Gold-Based Nanostructures for Photo-Triggered Cancer Theranostics: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 1 by Nagaraj Basavegowda.

Cancer is one of the most dangerous health problems in the millennium and it is the third foremost human cause of death in the universe. Traditional cancer treatments face several disadvantages and cannot often afford adequate outcomes. It has been exhibited that the outcome of several therapies can be improved when associated with nanostructures. In addition, a modern tendency is being developed in cancer therapy to convert single-modal into multi-modal therapies with the help of existing various nanostructures. Among them, gold is the most successful nanostructure for biomedical applications due to its flexibility in preparation, stabilization, surface modifications, less cytotoxicity, and ease of bio-detection.

  • cancer therapy
  • gold nanostructures
  • synergetic therapies
  • nanomaterials
  • biomedical applications

1. Introduction

Cancer is a leading public health issue and the second primary reason for death at the global level. In 2020, nearly 10 million deaths were reasoned due to cancer as reported by the World Health Organization. Incredibly, about six out of one death globally is thought to be because of cancer [1]. The rising awareness of the molecular and cellular facts that reason cancer has permitted the advancement of novel approaches to handle this disease [2]. Still, the most general and traditional therapy surgery for clearing the tumor tissue is tailed by chemo/radiotherapy, either single or multi-modal, which is commonly based on the category and spreading of cancer. There is no united method to handle all categories of cancer, however, single-mode treatment not exhibiting a cent percentage efficiency for each case [3,4][3][4]. The conventional interventions for cancer have exhibited certain boundaries because of the absence of targeting ability toward the malignant consequently it rises several side effects. Moreover, these conventional treatments are incapable to eliminate the tumors from the body. These traditional approaches can also generate strong specific pressure aids to develop resistance against the therapy [5,6,7][5][6][7]. The golden standard method depends on the surgical clearances of cancer-affected parts, which is a promising way for the initial stage of the disease. Though, imperfect resection may again seed the malignant cells at the infected region, which then are prone to cancer regeneration [8]. Chemotherapy utilizes organic molecules-based drugs to control and abolish the cancer cell multiplications, whose restricted efficiency, heavy cytotoxicity, and multi-drug resistance of cancer cells have induced the advancement of novel organic/inorganic compounds [9,10,11][9][10][11]. Commonly, chemotherapy is jointly applied with surgery, radiotherapy, and immunotherapy [7]. Radiotherapy utilizes high-energy ionizing radiation to destroy the tumor sites. Usually, radiotherapy is a supportive approach to chemotherapy to enhance the ablation of tumor sites [8,12,13][8][12][13]. Based on these limitations of these traditional therapies have led to the development of modern approaches, from synergetic therapies that depend on regular anticancer drugs to radial novel strategies that make use of advanced equipment [14].
Among these modern treatments, phototherapy has gained enormous attention and emerged as an alternative to traditional cancer therapies. Phototherapy abolishes cancer cells by selectively triggering photochemical and/or photothermal processes. The purpose of phototherapy is to abolish cancer cells by generating reactive oxygen species (ROS) or heat using photoactive agents. Phototherapy can be divided into two types according to their different mechanisms of action: photodynamic therapy (PDT) and photothermal therapy (PTT) [15]. Nanoparticle-based phototherapies have encouraged novel strategies that involve the specific targeting ability of biomolecules to the tumor microenvironments, the prospect of applying novel forms to combine various treatments such as photodynamic therapy and photothermal ability, in combination or single mode. Nanotechnology-based cancer therapies may provide various exciting opportunities for the advancement of novel approaches for imaging and therapy as well as enhances the efficiency of currently existing treatments [5,7,8,15,16][5][7][8][15][16].
In recent years, many multifunctional nanocarriers have been reported applying numerous kinds of organic/inorganic nanoparticles in which different components may be joined into one nanoplatforms for synergetic imaging and therapy of cancer. Commonly, organic-based nanocarriers comprise dendrimers, polymers, and liposomes nanoparticles; whereas inorganic-based nanocarriers prominently consist of magnetic, plasmonic, mesoporous silica nanoparticles, quantum dots, and carbon-mediated nanomaterials, among others [17,18][17][18]. Heterocyclic structure-based organic nanoparticles have several for clinical photodynamic treatments, such as quick chemical functionalization, minimum cytotoxicity, hemocompatibility, and biodegradability in physiological conditions [18,19][18][19]. Though organic nanoparticles are only triggered by ultraviolet-visible sources and in some specific cases include near-infrared radiation also, which may restrict direct application and specific targeting ability to therapy [17]. On the other hand, inorganic-based nanoparticles particularly noble metals nanoparticles (Au, Ag, Pt, Pd) usually have good near-infrared (NIR) radiation absorption, which is more promising for deep penetration on phototherapies.

2. Light-Based Cancer Therapies

Light develops physiological changes in cells and stimulates an endogenous biological chemical reaction either directly or indirectly [8,24][8][20]. The light-based biochemical activity can be extensively classified into those relying on the applications of photoconversion efficiency. In PDT, organic/inorganic nanoparticles can absorb light, this will then stimulate the chemical changes leading to the therapeutic outcome. While in PTT, the absorbed light by inorganic nanoparticles will stimulate the transformation of irradiating laser into localized thermal energy [8,19][8][19]. Other approaches utilize the capacity of GNPs to develop high energetic radiation (radiotherapy) to focus therapy on the cancer region. The potential merits of phototherapies are minimal cytotoxicity, invasive, and specific tumor ability [17]. Commonly, lasers are applied as radiation sources for photo-based cancer theranostics which can produce a monochromatic light that can be allowed via an optical fiber and attainted the target region directly [25][21]. The wavelength of the laser irradiation should depend on the capability to absorb the light by photosensitizers (PS), the location of the tumor, size, and various other factors to standardize the activation of the PS [27,28][22][23]. For effective clinical practice, phototherapies are strongly dependent on wavelength, exposure duration, penetration power, mode of delivery, and total dose of the light [29][24]. From 600 to 800 nm is the common wavelength range applied for phototherapies named a therapeutic window, within this wavelength, the energy of radiation can excite the PS permitting efficient tissue penetration but restricting the absorption of light by other cellular parts (cytochromes) [28][23]. PDT consists of three potential components as Oxygen, PS, and light source for cancer therapy [30][25]. One major advantage of PDT, it can proceed in the repetitive mode without generating immune/myelosuppressive effects and is also administered even after traditional therapies. The standard PS ligand should be a single pure molecule that permits quality assurance research with resonance stability. The PS can administer via topical or intravenous injections. However, the alteration in biodistribution over a long duration gets affected; the alternative path to regulate the impact of PDT is the duration of light exposure. The laser absorption, the sensitizer, is transformed from a single state (short-lived) to a triplet state (long-lived). This triple state responds in two types such as (i) triple state reacts directly to the cell membrane and transfers the molecules into free radicals. When these free radicals react with oxygen molecules to form the oxygenated products (type I). (ii) Another way, the triple state can transfer its energy to the oxygen molecules and transform the singlet oxygen into a reactive oxygen species (type II). However, commonly all PDT drugs are oxygen-dependent, this therapy does not work in the anoxia region of the infected organ. Near-infrared radiation has been widely applied to produce thermal spots and serve the resolution. Compared to other techniques, PTT has a great attraction to offer excellent therapeutic value because of its low invasiveness, enhanced therapeutic efficiency, low side effects, no need for long-lasting therapy, and fast recovery [38][26]. A photothermal agent can be allowed in the tumor microenvironment region and absorb the NIR laser lights to generate kinetic energy that releases thermal energy on the cancer cells and lead to cell death [39][27]. The permitted capacity of NIR absorption on healthy organs in the wavelength between 650–1350 nm develops crucial interpenetration in the patient’s body and destroys the tumors [40][28]. Additionally, this technique develops thermal waves to generate trouble in the cell membrane of the adjacent cancer cells [41][29]. Hyperthermia via absorption of NIR laser develops some problems such as short inter-penetrating power and strong absorption which leads to developing injury on normal cells and reducing the therapeutic efficiency. In the case of surface plasma resonances holding nanostructures, showed the electrons on the conduction band were localized on the surface of the nanomaterials upon NIR applications, and subsequently the localization attaints the target at the resonant frequency to develop resonances, thereafter absorbed NIR transforming into thermal waves. Therefore, it is probable to control NPS via local intravenous injection and consequently, exciting them in the NIR open windows I and II. Additionally, the nanomaterials incline to deposits in the spleen, kidney, and liver inducing injury irreversibly [42,43][30][31]. Hence, it is very essential to develop the PTT approach more safely and effectively.

3. Gold Nanostructures in Nanomedicine

Inorganic nanomaterials have more potential properties than organic nanomaterials due to colloidal stability, ease of synthesis in various adjustable sizes, and optical, magnetic, and quick surface modifications make them biocompatible materials in the area of biomedical applications [44,45][32][33]. Moreover, organic-based nanoparticles showed a higher degradation rate when compared to inorganic nanomaterials, this advantage makes them more colloidal stable in physiological conditions [46][34]. Among several types of inorganic nanoparticles, GNPs are promising candidates for photo therapies, due to their bioinertness, low cytotoxicity, as well as ease of preparation, and surface modifications [47][35]. Additionally, gold nanostructures are capable to improve the passive cellular uptake of phototherapies carriers in cancer sites through the enhanced permeability and retention (EPR) effect [48,49][36][37]. Further, GNPs hold a great surface area, which can aid in quick surface modifications with different bioactive molecules for active targeting in cancer therapy [50][38]. With the high binding affinity of GNPs with thiol and amine groups, the surface of GNPs can be modified with nucleic acids, proteins, and antibodies, which enable specific targeting ability and improve PSs delivery in tumor microenvironments [51][39]. The photostability of the GNPs leads to the high efficiency of absorbed laser radiation to thermal conversion which is photothermal conversion efficiency. The most important characteristics of GNPs is optical property originating from the localized surface plasmon resonance (LSPR) and huge surface-to-volume ratio that permits bioconjugation to bioactive molecules by a combination of various strategies of surface chemistry leads to develop conjugation of anticancer drug, targeting agent, and imaging probe in single nanoplatform [52][40]. In 1857, first time Faraday reported the synthesis of a colloidal solution of GNPs from the reduction of gold chloride (AuCl4) by phosphorus [44][32]. Turkevich and co-authors reported the colloidal GNPs through the chemical reduction of gold salt with trisodium citrate [53][41]. Currently, apart from spherical GNPs, various types of gold nanostructures of different morphology have been reported, such as nanoshells, nanostars, nanorods, and nanocages [54,55,56,57][42][43][44][45]. The LSPR indicates the coherent oscillation of excited electrons from the surface of the metals by absorbing strong NIR radiation [54][42]. The rise of this oscillation increased absorption of the electromagnetic radiation in the combination of SPR of the metallic nanomaterials, which is estimated by the morphology and size of the nanostructures. The optical properties of metal nanomaterials have been extensively used in nanomedicine, from real-time molecular imaging to multimodal therapeutic based on the use of light, for instance, gaining from the efficient light to thermal conversion gold nanostructures [58][46]. One major issue that was raised on the usage of nanomaterials in clinical practice is the potential cytotoxicity of these nanoparticles. Many nanoparticles have been shown to indicate cytotoxicity for both cancer and healthy cells, because of oxidative stress and the stimulation of inflammatory effects [58,59,60,61][46][47][48][49]. These cytotoxicity effects are commonly based on the structure and size of the nanomaterials [62][50]. The GNPs with sizes ranging from 20–60 nm showed low cytotoxicity for biomedical applications. Their use has been progressively permitted by regulatory frameworks advanced by the Federal Food and Drug Administration [63,64][51][52]. Moreover, surface charge shows a strong influence on acute cytotoxicity, in GNPs with positive charges showed much more toxicity to tissues when compared to negatively charged nanomaterials [65,66][53][54]. Currently, GNPs are extensively applied in biomedical science, particularly in molecular diagnosis, biosensing, nanocarrier for drug delivery, and phototherapy agents owing to their electronic, optical, and colloidal stability. In presence of LSPR, GNPs may be suitable for therapy depending on the variety of wavelengths in the NIR, adapting them for nanomedicine as carriers in PDT or photothermal agents in cancer phototherapy. Indeed, when GNPs are treated by NIR laser radiation, most often in the NIR region (650–900 nm), they appear to be effective in converting photons to thermal waves. When GNPs are irradiated, they will scatter thermal waves in a localized path, generating an effective flow in temperature (40–42 °C), which sequentially has a deep influence on the survival of cancer cells that are lowly resistant to localized thermal waves when compared to healthy cells. From these results, hyperthermia has been suggested as a unique non-invasive cancer theranostics with localized thermal effects and without side effects to healthy tissues or cells. Many light sources have been suggested, from radiofrequency to laser are applied to stimulate the irradiation in gold nanostructures for phototherapy [67,68,69,70,71,72][55][56][57][58][59][60].

4. Optical Properties

Effects of Size and Shape on Optical Properties of Gold Nanostructure

4.1. Effects of Size and Shape on Optical Properties of Gold Nanostructure

The optical properties of gold nanostructures differ from other metallic nanomaterials. When gold nanostructures are treated with light, the excited electrons from the GNPs surface generated coherent oscillations which are named LSPR [73][61]. Due to this LSPR, deep amplified and localized electromagnetic fields are produced in the gold nanostructure surface upon irradiation with a suitable wavelength of the laser. The absorbed NIR radiation by GNPs would decay radiatively by dispersing the emitting light with the same wavelength as the incident light, meanwhile the nonradiative relaxation, potentially the delivery of localized thermal energy. Hence, GNPs concentrate the orders of magnitude optical absorption, which in turn open the window for using the gold nanostructures as various bioimaging and biosensing agent beyond photo-associated therapies. As LSPR is influenced by the density and motion of electrons on the GNPs surface, the distinct size and variety of morphology of gold nanostructures depend on their scattering and plasmonic absorption. Gold nanostructures are commonly prepared through the reduction of gold salt using various reducing agents, and suitable reaction methodology has been widely applied to attain different sizes and shapes. Gold nanorods, nanoshells, nanocages, and nanorings have been widely studied as promising candidates for phototherapy. Based on biomedical applications, the NIR ranging from 750 to 1700 nm is better for tissue penetration because of low tissue scattering. Especially, the radiation ranging from 1000 to 1350 nm (second NIR window) can penetrate deepest than the range from 750 to 1000 nm (first NIR window). However, very low assessable biocompatible probes exist for pre-clinical applications in the NIR window. Incidentally, the probability of shifting the laser absorption and the plasmonic band of gold nanostructures from visible to NIR region by tuning their sizes and varying the shapes makes them hopeful replacements for in vivo applications in the phototherapy of cancer [21][62]. Currently, the appreciation of translation of LSPR within the suitable NIR region from anisotropic gold nanostructures has stimulated the preparation of a variety of gold nanostructures such as gold nanoclusters, nanostars, and nanoplates. Among many, certain common functional groups are associated with the gold nanostructures to attain favorable multi-functionality for cancer theranostics. Advanced nanotechnology area provides the chance to build GNPs of different sizes and morphology. The physicochemical characteristics of GNPs differ among various functionalization and these characteristics could be accurately regulated through nanotechnology with the drive of connecting interdisciplinary fields (chemistry, biological, and physical) needs of the multifunctional phototherapies. Each of these gold nanostructures of various shapes holds certain special natures and their feature change openly depending on any rule. Important features of phototherapies, for instance, NIR absorbance and clearance rate, are majorly based on the shapes and sizes of these gold nanostructures. Materials science scientists must conduct various investigations to analyze the properties of these gold nanostructures to find those GNPs holds most desirable for phototherapies. Commonly, for a specific type of GNPs, the maximum extinction wavelength (λmax) increases with the increasing size of the gold nanostructures. GNPs with smaller sizes exhibit much better absorption capacity and a minimum scattering-to-absorption efficacy ratio. Hence, the GNPs have larger size that exhibits higher scattering efficiency. In this regard, large-sized GNPs are recommended for good resolution and sensitive imaging-guided phototherapies in cancer and small-sized GNPs have higher absorption capability that is suggested for good photothermal conversion efficiency in phototherapies. Determined maximum wavelength (λmax) and the ratio of scattering efficiency (µs) to absorption efficiency (µa) for GNPs with various shapes and sizes [74,75][63][64].

5. Phototherapies-Based Clinical Trials in Gold Nanostructure

Over the past many eras, ongoing efforts have been made toward achieving an optimum association of nanoparticles and hyperthermia to overwhelm the issues of conventional thermal therapies. Among several reported nanoparticles for phototherapies applications, GNPs have been widely studied because of their high photothermal conversion efficiency and rapid surface modifications for improved targeted delivery of PS [79][65]. Plasmon resonance is a combined motion of a more number of mobile electrons [80,81,82][66][67][68]. In nanoparticle-mediated hyperthermia, GNPs can absorb strong NIR radiation and then produce the primary source of heat and reverse the direction of heat loss to stimulate thermal ablation on tumor cell [83,84,85,86][69][70][71][72]. Currently, various gold nanostructures have been translated for clinical trials, potential struggle has resulted in the reports of multiple research works on gold-based phototherapies in the recent literature such as gold nanocages, nanobipyramids, nanoflowers, nanoshell, nanorods, hollow nanospheres, nanostars, nanoechius and plasmonic blackbodies [87,88,89,90,91,92,93,94,95,96,97,98,99,100,101][73][74][75][76][77][78][79][80][81][82][83][84][85][86][87]. However, significant translations of these nanomaterials to pre-clinical trials have not yet been attained. The potential clinical trial on phototherapies is mostly based on irradiation duration, gross irradiation dose, and mode of radiation delivery. Moreover, traditional passive targeting phototherapies based on gold nanostructures have been circumscribed in clinical trials because of their non-specific ability and generate side effects on normal tissues. Hence, currently designing active targeting GNPs-based phototherapies are under clinical trials.

6. PS Conjugated Gold Nanocarriers for PDT

Even though the PDT has significant efficiency for cancer therapy, still it’s mainly limited to in vivo investigations and loss-efficient translation into the clinical trial [110][88]. In this regard, nanomaterials conjugated PS-based PDT has developed as a modern approach in which nanoparticles act as carriers for PS gaining from the special properties that generate them as PS themselves [111][89]. Various chemotherapeutic agents have been delivered by functionalizing the surface of GNPs. Thus, GNPs have aided the specific-targeted delivery to cancer sites of different biomolecules utilized as therapeutic agents. PS-conjugated GNPs result in significantly increased electron transfer between the gold nanostructure and the photoactive dyes which improves the photodynamic efficiency [112][90]. Up to date only a few of these gold-conjugated PS-mediated PDT nanoplatforms achieved clinical trials. Recently, Anine Crous reported the application of AuNPs as a carrier for the AlPcS4Cl PS. In this study, synthesized gold nanoparticles were conjugated AlPcS4Cl, and in vitro models revealed that phototherapy using the AlPcS4Cl-gold nano bioconjugate was potential against human lung (A549) cancer cells [113][91]. In another approach, specific targeted PDT was carried out with folic acid (FA) and protoporphyrin IX (PPIX) conjugation on surface-modified AuNPs. The authors initially applied 6-mercapto-1-hexanol (MH) on the surface of AuNPs and it enhances the bioconjugation of FA/PPIX on nanoparticles to form a nanosystem (PPIX/FA-MH-AuNP). In this study, the synthesized nanosystem showed high cytotoxicity against HeLa cancer cells, due to the presence of targeting ligands FA and folate-mediated endocytosis. The results show that the PPIX/FA-MH-AuNP nanosystem increases the targeting nature and also phototoxicity efficiency of HeLa cells when compared with the traditional PDT [114][92]. In another approach, gold-based core@shell nanostructure was utilized to conjugate with PS for effective PDT. Ping and co-authors developed the nanosystem of Au@TiO2 nanostructure with hematoporphyrin monomethyl ether (HMME). The PDT efficiency of Au@TiO2-HMME was studied in KB cancer cells [115][93]. Dimakatso and co-authors fabricated the physically loaded hypericin (Hyp) on gold nanoparticles through sonication. The non-covalent bonds between the AuNPs and Hyp increase the PS accumulation in MCF-7 breast cancer cells and hence improve PDT efficiency with the minimum concentrations. From this study, it is revealed that physically loaded Hyp PS on AuNPs is a hopeful strategy for hydrophobic PS drug delivery to improve PDT efficiency [116][94]

7. Gold Nanostructures-Based Targeting Hypoxia for PDT

Hypoxia commonly exists in the tumor microenvironment due to the excess proliferation of cancer cells that surpass the blood circulation and oxygen supply for the growth of weak vasculature tumors. It has been revealed that hypoxic efficiently promotes cancer development and invasion [126][95]. The potentiality of PDT is restricted by hypoxia since the traditional PDT is oxygen dependent to generate reactive oxygen species (ROS). Hence, targeting hypoxia in the tumor sites in absence of oxygen-based PDT has developed as a potential tool to enhance PDT. A recent review by Lou-Franco reported that gold nanostructures hold efficient enzymatic activities for instance reductase, peroxidase, oxidase, and superoxide dismutase, which can be leveraged to relieve hypoxia [127][96]. In a recent study, an oxygen self-producing nanosystem containing metal-organic frameworks, AuNPs and Ce6 PS was utilized to diminish the hypoxic which consequently improves PDT. In this approach, AuNPs are applied to catalase efficiently by catalyzing the high H2O2 developed in the tumor sites to generate oxygen molecules to reduce hypoxia and improve ROS generation with high cytotoxicity [128][97]. Moreover, to reduce hypoxia and improve PDT, core@shell Au@Rh nanoparticles based on Au and rhodium (Rh) with catalase activities were applied as nanoenzyme. The ICG was conjugated with Au@Rh and functionalized with a cancer cell membrane (CM) to develop Au@Rh-ICG-CM nanosystem. The nanosystem catalyzed the H2O2 in the tumor sites to form oxygen via the action of Au@Rh to improve ROS generation for efficient PDT. The functionalization with CM permits specific targeting via homology adsorption [129][98]. In a very recent study, Yin and co-authors reported gold nanoclusters (AuNCs), mesoporous silica (mSiO2), and MnO2-based nanoenzyme system with oxygen generators for improved PDT and magnetic resonance imaging. In this study, the nanosystem made up of AuNCs doped on mSiO2 was coated with MnO2 and acted as a shell (AuNCs@mSiO2@MnO2). In lower pH, becomes turn ‘on’ the nanoenzyme is treated with H2O2 consequently resulting in the degradation of MnO2. The slow degradation of MnO2 generates massive oxygen molecules, which in turn enhances Magnetic Resonance Imaging and PDT [131][99].

8. Synergetic Phototherapies Based on Gold Nanostructures

The boundaries of cancer nanotheranostics are constantly being altered as better thought of biomedical characteristics of nanomaterials is expanding. Over the last two decades, there is an outstanding evolution in the fabrication and advantages of various nanostructures in cancer nanotheranostics [132][100]. In this regard, GNPs have currently emerged as the most hopeful theranostic agent [133,134][101][102]. The gold nanostructures are the potential ability for passive accumulation and preferentially reach the tumor microenvironment through enhanced permeability and retention effect of the infected organs [135][103]. In addition to that, the surface of gold nanostructures can be readily modified with biomolecules such as peptides, monoclonal antibodies, and proteins to overcome the non-specific targeting ability of theranostics agents [136,137,138,139][104][105][106][107]. Due to their natural biointerness, surface area-to-volume ratio, and surface chemistry, gold nanostructures are widely applied as nanocarriers for targeted drug delivery [140][108]. The high atomic numbers in gold are capable to afford a higher X-ray absorption cross-section, making them promising nanomaterials to act as potential radiosensitizers for improving radiotherapy (RT) [141][109]. Moreover, the photothermal conversion efficiency of gold nanostructures can be used to produce localized thermal for tumor ablation [142][110]. In recent years, the thermal efficiency of gold nanomaterials has been discovered and utilized to help hyperthermia in curing cancer. According to this, the radiofrequency (RF) electrical field absorption efficiency of gold nanostructure combined with their sonosensitizing characteristics has been employed to enhance the RF and ultrasound-stimulated hyperthermia therapeutic efficiency [143][111]. In addition to these, the thermal efficiency of gold nanostructures can be applied to stimulate responsive drug payload delivery with good specific and resolution upon external triggering through hyperthermia materials [144,145][112][113]. Currently, many biomaterials scientists have also revealed that gold nanostructures are capable to produce cytotoxic ROS upon laser and ultrasound treatments, and could therefore be applied to promote PDT and sonodynamic therapy simultaneously [146,147][114][115]. With these characteristics combined into a single nanoplatform, gold nanostructures have emerged as an entirely irreplaceable nanostructure ability to combine various therapies to attain improved therapeutic outcomes. Hence, they could deliver a golden choice to combine various therapies in a single mode, thus emphasizing the influence of gold nanostructure in synergetic cancer theranostics. Taking the benefit of gold nanostructures for real-time co-delivery of various therapies cannot only improve the potentiality of every individual cancer treatment but fascinatingly may also deliver an extra advantage by the synergistic interactions that happen among different therapies, which results in very powerful therapeutic outcomes. This synergistic therapy also aids to limits high dose concentrations and associated side effects by permitting a reduction in administered drugs level for cancer treatments [148][116]. Table 1, provides a summary of the therapeutic aids of various gold nanostructures for synergetic phototherapies for cancer theranostics.
Table 1.
The summary of the therapeutic aids of various gold nanostructures for synergetic phototherapies for cancer theranostics.

Gold Nanostructures

Multifunctional

Nano Platforms

Therapeutic Agents

Imaging Model

Therapy Model

In Vitro Models

Ref.

Pharmaceutics 15 00433 i006

Nanocages

BPQD-AuNCs

DOX and QUR

FL

PDT-CT

MCF-7/ADR

[149][117]

DOX/ICG-biotin-PEG-

AuNC-PCM

DOX and ICG

FL

PDT-CT-PTT

MCF-7/ADR

[150][118]

AuNCs-HA

HA

PA

PDT-RT-CT

4T1

[151][119]

Pharmaceutics 15 00433 i007

Nanocluster

Ce6-GNCs-DOX

Ce6 and DOX

FL

PDT-CT

A549

[152][120]

Ce6-GNCs-Ab-CIK

Ce6 and CD3 antibody

FL

PDT-IT

MGC-803

[153][121]

AuS-U11

5-ALA and Cy5.5

FL

PDT-PTT

PANC1-CTSE

[154][122]

Gd2O3-AuNCs-ICG

ICG

MRI/CT

PDT-PTT

HeLa

[155][123]

Au NCs-INPs

ICG

NIRF/PA

PDT-PTT

4T1

[156][124]

Pharmaceutics 15 00433 i008

Nanorods

GNRs-MPH-ALA/DOX-PEG

5-ALA and DOX

NIRF/PA

PDT-CT-PTT

MCF-7

[157][125]

FA-PEG-P(Asp)-

DHLA-AuNR100-SS-Ce6

Ce6

-

PDT-PTT

MCF7 and A549

[158][126]

AuNR-PEG-PEI-APP/Ce6—ADSC

Ce6

-

PDT-PTT

MCF-7

[159][127]

AuNRs-Ce6-MSNRs

Ce6

NIRF/PA

PDT-PTT

4T1

[160][128]

PEG-GNR-ACPI

PpIX

FL

PDT-PTT

SCC-7

[161][129]

Pharmaceutics 15 00433 i009

Nanoshells

GGS-ICG

ICG

 

PDT-PTT

4T1

[162][130]

Pt@UiO-66-NH2-Aushell-Ce6

Ce6

FL

PDT-PTT

MCF-7

[163][131]

ICG-Au@BSA-Gd

ICG

NIRF/PA/CT/MR

PDT-PTT

4T1

[164][132]

Pharmaceutics 15 00433 i010

Nanostars

GNS@CaCO3/Ce6-NK

Ce6

NIRF/PA

PDT-IT-PTT

A549

[165][133]

GNS-PEG-Ce6

Ce6

FL

PDT-PTT

A549

[102][134]

GNS@BSA/I-MMP2

MMP2

NIRF/PA

PDT-PTT

A549

[166][135]

GNS@CaCO3/ICG

ICG

NIRF/PA

PDT-PTT

MGC803

[167][136]

References

  1. Ritchie, H.; Roser, M. Drug Use. Our World in Data. 2020. Available online: https://ourworldindata.org/cancer (accessed on 30 October 2022).
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424, Erratum in CA Cancer J. Clin. 2020, 70, 313.
  3. Phung, D.C.; Nguyen, H.T.; Phuong Tran, T.T.; Jin, S.G.; Yong, C.S.; Truong, D.H.; Tran, T.H.; Kim, J.O. Combined hyperthermia and chemotherapy as a synergistic anticancer treatment. J. Pharm. Investig. 2019, 49, 519.
  4. Roma-Rodrigues, C.; Mendes, R.; Baptista, P.V.; Fernandes, A.R. Targeting tumor microenvironment for cancer therapy. Int. J. Mol. Sci. 2019, 20, 840.
  5. Roma-Rodrigues, C.; Fernandes, A.R.; Baptista, P.V. Exosome in tumour microenvironment: Overview of the crosstalk between normal and cancer cells. Biomed Res. Int. 2014, 2014, 179486.
  6. Zhang, J.; Jiang, C.; Longo, J.P.F.; Azevedo, R.B.; Zhang, H.; Muehlmann, L.A. An updated overview on the development of new photosensitizers for anticancer photodynamic therapy. Acta Pharm. Sin. B 2018, 8, 137.
  7. Chen, X.; Wong, S. (Eds.) Cancer Theranostics; Academic Press: Cambridge, MA, USA, 2014.
  8. Cabral, R.M.; Baptista, P.V. The chemistry and biology of gold nanoparticle-mediated photothermal therapy: Promises and challenges. Nano LIFE 2013, 3, 1330001.
  9. Elahi, N.; Kamali, M.; Baghersad, M.H. Recent biomedical applications of gold nanoparticles: A review. Talanta 2018, 184, 537–556.
  10. Martins, P.; Marques, M.; Coito, L.; Pombeiro, A.J.L.; Viana Baptista, P.; Fernandes, A.R. Organometallic compounds in cancer therapy: Past lessons and future directions. Anti-Cancer Agents Med. Chem. 2014, 14, 1199.
  11. Silva, T.F.; Smoleński, P.; Martins, L.M.; Guedes da Silva, M.F.C.; Fernandes, A.R.; Luis, D.; Silva, A.; Santos, S.; Borralho, P.M.; Rodrigues, C.M.; et al. Cobalt and Zinc Compounds Bearing 1, 10-Phenanthroline-5, 6-dione or 1, 3, 5-Triaza-7-phosphaadamantane Derivatives–Synthesis, Characterization, Cytotoxicity, and Cell Selectivity Studies. Eur. J. Inorg. Chem. 2013, 2013, 3651–3658.
  12. Haume, K.; Rosa, S.; Grellet, S.; Śmiałek, M.A.; Butterworth, K.T.; Solov’yov, A.V.; Prise, K.M.; Golding, J.; Mason, N.J. Gold nanoparticles for cancer radiotherapy: A review. Cancer Nanotechnol. 2016, 7, 1–20.
  13. Huang, X.; El-Sayed, M.A. Plasmonic photo-thermal therapy (PPTT). Alex. J. Med. 2011, 47, 1–9.
  14. Beik, J.; Abed, Z.; Ghoreishi, F.S.; Hosseini-Nami, S.; Mehrzadi, S.; Shakeri-Zadeh, A.; Kamrava, S.K. Nanotechnology in hyperthermia cancer therapy: From fundamental principles to advanced applications. J. Control. Release 2016, 235, 205.
  15. Hu, Y.; Fine, D.H.; Tasciotti, E.; Bouamrani, A.; Ferrari, M. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2012, 4, 638.
  16. Mendes, R.; Carreira, B.; Baptista, P.V.; Fernandes, A.R. Non-small cell lung cancer biomarkers and targeted therapy-two faces of the same coin fostered by nanotechnology. Exp. Rev. Precis. Med. Drug Dev. 2016, 1, 227.
  17. Vankayala, R.; Hwang, K.C. Near-infrared-light-activatable nanomaterial-mediated photo theranostic nanomedicines: An emerging paradigm for cancer treatment. Adv. Mater. 2018, 30, 1706320.
  18. Cai, Y.; Si, W.; Huang, W.; Chen, P.; Shao, J.; Dong, X. Organic dye based nanoparticles for cancer phototheranostics. Small 2018, 14, 1704247.
  19. Zhu, X.M.; Wan, H.Y.; Jia, H.; Liu, L.; Wang, J. Porous Pt Nanoparticles with High Near-Infrared Photothermal Conversion Efficiencies for Photothermal Therapy. Adv. Healthc. Mater. 2016, 5, 3165–3172.
  20. Vines, J.B.; Yoon, J.H.; Ryu, N.E.; Lim, D.J.; Park, H. Gold nanoparticles for photothermal cancer therapy. Front. Chem. 2019, 7, 167.
  21. Gao, D.; Guo, X.; Zhang, X.; Chen, S.; Wang, Y.; Chen, T.; Huang, G.; Gao, Y.; Tian, Z.; Yang, Z. Multifunctional phototheranostic nanomedicine for cancer imaging and treatment. Mater. Today Bio. 2020, 5, 100035.
  22. Srivatsan, A.; Missert, J.R.; Upadhyay, S.K.; Pandey, R.K. Porphyrin-based photosensitizers and the corresponding multifunctional nanoplatforms for cancer-imaging and phototherapy. J. Porphyr. Phthalocyanines 2015, 19, 109–134.
  23. Banerjee, S.M.; MacRobert, A.J.; Mosse, C.A.; Periera, B.; Bown, S.G.; Keshtgar, M.R.S. Photodynamic therapy: Inception to application in breast cancer. Breast 2017, 31, 105–113.
  24. Agostinis, P.; Berg, K.; Cengel, K.A.; Foster, T.H.; Girotti, A.W.; Gollnick, S.O.; Hahn, S.M.; Hamblin, M.R.; Juzeniene, A.; Kessel, D.; et al. Photodynamic therapy of cancer: An update. CA Cancer J. Clin. 2011, 61, 250–281.
  25. Dobson, J.; de Queiroz, G.F.; Golding, J.P. Photodynamic therapy and diagnosis: Principles and comparative aspects. Vet. J. 2018, 233, 8–18.
  26. Cheng, L.; Wang, C.; Feng, L.; Yang, K.; Liu, Z. Functional nanomaterials for phototherapies of cancer. Chem. Rev. 2014, 114, 10869–10939.
  27. Camerin, M.; Rello, S.; Villanueva, A.; Ping, X.; Kenney, M.E.; Rodgers, M.A.; Jori, G. Photothermal sensitisation as a novel therapeutic approach for tumours: Studies at the cellular and animal level. Eur. J. Cancer 2005, 41, 1203–1212.
  28. Robinson, J.T.; Tabakman, S.M.; Liang, Y.; Wang, H.; Sanchez Casalongue, H.; Vinh, D.; Dai, H. Ultrasmall reduced graphene oxide with high near-infrared absorbance for photothermal therapy. J. Am. Chem. Soc. 2011, 133, 6825–6831.
  29. Van der Zee, J. Heating the patient: A promising approach? Ann. Oncol. 2002, 13, 1173–1184.
  30. Longmire, M.; Choyke, P.L.; Kobayashi, H. Clearance properties of nano-sized particles and molecules as imaging agents: Considerations and caveats. Nanomedicine 2008, 3, 703–717.
  31. Chithrani, D.B.; Dunne, M.; Stewart, J.; Allen, C.; Jaffray, D.A. Cellular uptake and transport of gold nanoparticles incorporated in a liposomal carrier. Nanomed. NBM 2010, 6, 161–169.
  32. Dos Santos, A.F.; De Almeida, D.R.Q.; Terra, L.F.; Baptista, M.S.; Labriola, L. Photodynamic therapy in cancer treatment—An update review. J. Cancer Metastasis Treat. 2019, 5, 25.
  33. Cheng, S.H.; Lo, L.W. Inorganic nanoparticles for enhanced photodynamic cancer therapy. Curr. Drug Discov. Technol. 2011, 8, 269–276.
  34. Colombeau, L.; Acherar, S.; Baros, F.; Arnoux, P.; Gazzali, A.M.; Zaghdoudi, K.; Toussaint, M.; Vanderesse, R.; Frochot, C. Inorganic Nanoparticles for Photodynamic Therapy. Top. Curr. Chem. 2016, 370, 113–134.
  35. Moreno Vega, A.I.; Gomez Quintero, T.; Nunez Anita, R.E.; Acosta Torres, L.S.; Castaño, V. Polymeric and ceramic nanoparticles in biomedical applications. J. Nanotechnol. 2012, 2012, 936041.
  36. Lee, J.; Chatterjee, D.K.; Lee, M.H.; Krishnan, S. Gold nanoparticles in breast cancer treatment: Promise and potential pitfalls. Cancer Lett. 2014, 347, 46–53.
  37. Penon, O.; Patiño, T.; Barrios, L.; Nogués, C.; Amabilino, D.B.; Wurst, K.; Pérez-García, L.A. New Porphyrin for the Preparation of Functionalized Water-Soluble Gold Nanoparticles with Low Intrinsic Toxicity. ChemistryOpen 2014, 4, 127–136.
  38. Calavia, P.G.; Chambrier, I.; Cook, M.J.; Haines, A.H.; Field, R.A.; Russell, D.A. Targeted photodynamic therapy of breast cancer cells using lactose-phthalocyanine functionalized gold nanoparticles. J. Colloid Interface Sci. 2018, 512, 249–259.
  39. Wang, S.; Lu, G. Applications of Gold Nanoparticles in Cancer Imaging and Treatment. In Noble and Precious Metals—Properties, Nanoscale Effects and Applications; IntechOpen: London, UK, 2018.
  40. Sharma, H.; Mishra, P.K.; Talegaonkar, S.; Vaidya, B. Metal nanoparticles: A theranostic nanotool against cancer. Drug Discov. Today 2015, 20, 1143.
  41. Faraday, M.X. The Bakerian Lecture.—Experimental relations of gold (and other metals) to light. Philos. Trans. R. Soc. Lond. 1857, 147, 145–181.
  42. Turkevich, J.; Stevenson, P.C.; Hillier, J. A study of the nucleation and growth processes in the synthesis of colloidal gold. Faraday Soc. 1951, 11, 55.
  43. Guo, J.; Rahme, K.; He, Y.; Li, L.L.; Holmes, J.D.; O’Driscoll, C.M. Gold nanoparticles enlighten the future of cancer theranostics. Int. J. Nanomed. 2017, 12, 6131.
  44. Lu, F.; Doane, T.L.; Zhu, J.J.; Burda, C. Gold nanoparticles for diagnostic sensing and therapy. Inorg. Chim. Acta 2012, 393, 142.
  45. Kohout, C.; Santi, C.; Polito, L. Anisotropic gold nanoparticles in biomedical applications. Int. J. Mol. Sci. 2018, 19, 3385.
  46. Onaciu, A.; Braicu, C.; Zimta, A.A.; Moldovan, A.; Stiufiuc, R.; Buse, M.; Ciocan, C.; Buduru, S.; Berindan-Neagoe, I. Gold nanorods: From anisotropy to opportunity. An evolution update. Nanomedicine 2019, 14, 1203.
  47. Pedrosa, P.; Vinhas, R.; Fernandes, A.; Baptista, P.V. Gold nanotheranostics: Proof-of-concept or clinical tool? Nanomaterials 2015, 5, 1853.
  48. Fratoddi, I.; Venditti, I.; Cametti, C.; Russo, M.V. How toxic are gold nanoparticles? The state-of-the-art. Nano Res. 2015, 8, 1771.
  49. Yildirimer, L.; Thanh, N.T.; Loizidou, M.; Seifalian, A.M. Toxicology and clinical potential of nanoparticles. Nano Today 2011, 6, 585.
  50. Hornos Carneiro, M.F.; Barbosa, F., Jr. Gold nanoparticles: A critical review of therapeutic applications and toxicological aspects. J. Toxicol. Environ. Health Part B 2016, 19, 129.
  51. Mironava, T.; Hadjiargyrou, M.; Simon, M.; Jurukovski, V.; Rafailovich, M.H. Gold nanoparticles cellular toxicity and recovery: Effect of size, concentration and exposure time. Nanotoxicology 2010, 4, 120.
  52. Pillai, G. Nanomedicines for cancer therapy: An update of FDA approved and those under various stages of development. SOJ Pharm. Pharm. Sci. 2014, 1, 13.
  53. Ventola, C.L. Progress in nanomedicine: Approved and investigational nanodrugs. Pharm. Ther. 2017, 42, 742.
  54. Hillyer, J.F.; Albrecht, R.M. Gastrointestinal persorption and tissue distribution of differently sized colloidal gold nanoparticles. J. Pharm. Sci. 2001, 12, 1927.
  55. Aryal, S.; Bisht, G. New paradigm for a targeted cancer therapeutic approach: A short review on potential synergy of gold nanoparticles and cold atmospheric plasma. Biomedicines 2017, 5, 38.
  56. Kaur, P.; Aliru, M.L.; Chadha, A.S.; Asea, A.; Krishnan, S. Hyperthermia using nanoparticles—Promises and pitfalls. Int. J. Hyperth. 2016, 32, 76.
  57. Kennedy, L.C.; Bickford, L.R.; Lewinski, N.A.; Coughlin, A.J.; Hu, Y.; Day, E.S.; West, J.L.; Drezek, R.A. A new era for cancer treatment: Gold-nanoparticle-mediated thermal therapies. Small 2011, 7, 169.
  58. Cherukuri, P.; Glazer, E.S.; Curley, S.A. Targeted hyperthermia using metal nanoparticles. Adv. Drug Delivery Rev. 2010, 62, 339.
  59. Yang, Z.; Gao, D.; Zhao, J.; Yang, G.; Guo, M.; Wang, Y.; Ren, X.; Kim, J.S.; Jin, L.; Tian, Z.; et al. Thermal immuno-nanomedicine in cancer. Nat. Rev. Clin. Oncol. 2023, 20, 116–134.
  60. Wang, Y.; Gao, D.; Jin, L.; Ren, X.; Ouyang, Y.; Zhou, Y.; He, X.; Jia, L.; Tian, Z.; Wu, D.; et al. NADPH Selective Depletion Nanomedicine-Mediated Radio-Immunometabolism Regulation for Strengthening Anti-PDL1 Therapy against TNBC. Adv. Sci. 2022, 10, 2203788.
  61. Amini, S.M. Gold nanostructures absorption capacities of various energy forms for thermal therapy applications. J. Therm. Biol. 2019, 79, 81.
  62. Huang, X.; El-Sayed, M.A. Gold nanoparticles: Optical properties and implementations in cancer diagnosis and photothermal therapy. J. Adv. Res. 2010, 1, 13.
  63. Park, J.E.; Kim, M.; Hwang, J.H.; Nam, J.M. Golden opportunities: Plasmonic gold nanostructures for biomedical applications based on the second near-infrared window. Small Methods 2017, 1, 1600032.
  64. Jain, P.K.; Lee, K.S.; El-Sayed, I.H.; El-Sayed, M.A. Calculated absorption and scattering properties of gold nanoparticles of different size, shape, and composition: Applications in biological imaging and biomedicine. J. Phys. Chem. B 2006, 110, 7238–7248.
  65. Bansal, S.A.; Kumar, V.; Karimi, J.; Singh, A.P.; Kumar, S. Role of Gold Nanoparticles in Advanced Biomedical Applications. Nanoscale Adv. 2020, 2, 3764–3787.
  66. Govorov, A.O.; Richardson, H.H. Generating heat with metal nanoparticles. NanoToday 2007, 2, 30–38.
  67. Linic, S.; Aslam, U.; Boerigter, C.; Morabito, M. Photochemical transformations on plasmonic metal nanoparticles. Nat. Mater. 2015, 14, 567–576.
  68. Ji, M.; Liu, H.; Cheng, M.; Huang, L.; Yang, G.; Bao, F.; Huang, G.; Huang, Y.; Hu, Y.; Cong, G.; et al. Plasmonic metal nanoparticle loading to enhance the photothermal conversion of carbon fibers. J. Phys. Chem. C 2022, 126, 2454–2462.
  69. Tsai, M.F.; Chang, S.H.G.; Cheng, F.Y.; Shanmugam, V.; Cheng, Y.S.; Su, C.H.; Yeh, C.S. Au Nanorod Design as Light-Absorber in the First and Second Biological Near-Infrared Windows for in Vivo Photothermal Therapy. ACS Nano 2013, 7, 5330–5342.
  70. Cai, K.; Zhang, W.; Zhang, J.; Li, H.; Han, H.; Zhai, T. Design of Gold Hollow Nanorods with Controllable Aspect Ratio for Multimodal Imaging and Combined Chemo-Photothermal Therapy in the Second Near-Infrared Window. ACS Appl. Mater. Interfaces 2018, 10, 36703–36710.
  71. Du, C.; Wang, A.; Fei, J.; Zhao, J.; Li, J. Polypyrrole-Stabilized Gold Nanorods with Enhanced Photothermal Effect towards Two-Photon Photothermal Therapy. J. Mater. Chem. B 2015, 3, 4539–4545.
  72. Lindley, S.A.; Zhang, J.Z. Bumpy Hollow Gold Nanospheres for Theranostic Applications: Effect of Surface Morphology on Photothermal Conversion Efficiency. ACS Appl. Mater. Interfaces 2019, 2, 1072–1081.
  73. Skrabalak, S.E.; Chen, J.; Sun, Y.; Lu, X.; Au, L.; Cobley, C.M.; Xia, Y. Gold Nanocages: Synthesis, Properties, and Applications. Acc. Chem. Res. 2008, 41, 1587–1595.
  74. Xu, Q.; Wan, J.; Bie, N.; Song, X.; Yang, X.; Yong, T.; Zhao, Y.; Yang, X.; Gan, L. A Biomimetic Gold Nanocages-Based Nanoplatform for Efficient Tumor Ablation and Reduced Inflammation. Theranostics 2018, 8, 5362–5378.
  75. Xu, Y.; Wang, X.; Cheng, L.; Liu, Z.; Zhang, Q. High-Yield Synthesis of Gold Bipyramids for In Vivo CT Imaging and Photothermal Cancer Therapy with Enhanced Thermal Stability. Chem. Eng. Sci. 2019, 378, 122025.
  76. Campu, A.; Focsan, M.; Lerouge, F.; Borlan, R.; Tie, L.; Rugina, D.; Astilean, S. ICG-Loaded Gold Nano-Bipyramids with NIR Activatable Dual PTT-PDT Therapeutic Potential in Melanoma Cells. Colloids Surf. B 2020, 194, 111213.
  77. Huang, P.; Lin, J.; Li, W.; Rong, P.; Wang, Z.; Wang, S.; Wang, X.; Sun, X.; Aronova, M.; Niu, G.; et al. Biodegradable Gold Nanovesicles with an Ultrastrong Plasmonic Coupling Effect for Photoacoustic Imaging and Photothermal Therapy. Angew. Chem. Int. Ed. Engl. 2013, 52, 13958–13964.
  78. Chung, U.S.; Kim, J.H.; Kim, B.; Kim, E.; Jang, W.D.; Koh, W.G. Dendrimer Porphyrin-Coated Gold Nanoshells for the Synergistic Combination of Photodynamic and Photothermal Therapy. Chem. Commun. 2016, 52, 1258–1261.
  79. Tam, J.M.; Tam, J.O.; Murthy, A.; Ingram, D.R.; Ma, L.L.; Travis, K.; Johnston, K.P.; Sokolov, K.V. Controlled Assembly of Biodegradable Plasmonic Nanoclusters for Near-Infrared Imaging and Therapeutic Applications. ACS Nano 2010, 4, 2178–2184.
  80. Chuang, Y.C.; Hsia, Y.; Chu, C.H.; Lin, L.J.; Sivasubramanian, M.; Lo, L.W. Precision Control of the Large-Scale Green Synthesis of Biodegradable Gold Nanodandelions as Potential Radiotheranostics. Biomater. Sci. 2019, 7, 4720–4729.
  81. Jiang, T.; Yin, N.; Liu, L.; Song, J.; Huang, Q.; Zhu, L.; Xu, X. An Au @CdTe/CdS/ZnS Quantum Dot Multifunctional Nanoprobe for Photothermal Treatment and Cellular Imaging. RSC Adv. 2014, 4, 23630–23636.
  82. Lu, W.; Singh, A.K.; Khan, S.A.; Senapati, D.; Yu, H.; Ray, P.C. Gold Nano-Popcorn-based Targeted Diagnosis, Nanotherapy Treatment, and In Situ Monitoring of Photothermal Therapy Response of Prostate Cancer Cells Using Surface-Enhanced Raman Spectroscopy. J. Am. Chem. Soc. 2010, 132, 18103–18114.
  83. Liu, Y.; Maccarini, P.; Palmer, G.M.; Etienne, W.; Zhao, Y.; Lee, C.T.; Ma, X.; Inman, B.A.; Vo-Dinh, T. Synergistic Immuno Photothermal Nanotherapy (SYMPHONY) for the Treatment of Unresectable and Metastatic Cancers. Sci. Rep. 2017, 7, 8606.
  84. 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.
  85. Vijayaraghavan, P.; Liu, C.H.; Vankayala, R.; Chiang, C.S.; Hwang, K.C. Designing Multi-Branched Gold Nanoechinus for NIR Light Activated Dual Modal Photodynamic and Photothermal Therapy in the Second Biological Window. Adv. Mater. 2014, 26, 6689–6695.
  86. Zhang, B.; Wang, J.; Sun, J.; Wang, Y.; Chou, T.; Zhang, Q.; Shah, H.R.; Ren, L.; Wang, H. Self-Reporting Gold Nanourchins for Tumor-Targeted Chemo-Photothermal Therapy Integrated with Multimodal Imaging. Adv. Ther. 2020, 3, 2000114.
  87. Zhou, J.; Jiang, Y.; Hou, S.; Upputuri, P.K.; Wu, D.; Li, J.; Wang, P.; Zhen, X.; Pramanik, M.; Pu, K.; et al. Compact Plasmonic Blackbody for Cancer Theranosis in the Near-Infrared II Window. ACS Nano 2018, 12, 2643–2651.
  88. Hong, E.J.; Choi, D.G.; Shim, M.S. Targeted and effective photodynamic therapy for cancer using functionalized nanomaterials. Acta Pharm. Sin. B 2016, 6, 297.
  89. Hou, X.; Tao, Y.; Pang, Y.; Li, X.; Jiang, G.; Liu, Y. Nanoparticle-based photothermal and photodynamic immunotherapy for tumor treatment. Int. J. Cancer 2018, 143, 3050.
  90. Nazir, S.; Hussain, T.; Ayub, A.; Rashid, U.; MacRobert, A.J. Nanomaterials in combating cancer: Therapeutic applications and developments. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 19.
  91. Crous, A.; Abrahamse, H. Photodynamic Therapy with an AlPcS4Cl Gold Nanoparticle Conjugate Decreases Lung Cancer’s Metastatic Potential. Coatings 2022, 12, 199.
  92. Imanparast, A.; Attaran, N.; Eshghi, H.; Sazgarnia, A. Surface modification of gold nanoparticles with 6-mercapto-1-hexanol to facilitate dual conjugation of protoporphyrin IX and folic acid for improving the targeted photochemical internalization. Iran. J. Basic Med. Sci. 2022, 25, 970–979.
  93. Wang, P.; Zhang, L.; Zhang, Z.; Wang, S.; Yao, C. Influence of Parameters on Photodynamic Therapy of 2–HMME Core-Shell Nanostructures. Nanomaterials 2022, 12, 1358.
  94. Mokoena, D.; George, B.P.; Abrahamse, H. Conjugation of Hypericin to Gold Nanoparticles for Enhancement of Photodynamic Therapy in MCF-7 Breast Cancer Cells. Pharmaceutics 2022, 14, 2212.
  95. Emami Nejad, A.; Najafgholian, S.; Rostami, A.; Sistani, A.; Shojaeifar, S.; Esparvarinha, M.; Nedaeinia, R.; Haghjooy Javanmard, S.; Taherian, M.; Ahmadlou, M.; et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: A novel approach to developing treatment. Cancer Cell Int. 2021, 21, 62.
  96. Lou-Franco, J.; Das, B.; Elliott, C.; Cao, C. Gold Nanozymes: From Concept to Biomedical Applications. Nanomicro Lett. 2020, 13, 10.
  97. Ma, Y.C.; Zhu, Y.H.; Tang, X.F.; Hang, L.F.; Jiang, W.; Li, M.; Khan, M.I.; You, Y.Z.; Wang, Y.C. Au nanoparticles with enzyme mimicking activity-ornamented ZIF-8 for highly efficient photodynamic therapy. Biomater. Sci. 2019, 7, 2740–2748.
  98. Wang, J.; Sun, J.; Hu, W.; Wang, Y.; Chou, T.; Zhang, B.; Zhang, Q.; Ren, L.; Wang, H. A Porous Bimetallic Core-Shell Nanostructure as an H(2)O(2)-Driven Oxygenerator to Alleviate Tumor Hypoxia for Simultaneous Bimodal Imaging and Enhanced Photodynamic Therapy. Adv. Mater. 2020, 32, 2001862.
  99. Yin, Z.; Ji, Q.; Wu, D.; Li, Z.; Fan, M.; Zhang, H.; Zhao, X.; Wu, A.; Cheng, L.; Zeng, L. H(2)O(2)-Responsive Gold Nanoclusters @Mesoporous Dioxide Nanozyme for “Off/On” Modulation and Enhancement of Magnetic Resonance Imaging and Photodynamic Therapy. ACS Appl. Mater. Interfaces 2021, 13, 14928–14937.
  100. Ferrari, M. Cancer nanotechnology: Opportunities and challenges. Nat. Rev. Cancer 2005, 5, 161–171.
  101. Yang, X.; Yang, M.; Pang, B.; Vara, M.; Xia, Y. Gold nanomaterials at work in biomedicine. Chem. Rev. 2015, 115, 10410–10488.
  102. Dreaden, E.C.; Alkilany, A.M.; Huang, X.; Murphy, C.J.; El-Sayed, M.A. The golden age: Gold nanoparticles for biomedicine. Chem. Soc. Rev. 2012, 41, 2740–2779.
  103. Maeda, H.; Wu, J.; Sawa, T.; Matsumura, Y.; Hori, K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. J. Control. Release 2000, 65, 271–284.
  104. Shakeri-Zadeh, A.; Ghasemifard, M.; Mansoori, G.A. Structural and optical characterization of folate-conjugated gold-nanoparticles. Phys. E 2010, 42, 1272–1280.
  105. Shakeri-Zadeh, A.; Mansoori, G.A.; Hashemian, A.R.; Eshghi, H.; Sazgarnia, A.; Montazerabadi, A.R. Cancerous cells targeting and destruction using folate conjugated gold nanoparticles. Dyn. Biochem. Process. Biotechnol. Mol. Biol. 2010, 4, 6–12.
  106. Sun, T.; Zhang, Y.S.; Pang, B.; Hyun, D.C.; Yang, M.; Xia, Y. Engineered nanoparticles for drug delivery in cancer therapy. In Nanomaterials and Neoplasms; Jenny Stanford Publishing: Dubai, United Arab Emirates, 2021; pp. 31–142.
  107. Samadian, H.; Hosseini-Nami, S.; Kamrava, S.K.; Ghaznavi, H.; Shakeri-Zadeh, A. Folate-conjugated gold nanoparticle as a new nanoplatform for targeted cancer therapy. J. Cancer Res. Clin. Oncol. 2016, 142, 2217–2229.
  108. 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.
  109. Her, S.; Jaffray, D.A.; Allen, C. Gold nanoparticles for applications in cancer radiotherapy: Mechanisms and recent advancements. Adv. Drug Deliv. Rev. 2017, 109, 84–101.
  110. Ghaznavi, H.; Hosseini-Nami, S.; Kamrava, S.K.; Irajirad, R.; Maleki, S.; Shakeri-Zadeh, A.; Montazerabadi, A. Folic acid conjugated PEG coated gold–iron oxide core–shell nanocomplex as a potential agent for targeted photothermal therapy of cancer. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1594–1604.
  111. Liu, X.; Chen, H.J.; Chen, X.; Parini, C.; Wen, D. Low frequency heating of gold nanoparticle dispersions for non-invasive thermal therapies. Nanoscale 2012, 4, 3945–3953.
  112. Shi, J.; Chen, Z.; Wang, L.; Wang, B.; Xu, L.; Hou, L.; Zhang, Z. A tumor-specific cleavable nanosystem of PEG-modified Au hybrid aggregates for radio frequency-controlled release, hyperthermia, photodynamic therapy and X-ray imaging. Acta Biomater. 2016, 29, 282–297.
  113. Deng, X.; Li, K.; Cai, X.; Liu, B.; Wei, Y.; Deng, K.; Xie, Z.; Wu, Z.; Ma, P.A.; Hou, Z.; et al. A hollow-structured 2 nanohybrid: Synergistically enhanced photothermal efficiency and photoswitchable targeting effect for cancer theranostics. Adv. Mater. 2017, 29, 1701266.
  114. Camerin, M.; Magaraggia, M.; Soncin, M.; Jori, G.; Moreno, M.; Chambrier, I.; Cook, M.J.; Russell, D.A. The in vivo efficacy of phthalocyanine–nanoparticle conjugates for the photodynamic therapy of amelanotic melanoma. Eur. J. Cancer 2010, 46, 1910–1918.
  115. Brazzale, C.; Canaparo, R.; Racca, L.; Foglietta, F.; Durando, G.; Fantozzi, R.; Caliceti, P.; Salmaso, S.; Serpe, L. Enhanced selective sonosensitizing efficacy of ultrasound-based anticancer treatment by targeted gold nanoparticles. Nanomedicine 2016, 12, 3053–3070.
  116. Fan, W.; Yung, B.; Huang, P.; Chen, X. Nanotechnology for multimodal synergistic cancer therapy. Chem. Rev. 2017, 117, 13566–13638.
  117. Zhang, Z.; Xu, S.; Wang, Y.; Yu, Y.; Li, F.; Zhu, H.; Shen, Y.; Huang, S.; Guo, S. Near-infrared triggered co-delivery of doxorubicin and quercetin by using gold nanocages with tetradecanol to maximize anti-tumor effects on MCF-7/ADR cells. J. Colloid Interface Sci. 2018, 509, 47–57.
  118. Yu, Y.; Zhang, Z.; Wang, Y.; Zhu, H.; Li, F.; Shen, Y.; Guo, S. A new NIR-triggered doxorubicin and photosensitizer indocyanine green co-delivery system for enhanced multidrug resistant cancer treatment through simultaneous chemo/photothermal/photodynamic therapy. Acta Biomater. 2017, 59, 170–180.
  119. Xu, X.; Chong, Y.; Liu, X.; Fu, H.; Yu, C.; Huang, J.; Zhang, Z. Multifunctional nanotheranostic gold nanocages for photoacoustic imaging guided radio/photodynamic/photothermal synergistic therapy. Acta Biomater. 2019, 84, 328–338.
  120. Xia, F.; Hou, W.; Zhang, C.; Zhi, X.; Cheng, J.; Jesús, M.; Song, J.; Cui, D. pH-responsive gold nanoclusters-based nanoprobes for lung cancer targeted near-infrared fluorescence imaging and chemo-photodynamic therapy. Acta Biomater. 2018, 68, 308–319.
  121. Xia, F.; Hou, W.; Liu, Y.; Wang, W.; Han, Y.; Yang, M.; Zhi, X.; Li, C.; Qi, D.; Li, T.; et al. Cytokine induced killer cells-assisted delivery of chlorin e6 mediated self-assembled gold nanoclusters to tumors for imaging and immuno-photodynamic therapy. Biomaterials 2018, 170, 1–11.
  122. Li, H.; Wang, P.; Deng, Y.; Zeng, M.; Tang, Y.; Zhu, W.H.; Cheng, Y. Combination of active targeting, enzyme-triggered release and fluorescent dye into gold nanoclusters for endomicroscopy-guided photothermal/photodynamic therapy to pancreatic ductal adenocarcinoma. Biomaterials 2017, 139, 30–38.
  123. Han, L.; Xia, J.M.; Hai, X.; Shu, Y.; Chen, X.W.; Wang, J.H. Protein-stabilized gadolinium oxide-gold nanoclusters hybrid for multimodal imaging and drug delivery. ACS Appl. Mater. Interfaces 2017, 9, 6941–6949.
  124. Cui, H.; Hu, D.; Zhang, J.; Gao, G.; Chen, Z.; Li, W.; Gong, P.; Sheng, Z.; Cai, L. Gold nanoclusters–indocyanine green nanoprobes for synchronous cancer imaging, treatment, and real-time monitoring based on fluorescence resonance energy transfer. ACS Appl. Mater. Interfaces 2017, 9, 25114–25127.
  125. Xu, W.; Qian, J.; Hou, G.; Wang, Y.; Wang, J.; Sun, T.; Ji, L.; Suo, A.; Yao, Y. PEGylated hydrazided gold nanorods for pH-triggered chemo/photodynamic/photothermal triple therapy of breast cancer. Acta Biomater. 2018, 82, 171–183.
  126. Choi, J.; Lee, S.E.; Park, J.S.; Kim, S.Y. Gold nanorod-photosensitizer conjugates with glutathione sensitive linkages for synergistic cancer photodynamic/photothermal therapy. Biotechnol. Bioeng. 2018, 115, 1340–1354.
  127. Chuang, C.C.; Chen, Y.N.; Wang, Y.Y.; Huang, Y.C.; Lin, S.Y.; Huang, R.Y.; Jang, Y.Y.; Yang, C.C.; Huang, Y.F.; Chang, C.W. Stem cell-based delivery of gold/chlorin e6 nanocomplexes for combined photothermal and photodynamic therapy. ACS Appl. Mater. Interfaces 2020, 12, 30021–30030.
  128. 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.
  129. Qiu, W.X.; Liu, L.H.; Li, S.Y.; Lei, Q.; Luo, G.F.; Zhang, X.Z. ACPI conjugated gold nanorods as nanoplatform for dual image guided activatable photodynamic and photothermal combined therapy in vivo. Small 2017, 13, 603956.
  130. Ghorbani, F.; Attaran-Kakhki, N.; Sazgarnia, A. The synergistic effect of photodynamic therapy and photothermal therapy in the presence of gold-gold sulfide nanoshells conjugated Indocyanine green on HeLa cells. Photodiagn. Photodyn. Ther. 2017, 17, 48–55.
  131. Liu, C.; Luo, L.; Zeng, L.; Xing, J.; Xia, Y.; Sun, S.; Zhang, L.; Yu, Z.; Yao, J.; Yu, Z.; et al. Porous gold nanoshells on functional NH2-MOFs: Facile synthesis and designable platforms for cancer multiple therapy. Small 2018, 14, 1801851.
  132. You, Q.; Sun, Q.; Yu, M.; Wang, J.; Wang, S.; Liu, L.; Cheng, Y.; Wang, Y.; Song, Y.; Tan, F.; et al. BSA–bioinspired gadolinium hybrid-functionalized hollow gold nanoshells for NIRF/PA/CT/MR quadmodal diagnostic imaging-guided photothermal/photodynamic cancer therapy. ACS Appl. Mater. Interfaces 2017, 9, 40017–40030.
  133. Liu, B.; Cao, W.; Cheng, J.; Fan, S.; Pan, S.; Wang, L.; Niu, J.; Pan, Y.; Liu, Y.; Sun, X.; et al. Human natural killer cells for targeting delivery of gold nanostars and bimodal imaging directed photothermal/photodynamic therapy and immunotherapy. Cancer Biol. Med. 2019, 16, 756.
  134. Kharlamov, A.N.; Feinstein, J.A.; Cramer, J.A.; Boothroyd, J.A.; Shishkina, E.V.; Shur, V. Plasmonic Photothermal Therapy of Atherosclerosis with Nanoparticles: Long-term Outcomes and Safety in NANOM-FIM Trial. Future Cardiol. 2017, 13, 345–363.
  135. Xia, F.; Niu, J.; Hong, Y.; Li, C.; Cao, W.; Wang, L.; Hou, W.; Liu, Y.; Cui, D. Matrix metallo peptidase 2 targeted delivery of gold nanostars decorated with IR-780 iodide for dual-modal imaging and enhanced photothermal/photodynamic therapy. Acta Biomater. 2019, 89, 289–299.
  136. Liu, Y.; Zhi, X.; Yang, M.; Zhang, J.; Lin, L.; Zhao, X.; Hou, W.; Zhang, C.; Zhang, Q.; Pan, F.; et al. Tumor-triggered drug release from calcium carbonate-encapsulated gold nanostars for near-infrared photodynamic/photothermal combination antitumor therapy. Theranostics 2017, 7, 1650.
More
Video Production Service