Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Adv Healthc Mater ; 13(5): e2302564, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38073257

ABSTRACT

Multidrug resistance (MDR) limits the application of clinical chemotherapeutic drugs. There is an urgent need to develop non-apoptosis-inducing agents that circumvent drug resistance. Herein, four therapeutic copper complexes encapsulated in natural nanocarrier apoferritin (AFt-Cu1-4) are reported. Although they are isomers, they exhibit significantly different organelle distributions and cell death mechanisms. AFt-Cu1 and AFt-Cu3 accumulate in the cytoplasm and induce autophagy, whereas AFt-Cu2 and AFt-Cu4 can quickly enter the nucleus and trigger oncosis. Excitedly, AFt-Cu2 and AFt-Cu4 show a strong tumor growth inhibition effect in mice models bearing multidrug-resistant colon xenograft via intravenous injection. To the best of the authors' knowledge, this is the first example of metal-based nucleus-targeted oncosis inducers overcoming multidrug resistance in vivo.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Nanoparticles , Humans , Mice , Animals , Copper/pharmacology , Apoferritins , Drug Resistance, Multiple , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Antineoplastic Agents/pharmacology
2.
Chem Sci ; 14(25): 7005-7015, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37389267

ABSTRACT

Drug resistance and metastases are the leading causes of death in clinics. To overcome this limitation, there is an urgent need for new therapeutic agents and drug formulations that are able to therapeutically intervene by non-traditional mechanisms. Herein, the physical adsorption and oxidative polymerization of Pt(iv) prodrugs in pore-confined spaces of CaCO3 nanoparticles is presented, and the nanomaterial surface was coated with DSPE-PEG2000-Biotin to improve aqueous solubility and tumor targeting. While the nanoparticle scaffold remained stable in an aqueous solution, it quickly degraded into Ca2+ in the presence of acid and into cisplatin in the presence of GSH. The nanoparticles were found to interact in cisplatin-resistant non-small lung cancer cells by a multimodal mechanism of action involving mitochondrial Ca2+ overload, dual depletion of GSH, nuclear DNA platination, and amplification of ROS and lipid peroxide generation, resulting in triggering cell death by a combination of apoptosis, ferroptosis and immunogenic cell death in vitro and in vivo. This study could present a novel strategy for the treatment of drug-resistant and metastatic tumors and therefore overcome the limitations of currently used therapeutic agents in the clinics.

3.
Chem Commun (Camb) ; 59(46): 6956-6968, 2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37184685

ABSTRACT

Metal complexes have shown promise as photosensitizers for cancer diagnosis and therapeutics. However, the vast majority of metal photosensitizers are not ideal and associated with several limitations including pharmacokinetic limitations, off-target toxicity, fast systemic clearance, poor membrane permeability, and hypoxic tumour microenvironments. Metal complex functionalized nanomaterials have the potential to construct multifunctional systems, which not only overcome the above defects of metal complexes but are also conducive to modulating the tumour microenvironment (TME) and employing combination therapies to boost photodynamic therapy (PDT) efficacy. In this review, we first introduce the current challenges of photodynamic therapy and summarize the recent research strategies (such as metal coordination bonds, self-assembly, π-π stacking, physisorption, and so on) used for preparing metal complexes functionalized nanomaterials in the application of PDT.


Subject(s)
Coordination Complexes , Nanostructures , Neoplasms , Photochemotherapy , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Coordination Complexes/chemistry , Nanostructures/chemistry , Combined Modality Therapy , Neoplasms/pathology , Tumor Microenvironment
4.
Dalton Trans ; 52(21): 6978-6986, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37067849

ABSTRACT

Four novel PSs (photosensitizers) of nitrogen-heterocyclic ruthenium polypyridyl complexes Ru(dip)2(o-pipppz)(PF6)2 (Ru1) (dip = 4,7-diphenyl-1,10-phenanthroline; o-pipppz = 1-(4-aldehydephenyl)-3-(pyridazyl-2-yl)-1H-pyrazole), Ru(dip)2(o-pipp) (PF6)2 (Ru2) (o-pipp = 1-(4-aldehydephenyl)-3-(pyrid-2-yl)-1H-pyrazole), Ru(dip)2(m-pipp)(PF6)2 (Ru3) (m-pipp = 1-(4-aldehydephenyl)-3-(pyrid-3-yl)-1H-pyrazole) and Ru(dip)2(p-pipp)(PF6)2 (Ru4) (p-pipp = 1-(4-aldehydephenyl)-3-(pyrid-4-yl)-1H-pyrazole) were reported, and the photodynamic activities of these complexes were studied on 2D and 3D HeLa cancer models. The longest visible absorption wavelength of these complexes was approximately 622 nm. The four Ru(II) complexes show preferable photodynamic activity and low dark toxicity (0.2-0.4 µM) in vitro against 2D HeLa tumor cells. These complexes exhibit very high singlet oxygen quantum yields in methanol (0.70-0.95), TPA cross-sections (7-31 GM), and high penetration depth. Thus, Ru1-Ru4 were utilized as one-photon and two-photon absorbing photosensitizers in both monolayer cells and 3D multicellular spheroids (MCSs). Among them, Ru2 revealed a higher singlet oxygen yield (0.95), a larger TPA cross-section (31 GM), and the strongest phototoxicity (EC50 = 0.20 µM). Moreover, flow cytometry shows that the four Ru(II) complexes can induced cell death mainly through apoptosis upon singlet oxygen-dependent reaction.


Subject(s)
Photochemotherapy , Ruthenium , Humans , Photosensitizing Agents/pharmacology , Ruthenium/pharmacology , Singlet Oxygen , Photons
5.
Chem Sci ; 14(6): 1461-1471, 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36794192

ABSTRACT

Conventional photodynamic therapy mainly causes a therapeutic effect on the primary tumor through the localized generation of reactive oxygen species, while metastatic tumors remain poorly affected. Complementary immunotherapy is effective in eliminating small, non-localized tumors distributed across multiple organs. Here, we report the Ir(iii) complex Ir-pbt-Bpa as a highly potent immunogenic cell death inducing photosensitizer for two-photon photodynamic immunotherapy against melanoma. Ir-pbt-Bpa can produce singlet oxygen and superoxide anion radicals upon light irradiation, causing cell death by a combination of ferroptosis and immunogenic cell death. In a mouse model with two physically separated melanoma tumors, although only one of the primary tumors was irradiated, a strong tumor reduction of both tumors was observed. Upon irradiation, Ir-pbt-Bpa not only induced the immune response of CD8+ T cells and the depletion of regulatory T cells, but also caused an increase in the number of the effector memory T cells to achieve long-term anti-tumor immunity.

6.
Small Methods ; 7(5): e2201403, 2023 05.
Article in English | MEDLINE | ID: mdl-36549671

ABSTRACT

Cancer ranks as a leading cause of death. There is an urgent need to develop minimally invasive methods to eradicate tumors and prevent their recurrence. As a light-driven modality, photodynamic therapy takes advantage of high tumor selectivity and low normal tissue damage. However, it shows poor potential for preventing tumor recurrence. Immunotherapy is currently being used as an alternative treatment for the control of malignant diseases. Although immunotherapy can establish long-time immune memory and efficiently protects treated patients from cancer relapse, its clinical efficacy is limited by the minority of patients' responding rate. Recently, photodynamic immunotherapy, which utilizes photosensitizers as an immunotherapy trigger to exert synergistic effects of photodynamic therapy and tumor immunotherapy, has attracted considerable interest. Like all the newly proposed treatments, there is still room for improvement. In this mini review, the progress in photodynamic immunotherapy with metal-based photosensitizers is summarized. It is hoped that this review can give a broad update on photodynamic immunotherapy and inspire readers.


Subject(s)
Neoplasms , Photochemotherapy , Humans , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Neoplasms/drug therapy , Immunotherapy , Treatment Outcome
7.
Angew Chem Int Ed Engl ; 61(28): e202205429, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35532958

ABSTRACT

The clinical application of photodynamic therapy is hindered by the high glutathione concentration, poor cancer-targeting properties, poor drug loading into delivery systems, and an inefficient activation of the cell death machinery in cancer cells. To overcome these limitations, herein, the formulation of a promising IrIII complex into a biodegradable coordination polymer (IrS NPs) is presented. The nanoparticles were found to remain stable under physiological conditions but deplete glutathione and disintegrate into the monomeric metal complexes in the tumor microenvironment, causing an enhanced therapeutic effect. The nanoparticles were found to selectively accumulate in the mitochondria where these trigger cell death by hybrid apoptosis and ferroptosis pathways through the photoinduced production of singlet oxygen and superoxide anion radicals. This study presents the first example of a coordination polymer that can efficiently cause cancer cell death by apoptosis and ferroptosis upon irradiation, providing an innovative approach for cancer therapy.


Subject(s)
Coordination Complexes , Ferroptosis , Photochemotherapy , Apoptosis , Cell Line, Tumor , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Glutathione , Iridium/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Polymers/pharmacology
8.
J Am Chem Soc ; 144(9): 4091-4101, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35171598

ABSTRACT

Despite the clinical success of photodynamic therapy (PDT), the application of this medical technique is intrinsically limited by the low oxygen concentrations found in cancer tumors, hampering the production of therapeutically necessary singlet oxygen (1O2). To overcome this limitation, we report on a novel mitochondria-localized iridium(III) endoperoxide prodrug (2-O-IrAn), which, upon two-photon irradiation in NIR, synergistically releases a highly cytotoxic iridium(III) complex (2-IrAn), singlet oxygen, and an alkoxy radical. 2-O-IrAn was found to be highly (photo-)toxic in hypoxic tumor cells and multicellular tumor spheroids (MCTS) in the nanomolar range. To provide cancer selectivity and improve the pharmacological properties of 2-O-IrAn, it was encapsulated into a biotin-functionalized polymer. The generated nanoparticles were found to nearly fully eradicate the tumor inside a mouse model within a single treatment. This study presents, to the best of our knowledge, the first example of an iridium(III)-based endoperoxide prodrug for synergistic photodynamic therapy/photoactivated chemotherapy, opening up new avenues for the treatment of hypoxic tumors.


Subject(s)
Neoplasms , Photochemotherapy , Prodrugs , Animals , Cell Line, Tumor , Hypoxia/drug therapy , Iridium/pharmacology , Mice , Mitochondria , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Singlet Oxygen/therapeutic use
9.
ACS Appl Mater Interfaces ; 13(33): 38959-38968, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34379404

ABSTRACT

Chemotherapy continues to be the most commonly applied strategy for cancer. Despite the impressive clinical success obtained with several drugs, increasing numbers of (multi)drug-resistant tumors are reported. To overcome this shortcoming, novel drug candidates and delivery systems are urgently needed. Herein, a therapeutic copper polypyridine complex encapsulated in natural nanocarrier apoferritin is reported. The generated nanoparticles showed higher cytotoxicity toward various (drug-resistant) cancer cell lines than noncancerous cells. The study of the mechanism revealed that the compound triggers cell autophagy-dependent apoptosis. Promisingly, upon injection of the nanodrug conjugate into the bloodstream of a mouse model bearing a multidrug-resistant colon tumor, a strong tumor growth inhibition effect was observed. To date, this is the first study describing the encapsulation of a copper complex in apoferritin that acts by autophagy-dependent apoptosis.


Subject(s)
Antineoplastic Agents/chemistry , Apoferritins/chemistry , Colonic Neoplasms/drug therapy , Coordination Complexes/chemistry , Copper/chemistry , Nanocapsules/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoferritins/metabolism , Autophagic Cell Death/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Membrane Permeability , Coordination Complexes/pharmacology , Drug Compounding , Drug Liberation , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice, Inbred BALB C , Neoplasms, Experimental
10.
Biomaterials ; 276: 121064, 2021 09.
Article in English | MEDLINE | ID: mdl-34391019

ABSTRACT

The photodynamic therapy (PDT) of cancer is limited by tumor hypoxia as PDT efficiency depends on O2 concentration. A novel oxygen self-sufficient photosensitizer (Ru-g-C3N4) was therefore designed and synthesized via a facile one-pot method in order to overcome tumor hypoxia-induced PDT resistance. The photosensitizer is based on [Ru(bpy)2]2+ coordinated to g-C3N4 nanosheets by Ru-N bonding. Compared to pure g-C3N4, the resulting nanosheets exhibit increased water solubility, stronger visible light absorption, and enhanced biocompatibility. Once Ru-g-C3N4 is taken up by hypoxic tumor cells and exposed to visible light, the nanosheets not only catalyze the decomposition of H2O2 and H2O to generate O2, but also catalyze H2O2 and O2 concurrently to produce multiple ROS (•OH, •O2-, and 1O2). In addition, Ru-g-C3N4 affords luminescence imaging, while continuously generating O2 to alleviate hypoxia greatly improving PDT efficacy. To the best of our knowledge, this oxygen self-sufficient photosensitizer produced via grafting a metal complex onto g-C3N4 is the first of its type to be reported.


Subject(s)
Photochemotherapy , Ruthenium , Graphite , Humans , Hydrogen Peroxide , Hypoxia/drug therapy , Nitrogen Compounds , Oxygen , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species
11.
Nanoscale ; 13(16): 7590-7599, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33884385

ABSTRACT

Photodynamic therapy (PDT) is a promising noninvasive cancer treatment. PDT in the clinic faces several hurdles due to the unique tumor environment, a feature of which is high levels of glutathione (GSH). An excess amount of GSH consumes reactive oxygen species (ROS) generated by photosensitizers (PSs), reducing PDT efficiency. Herein, nano-photosensitizers (RuS1 NPs and RuS2 NPs) are reported. These consist of ruthenium complexes joined by disulfide bonds forming GSH sensitive polymer nanoparticles. The NPs achieve enhanced uptake compared to their constituent monomers. Inside cancer cells, high levels of GSH break the S-S bonds releasing PS molecules in the cell. The level of GSH is also then reduced leading to excellent PDT activity. Furthermore, RuS2 NPs functionalized with tumor targeting hyaluronic acid (HA@RuS2 NPs) assessed in vivo were highly effective with minimal side effects. To the best of our knowledge, RuS NPs are the first metal complex-based nano-assembled photosensitizers which exhibit enhanced specificity and consume endogenous GSH simultaneously, thus achieving excellent two-photon PDT efficiency in vitro and in vivo.


Subject(s)
Nanoparticles , Photochemotherapy , Photosensitizing Agents , Ruthenium , Cell Line, Tumor , Glutathione , Humans , Photosensitizing Agents/pharmacology
12.
Chem Commun (Camb) ; 57(14): 1790-1793, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33475648

ABSTRACT

A Ru(ii)-BODIPY conjugate has been rationally designed and exhibits an intense absorption in the NIR region to boost lysosome-targeted PDT in vitro and in vivo. The advantages of Ru(ii) and BODIPY were successfully instilled into the conjugate to yield highly effective PDT efficacy against malignant melanoma A375 cells (PI = 3448) and A375 mice xenografts.


Subject(s)
Boron Compounds/chemistry , Lysosomes/radiation effects , Photochemotherapy , Ruthenium Compounds/chemistry , Animals , Cell Line, Tumor , Humans , Infrared Rays , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/therapy , Photosensitizing Agents , Xenograft Model Antitumor Assays
13.
ACS Appl Mater Interfaces ; 10(21): 18036-18049, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29745229

ABSTRACT

Design and development of photosensitizers that can efficiently convert energy of near-infrared (NIR) laser irradiation are of major importance for cancer photoassisted therapeutics. Herein, for the first time, it is demonstrated that Prussian blue (PB), a classic coordination compound, can act as a novel photosensitizer with efficient generation of singlet oxygen and excellent photothermal conversion via NIR photoirradiation-induced energy transfer. After modification with hyaluronic acid (HA), the as-prepared HA-modified PB nanocubes (HA@PB) are highly dispersible in aqueous and physiological solutions, as well as show excellent photothermal/photodynamic activities under NIR (808 nm) photoexcitation. On the basis of these features, HA@PB is used to study their in vitro and in vivo combined therapeutic effect. Owing to the CD44 ligand of HA, HA@PB have specific uptake by CD44-positive cells in vitro and can be precisely in vivo delivered to the tumor site. HA@PB as one of the synergistically photodynamic/photothermal combination nanoplatforms could achieve excellent therapeutic efficacy with targeted specificity under the guidance of dual-modality photoacoustic/infrared thermal imaging. Hence, this work is expected to pave the way for using PB-based nanomaterials as a promising multifunctional theranostic nanoplatform in biomedical fields.

SELECTION OF CITATIONS
SEARCH DETAIL
...