Phototherapeutic Agents for Fabricating High Drug-Loading Nanomedicines: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , ,

The key requirement of the chemo–photo combination therapy is the high drug-loading nanomedicines, which can load either chemotherapy drugs or phototherapy agents at the same nanomedicines and simultaneously deliver them to tumors, and play a multimode therapeutic role for tumor treatment. Many kinds of photothermal materials or photosensitizers have been used as carrier materials to construct high drug-loading nanomedicines for tumor combination therapy.

  • high drug-loading nanomedicines
  • chemotherapy
  • phototherapy
  • combination therapy

1. Introduction

On the premise of a stable system, the lesser amount of carrier material and the higher drug-loading ratio are pursued to prepare the high drug-loading nanomedicines. For tumor chemo–photo combination therapy, chemotherapeutic drugs and phototherapeutic agents (photothermal agents and photosensitizers) are equally important in the nanomedicines, and their proportion determines the final therapeutic effect on tumor to a certain extent. However, the construction of nanomedicines to realize the high loading of chemotherapeutic drugs and ensure the effective dose of phototherapeutic agents in the same nanomedicine is a challenge. After long-term exploration, researchers have found that using a phototherapeutic agent as a carrier to load chemotherapeutic drugs is an effective method of realizing high drug-loading for chemo–photo combination therapy. Now, many kinds of photothermal materials or photosensitizers (PSs) have been used as carrier materials to construct high drug-loading nanomedicines for tumor combination therapy of chemo-PDT (photodynamic therapy), chemo-PTT (photothermal therapy), and chemo-PDT-PTT.

2. Photothermal Agents as Carriers for Fabricating High Drug-Loading Nanomedicines

Photothermal agents absorb the energy of near-infrared (NIR) light and convert it into heat. Consequently, local tumors are induced to generate high temperatures, leading to their thermal ablation or thermal damage, thereby killing tumor cells with less harm to healthy tissues. At present, many photothermal agents, especially nanomaterials with strong absorption capacity and high photothermal-conversion efficiency in the NIR, have been widely studied and explored with some already undergoing clinical trials [1]. These photothermal agents can be primarily divided into inorganic and organic materials. Inorganic materials also include noble-metal nanomaterials, transition-metal chalcogenides and oxides, and carbon-based materials. Numerous studies have shown that nanomedicines prepared using a photothermal agent as a nanocarrier loaded with chemotherapy drugs have high drug-loading and high combination efficiency of chemotherapy-PTT [2]. In this section, chemo-PTT combination therapy with common photothermal agents used as carrier materials are summarized in Table 1.
Table 1. A summary of photothermal agents as carriers for high drug-loading nanomedicines.

2.1. Inorganic Materials

Noble-metal-based materials (Au, Ag, and Pb) are extensively used in tumor imaging and PTT due to their strong localized surface plasmon resonance (LSPR) and enhanced photothermal-conversion efficiency in the NIR-light region [30]. Among them, gold (Au)-based nanomaterials have excellent performance for clinical translation owing to their high X-ray attenuation coefficient, excellent biocompatibility, and bio-inertness. Accordingly, the preparation of high drug-loading nanomedicines with Au-based materials such as gold NPs (Au NPs) [31][32][33], Au nanorods (Au NRs) [34][35], and Au nanocages (Au NCs) [36][37], Au nanoflowers (Au NFs) [38][39], and Au nanoclusters (Au NCSs) [40][41] are attracting extensive attention for chemo–photo combination tumor therapy. Huang et al. used crystalline zeolitic imidazolate framework-8 (ZIF-8) to grow on Au NRs and prepare biocompatible and biodegradable metal–organic frameworks (MOFs), i.e., Au@ZIF-8 [42]. Due to the large surface area and guest-matching pore size of ZIF-8, DOX is successfully loaded into Au@ZIF-8 with a high drug load efficiency of ~37%. Au@ZIF-8 shows a high photothermal effect upon NIR irradiation, and the ZIF-8 layer quickly degrades with laser irradiation, resulting in on-demand drug release at the tumor site. Furthermore, in vivo therapeutic results confirm that the combination of chemo–photothermal with Au@ZIF-8/DOX + NIR achieves much higher treatment efficacy than that of PTT with Au@ZIF-8 + NIR or chemotherapy with DOX only, even resulting in complete tumor elimination without obvious adverse effect. Xu et al. fabricated PDA-coated Au nanobone (NB) nanocomplexes (AuNBs@PDA) through the in situ polymerization of dopamine on the surface of Au NBs. The DOX is loaded through π-π stacking and hydrogen-binding interactions with PDA [5]. The AuNBs@PDA nanocomplexes exhibit higher photothermal-conversion efficiency (75.48%) than Au NBs alone, which is beneficial to photoacoustic imaging and PTT.
Moreover, the load efficiency (LE) of DOX by AuNBs@PDA nanomedicines could reach up to about 70%, showing significant cytotoxicity and antitumor effect. He et al. prepared metal@MOFs (Au@Cu3(BTC)2NPs) with a core–shell structure by assembling Cu3(BTC)2 on Au NPs with 4-mercaptobenzoic acid (4-MBA) as bridging molecule [43]. The Cu3(BTC)2 shell can provide sites for aptamer functionalization and drug loading. The Au@Cu3(BTC)2NPs exhibit high drug-loading efficiency (57%) and good photothermal-conversion efficiency.
Carbon nanomaterials primarily include the zero-dimensional (0D) fullerenes, the one-dimensional (1D) carbon nanotubes (CNTs), and the two-dimensional (2D) graphene [1]. Given the high NIR absorption coefficient (6.2 × 106 M−1 cm−1) and ultra-high specific surface area of CNTs and graphene, the preparation of high drug-loading nanomedicines based on these materials for tumor chemo–photothermal combination therapy is attracting considerable attention [44][45]. However, CNTs and graphene without appropriate surface functionalization cause toxicity and side effects because of the poor dispersibility and stability of carbon-based materials in physiological solutions. Accordingly, researchers have developed various surface-functionalized CNTs or graphene nanocarriers to load chemotherapeutic drugs and prepared high drug-loading nanomedicines for tumor chemo–photothermal combination therapy. Xing et al. synthesized a three-dimensional framework with good stability and large specific surface area through the amide reaction of graphdiyne oxide (GDYO) and cisplatin (CDDP) and then loaded DOX by π-π stacking [15]. Self-assembly is then performed with the targeting group DSPE-PEG2000-methotrexate (MTX) and GDYO-CDDP to obtain multifunctional nanomedicines of GDYO-CDDP/DOX@DSPE-PEG-MTX (GCDM) for diagnosis and targeted cancer chemo–photothermal combination therapy. In this nanomedicines, three traditional anticancer drugs (CDDP, DOX, and MTX) play new roles and can reduce multidrug resistance through combined antitumor effects. Although only 40.3% of DOX is given, the load efficiency of the three chemotherapy drugs is actually higher. Wang et al. modified the surface of graphene oxide (GO) nanosheets with PEG derivatives and lactobionic acid (LA), and then loaded curcumin (CUR) on graphene nanosheets by π-π stacking to prepare GO-PEG/LA-CUR composite nanomedicines with a drug-loading efficiency of 56.8% [46]. In vivo experiments show that tumor growth is significantly inhibited in subcutaneous hepatocellular carcinoma tumor-bearing mice treated with GO-PEG/LA-CUR after NIR irradiation with a tumor inhibition rate of 86%, demonstrating the higher efficacy of the chemo-PTT combination therapy. Yang et al. designed a highly potent chemo–photothermal theragnostic system by sequentially loading DOX and CDDP onto dual polymer-modified single-wall carbon nanohorns (SWNHs) [19]. SWNHs are first modified simultaneously with poly(maleic anhydride-alt-1-octadecene) (C18PMH) and methoxyPEG-b-poly-D,L-lactide (mPEG-PLA) through hydrophobic–hydrophobic interactions and π-π stacking. Among various carbonaceous materials, SWNHs are advantageous for drug loading and have strong potential for chemo–photothermal combination therapy owing to their unique structure with a horn shape. This closed-tip nanotube possesses large inner voids and can be filled with suitable drugs ranging from small molecules to proteins. In this nanomedicines, the drug-loading efficiency of DOX and CDDP is 44% and 66%, respectively. In vivo treatment results show that SWNHs/C18PMH/MPEG-PLA-DOX-Pt-mediated chemo–photothermal combination therapy with the guidance of photoacoustic imaging completely eliminates the primary breast tumor and inhibits its lung metastasis.
A wide variety of transition-metal compound nanomaterials have opened up a new regime in PTT owing to their excellent NIR absorption and efficient heat-generation abilities. These nanomaterials primarily contain transition-metal chalcogenides (e.g., CuxSy [9][47][48], Cu2−xSe [49], CoS [11], MoS2 [50], WS2 [51], FeSe2 [52], FeS [53], and TiS2 [54]) and oxides (e.g., MoOx [55], WxOy [56], and Ti8O15 [57]) with different structures of nanosheets, nanodots, and nanoparticles. Among them, copper chalcogenide semiconductors characterized by low cost and toxicity are new kinds of promising photothermal agents. Wu et al. synthesized hollow copper sulfide NPs (HCuSNP) loaded with DOX and a photothermal agent (ICG). They coated the surface of B16 membrane to prepare HCuSNP-ICG-DOX@B16F10 (ID-HCuSNP@B16F10) nanomedicines, which serve as a biomimetic platform for in vivo chemo–photothermal combination therapy of tumors [58]. The hollow mesoporous structure of HCuSNPs makes them an ideal drug carrier. The loading efficiency of ICG and DOX can reach 98% and 85%, respectively. ID-HCuSNP@B16F10 exhibits an excellent photothermal effect in melanoma animal models and achieves a high tumor-ablation rate. As an emerging two-dimensional nanomaterial, MXenes have a large specific surface area and various surface groups, which enable them to have high drug-loading efficiency and the possibility of surface functionalization. Some recent reports have also confirmed the high photothermal-conversion performance of MXenes in NIR PTT. Li et al. developed multifunctional Ti2N MXene-based nanomedicines (Ti2N@oSi) for chemo–photothermal combination anticancer therapy [59]. Ti2N nanosheets are stabilized with soybean phospholipids and loaded with DOX, followed by coating with biodegradable silica shells to prevent drug leakage, and finally loading with the second drug (CDDP) and the target agent (bombesin) in the outer layer. The unique structure of Ti2N nanosheets endows the drug carriers with an ultrahigh drug-loading efficiency of 88.8% and an excellent NIR photothermal-conversion efficiency of 41.6% for chemo–photothermal combination therapy. Although inorganic nanomaterials have been developed for the PTT treatment of tumors, most of the inorganic photothermal agents currently used are nonbiodegradable and have potential long-term toxicity, hindering their further application in clinical tumor therapy.

2.2. Organic Materials

Organic photothermal agents have excellent biodegradability and biocompatibility. Compared with inorganic photothermal agents, organic agents have fewer safety problems. Thus, the development and application of organic photothermal agents have also been extensively studied for many years.
Common organic photothermal agents include NIR-absorbing dye containing nanocomplexes, NIR-absorbing conjugated polymers, and melanin-based photothermal materials. Small-molecule organic dyes such as ICG, IR780, and IR808 are difficult to be loaded with drugs as nanomedicines owing to some of their physicochemical characteristics, such as concentration-dependent aggregation and poor aqueous stability. Accordingly, other carrier materials are often needed. Lu et al. designed two-component NPs by π-π stacking and hydrophobic interactions between amino acid-conjugated camptothecin (CPT-RT) and canine dyes (IR-783) [21]. Then, the positively charged Angiopep-2-modified PEGlyated poly-l-lysine (Ang-PEG-g-PLL) is coated onto the negatively charged two-component NPs by electrostatic interaction to form the three-component nanomedicines Ang-PEG-g-PLL@CPT-RT@IR783 (APCI) for synergistic chemo–photothermal anti-glioma therapy. The drug-loading efficiency of APCI reaches about 62%. In vivo results reveal that the nanomedicines mediated by chemo–photothermal combination therapy achieve a better therapeutic effect, longer survival time, and minimal toxic side effects in orthotopic glioblastoma tumor-bearing nude mice.
Compared with NIR dyes, conductive polymers such as polypyrrole and polyaniline have better photothermal stability, are not easy to photobleach and have a longer circulation time in vivo [25]. Thus, as a photothermal agent, the conductive polymer is also an ideal drug-delivery platform. Liu et al. fabricated poly(acrylic acid) (PAA)-stabilized poly(pyrrole-3-COOH) NPs (PAA@PPyCOOH NPs) as nanomedicines loaded with DOX for chemo–photothermal combination therapy [26]. The PAA@PPyCOOH NPs are found to be ideal nanomedicines with good dispersity, excellent biocompatibility, high drug-loading efficiency (43.3%), and photothermal-conversion efficiency (56%). In vivo experiments demonstrate that the PAA@PPyCOOH@DOX nanomedicines can be specifically degraded by excess H2O2 in the tumor, and the tumor growth of 4T1 breast-cancer model is drastically inhibited by chemo-PTT.
PDA is a kind of melanin-like polymer that has been developed for photothermal agents and nanocarriers because of its excellent biocompatibility, biodegradability, simple preparation conditions, and high photothermal-conversion efficiency. PDA has many functional groups such as benzene ring, catechol group, and quinone group, enabling it to load a large number of antitumor drugs through π-π stacking or hydrogen bonding. In previous work, researchers have developed lollipop-like dual-drug-loaded NPs (DOX–PDA–gossypol NPs) based on the self-assembly of gossypol, DOX, and PDA through π-π stacking [60]. The DOX–PDA–gossypol NPs have a high drug-loading efficiency of 91%. In vivo antitumor experiments show that the TIRs of DOX-PDA-gossypol NPs are more than 90% at both low doses, which is beneficial for widening the drug therapeutic window. In another study, the researchers used PDA as a carrier for loaded hemoglobin and Ce6 to construct small NPs (PHC NPs; about 10 nm) for combined PTT/PDT [61]. The PHC NPs were encapsulated into the micelles formed by aldehyde-modified PEG and polyethyleneimine through benzoic-imine bonds, and the surface was modified with hyaluronic acid to form larger-size composite NPs ([PHC]PP@HA NPs; about 140 nm). The photothermal-conversion efficiency of [PHC]PP@HA NPs is 47.09%, and the load contents of Ce6 and Hb in PHC NPs were 27.3% and 54.8%, respectively. In vivo PTT/PDT experiments show that the TIR in mice is close to 100% within 30 days, and the tumor-recurrence rate is only 8.3% within 60 days.
In addition to the common nanomaterials mentioned above, researchers have developed photothermal agents such as mesoporous Prussian blue NPs [62][63], Mo-based polyoxometalate clusters [10][64], PbS/CdS/ZnS quantum dots [8][65], and so on. To prepare multifunctional nanomedicines with good biocompatibility, high stability, high drug load, degradability in vivo, nontoxic side effects, and high photothermal-conversion efficiency, it is a popular trend to hybridize inorganic and organic materials [66]. However, developing the perfect photothermal agent mentioned above for nanomedicines, and in vivo toxicity and photothermal-conversion efficiency are the most important issues.

3. Photosensitizers as Carriers for Fabricating High Drug-Loading Nanomedicine

PDT has become an important method in preclinical research and clinical practice in tumor treatment because of its minimally invasive and local tumor-killing ability without damaging surrounding healthy tissues and cells. The laser, PS, and oxygen are three indispensable elements in PDT. An ideal PS has high absorption coefficient in the 650–850 nm region, high 1O2 yield, solubility under physiological conditions, high tumor selectivity, low damage to healthy tissues, and few side effects. However, the PSs developed at present have some disadvantages. Hematoporphyrin isolated from hemoglobin is the first-generation PS, which has obvious toxicity. The second-generation PSs such as Ce6 and methylene blue are mostly hydrophobic drugs, which easily aggregate in the aqueous medium, and have no tumor selectivity and poor bioavailability. Therefore, the third-generation PS combining PS with a nanosized delivery system or nanomedicines has been developed. Those nanoformulated PSs are also called smart PSs because of their specificity and good therapeutic effects [67][68]Table 2 summarizes the different photosensitizers as carriers for fabricating high drug-loading nanomedicines.

3.1. Inorganic Materials

In recent years, the development of inorganic material synthesis technology has been booming. Naturally, different kinds of inorganic materials with appropriate sizes and surface properties have been prepared and used as photosensitizers in the field of PDT. At present, the reported inorganic materials that can be equipped with chemotherapeutic drugs and enhanced PDT include CdSe/ZnS quantum dots [69], gold nanorods [70], reduced graphene oxide [71], and black phosphorus [72]. Those inorganic nanomedicines can not only directly generate singlet oxygen, but also change the microenvironment of tumor cells and promote the PDT effect.
Wang et al. constructed novel stimulation-responsive multifunctional nanomedicines by inserting folate-linked ZnPcG4 molecules (ZnPcG4-FA) into layered dihydroxide (LDH) via a simple coprecipitate method [73]. Chemotherapy drugs (DOX) are loaded on the surface of LDH, and the loading efficiency of DOX was calculated to be 41.7%. In the acidic tumor microenvironment, the release of DOX can be controlled with the degradation of LDH. The LDH is an interesting series of layered inorganic materials with the chemical formula as [M2+−xM3+x(OH)2]x+ An−x/n·uH2O, where M2+, M3+ and An− represent divalent, trivalent metal ions and charge-balancing interlayer anion. Due to the special structure and pH response performance, LDH has been used as a carrier in drug delivery. Upon the 650 nm laser irradiation, the nanomedicines demonstrated excellent anticancer efficiency in vitro and in vivo because of the chemo–photodynamic combination therapy derived from the released DOX and ZnPcG4 photosensitizer.
Table 2. A summary of different PSs as carriers for fabricating high drug-loading nanomedicines.
Recently, dipyrromethene boron difluoride (BODIPY) has been widely employed as an ideal photosensitizer for PDT, which is attributed to its strong absorption coefficient, significant photostability and high ROS yield. Meng et al. synthesized a novel two-fold interpenetration pillar-layered metal–organic framework (MOF) material (BBP-MOFs) using BODIPY-derivate bipyridine as a linker and photosensitizer, biphenyl-4,4′-dicarboxylic acid (BPDC), and Cd (NO3)2 [74]. BBP-MOFs are not only photodynamic agents that generate singlet oxygen (1O2) to perform PDT under 660 nm light irradiation, but also are drug carriers by loading DOX for chemotherapy. The results showed that BBP-MOFs exhibited high drug-loading efficiency of 49.7% as well as controlled drug release and ideal biocompatibility.
Zou et al. designed and constructed multifunctional hybrid mesoporous nanomedicines (HMNPs) that integrate near-infrared persistent luminescence nanoparticle, magnetic nanoparticles (Gd2O3), and radionuclides (68Ga) via a large-pore (mesoporous silica nanoparticle) MSN-templated strategy (Figure 5B) [75]. The prepared HMNPs not only have good morphology, mesoporous structure, and surface properties, but also can load a variety of therapeutic agents at the same time. In the nanomedicines, the loading efficiency of DOX and photosensitizer (Si-Pc) were 34.5% and 51.2%, respectively. The studies on mice tumor models demonstrate that the DOX/Si-Pc-loaded HMNPs possess excellent cancer-cell-killing ability and an outstanding tumor suppression effect without systemic toxicity.
In addition to a variety of inorganic materials mentioned above, metal nanoparticles can be used in photodynamic therapy by conjugation or loading photosensitizer on their surfaces. For example, it is possible to modify the Au NPs either covalently or noncovalently with photosensitizer molecules [78][99]. Magnetic nanoparticles (MNPs) are one of the few inorganic materials approved by the FDA for therapeutic or imaging use in vivo. Photosensitizer molecules could be conjugated to MNPs for simultaneous magnetic resonance imaging and PDT [100][101]. Other carbon nanomaterials, including CNTs, graphene, and carbon dots, have also been used as carriers in PDT [102][103][104]. Moreover, upconversion nanoparticles (UCNPs) can emit high-energy light words under lower energy radiation, which has the advantages of better tissue penetration depth and higher photochemical stability. Therefore, nanomedicines of photosensitizer materials based on UCNPs have been gradually developed for photodynamic therapy [105][106][107].

3.2. Organic Materials

Biodegradable organic-based nanomedicines have been widely used in drug delivery, including photosensitizers loaded with organic materials for photodynamic tumor therapy. For example, coupling or loading photosensitizer with poly(ethylene glycol) (PEG) [86][108], amphiphilic block polymers [94][109], polysaccharides [91][110], folic acid [93], peptides [90][95], liposomes [88][97], and polystyrene [104] have been studied. Phycocyanin (PC) is considered to be an effective natural photosensitizer, but it has not been well utilized due to its low biostability and intracellular accumulation. In order to overcome these limitations, nanosized PC particles (LAPC/DOX) were prepared by grafting lactonic acid (LA) and loading adriamycin (DOX). Compared with PC solution, the storage stability and photostability of PC particles are significantly improved, and the formation of nanomedicines further improves their biological stability [111]. Zhang et al. synthesized pH/reactive ROS dual-responsive PEG-DOX conjugate (labeled TPD) through acyl alkynylamine click reaction by PEG dipropiolate (PEGB), amine-terminated thioketal (TKL), and DOX [108]. The prepared amphiphilic TPD not only has high drug-loading efficiency for photosensitizer chlorine e6 (Ce6), but is also sensitive to acidic tumor microenvironment (TME) and ROS. Under laser irradiation, Ce6 produces abundant singlet oxygen (1O2), enabling programmable accelerated release of DOX and more Ce6 at tumor sites. Luo et al. prepared a prodrug nanoparticle (CDC NPs) by co-encapsulation of single thioether-linked dihydroartemisinin (DHA) dimer and Ce6, then stabilized by albumin-capturing maleimide- and hypoxia-sensitive 2-nitroimidazole-modified carboxymethyl chitosan [110]. DHA-S-DHA served as a ROS-responsive carrier for Ce6 and a chemotherapeutic drug. Upon laser irradiation, Ce6 could generate reactive oxygen species (ROS), which not only exerted the effect of the PDT but also broke the ROS-sensitive single thioether bridge in the dimeric prodrug DHA-S-DHA, thus accelerating the disassembly of the nanomedicines. Moreover, some researchers attempted to functionalize Ce6 with amino acid [112] and poly (amido amine) generation dendrimer [113] for better therapeutic effect.
The carrier-free nanomedicines, made up of photosensitizers and antitumor drug molecules, have become a kind of common nanomedicines for chemo–photodynamic combination therapy. Liu et al. fabricated porphyrin-based drug self-framed delivery systems without any carrier materials, in which water-soluble photosensitizer (tetra sodium mesotetra (sulfonatophenyl)-porphyrin, H2TPPS) and DOX could self-assemble to form H2TPPS@DOX nanomedicines by supramolecular interaction such as hydrophobic, electrostatic and π-π stacking interactions [87]. Owing to the higher drug-loading efficiency of 42.4%, the H2TPPS@DOX could effectively generate singlet oxygen to obviously block DOX efflux and ultimately induce apoptosis to effectively reverse multidrug resistance of tumor cells under light irradiation.

This entry is adapted from the peer-reviewed paper 10.3390/pharmaceutics14081735

References

  1. Cheng, L.; Wang, C.; Feng, L.; Yang, K.; Liu, Z. Functional Nanomaterials for Phototherapies of Cancer. Chem. Rev. 2014, 114, 10869–10939.
  2. Cao, J.; Chen, Z.; Chi, J.; Sun, Y.; Sun, Y. Recent progress in synergistic chemotherapy and phototherapy by targeted drug delivery systems for cancer treatment. Artif. Cells Nanomed. Biotechnol. 2018, 46, 817–830.
  3. Li, W.; Cao, Z.; Yu, L.; Huang, Q.; Zhu, D.; Lu, C.; Lu, A.; Liu, Y. Hierarchical drug release designed Au @PDA-PEG-MTX NPs for targeted delivery to breast cancer with combined photothermal-chemotherapy. J. Nanobiotechnol. 2021, 19, 143.
  4. Awan, U.A.; Raza, A.; Ali, S.; Saeed, R.F.; Akhtar, N. Doxorubicin-loaded gold nanorods: A multifunctional chemo–photothermal nanoplatform for cancer management. Beilstein J. Nanotechnol. 2021, 12, 295–303.
  5. Xu, J.; Cheng, X.; Chen, F.; Li, W.; Xiao, X.; Lai, P.; Xu, G.; Xu, L.; Pan, Y. Fabrication of multifunctional polydopamine-coated gold nanobones for PA/CT imaging and enhanced synergistic chemo–photothermal therapy. J. Mater. Sci. Technol. 2021, 63, 97–105.
  6. Wang, J.; Sun, J.; Wang, Y.; Chou, T.; Zhang, Q.; Zhang, B.; Ren, L.; Wang, H. Gold Nanoframeworks with Mesopores for Raman–Photoacoustic Imaging and Photo-Chemo Tumor Therapy in the Second Near-Infrared Biowindow. Adv. Funct. Mater. 2020, 30, 1908825.
  7. Song, Y.; Qu, Z.; Li, J.; Shi, L.; Zhao, W.; Wang, H.; Sun, T.; Jia, T.; Sun, Y. Fabrication of the biomimetic DOX/ nanoparticles hybrid nanostructures for the combinational chemo/photothermal cancer therapy. J. Alloy. Compd. 2021, 881, 160592.
  8. Sun, C.; Liu, Y.; Zhou, R.; Yao, L.; Wang, R.; Zang, W.; Meng, W. Nano Pd-Decorated Manganese Dioxide Nanosheets for Effective Photothermal Chemotherapy. ACS Appl. Bio Mater. 2019, 2, 4747–4755.
  9. Li, X.; Yuan, H.J.; Tian, X.M.; Tang, J.; Liu, L.F.; Liu, F.Y. Biocompatible copper sulfide–based nanocomposites for artery interventional chemo–photothermal therapy of orthotropic hepatocellular carcinoma. Mater. Today Bio 2021, 12, 100128.
  10. Xie, M.; Li, J.; Deng, T.; Yang, N.; Yang, M. Modification of magnetic molybdenum disulfide by chitosan/carboxymethylcellulose with enhanced dispersibility for targeted photothermal-/chemotherapy of cancer. J. Mater. Chem. B 2021, 9, 1833–1845.
  11. Hou, M.; Zhong, Y.; Zhang, L.; Xu, Z.; Kang, Y.; Xue, P. Polydopamine (PDA)-activated cobalt sulfide nanospheres responsive to tumor microenvironment (TME) for chemotherapeutic-enhanced photothermal therapy. Chin. Chem. Lett. 2021, 32, 1055–1060.
  12. Xie, M.; Yang, M.; Sun, X.; Yang, N.; Deng, T.; Li, Y.; Shen, H. WS2 nanosheets functionalized by biomimetic lipids with enhanced dispersibility for photothermal and chemo combination therapy. J. Mater. Chem. B 2020, 8, 2331–2342.
  13. Chen, Y.; Khan, A.R.; Yu, D.; Zhai, Y.; Ji, J.; Shi, Y.; Zhai, G. Pluronic F127-functionalized molybdenum oxide nanosheets with pH-dependent degradability for chemo–photothermal cancer therapy. J. Colloid Interface Sci. 2019, 553, 567–580.
  14. Zhan, X.; Teng, W.; Sun, K.; He, J.; Yang, J.; Tian, J.; Huang, X.; Zhou, L.; Zhou, C. CD47-mediated DTIC-loaded chitosan oligosaccharide-grafted nGO for synergistic chemo–photothermal therapy against malignant melanoma. Mater. Sci. Eng. C 2021, 123, 112014.
  15. Xing, E.; Du, Y.; Yin, J.; Chen, M.; Zhu, M.; Wen, X.; Xu, J.; Feng, Y.; Meng, S. Multi-functional Nanodrug Based on a Three-dimensional Framework for Targeted Photo-chemo Synergetic Cancer Therapy. Adv. Heal. Mater. 2021, 10, e2001874.
  16. Wang, Y.; Zhang, H.; Xie, J.; Liu, Y.; Wang, S.; Zhao, Q. Three dimensional mesoporous carbon nanospheres as carriers for chemo–photothermal therapy compared with two dimensional graphene oxide nanosheets. Colloids Surf. A Physicochem. Eng. Asp. 2020, 590, 124498.
  17. Dong, X.; Sun, Z.; Wang, X.; Leng, X. An innovative MWCNTs/DOX/TC nanosystem for chemo–photothermal combination therapy of cancer. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2271–2280.
  18. Wang, C.; Chen, Y.; Xu, Z.; Chen, B.; Zhang, Y.; Yi, X.; Li, J. Fabrication and characterization of novel cRGD modified graphene quantum dots for chemo–photothermal combination therapy. Sens. Actuators B Chem. 2020, 309, 127732.
  19. Yang, J.; Su, H.; Sun, W.; Cai, J.; Liu, S.; Chai, Y.; Zhang, C. Dual Chemodrug-Loaded Single-Walled Carbon Nanohorns for Multimodal Imaging-Guided Chemo–photothermal Therapy of Tumors and Lung Metastases. Theranostics 2018, 8, 1966–1984.
  20. Wei, X.; Zhang, L.; Li, S.; Chen, X.; Zhang, M.; Wang, C.; Wang, T.; Li, L. A designed synthesis of multifunctional carbon nanoframes for simultaneous imaging and synergistic chemo–photothermal cancer therapy. New J. Chem. 2018, 42, 923–929.
  21. Lu, L.; Wang, K.; Lin, C.; Yang, W.; Duan, Q.; Li, K.; Cai, K. Constructing nanocomplexes by multicomponent self-assembly for curing orthotopic glioblastoma with synergistic chemo–photothermal therapy. Biomaterials 2021, 279, 121193.
  22. Gao, H.; Bai, Y.; Chen, L.; Fakhri, G.E.; Wang, M. Self-Assembly Nanoparticles for Overcoming Multidrug Resistance and Imaging-Guided Chemo–photothermal Synergistic Cancer Therapy. Int. J. Nanomed. 2020, 15, 809–819.
  23. He, L.; Qing, F.; Li, M.; Lan, D. Paclitaxel/IR1061-Co-Loaded Protein Nanoparticle for Tumor-Targeted and pH/NIR-II-Triggered Synergistic Photothermal-Chemotherapy. Int. J. Nanomed. 2020, 15, 2337–2349.
  24. Liu, S.; Pan, J.; Liu, J.; Ma, Y.; Qiu, F.; Mei, L.; Zeng, X.; Pan, G. Dynamically PEGylated and Borate-Coordination-Polymer-Coated Polydopamine Nanoparticles for Synergetic Tumor-Targeted, Chemo–photothermal Combination Therapy. Small 2018, 14, e1703968.
  25. Zhang, Y.; Fang, F.; Chen, Y.; Li, M.; Li, L.; Li, W.; Zhang, J. Hollow mesoporous polyaniline nanoparticles with high drug payload and robust photothermal capability for cancer combination therapy. Chin. J. Chem. Eng. 2021, 38, 221–228.
  26. Liu, X.; Liu, Y.; Guo, Y.; Shi, W.; Sun, Y.; He, Z.; Shen, Y.; Zhang, X.; Xiao, H.; Ge, D. Metabolizable pH/H2O2 dual-responsive conductive polymer nanoparticles for safe and precise chemo–photothermal therapy. Biomaterials 2021, 277, 121115.
  27. Fathy, M.M.; Nasser, L.; El-Sokkary, G.; Rasheedy, M.S. Combined Chemo–photothermal Therapy of Metastatic Mammary Adenocarcinoma Using Curcumin-Coated Iron Oxide Nanoparticles. BioNanoScience 2021, 11, 447–453.
  28. Liu, Y.; Han, Q.; Yang, W.; Gan, X.; Yang, Y.; Xie, K.; Xie, L.; Deng, Y. Two-dimensional MXene/cobalt nanowire heterojunction for controlled drug delivery and chemo–photothermal therapy. Mater. Sci. Eng. C 2020, 116, 111212.
  29. Huang, Y.; Xue, Z.; Zeng, S. Hollow Mesoporous Nanoshell as a Novel Dual-Stimuli-Responsive Nanocarrier for Synergistic Chemo–photothermal Cancer Therapy. ACS Appl. Mater. Interfaces 2020, 12, 31172–31181.
  30. Wang, J.; Wu, X.; Shen, P.; Wang, J.; Shen, Y.; Shen, Y.; Webster, T.J.; Deng, J. Applications of Inorganic Nanomaterials in Photothermal Therapy Based on Combinational Cancer Treatment. Int. J. Nanomed. 2020, 15, 1903–1914.
  31. Connor, D.M.; Broome, A.-M. Chapter Seven—Gold Nanoparticles for the Delivery of Cancer Therapeutics. In Advances in Cancer Research; Broome, A.-M., Ed.; Academic Press: Cambridge, MA, USA, 2018; Volume 139, pp. 163–184.
  32. Zhang, L.; Shen, S.; Cheng, L.; You, H.; Lu, L.; Ma, C.; Dai, Y.; Fang, J. Mesoporous gold nanoparticles for photothermal controlled anticancer drug delivery. Nanomedicine 2019, 14, 1443–1454.
  33. Lee, C.-S.; Kim, H.; Yu, J.; Yu, S.H.; Ban, S.; Oh, S.; Jeong, D.; Im, J.; Baek, M.J.; Kim, T.H. Doxorubicin-loaded oligonucleotide conjugated gold nanoparticles: A promising in vivo drug delivery system for colorectal cancer therapy. Eur. J. Med. Chem. 2017, 142, 416–423.
  34. 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 2018, 83, 90–98.
  35. 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, 1705451.
  36. 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.
  37. Xia, Y.; Li, W.; Cobley, C.M.; Chen, J.; Xia, X.; Zhang, Q.; Yang, M.; Cho, E.C.; Brown, P.K. Gold Nanocages: From Synthesis to Theranostic Applications. Accounts Chem. Res. 2011, 44, 914–924.
  38. Li, S.; Zhang, L.; Wang, T.; Li, L.; Wang, C.; Su, Z. The facile synthesis of hollow Au nanoflowers for synergistic chemo–photothermal cancer therapy. Chem. Commun. 2015, 51, 14338–14341.
  39. Song, C.; Dou, Y.; Yuwen, L.; Sun, Y.; Dong, C.; Li, F.; Yang, Y.; Wang, L. A gold nanoflower-based traceable drug delivery system for intracellular SERS imaging-guided targeted chemo–phototherapy. J. Mater. Chem. B 2018, 6, 3030–3039.
  40. Wang, C.; Li, J.; Amatore, C.; Chen, Y.; Jiang, H.; Wang, X.-M. Gold Nanoclusters and Graphene Nanocomposites for Drug Delivery and Imaging of Cancer Cells. Angew. Chem. Int. Ed. 2011, 50, 11644–11648.
  41. Cui, H.; Shao, Z.-S.; Song, Z.; Wang, Y.-B.; Wang, H.-S. Development of gold nanoclusters: From preparation to applications in the field of biomedicine. J. Mater. Chem. C 2020, 8, 14312–14333.
  42. Huang, J.; Xu, Z.; Jiang, Y.; Law, W.C.; Dong, B.; Zeng, X.; Ma, M.; Xu, G.; Zou, J.; Yang, C. Metal organic framework-coated gold nanorod as an on-demand drug delivery platform for chemo–photothermal cancer therapy. J. Nanobiotechnol. 2021, 19, 219.
  43. He, J.; Dong, J.; Hu, Y.; Li, G.; Hu, Y. Design of Raman tag-bridged core–shell (BTC)2 nanoparticles for Raman imaging and synergistic chemo–photothermal therapy. Nanoscale 2019, 11, 6089–6100.
  44. Shibu, E.S.; Hamada, M.; Murase, N.; Biju, V. Nanomaterials formulations for photothermal and photodynamic therapy of cancer. J. Photochem. Photobiol. C Photochem. Rev. 2013, 15, 53–72.
  45. Liu, Z.; Tabakman, S.M.; Chen, Z.; Dai, H. Preparation of carbon nanotube bioconjugates for biomedical applications. Nat. Protoc. 2009, 4, 1372–1382.
  46. Wang, Y.; Hu, J.; Xiang, D.; Peng, X.; You, Q.; Mao, Y.; Hua, D.; Yin, J. Targeted nanosystem combined with chemo–photothermal therapy for hepatocellular carcinoma treatment. Colloids Surf. A Physicochem. Eng. Asp. 2020, 596, 124711.
  47. Ortiz de Solorzano, I.; Prieto, M.; Mendoza, G.; Alejo, T.; Irusta, S.; Sebastian, V.; Arruebo, M. Microfluidic Synthesis and Biological Evaluation of Photothermal Biodegradable Copper Sulfide Nanoparticles. ACS Appl. Mater. Interfaces 2016, 8, 21545–21554.
  48. Gayathri, R.; Rajalakshmi, P.S.; Thomas, A.; Rengan, A.K. Doxorubicin loaded polyvinylpyrrolidone-copper sulfide nanoparticles enabling mucoadhesiveness and chemo–photothermal synergism for effective killing of breast cancer cells. Materialia 2021, 19, 101195.
  49. Zhang, S.; Sun, C.; Zeng, J.; Sun, Q.; Wang, G.; Wang, Y.; Wu, Y.; Dou, S.; Gao, M.; Li, Z. Ambient Aqueous Synthesis of Ultrasmall PEGylated Cu2−xSe Nanoparticles as a Multifunctional Theranostic Agent for Multimodal Imaging Guided Photothermal Therapy of Cancer. Adv. Mater. 2016, 28, 8927–8936.
  50. Chou, S.S.; Kaehr, B.; Kim, J.; Foley, B.M.; De, M.; Hopkins, P.E.; Huang, J.; Brinker, C.J.; Dravid, V.P. Chemically Exfoliated MoS2 as Near-Infrared Photothermal Agents. Angew. Chem. Int. Ed. 2013, 52, 4160–4164.
  51. Wu, C.; Wang, S.; Zhao, J.; Liu, Y.; Zheng, Y.; Luo, Y.; Ye, C.; Huang, M.; Chen, H. Biodegradable Fe(III)@WS2-PVP Nanocapsules for Redox Reaction and TME-Enhanced Nanocatalytic, Photothermal, and Chemotherapy. Adv. Funct. Mater. 2019, 29, 1901722.
  52. Fang, L.; Wang, J.; Ouyang, X.; Liang, B.; Zhao, L.; Huang, D.; Deng, D. FeSe2 nanosheets as a bifunctional platform for synergistic tumor therapy reinforced by NIR-II light. Biomater. Sci. 2021, 9, 5542–5550.
  53. She, D.; Peng, S.; Liu, L.; Huang, H.; Zheng, Y.; Lu, Y.; Geng, D.; Yin, B. Biomimic FeS2 nanodrug with hypothermal photothermal effect by clinical approved NIR-Ⅱ light for augmented chemodynamic therapy. Chem. Eng. J. 2020, 400, 125933.
  54. Qian, X.; Shen, S.; Liu, T.; Cheng, L.; Liu, Z. Two-dimensional TiS2 nanosheets for in vivo photoacoustic imaging and photothermal cancer therapy. Nanoscale 2015, 7, 6380–6387.
  55. Chen, Y.; Gao, M.; Zhang, L.; Ha, E.; Hu, X.; Zou, R.; Yan, L.; Hu, J. Tumor Microenvironment Responsive Biodegradable Fe-Doped MoOx Nanowires for Magnetic Resonance Imaging Guided Photothermal-Enhanced Chemodynamic Synergistic Antitumor Therapy. Adv. Healthc. Mater. 2021, 10, 2001665.
  56. Sharker, S.M.; Kim, S.M.; Lee, J.E.; Choi, K.H.; Shin, G.; Lee, S.; Lee, K.D.; Jeong, J.H.; Lee, H.; Park, S.Y. Functionalized biocompatible WO3 nanoparticles for triggered and targeted in vitro and in vivo photothermal therapy. J. Control. Release 2015, 217, 211–220.
  57. Ou, G.; Li, Z.; Li, D.; Cheng, L.; Liu, Z.; Wu, H. Photothermal therapy by using titanium oxide nanoparticles. Nano Res. 2016, 9, 1236–1243.
  58. Wu, M.; Mei, T.; Lin, C.; Wang, Y.; Chen, J.; Le, W.; Sun, M.; Xu, J.; Dai, H.; Zhang, Y.; et al. Melanoma Cell Membrane Biomimetic Versatile CuS Nanoprobes for Homologous Targeting Photoacoustic Imaging and Photothermal Chemotherapy. ACS Appl. Mater. Interfaces 2020, 12, 16031–16039.
  59. Li, L.; Lu, Y.; Qian, Z.; Yang, Z.; Zong, S.; Wang, Z.; Cui, Y. A Ti2N MXene-based nanosystem with ultrahigh drug loading for dual-strategy synergistic oncotherapy. Nanoscale 2021, 13, 18546–18557.
  60. Wang, Y.; Wu, Y.; Li, K.; Shen, S.; Liu, Z.; Wu, D. Ultralong Circulating Lollipop-Like Nanoparticles Assembled with Gossypol, Doxorubicin, and Polydopamine via π-π Stacking for Synergistic Tumor Therapy. Adv. Funct. Mater. 2019, 29, 1805582.
  61. Wang, Y.; Luo, S.; Wu, Y.; Tang, P.; Liu, J.; Liu, Z.; Shen, S.; Ren, H.; Wu, D. Highly Penetrable and On-Demand Oxygen Release with Tumor Activity Composite Nanosystem for Photothermal/Photodynamic Synergetic Therapy. ACS Nano. 2020, 14, 17046–17062.
  62. Lin, X.; Cao, Y.; Li, J.; Zheng, D.; Lan, S.; Xue, Y.; Yu, F.; Wu, M.; Zhu, X. Folic acid-modified Prussian blue/polydopamine nanoparticles as an MRI agent for use in targeted chemo/photothermal therapy. Biomater. Sci. 2019, 7, 2996–3006.
  63. Chen, W.; Zeng, K.; Liu, H.; Ouyang, J.; Wang, L.; Liu, Y.; Wang, H.; Deng, L.; Liu, Y.-N. Cell Membrane Camouflaged Hollow Prussian Blue Nanoparticles for Synergistic Photothermal-/Chemotherapy of Cancer. Adv. Funct. Mater. 2017, 27, 1605795.
  64. Tian, Y.; Yi, W.; Bai, L.; Cheng, X.; Yi, T.; Mu, M.; Zhang, P.; Si, J.; Hou, X.; Hou, J. One-step in situ growth of as a dual-stimuli-responsive nanocarrier for synergistic chemo–photothermal therapy. New J. Chem. 2021, 45, 17966–17975.
  65. Zhang, Y.; Guo, R.; Wang, D.; Sun, X.; Xu, Z. Pd nanoparticle-decorated hydroxy boron nitride nanosheets as a novel drug carrier for chemo–photothermal therapy. Colloids Surf. B Biointerfaces 2019, 176, 300–308.
  66. Dinçer, C.A.; Getiren, B.; Gökalp, C.; Ciplak, Z.; Karakeçili, A.; Yildiz, N. An anticancer drug loading and release study to ternary GO-Fe3O4-PPy and Fe3O4 @PPy-NGQDs nanocomposites for photothermal chemotherapy. Colloids Surf. A Physicochem. Eng. Asp. 2022, 633, 127791.
  67. Hak, A.; Ravasaheb Shinde, V.; Rengan, A.K. A review of advanced nanoformulations in phototherapy for cancer therapeutics. Photodiagnosis Photodyn. Ther. 2021, 33, 102205.
  68. Kim, J.; Jo, Y.U.; Na, K. Photodynamic therapy with smart nanomedicine. Arch. Pharm. Res. 2020, 43, 22–31.
  69. Sewid, F.A.; Skurlov, I.D.; Kurshanov, D.A.; Orlova, A.O. Singlet oxygen generation by hybrid structures based on CdSe/ZnS quantum dots and tetraphenylporphyrin in organic medium. Chem. Phys. Lett. 2021, 765, 138303.
  70. 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.
  71. Liang, J.; Chen, B.; Hu, J.; Huang, Q.; Zhang, D.; Wan, J.; Hu, Z.; Wang, B. pH and Thermal Dual-Responsive Graphene Oxide Nanocomplexes for Targeted Drug Delivery and Photothermal-Chemo/Photodynamic Synergetic Therapy. ACS Appl. Bio Mater. 2019, 2, 5859–5871.
  72. Liu, J.; Du, P.; Liu, T.; Córdova Wong, B.J.; Wang, W.; Ju, H.; Lei, J. A black phosphorus/manganese dioxide nanoplatform: Oxygen self-supply monitoring, photodynamic therapy enhancement and feedback. Biomaterials 2019, 192, 179–188.
  73. Wang, Z.; Zhu, H.; He, F.; Yan, D.; Jia, T.; Sun, Q.; Wang, X.; Wang, X.; Yang, D.; Gai, S.; et al. Layer structured LDH_ZnPcG4-FA nanoplatform for targeted and imaging guided chemo–photodynamic therapy mediated by 650 nm light. Chem. Eng. J. 2020, 382, 122847.
  74. Meng, Y.; Du, Y.; Lin, Y.; Su, Y.; Li, R.; Feng, Y.; Meng, S. A two-fold interpenetration pillar-layered metal-organic frameworks based on BODIPY for chemo–photodynamic therapy. Dye. Pigment. 2021, 188, 109174.
  75. Zou, R.; Gao, Y.; Zhang, Y.; Jiao, J.; Wong, K.-L.; Wang, J. 68Ga-Labeled Magnetic-NIR Persistent Luminescent Hybrid Mesoporous Nanoparticles for Multimodal Imaging-Guided Chemotherapy and Photodynamic Therapy. ACS Appl. Mater. Interfaces 2021, 13, 9667–9680.
  76. Wu, X.; Zhang, Y.; Wang, Z.; Wu, J.; Yan, R.; Guo, C.; Jin, Y. Near-Infrared Light-Initiated Upconversion Nanoplatform with Tumor Microenvironment Responsiveness for Improved Photodynamic Therapy. ACS Appl. Bio Mater. 2020, 3, 5813–5823.
  77. Huang, Y.; Liu, D.-E.; An, J.; Liu, B.; Sun, L.; Fu, H.; Yan, S.; Sun, W.; Gao, H. Reactive Oxygen Species Self-Sufficient Multifunctional Nanoplatform for Synergistic Chemo–Photodynamic Therapy with Red/Near-Infrared Dual-Imaging. ACS Appl. Bio Mater. 2020, 3, 9135–9144.
  78. Cui, Y.-X.; Sun, G.-Y.; Li, Y.-H.; Tang, A.-N.; Zhu, L.-N.; Kong, D.-M. DNA-Based pH-Responsive Core–Shell Drug Nanocarrier for Tumor-Targeted Chemo–Photodynamic Therapy. Adv. Mater. Interfaces 2020, 7, 2000292.
  79. Ren, S.-Z.; Wang, B.; Zhu, X.-H.; Zhu, D.; Liu, M.; Li, S.-K.; Yang, Y.-S.; Wang, Z.-C.; Zhu, H.-L. Oxygen Self-Sufficient Core–Shell Metal–Organic Framework-Based Smart Nanoplatform for Enhanced Synergistic Chemotherapy and Photodynamic Therapy. ACS Appl. Mater. Interfaces 2020, 12, 24662–24674.
  80. Zhu, K.; Liu, G.; Hu, J.; Liu, S. Near-Infrared Light-Activated Photochemical Internalization of Reduction-Responsive Polyprodrug Vesicles for Synergistic Photodynamic Therapy and Chemotherapy. Biomacromolecules 2017, 18, 2571–2582.
  81. Liu, J.; Liu, K.; Feng, L.; Liu, Z.; Xu, L. Comparison of nanomedicine-based chemotherapy, photodynamic therapy and photothermal therapy using reduced graphene oxide for the model system. Biomater. Sci. 2017, 5, 331–340.
  82. Liu, W.; Wang, Y.-M.; Li, Y.-H.; Cai, S.-J.; Yin, X.-B.; He, X.-W.; Zhang, Y.-K. Fluorescent Imaging-Guided Chemotherapy-and-Photodynamic Dual Therapy with Nanoscale Porphyrin Metal–Organic Framework. Small 2017, 13, 1603459.
  83. Lan, J.-S.; Liu, L.; Zeng, R.-F.; Qin, Y.-H.; Hou, J.-W.; Xie, S.-S.; Yue, S.; Yang, J.; Ho, R.J.Y.; Ding, Y.; et al. Tumor-specific carrier-free nanodrugs with GSH depletion and enhanced ROS generation for endogenous synergistic anti-tumor by a chemotherapy-photodynamic therapy. Chem. Eng. J. 2021, 407, 127212.
  84. Huang, L.; Wan, J.; Wu, H.; Chen, X.; Bian, Q.; Shi, L.; Jiang, X.; Yuan, A.; Gao, J.; Wang, H. Quantitative self-assembly of photoactivatable small molecular prodrug cocktails for safe and potent cancer chemo–photodynamic therapy. Nano Today 2021, 36, 101030.
  85. Choi, J.; Shim, M.K.; Yang, S.; Hwang, H.S.; Cho, H.; Kim, J.; Yun, W.S.; Moon, Y.; Kim, J.; Yoon, H.Y.; et al. Visible-Light-Triggered Prodrug Nanoparticles Combine Chemotherapy and Photodynamic Therapy to Potentiate Checkpoint Blockade Cancer Immunotherapy. ACS Nano 2021, 15, 12086–12098.
  86. Liu, L.; Bao, Y.; Zhang, Y.; Xiao, C.; Chen, L. Acid-responsive dextran-based therapeutic nanoplatforms for photodynamic-chemotherapy against multidrug resistance. Int. J. Biol. Macromol. 2020, 155, 233–240.
  87. Liu, L.; Bao, Y.; Wang, J.; Xiao, C.; Chen, L. Construction of carrier-free porphyrin-based drug self-framed delivery system to reverse multidrug resistance through photodynamic-chemotherapy. Dye. Pigment. 2020, 177, 107922.
  88. Wang, X.; Tong, J.; He, Z.; Yang, X.; Meng, F.; Liang, H.; Zhang, X.; Luo, L. Paclitaxel-Potentiated Photodynamic Theranostics for Synergistic Tumor Ablation and Precise Anticancer Efficacy Monitoring. ACS Appl. Mater. Interfaces 2020, 12, 5476–5487.
  89. Jiang, M.; Mu, J.; Jacobson, O.; Wang, Z.; He, L.; Zhang, F.; Yang, W.; Lin, Q.; Zhou, Z.; Ma, Y.; et al. Reactive Oxygen Species Activatable Heterodimeric Prodrug as Tumor-Selective Nanotheranostics. ACS Nano 2020, 14, 16875–16886.
  90. Cheng, Y.-J.; Qin, S.-Y.; Liu, W.-L.; Ma, Y.-H.; Chen, X.-S.; Zhang, A.-Q.; Zhang, X.-Z. Dual-Targeting Photosensitizer-Peptide Amphiphile Conjugate for Enzyme-Triggered Drug Delivery and Synergistic Chemo–Photodynamic Tumor Therapy. Adv. Mater. Interfaces 2020, 7, 2000935.
  91. Wang, Y.; Yang, M.; Qian, J.; Xu, W.; Wang, J.; Hou, G.; Ji, L.; Suo, A. Sequentially self-assembled polysaccharide-based nanocomplexes for combined chemotherapy and photodynamic therapy of breast cancer. Carbohydr. Polym. 2019, 203, 203–213.
  92. Luo, C.; Sun, B.; Wang, C.; Zhang, X.; Chen, Y.; Chen, Q.; Yu, H.; Zhao, H.; Sun, M.; Li, Z.; et al. Self-facilitated ROS-responsive nanoassembly of heterotypic dimer for synergistic chemo–photodynamic therapy. J. Control. Release 2019, 302, 79–89.
  93. Wang, Z.; Sun, M.; Liu, T.; Tan, X.; Zhang, H.; Zhang, X.; He, Z.; Sun, J. A surfactant-like chemotherapeutic agent as a nanocarrier for delivering photosensitizers against cancer: A facile drug-delivering-drug strategy. Int. J. Pharm. 2019, 562, 313–320.
  94. Wang, M.; Zhai, Y.; Ye, H.; Lv, Q.; Sun, B.; Luo, C.; Jiang, Q.; Zhang, H.; Xu, Y.; Jing, Y.; et al. High Co-loading Capacity and Stimuli-Responsive Release Based on Cascade Reaction of Self-Destructive Polymer for Improved Chemo–Photodynamic Therapy. ACS Nano 2019, 13, 7010–7023.
  95. Liu, R.; Yu, M.; Yang, X.; Umeshappa, C.S.; Hu, C.; Yu, W.; Qin, L.; Huang, Y.; Gao, H. Linear Chimeric Triblock Molecules Self-Assembled Micelles with Controllably Transformable Property to Enhance Tumor Retention for Chemo–Photodynamic Therapy of Breast Cancer. Adv. Funct. Mater. 2019, 29, 1808462.
  96. Luo, Y.; Song, Y.; Wang, M.; Jian, T.; Ding, S.; Mu, P.; Liao, Z.; Shi, Q.; Cai, X.; Jin, H.; et al. Bioinspired Peptoid Nanotubes for Targeted Tumor Cell Imaging and Chemo-Photodynamic Therapy. Small 2019, 15, e1902485.
  97. Lee, H.; Han, J.; Shin, H.; Han, H.; Na, K.; Kim, H. Combination of chemotherapy and photodynamic therapy for cancer treatment with sonoporation effects. J. Control. Release 2018, 283, 190–199.
  98. Pei, Q.; Hu, X.; Zheng, X.; Liu, S.; Li, Y.; Jing, X.; Xie, Z. Light-Activatable Red Blood Cell Membrane-Camouflaged Dimeric Prodrug Nanoparticles for Synergistic Photodynamic/Chemotherapy. ACS Nano 2018, 12, 1630–1641.
  99. Liu, D.; Liu, L.; Liu, F.; Zhang, M.; Wei, P.; Yi, T. HOCl-Activated Aggregation of Gold Nanoparticles for Multimodality Therapy of Tumors. Adv. Sci. 2021, 8, 2100074.
  100. Zhang, F.; Lu, G.; Wen, X.; Li, F.; Ji, X.; Li, Q.; Wu, M.; Cheng, Q.; Yu, Y.; Tang, J.; et al. Magnetic nanoparticles coated with polyphenols for spatio-temporally controlled cancer photothermal/immunotherapy. J. Control. Release 2020, 326, 131–139.
  101. Yan, L.; Amirshaghaghi, A.; Huang, D.; Miller, J.; Stein, J.M.; Busch, T.M.; Cheng, Z.; Tsourkas, A. Protoporphyrin IX (PpIX)-Coated Superparamagnetic Iron Oxide Nanoparticle (SPION) Nanoclusters for Magnetic Resonance Imaging and Photodynamic Therapy. Adv. Funct. Mater. 2018, 28, 1707030.
  102. Wang, L.; Shi, J.; Liu, R.; Liu, Y.; Zhang, J.; Yu, X.; Gao, J.; Zhang, C.; Zhang, Z. Photodynamic effect of functionalized single-walled carbon nanotubes: A potential sensitizer for photodynamic therapy. Nanoscale 2014, 6, 4642–4651.
  103. Taheri, H.; Unal, M.A.; Sevim, M.; Gurcan, C.; Ekim, O.; Ceylan, A.; Syrgiannis, Z.; Christoforidis, K.C.; Bosi, S.; Ozgenç, O.; et al. Photocatalytically Active Graphitic Carbon Nitride as an Effective and Safe 2D Material for In Vitro and In Vivo Photodynamic Therapy. Small 2020, 16, 1904619.
  104. Murali, G.; Kwon, B.; Kang, H.; Modigunta, J.K.R.; Park, S.; Lee, S.; Lee, H.; Park, Y.H.; Kim, J.; Park, S.Y.; et al. Hematoporphyrin Photosensitizer-Linked Carbon Quantum Dots for Photodynamic Therapy of Cancer Cells. ACS Appl. Nano Mater. 2022, 5, 4376–4385.
  105. Shi, Z.; Zhang, K.; Zada, S.; Zhang, C.; Meng, X.; Yang, Z.; Dong, H. Upconversion Nanoparticle-Induced Multimode Photodynamic Therapy Based on a Metal–Organic Framework/Titanium Dioxide Nanocomposite. ACS Appl. Mater. Interfaces 2020, 12, 12600–12608.
  106. Jin, F.; Qi, J.; Liu, D.; You, Y.; Shu, G.; Du, Y.; Wang, J.; Xu, X.; Ying, X.; Ji, J.; et al. Cancer-cell-biomimetic Upconversion nanoparticles combining chemo–photodynamic therapy and CD73 blockade for metastatic triple-negative breast cancer. J. Control. Release 2021, 337, 90–104.
  107. Liang, S.; Sun, C.; Yang, P.; Ma, P.A.; Huang, S.; Cheng, Z.; Yu, X.; Lin, J. Core-shell structured upconversion nanocrystal-dendrimer composite as a carrier for mitochondria targeting and catalase enhanced anti-cancer photodynamic therapy. Biomaterials 2020, 240, 119850.
  108. Zhang, N.; Wang, D.; Jing, X.; Yang, T.; Yang, H.; Meng, L. pH/ROS Dual-Responsive Polymer–Drug-Based Nanocarriers: Click-Reaction Preparation and Fluorescence Imaging-Guided Chemotherapy and Photodynamic Therapy. ACS Appl. Bio. Mater. 2021, 4, 6294–6303.
  109. Ma, Q.; Zhao, Y.; Guan, Q.; Zhao, Y.; Zhang, H.; Ding, Z.; Wang, Q.; Wu, Y.; Liu, M.; Han, J. Amphiphilic block polymer-based self-assembly of high payload nanoparticles for efficient combinatorial chemo–photodynamic therapy. Drug Deliv. 2020, 27, 1656–1666.
  110. Luo, R.; Zhang, Z.; Han, L.; Xue, Z.; Zhang, K.; Liu, F.; Feng, F.; Xue, J.; Liu, W.; Qu, W. An albumin-binding dimeric prodrug nanoparticle with long blood circulation and light-triggered drug release for chemo–photodynamic combination therapy against hypoxia-induced metastasis of lung cancer. Biomater. Sci. 2021, 9, 3718–3736.
  111. Liu, X.; Du, J.; Xie, Z.; Wang, L.; Liu, X.; Hou, Z.; Wang, X.; Tang, R. Lactobionic acid-modified phycocyanin nanoparticles loaded with doxorubicin for synergistic chemo–photodynamic therapy. Int. J. Biol. Macromol. 2021, 186, 206–217.
  112. Chibh, S.; Katoch, V.; Singh, M.; Prakash, B.; Panda, J.J. Miniatured Fluidics-Mediated Modular Self-Assembly of Anticancer Drug–Amino Acid Composite Microbowls for Combined Chemo–Photodynamic Therapy in Glioma. ACS Biomater. Sci. Eng. 2021, 7, 5654–5665.
  113. Díaz, C.F.; Guzmán, L.; Jiménez, V.A.; Alderete, J.B. Polyamidoamine dendrimers of the third generation–chlorin e6 nanoconjugates: Nontoxic hybrid polymers with photodynamic activity. J. Appl. Polym. Sci. 2022, 139, 51835.
More
This entry is offline, you can click here to edit this entry!