu@Ag core–shell nanorod coated by zeolitic imidazolate framework (ZIF-8) with 4-aminothiophenol (4-ATP) as reporter molecule, loaded by DOXCell entrance through endocytosis, Enhanced drug’s therapeutic effectsCa[110]
DDDOX loaded Chlorella, GNRs and BSA-based GelIrradiation of 660 nm and 808 nm for oxygen and heat generation by Chlorella and GNR, respectivelyPT[111]
DDGNRs incorporated with Poly (N-isopropylacrylamide)-co-acrylamide polymer, loaded with a model drugPhotothermal hyperthermia and local drug releasePT[112]
DDGNR core, mesoporous silica shell, functionalized with d-α tocopherol PEG 1000 succinate and PEI, loaded by DOXNIR-activated drug releasePT[113]
DDPEGylated graphene oxide capped-GNR/SiO2 core–shell, loaded by DOXDual NIR- and Ph-activated drug releasePT[114]
DD, GDC(HK)4-mini PEG-RGD peptide-functionalized GNRsαvβ3 integrin targeted, Cell uptake by RGD (Arg-Gly-Asp) peptide, HK (His-Lys) peptide-conjugated deliveryCa[115]
DD, PTTcRGD peptide-modified GNRs loaded by paclitaxel and curcuminαvβ3 integrin targeted by cRGD, NIR-responsive drug release and tumor growth inhibition by apoptosisPT[116]
DD, PTTGNR @ silica coated/poly histidine/d-α-tocopherol PEG succinate/DOX-loaded PEGylated nanocompositeNIR thermal-/pH-triggered drug release and PTTPT[117]
DD, PTTGNRs coated by folate-α,β-poly(N-2 hydroxyethyl)-d,l-aspartamide, loaded by Antineoplastic drug irinotecan (Iri)Folate-targeted drug release, Photothermal hyperthermiaPT[118]
DD, PTTGNRs@Kr (Keratine), loaded by DOXNIR-triggered and redox-responsive drug releasePT[119]
DD, PTTHollow H-apatite capped (poly(N-isopropylacrylamide-co-acrylic acid) GNRs-DOX loaded hybrids microcapsulespH/NIR-responsive DOX releasePT[120]
DDGNR-polydopamine capped hollow h-apatite hybrid microcapsulespH/NIR-responsive drug releasePT[121]
DDGNR-chitosan modified by poly(l-lactide), poly(N-isopropylacrylamide) and poly (acrylamide) micelles, loaded by paclitaxellight triggered DD and PTTPT[122]
DDAlginate/polyacrylamide hydrogel patch -hybridized by polyvinylpyrrolidone- graphene oxide@GNRsNIR-stimulated thermoresponsive methotrexate and rhodamine B co-deliveryPT[123]
DDMulti-layer allylamine hydrochloride/poly methacrylic acid
capsules –DOX/nimbin co-delivery
NIR-stimulated drugs releasePT[124]
DD, PTTFolate-modified liposomes encapsulating NF-κB decoy oligodeoxynucleotides, GNRs, and dexamethasoneTherapeutic effects by anti-inflammatory agents delivery and PTTPT[125]
DD, ThCore–shell mesoporous silica-coated GNRs, deposited by the metal-organic framework, modified by hyaluronic acid, loaded by DOXLaser irradiation-mediated imaging and drug releasePA/PT[126]
DD, ImGNRs coated by poly platinum (polyPt(IV))Photothermal hyperthermia, redox-triggered polyPt(IV) releasePA/PT[127]
DD, PTTPLGA-PEG polymeric corona loaded by vincristine encapsulating silica-coated GNRspH-responsive sustained drug release and PTTPT[128]
DD, PTT, PDTHyaluronic acid encapsulated, GNRs coated with mesoporous and degradable silica, loaded with DOX and IR820 (a photosensitizer)Hyaluronidase and high glutathione concentration-targeted, triple therapy by NIR irradiation (PDT, PTT and chemotherapy)PT[129]
DDGNRs decorated Polymethacrylate co-polymers and ciprofloxacinTargeted delivery to bacteria and biofilms, PT effect and antibiotic deliveryPT[130]
DD, GDDOX, YAP-siRNA and GNRs loaded cationic liposomeTargeted chemo, PT and gene combination therapy using NIR irradiationPT[131]
DDGNRs-doped hydrogel, made of hyaluronic acid, PEG diacrylate/4-vinyl phenylboronic acid and Astragaloside IV nanoparticlesSustained drug release, as conductive as native myocardiumCo[99]
DD, PTTDOX, Mesoporous silica-capped GNRs covered with selenium nanoparticles shell (Se@Au@mSiO2)Chemo-drug, selenium nanoparticles as anticancer agent, and PTT ability of GNRs as cancer therapy systemPT[132]
Abbreviations: PT: photothermal, DD: drug delivery, PD: protein delivery, PTT: photothermal therapy, PA: photoacoustic, Co: conductivity, Ca: carrier, St: stability, PDT: photodynamic therapy, Im: imaging, GD: gene delivery, Th: theranostic, siRNA: small interfering RNA, YAP: yes-associated protein, PLGA: poly(lactic-co-glycolic acid).
Table 3. GNR-based photothermal (PT) effects in photothermal therapeutic systems (research-based studies).
Table 3. GNR-based photothermal (PT) effects in photothermal therapeutic systems (research-based studies).
PurposeSystem’s StructureMechanism of FunctionRef.
TD, PTTCD11b antibody @ GNRsNIR-induced targeted protozoan cell (tachyzoites) death[155]
PTTBSA-coated GNRs-laden-macrophagesRAW 264.7 macrophages, hepatic satellite cells (HSCs), HepG2 photo-induced cell damage, leading to lower tumor recurrence[159]
PTT, PDTGNRs @ silicon 2,3-naphthalocyanine dihydroxide stabilized with alkylthiol-conjugated polyethylene glycolPD & PT synergistic therapy, using a single specific laser wavelength[160]
PTTEncapsulated GNRs by lipid-based envelopeGood cellular uptake, enhanced NIR-induced PTT, caused cancer cells apoptosis [161]
TD, PTTVirus inspired therapeutic system (RVG 29-binded PEG-SiO2@ GNRs)Rabies virus glycoprotein (RVG)-guided for BBB crossing, brain glioma targeted PTT[156]
TD, Cancer PTTGNRs combined with a Nestin binding peptidePT-induced Nestin positive of Glioblastoma Multiform tumors cell apoptosis [162]
PTTSilica coated Au-TPGS (vitamin E polyethylene glycol succinate)-HA co-polymer NIR-induced thermal cancer cell death[163]
TD, PTTACE-2-functionalized GNRsNIR-induced thermal SARS-CoV-2 cell damage[157]
Gas synchronous PTTGNR- polydopamine (PDA) core–shell nanostructure doped by Benzothiazole sulfinate (BTS) pH triggered BTS (SO2 prodrug) release, resulting in deep tumor gas therapy and PTT [164]
PTTPLNP-GNR nanocomposite platforms consisting of ZGGO:Cr3+@CTAB@PW12 and GNR@CTAB, PW12- encapsulated Persistent luminescent nanoparticles combined with GNRs as biocompatible platform for photo-induced therapy.[158]
Abbreviations: TD: targeted delivery, PT: photothermal, PTT: photothermal therapy, PDT: photodynamic therapy.
Table 4. GNR applications in theranostics systems (research-based studies).
Table 4. GNR applications in theranostics systems (research-based studies).
PurposeSystem’s StructureMechanism of FunctionGNR’s Applied FeatureRef.
DD, TRKGNRs@folic acid as the targeting ligand the anthracycline drug, DOXTargeted drug delivery to cancerous cells (expressing folate receptor cells), fluorescence lifetime imaging, using photoluminescence of GNRs and the innate fluorescence of DOXPL[174]
DD, TRKAu@mesoporous SiO2/rhodamine B isothiocyanate (Au@mSiO2/RBITC) nanocomposite Monitoring the photothermal therapy, drug release, cell tracking using an 808 nm laser and a confocal laser scanning microscopy system PT[175]
DD, In situ ATP TRKDNA-functionalized GNRs loaded DOXTargeted drug delivery, drug release in response to aptamer–ATP interaction, tumor growth inhibition and increasing fluorescence emission of DOX proportional to ATP concentrationPT[173]
PTT and ImPEG-peptide-modified GNRs containing peptide substrate, overexpressed in malignant tumor cellsFormation of GNR aggregates in response to uPA activity and PEG-chains release and absorption reduction in tumor’s sitePT[176]
PTT, PDT, ImCe6-doped mesoporous silica- nanorods (AuNRs-Ce6-MSNRs)Generation of hyperthermia by GNRs (PTT) and singlet oxygen (1O2) production by Ce6-mediated PDT. Dual-imaging by the photoacoustic and NIR-induced fluorescence of AuNRs & Ce6PT/PA[177]
Thchlorin e6 (Ce6)-PEG-GNRPTT and PDT, using pH-responsive Ce6 (fluorescent tag)-based local tumor tracking and dual IR imagingPT/PDT[72]
PTT, PDT, ImAuNR@MOFs nanocomposite loaded CPTphotothermal-induced CPT release and photodynamic combinational therapy PT/PDT/PA[172]
Th (Im, PDT & PTT synergy/TDPheophorbide-responsive glutathione functionalized GNRs, conjugated folic acid-PEG co-polymerLight triggered imaging and NIR-induced synergistic therapeutic PT/PDT[178]
Im, PTTGraphene oxide (GO)-GNR contrasting thermal conductivity capacity and heating therapy.NIR-induced imaging and thermal stone dissolution PA/PT[179]
Th (Im, PDT, PTT, TRK)(PNIPAM) coated SiO2@GNR loaded ICG (nanocom-ICG), thermal-induced targeted release for tumor therapyNIR-induced tracking and enhancing the thermal release of ICG-mediated photodynamic therapy PA/PT[180]
ThEu@SiO2@GNR complex as multiplexed PT agent, photo and radio sensitizerImage-Guided Photothermal and X-ray-induced Photodynamic Therapy of TumorsPT/PDT/PA[181]
Abbreviations: TRK: tracking, TD: targeted delivery, PL: photoluminescence, PT: photothermal, PA: photoacoustic, Im: imaging, PTT: photothermal therapy, PDT: photodynamic therapy, Th: theranostics.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jahangiri-Manesh, A.; Mousazadeh, M.; Taji, S.; Bahmani, A.; Zarepour, A.; Zarrabi, A.; Sharifi, E.; Azimzadeh, M. Gold Nanorods for Drug and Gene Delivery: An Overview of Recent Advancements. Pharmaceutics 2022, 14, 664. https://doi.org/10.3390/pharmaceutics14030664

AMA Style

Jahangiri-Manesh A, Mousazadeh M, Taji S, Bahmani A, Zarepour A, Zarrabi A, Sharifi E, Azimzadeh M. Gold Nanorods for Drug and Gene Delivery: An Overview of Recent Advancements. Pharmaceutics. 2022; 14(3):664. https://doi.org/10.3390/pharmaceutics14030664

Chicago/Turabian Style

Jahangiri-Manesh, Atieh, Marziyeh Mousazadeh, Shirinsadat Taji, Abbas Bahmani, Atefeh Zarepour, Ali Zarrabi, Esmaeel Sharifi, and Mostafa Azimzadeh. 2022. "Gold Nanorods for Drug and Gene Delivery: An Overview of Recent Advancements" Pharmaceutics 14, no. 3: 664. https://doi.org/10.3390/pharmaceutics14030664

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