Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
Add more filters

Publication year range
1.
Int J Biol Macromol ; 266(Pt 2): 131359, 2024 May.
Article in English | MEDLINE | ID: mdl-38580018

ABSTRACT

The combination of photothermal therapy (PTT) and photodynamic therapy (PDT) has emerged as a promising strategy for cancer treatment. However, the poor photostability and photothermal conversion efficiency (PCE) of organic small-molecule photosensitizers, and the intracellular glutathione (GSH)-mediated singlet oxygen scavenging largely decline the antitumor efficacy of PTT and PDT. Herein, a versatile nanophotosensitizer (NPS) system is developed by ingenious incorporation of indocyanine green (ICG) into the PEGylated chitosan (PEG-CS)-coated polydopamine (PDA) nanoparticles via multiple π-π stacking, hydrophobic and electrostatic interactions. The PEG-CS-covered NPS showed prominent colloidal and photothermal stability as well as high PCE (ca 62.8 %). Meanwhile, the Michael addition between NPS and GSH can consume GSH, thus reducing the GSH-induced singlet oxygen scavenging. After being internalized by CT26 cells, the NPS under near-infrared laser irradiation produced massive singlet oxygen with the aid of thermo-enhanced intracellular GSH depletion to elicit mitochondrial damage and lipid peroxide formation, thus leading to ferroptosis and apoptosis. Importantly, the combined PTT and PDT delivered by NPS effectively inhibited CT26 tumor growth in vivo by light-activated intense hyperthermia and redox homeostasis disturbance. Overall, this work presents a new tactic of boosting antitumor potency of ICG-mediated phototherapy by PEG-CS-covered NPS.


Subject(s)
Chitosan , Glutathione , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Photothermal Therapy , Polyethylene Glycols , Chitosan/chemistry , Photochemotherapy/methods , Animals , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Glutathione/metabolism , Polyethylene Glycols/chemistry , Mice , Nanoparticles/chemistry , Photothermal Therapy/methods , Cell Line, Tumor , Indocyanine Green/chemistry , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/pathology , Singlet Oxygen/metabolism , Humans , Apoptosis/drug effects , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry
2.
Small ; 20(31): e2310706, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38446096

ABSTRACT

Photothermal treatment (PTT) has emerged as a promising avenue for biofilm elimination, yet its potential drawbacks, such as local hyperpyrexia and bacterial heat resistance, have posed challenges. To address these concerns, an innovative nanoplatform (Au@mSiO2-arg/ICG) is devised that integrates phototherapeutic and gas therapeutic functionalities. This multifaceted nanoplatform is composed of mesoporous silica-coated Au nanorods (Au@mSiO2), supplemented with l-arginine (l-arg) and indocyanine green (ICG), and is engineered for mild temperature PTT aimed at biofilm eradication. Au@mSiO2-arg/ICG nanoparticles (NPs) show excellent antibacterial effects through the generation of nitric oxide (NO) gas, heat, and reactive oxygen species (ROS) under 808 nm light irradiation. The ROS generated by ICG initiates a cascade reaction with l-arg, ultimately yielding NO gas molecules. This localized release of NO not only effectively curbs the expression of heat shock proteins 70 mitigating bacterial thermoresistance, but also reduces extracellular polymeric substance allowing better penetration of the therapeutic agents. Furthermore, this nanoplatform achieves an outstanding biofilm elimination rate of over 99% in an abscess model under 808 nm light irradiation (0.8 W·cm-2), thereby establishing its potential as a dependable strategy for NO-enhanced mild PTT and antibacterial photodynamic therapy (aPDT) in clinical settings.


Subject(s)
Biofilms , Indocyanine Green , Infrared Rays , Nitric Oxide , Biofilms/drug effects , Nitric Oxide/metabolism , Nitric Oxide/chemistry , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Gold/chemistry , Silicon Dioxide/chemistry , Reactive Oxygen Species/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Nanoparticles/chemistry , Arginine/chemistry , Arginine/pharmacology , Animals , Nanotubes/chemistry
3.
Colloids Surf B Biointerfaces ; 235: 113770, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38330689

ABSTRACT

Significant progress has been made in cancer immunotherapy; however, challenges such as interpatient variability, limited treatment response, and severe side effects persist. Although nanoimmunotherapy has emerged as a promising approach, the construction of precise and efficient nanosystems remain formidable challenges. Herein, a multifunctional nanoplatform was developed using macrophage-derived cellular vesicles (MCVs) for NIR-II imaging-guided precise cancer photo-immunotherapy. MCVs exhibited excellent tumor targeting and TAMs re-education effects, serving as both delivery carriers and therapeutic agents. Through amide bond, indocyanine green (ICG) was conjugated to the surface of MCVs, enabling in vivo tracking of MCVs distribution. Notably, ICG exhibited dual functionality as a NIR-II fluorescent agent and possessed photodynamic and photothermal effects, enabling the conversion of light energy into chemical or heat energy to eliminate tumor cells. This precision phototherapy triggered immunogenic cell death (ICD) of tumor, thereby activating the anti-tumor immune response. Additionally, MCVs loaded with R848, a toll-like receptor agonist, augmented the ICD-induced anti-tumor immunity. Animal experiments confirmed that MCVs-mediated photoimmunotherapy promoted T cell infiltration, inhibited tumor growth, and improved survival rates. In conclusion, we have developed a promising precision immunotherapy strategy capable of enhancing the immune response while mitigating off-target effects. These findings offer encouraging prospects for clinical translation.


Subject(s)
Nanoparticles , Neoplasms , Animals , Cell Line, Tumor , Phototherapy , Indocyanine Green/chemistry , Neoplasms/diagnostic imaging , Neoplasms/therapy , Immunotherapy , Optical Imaging , Nanoparticles/chemistry
4.
J Mater Chem B ; 12(7): 1846-1853, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38284427

ABSTRACT

Combining phototherapy with other treatments has significantly advanced cancer therapy. Here, we designed and fabricated calcium-enriched carbon nanoparticles (Ca-CNPs) that could effectively deplete glutathione (GSH) and release calcium ions in tumors, thereby enhancing the efficacy of photodynamic therapy (PDT) and the calcium overload effect that leads to mitochondrial dysfunction. Due to the electrostatic interaction, π-π stacking interaction, multiple hydrogen bonds, and microporous structures, indocyanine green (ICG) was loaded onto the surface of Ca-CNPs with a high loading efficiency of 44.7 wt%. The obtained Ca-CNPs@ICG can effectively improve the photostability of ICG while retaining its ability to generate singlet oxygen (1O2) and undergo photothermal conversion (Ca-CNPs@ICG vs. ICG, 45.1% vs. 39.5%). In vitro and in vivo experiments demonstrated that Ca-CNPs@ICG could be used for near-infrared fluorescence imaging-guided synergistic calcium overload, photothermal therapy, and GSH depletion-enhanced PDT. This study sheds light on the improvement of 1O2 utilization efficiency and calcium overload-induced mitochondrial membrane potential imbalance in tumor cells.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Humans , Indocyanine Green/pharmacology , Indocyanine Green/chemistry , Calcium , Photothermal Therapy , Nanoparticles/chemistry , Neoplasms/therapy , Optical Imaging , Carbon/pharmacology
5.
Biomacromolecules ; 25(2): 964-974, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38232296

ABSTRACT

Thermosensitive nanoparticles can be activated by externally applying heat, either through laser irradiation or magnetic fields, to trigger the release of drug payloads. This controlled release mechanism ensures that drugs are specifically released at the tumor site, maximizing their effectiveness while minimizing systemic toxicity and adverse effects. However, its efficacy is limited by the low concentration of drugs at action sites, which is caused by no specific target to tumor sties. Herein, hyaluronic acid (HA), a gooey, slippery substance with CD44-targeting ability, was conjugated with a thermosensitive polymer poly(acrylamide-co-acrylonitrile) to produce tumor-targeting and thermosensitive polymeric nanocarrier (HA-P) with an upper critical solution temperature (UCST) at 45 °C, which further coloaded chemo-drug doxorubicin (DOX) and photosensitizer Indocyanine green (ICG) to prepare thermosensitive nanoreactors HA-P/DOX&ICG. With photosensitizer ICG acting as the "temperature control element", HA-P/DOX&ICG nanoparticles can respond to temperature changes when receiving near-infrared irradiation and realize subsequent structure depolymerization for burst drug release when the ambient temperature was above 45 °C, achieving programmable and on-demand drug release for effective antitumor therapy. Tumor inhibition rate increased from 61.8 to 95.9% after laser irradiation. Furthermore, the prepared HA-P/DOX&ICG nanoparticles possess imaging properties, with ICG acting as a probe, enabling real-time monitoring of drug distribution and therapeutic response, facilitating precise treatment evaluation. These results provide enlightenment for the design of active tumor targeting and NIR-triggered programmable and on-demand drug release of thermosensitive nanoreactors for tumor therapy.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Neoplasms , Humans , Photosensitizing Agents/therapeutic use , Hyperthermia, Induced/methods , Phototherapy/methods , Doxorubicin/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Indocyanine Green/pharmacology , Indocyanine Green/chemistry , Nanotechnology , Drug Liberation , Cell Line, Tumor
6.
J Colloid Interface Sci ; 657: 993-1002, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38104364

ABSTRACT

Phototherapy, encompassing photothermal therapy and photodynamic therapy, is gaining attention as an appealing cancer treatment modality. To enhance its clinical implementation, a comprehensive exploration of the pivotal factors influencing phototherapy is warranted. In this study, the L/d-cysteine (Cys)-copper ion (Cu2+) chiral nanoparticles, through the assembly of L/d-Cys-Cu2+ coordination complexes, were constructed. We found that these nanoparticles interacted with chiral liposomes in a chirality-dependent manner, with d-Cys-Cu2+ nanoparticles exhibiting more than three times stronger binding affinity than l-Cys-Cu2+ nanoparticles. Furthermore, we demonstrated that the d-Cys-Cu2+ nanoparticles were more efficiently internalized by Hela cells in contrast with l-Cys-Cu2+. On this basis, indocyanine green (ICG), acting as both photothermal and photodynamic agent, was encapsulated into L/d-Cys-Cu2+ nanoparticles. Experimental results showed that the l-Cys-Cu2+-ICG and d-Cys-Cu2+-ICG nanoparticles displayed almost identical photothermal performance and singlet oxygen (1O2) generation capability in aqueous solution. However, upon laser irradiation, the d-Cys-Cu2+-ICG nanoparticles achieved enhanced anti-tumor effects compared to l-Cys-Cu2+-ICG due to their chirality-promoted higher cellular uptake efficiency. These findings highlight the crucial role of chirality in phototherapy and provide new perspectives for engineering cancer therapeutic agents.


Subject(s)
Nanoparticles , Photochemotherapy , Humans , Copper/pharmacology , Cysteine , HeLa Cells , Phototherapy/methods , Indocyanine Green/chemistry , Nanoparticles/chemistry , Cell Line, Tumor
7.
J Control Release ; 366: 142-159, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38145660

ABSTRACT

Responsive heat resistance (by heat shock protein upregulation) and spontaneous reactive oxygen species (ROS) detoxification have been regarded as the major obstacles for photothermal/photodynamic therapy of cancer. To overcome the thermal resistance and improve ROS susceptibility in breast cancer therapy, Au ion-crosslinked hydrogels including indocyanine green (ICG) and polyphenol are devised. Au ion has been introduced for gel crosslinking (by catechol-Au3+ coordination), cellular glutathione depletion, and O2 production from cellular H2O2. ICG can generate singlet oxygen from O2 (for photodynamic therapy) and induce hyperthermia (for photothermal therapy) under the near-infrared laser exposure. (-)-Epigallocatechin gallate downregulates heat shock protein to overcome heat resistance during hyperthermia and exerts multiple anticancer functions in spite of its ironical antioxidant features. Those molecules are concinnously engaged in the hydrogel structure to offer fast gel transformation, syringe injection, self-restoration, and rheological tuning for augmented photo/chemotherapy of cancer. Intratumoral injection of multifunctional hydrogel efficiently suppressed the growth of primary breast cancer and completely eliminated the residual tumor mass. Proposed hydrogel system can be applied to tumor size reduction prior to surgery of breast cancer and the complete remission after its surgery.


Subject(s)
Breast Neoplasms , Hyperthermia, Induced , Photochemotherapy , Humans , Female , Reactive Oxygen Species/metabolism , Hydrogels/therapeutic use , Hydrogen Peroxide , Indocyanine Green/therapeutic use , Indocyanine Green/chemistry , Breast Neoplasms/drug therapy , Heat-Shock Proteins
8.
J Nanobiotechnology ; 21(1): 465, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38049882

ABSTRACT

Breast cancer treatment has been a global puzzle, and apoptosis strategies based on mitochondrial Ca2+ overload have attracted extensive attention. However, various limitations of current Ca2+ nanogenerators make it difficult to maintain effective Ca2+ overload concentrations. Here, we constructed a multimodal Ca2+ nano-modulator that, for the first time, combined photothermal therapy (PTT) and mitochondrial Ca2+ overload strategies to inhibit tumor development. By crosslinking sodium alginate (SA) on the surface of calcium carbonate (CaCO3) nanoparticles encapsulating with Cur and ICG, we prepared a synergistic Ca2+ nano-regulator SA/Cur@CaCO3-ICG (SCCI). In vitro studies have shown that SCCI further enhanced photostability while preserving the optical properties of ICG. After uptake by tumor cells, SCCI can reduce mitochondrial membrane potential and down-regulate ATP production by producing large amounts of Ca2+ at low pH. Near-infrared light radiation (NIR) laser irradiation made the tumor cells heat up sharply, which not only accelerated the decomposition of CaCO3, but also produced large amounts of reactive oxygen species (ROS) followed by cell apoptosis. In vivo studies have revealed that the Ca2+ nano-regulators had excellent targeting, biocompatibility, and anti-tumor effects, which can significantly inhibit the proliferation of tumor cells and play a direct killing effect. These findings indicated that therapeutic strategies based on ionic interference and PTT had great therapeutic potential, providing new insights into antitumor therapy.


Subject(s)
Breast Neoplasms , Nanoparticles , Photochemotherapy , Humans , Female , Breast Neoplasms/therapy , Indocyanine Green/chemistry , Phototherapy , Nanoparticles/chemistry , Homeostasis , Cell Line, Tumor
9.
ACS Appl Mater Interfaces ; 15(46): 53827-53834, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37944101

ABSTRACT

Antibacterial nanoagents with well-controlled structures are greatly desired to address the challenges of bacterial infections. In this study, a featherlike tellurium-selenium heterostructural nanoadjuvant (TeSe HNDs) was created. TeSe HNDs produced 1O2 and had high photothermal conversion efficiency when stimulated with 808 nm near-infrared (NIR) light. To create a synergistic treatment system (TeSe-ICG) with better photothermal and photodynamic capabilities, the photosensitizer indocyanine green (ICG) was then added. With a bactericidal rate of more than 99%, the NIR-mediated TeSe-ICG demonstrated an efficient bactericidal action against both Gram-negative bacteria (Escherichia coli) and Gram-positive bacteria (Staphylococcus aureus). In addition, TeSe-ICG was also effective in treating wound infections and could effectively promote wound healing without obvious toxic side effects. In conclusion, TeSe-ICG is expected to be a good candidate for the treatment of bacterial infections.


Subject(s)
Photochemotherapy , Selenium , Staphylococcal Infections , Humans , Selenium/pharmacology , Tellurium/pharmacology , Phototherapy , Indocyanine Green/chemistry , Escherichia coli , Anti-Bacterial Agents/pharmacology
10.
Nanoscale ; 15(42): 16947-16958, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37779508

ABSTRACT

Breast cancer has emerged as a leading cause of mortality among women. Photothermal therapy represents a recent therapeutic modality for eradicating localized tumors, albeit hindered by its limited penetration into tumor tissues. Recognizing the potential of photothermal therapy to induce immunogenic cell death in tumor cells, we explored a gene delivery approach utilizing small interfering RNA targeting programmed death ligand 1 (PD-L1), abbreviated as siPD-L1, to bolster the anti-tumor immune response elicited by this therapy. Nonetheless, the suboptimal release efficiency and inherent instability of RNA molecules have posed challenges to their therapeutic efficacy. In this study, we designed a glutathione (GSH)/pH-responsive micelle system, employing biocompatible and low-toxicity polyethyleneimine in conjunction with structurally robust pluronic P123, to encapsulate both indocyanine green (ICG) and siPD-L1 for precise targeting in breast cancer treatment. The resulting PSP/ICG/siPD-L1 nanocarrier demonstrated admirable biocompatibility and stability. Upon internalization into tumor cells, this nanocarrier exhibited rapid release of both ICG and siPD-L1, responding to the acidic tumor microenvironment and GSH conditions. The inclusion of siPD-L1 effectively downregulated the expression of PD-L1 on the tumor cell surface, thereby impeding tumor growth. Additionally, ICG demonstrated a photothermal effect when exposed to near-infrared light. Both in vitro and in vivo investigations substantiated the nanocarrier's efficacy against tumor cells, culminating in the complete ablation of 4T1 tumors in situ. Consequently, PSP/ICG/siPD-L1 emerges as a promising nanocarrier candidate for augmenting anti-tumor immunity through the synergistic combination of photothermal therapy and gene-based intervention.


Subject(s)
Breast Neoplasms , Hyperthermia, Induced , Nanoparticles , Female , Humans , B7-H1 Antigen , Phototherapy/methods , Hyperthermia, Induced/methods , Drug Delivery Systems/methods , Indocyanine Green/pharmacology , Indocyanine Green/chemistry , Breast Neoplasms/therapy , Immunotherapy , Glutathione , Hydrogen-Ion Concentration , Cell Line, Tumor , Nanoparticles/chemistry , Tumor Microenvironment
11.
ACS Appl Mater Interfaces ; 15(29): 34617-34630, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37437265

ABSTRACT

Indocyanine green (ICG) has been widely explored for the theranostics of tumors. However, ICG mainly accumulates in the liver, spleen, or kidney in addition to in tumors, causing inaccurate diagnoses and impaired therapeutic effects under NIR irradiation. Herein, a hybrid nanomicelle was constructed by integrating hypoxia-sensitive iridium(III) and ICG for precise tumor localization and photothermal therapy in sequence. In this nanomicelle, the amphiphilic iridium(III) complex (BTPH)2Ir(SA-PEG) was synthesized through the coordination substitution of hydrophobic (BTPH)2IrCl2 and hydrophilic PEGlyated succinylacetone (SA-PEG). Meanwhile, PEGlyated ICG (ICG-PEG) as a derivative of the photosensitizer ICG was also synthesized. (BTPH)2Ir(SA-PEG) and ICG-PEG were coassembled by dialysis to form the hybrid nanomicelle M-Ir-ICG. Hypoxia-sensitive fluorescence, ROS generation, and the photothermal effect of M-Ir-ICG were investigated in vitro and in vivo. The experimental results indicated that M-Ir-ICG nanomicelles could locate at the tumor site first and then perform photothermal therapy with 83.90% TIR, demonstrating great potential for clinical applications.


Subject(s)
Colonic Neoplasms , Nanoparticles , Humans , Indocyanine Green/chemistry , Iridium , Phototherapy/methods , Photosensitizing Agents , Cell Line, Tumor
12.
Adv Healthc Mater ; 12(27): e2300929, 2023 10.
Article in English | MEDLINE | ID: mdl-37300324

ABSTRACT

The purpose of this study is to down-regulate heat shock proteins and improve the mild photothermal therapy (mild-PTT) effect of polydopamine (PDA) by preparing the nanosystem of Cu2+ and indocyanine green (ICG)-loaded PDA nanospheres with surface modification of integrin-targeted cyclic peptide (cRGD) (PDA/Cu/ICG/R), which can limit ATP synthesis through the double mitochondrial destruction pathway. In vitro and in vivo experiments using PDA/Cu/ICG/R irradiated with an NIR laser demonstrate that when NIR is "OFF," Cu2+ can undergo Fenton-like reaction in tumor cells, producing a large amount of hydroxyl radicals (·OH), which leads to oxidative stress in cells. This oxidative stress can cause mitochondrial oxidative phosphorylation dysfunction, resulting in limited ATP synthesis. When NIR is "ON," mild-PTT can accelerate Cu2+ to produce ·OH. Simultaneously, NIR can activate ICG to produce reactive oxygen species (ROS) storm, amplify intracellular oxidative stress, and continuously damage mitochondria. The biodegradability of PDA greatly reduces the risk of toxicity caused by long-term retention of PDA/Cu/ICG/R in organisms. Finally, the improvement of the mild-PTT effect of PDA is successfully achieved through the double mitochondrial destruction pathway of Cu2+ and ICG controlled by NIR "switch."


Subject(s)
Indocyanine Green , Nanoparticles , Indocyanine Green/pharmacology , Indocyanine Green/chemistry , Photothermal Therapy , Down-Regulation , Adenosine Triphosphate , Nanoparticles/chemistry , Cell Line, Tumor , Phototherapy
13.
Angew Chem Int Ed Engl ; 62(28): e202305564, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37162307

ABSTRACT

Indocyanine green (ICG) is the only near-infrared (NIR) dye approved for clinical use. Despite its versatility in photonic applications and potential for photothermal therapy, its photobleaching hinders its application. Here we discovered a nanostructure of dimeric ICG (Nano-dICG) generated by using ICG to stabilize nanoemulsions, after which ICG enabled complete dimerization on the nanoemulsion shell, followed by J-aggregation of ICG-dimer, resulting in a narrow, red-shifted (780 nm→894 nm) and intense (≈2-fold) absorbance. Compared to ICG, Nano-dICG demonstrated superior photothermal conversion (2-fold higher), significantly reduced photodegradation (-9.6 % vs. -46.3 %), and undiminished photothermal effect (7 vs. 2 cycles) under repeated irradiations, in addition to excellent colloidal and structural stabilities. Following intravenous injection, Nano-dICG enabled real-time tracking of its delivery to mouse tumors within 24 h by photoacoustic imaging at NIR wavelength (890 nm) distinct from the endogenous signal to guide effective photothermal therapy. The unprecedented finding of nanostructure-driven ICG dimerization leads to an ultra-stable phototheranostic platform.


Subject(s)
Nanoparticles , Nanostructures , Mice , Animals , Indocyanine Green/chemistry , Dimerization , Nanoparticles/chemistry , Nanostructures/therapeutic use , Nanostructures/chemistry , Polymers , Phototherapy/methods , Cell Line, Tumor
14.
Int J Nanomedicine ; 18: 323-337, 2023.
Article in English | MEDLINE | ID: mdl-36700147

ABSTRACT

Background: Multifunctional stimuli-responsive nanoparticles with photothermal-chemotherapy provided a powerful tool for improving the accuracy and efficiency in the treatment of malignant tumors. Methods: Herein, photosensitizer indocyanine green (ICG)-loaded amorphous calcium-carbonate (ICG@) nanoparticle was prepared by a gas diffusion reaction. Doxorubicin (DOX) and ICG@ were simultaneously encapsulated into poly(lactic-co-glycolic acid)-ss-chondroitin sulfate A (PSC) nanoparticles by a film hydration method. The obtained PSC/ICG@+DOX hybrid nanoparticles were characterized and evaluated by Fourier transform infrared spectroscopy (FTIR), dynamic light scattering (DLS), transmission electron microscopy (TEM), and differential scanning calorimetry (DSC). The cellular uptake and cytotoxicity of PSC/ICG@+DOX nanoparticles were analyzed by confocal laser scanning microscopy (CLSM) and MTT assay in 4T1 cells. In vivo antitumor activity of the nanoparticles was evaluated in 4T1-bearing Balb/c mice. Results: PSC/ICG@+DOX nanoparticles were nearly spherical in shape by TEM observation, and the diameter was 407 nm determined by DLS. Owing to calcium carbonate and disulfide bond linked copolymer, PSC/ICG@+DOX nanoparticles exhibited pH and reduction-sensitive drug release. Further, PSC/ICG@+DOX nanoparticles showed an effective photothermal effect under near-infrared (NIR) laser irradiation, and improved cellular uptake and cytotoxicity in breast cancer 4T1 cells. Importantly, PSC/ICG@+DOX nanoparticles demonstrated the most effective suppression of tumor growth in orthotopic 4T1-bearing mice among the treatment groups. In contrast with single chemotherapy or photothermal therapy, chemo-photothermal treatment by PSC/ICG@+DOX nanoparticles synergistically inhibited the growth of 4T1 cells. Conclusion: This study demonstrated that PSC/ICG@+DOX nanoparticles with active targeting and stimuli-sensitivity would be a promising strategy to enhance chemo-photothermal cancer therapy.


Subject(s)
Hyperthermia, Induced , Multifunctional Nanoparticles , Nanoparticles , Neoplasms , Animals , Mice , Indocyanine Green/chemistry , Photothermal Therapy , Phototherapy/methods , Hyperthermia, Induced/methods , Doxorubicin , Neoplasms/drug therapy , Nanoparticles/chemistry , Cell Line, Tumor
15.
Nanoscale ; 15(4): 1925-1936, 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36625142

ABSTRACT

A simple yet effective strategy to enhance the properties of traditional dye indocyanine green (ICG) in all aspects was proposed and demonstrated. Specifically, indocyanine green-derived carbon dots (ICGCDs) were synthesized from ICG via a simple hydrothermal treatment. The ICGCDs exhibited significantly enhanced thermal stability and anti-photobleaching compared to ICG. Furthermore, their photothermal properties were also notably strengthened, in which a wider functional pH range, 50% improvement in photothermal conversion efficiency and superior photothermal cyclability were achieved. Thanks to these superior properties, ICGCDs were demonstrated as efficient NIR bioimaging and photothermal agents in both in vitro and in vivo experiments. Most excitingly, the strategy demonstrated in this study is likely to have broad applications in other systems.


Subject(s)
Indocyanine Green , Photothermal Therapy , Indocyanine Green/pharmacology , Indocyanine Green/chemistry , Carbon , Phototherapy , Cell Line, Tumor
16.
J Mater Chem B ; 11(1): 119-130, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36504220

ABSTRACT

Intracellular-synthesized chemo-drugs based on the inherent characteristics of the tumor microenvironment (TME) have been extensively applied in oncotherapy. However, combining other therapeutic strategies to convert nontoxic small molecules into toxic small-molecule chemo-drugs in the TME is still a huge challenge. To address this issue, herein we have developed a biomimetic dual-responsive bioengineered nanotheranostics system via the supramolecular co-assembly of the nontoxic small-molecule 1,5-dihydroxynaphthalene (DHN) and small-molecule photosensitizer indocyanine green (ICG) followed by surface cloaking through red blood cell membranes (RBCs) for intracellular cascade-synthesizing chemo-drugs and efficient oncotherapy. Such nanotheranostics with a suitable diameter, core-shell structure, ultrahigh dual-drug payload rate, and excellent stability can efficiently accumulate in tumor regions and then internalize into tumor cells. Under the dual stimulations of near-infrared laser irradiation and acidic lysosomes, the nanotheranostics system exhibited exceptional instability under heat-primed membrane rupture and pH decrease, thereby achieving rapid disassembly and on-demand drug release. Furthermore, the released ICG can efficiently convert 3O2 into 1O2. After that, the generated 1O2 can efficiently oxidize the released nontoxic DHN into the highly toxic chemo-drug juglone, thereby realizing intracellular cascade-synthesizing chemo-drugs and synergistic photodynamic-chemotherapy while reducing detrimental side effects on normal cells or tissues. Overall, it is envisioned that RBC-cloaked nanotheranostics with intracellular cascade-synthesizing chemo-drugs can provide a promising strategy for intracellular chemo-drug synthesis-based oncotherapy.


Subject(s)
Antineoplastic Agents , Biomimetics , Theranostic Nanomedicine , Antineoplastic Agents/pharmacology , Phototherapy , Photosensitizing Agents/chemistry , Indocyanine Green/pharmacology , Indocyanine Green/chemistry
17.
ACS Appl Mater Interfaces ; 14(51): 56597-56612, 2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36512413

ABSTRACT

Although phototherapy has attracted extensive attention in antitumor field in recent years, its therapeutic effect is usually unsatisfactory because of the complexity and variability of the tumor microenvironment (TME). Herein, we report novel CoSn(OH)6@CoOOH hollow carriers with oxidase properties that can enhance phototherapy. Hollow CoSn(OH)6@CoOOH nanocubes (NCs) with a particle size of ∼160 nm were synthesized via a two-step process of coprecipitation and etching. These NCs can react with O2 to generate singlet oxygen without hydrogen peroxide and consume glutathione, and their hollow structure can be utilized to carry drug molecules. After loading indocyanine green (ICG) and 1,2-bis(2-(4,5-dihydro-1H-imidazol-2-yl)propan-2-yl) diazene dihydrochloride (AIPH), the resulting nanosystem (HCIA) exhibited enhanced phototherapy effects through the catalytic activity of oxidase, production of alkyl radicals, and consumption of glutathione. Cell and mouse experiments showed that HCIA combined with near-infrared laser irradiation significantly inhibited the growth of 4T1 tumors. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that PI3K-Akt and MAPK signaling pathways were highly relevant to this therapeutic system. Such hollow NCs with oxidase activity have considerable potential for the design of multifunctional drug delivery vehicles for tumor therapy.


Subject(s)
Nanoparticles , Neoplasms , Mice , Animals , Phosphatidylinositol 3-Kinases , Phototherapy/methods , Neoplasms/drug therapy , Neoplasms/pathology , Indocyanine Green/pharmacology , Indocyanine Green/therapeutic use , Indocyanine Green/chemistry , Oxidoreductases/therapeutic use , Cell Line, Tumor , Nanoparticles/chemistry , Tumor Microenvironment
18.
Biomater Sci ; 11(1): 248-262, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36440665

ABSTRACT

Multimodality imaging-navigated precise phototherapy has been well-established as a promising strategy for enhancing the diagnostic and therapeutic efficiency of cancer in preclinical trials. However, proper theranostic agents with adequate biosafety and biological efficacy as well as simple components and preparations are still in great demand to promote the clinical translation of this regimen. Here, we developed a multifunctional nanosystem based on the self-assembly of FDA-approved indocyanine green (ICG) and 125I-labeled glycopeptides, which were composed of FDA-approved natural polysaccharide sodium alginate and endogenous tyrosine, for fluorescence imaging/single photon emission computed tomography (FLI/SPECT)-guided synergistic photothermal/photodynamic therapy (PTT/PDT) of breast cancer. The as-prepared ICG@ADY(125I) NPs possessed a stable nanostructure and radiolabel, an ICG-equivalent ROS and hyperthermia generation property, and a preferable photo/photothermal stability and biocompatibility, favoring its tumor homing, multimodality imaging, and phototherapy with high biosafety. Consequently, ICG@ADY(125I) NPs smoothly accumulated in tumors by virtue of their long blood circulation (t1/2 = 15.76 ± 1.34 h) and the EPR effect, thereby presenting highly sensitive FLI/SPECT images to realize cancer diagnosis. Guided by multimodality imaging, accurate PTT/PDT was performed using NIR laser irradiation, achieving a high tumor inhibition rate (81.8%) against 4T1 breast cancer models without appreciable side effects. Altogether, this theranostic nanosystem may have huge potential for the clinical diagnosis and treatment of breast cancer.


Subject(s)
Breast Neoplasms , Nanoparticles , Humans , Female , Indocyanine Green/chemistry , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Precision Medicine , Nanoparticles/chemistry , Phototherapy/methods , Theranostic Nanomedicine , Cell Line, Tumor
19.
Int J Mol Sci ; 23(18)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36142205

ABSTRACT

Despite its common side effects and varying degrees of therapeutic success, chemotherapy remains the gold standard method for treatment of cancer. Towards developing a new therapeutic approach, we have engineered nanoparticles derived from erythrocytes that contain indocyanine green as a photo-activated agent that enables near infrared photothermal heating, and doxorubicin hydrochloride (DOX) as a chemotherapeutic drug. We hypothesize that milliseconds pulsed laser irradiation results in rapid heating and photo-triggered release of DOX, providing a dual photo-chemo therapeutic mechanism for tumor destruction. Additionally, the surface of the nanoparticles is functionalized with folate to target the folate receptor-α on tumor cells to further enhance the therapeutic efficacy. Using non-contract infrared radiometry and absorption spectroscopy, we have characterized the photothermal response and photostability of the nanoparticles to pulsed laser irradiation. Our in vitro studies show that these nanoparticles can mediate photo-chemo killing of SKOV3 ovarian cancer cells when activated by pulsed laser irradiation. We further demonstrate that this dual photo-chemo therapeutic approach is effective in reducing the volume of tumor implants in mice and elicits an apoptotic response. This treatment modality presents a promising approach in destruction of small tumor nodules.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Neoplasms , Animals , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Erythrocytes/pathology , Folic Acid/chemistry , Indocyanine Green/chemistry , Lasers , Mice , Nanoparticles/chemistry , Neoplasms/pathology , Phototherapy
20.
Biomacromolecules ; 23(10): 4308-4317, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36146942

ABSTRACT

As a class of biocompatible and biodegradable naturally derived nanomaterials, cellulose nanocrystals (CNCs) with diverse surface functionalization have aroused considerable attention for a range of biomedical applications in drug or gene delivery, as a fluorescent nanoprobe, in cancer targeting, and in photothermal cancer therapy, among others. Herein, we construct the copolymer-functionalized CNCs as a pH- and near-infrared (NIR)-triggered drug carrier for simultaneous photothermal therapy and chemotherapy of cancer cells. Poly(ε-caprolactone)-b-poly(2-(dimethylamino)ethyl methacrylate) (PCL-b-PDMAEMA) was conjugated onto the surface of CNCs through ring-opening polymerization, followed by activators regenerated by electron transfer atom transfer radical polymerization (ARGET ATRP). The resultant CNC-based drug carrier can encapsulate doxorubicin (DOX) as a therapeutic agent and indocyanine green (ICG) as an NIR dye in the PCL core and the PDMAEMA shell, respectively, via hydrophobic and electrostatic interactions. In addition to the intrinsic pH response, the release profile of DOX can also be controlled by the duration of laser irradiation due to collapse of the crystal structure of the PCL domain with the increase of temperature induced by photothermal conversion. The drug carrier can exhibit enhanced cytotoxicity toward HepG2, human hepatocyte carcinoma, cells upon laser irradiation, which can be attributed to the synergistic effect arising from NIR-triggered burst release of DOX and photothermal heating. The rod-like morphology of the CNC-based drug carrier may help accelerate the endocytosis in cell membranes compared with its common spherical counterpart. Based on the abovementioned advantages, copolymer-functionalized CNCs can serve as a promising candidate for effective cancer treatment.


Subject(s)
Nanoparticles , Neoplasms , Cellulose/metabolism , Doxorubicin/chemistry , Drug Carriers , Drug Liberation , Humans , Hydrogen-Ion Concentration , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Methacrylates , Nanoparticles/chemistry , Neoplasms/drug therapy , Nylons , Phototherapy , Photothermal Therapy , Polymers/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL