Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 428
Filter
1.
J Inorg Biochem ; 251: 112427, 2024 02.
Article in English | MEDLINE | ID: mdl-37979498

ABSTRACT

Breast cancer is the most commonly diagnosed cancer and second­leading cause of cancer deaths in women. Signal transducer and activator of transcription 3 (STAT3) plays a critical role in promoting breast cancer cell proliferation, invasion, angiogenesis, and metastasis, and the high expression of STAT3 is related to the occurrence and poor chemotherapy sensitivity of breast cancer. Iridium(III) complexes Ir-PTS-1- 4 containing a pterostilbene-derived ligand were synthesized to inhibit the STAT3 pathway in breast cancer. Ir-PTS-4 inhibited the proliferation of breast cancer cells by suppressing the expression of phosphorylated STAT3 and STAT3-related cyclin D1, arresting cell cycle in the S-phase, inducing DNA damage and reactive oxygen species (ROS) generation, eventually leading to autophagic cell death. The cell metastasis and invasion were also inhibited after Ir-PTS-4 treatment. Besides, Ir-PTS-4 exhibited excellent anti-proliferation activity in 3D multicellular tumor spheroids, showing potential for the treatment of solid tumors. This work presents the rational design of metal-based anticancer agents to block the STAT3 pathway for simultaneously inhibiting breast cancer proliferation and metastasis.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Iridium , Female , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Iridium/pharmacology , Iridium/therapeutic use , STAT3 Transcription Factor/metabolism
2.
Acta Biomater ; 172: 369-381, 2023 12.
Article in English | MEDLINE | ID: mdl-37852456

ABSTRACT

Cancer stem cells (CSCs) are found in many solid tumors, which play decisive roles in the occurrence, recurrence and metastasis of tumors. However, drugs are difficult to kill CSCs due to their limited number and location in oxygen-deprived tissue far from the blood vessels. Meanwhile, the survival and stemness maintenance of CSCs strongly depend on the tumor microenvironment (TME). Herein, we developed a CD44 antibody modified iridium nanosheet with enzyme-like activity (defined as Ir Nts-Ab) that effectively eradicates CSCs for cancer therapy. We observe that Ir Nts-Ab can enrich tumor tissues to remove excessive reactive oxygen species and produce oxygen, thus alleviating hypoxia and the inflammatory TME to reduce the proportion of CSCs and inhibit metastasis. In addition, Ir Nts-Ab targets CSCs and normal cancer cells with near infrared II-region photothermal therapy (NIR-II PTT), and is easily taken up by CSCs due to recognition of the CD44 proteins. Moreover, photoacoustic imaging helps monitor drug accumulation and hypoxic TME improvement in tumor tissue. Importantly, Ir Nts-Ab has good biological safety, making it suitable for biomedical applications. This iridium nanozyme based on TME regulation as well as NIR-II PTT will be a promising strategy for the treatment of cancer. STATEMENT OF SIGNIFICANCE: Cancer stem cells (CSCs) are key factors that make tumors difficult to eradicate, and strongly depend on the hypoxic tumor microenvironment (TME), which plays a crucial role in the occurrence and metastasis of tumors. Herein, an antibody modified iridium nanosheet (definition as Ir Nts-Ab) was developed for targeted eradication of CSCs by photoacoustic imaging guided photothermal therapy (PTT) and TME regulation. Ir Nts-Ab with catalase-like activity could inhibit HIF-1α by producing oxygen, thus effectively reducing the proportion of CSCs and inhibiting tumor metastasis. Additionally, Ir Nts-Ab achieved the eradication of CSCs by PTT, and eliminated reactive oxygen species to decrease the inflammatory response, resulting in reduced tumor metastasis, which was promising for the cure of solid tumors in the clinics.


Subject(s)
Nanoparticles , Neoplasms , Photoacoustic Techniques , Humans , Photothermal Therapy , Iridium/pharmacology , Iridium/therapeutic use , Tumor Microenvironment , Photoacoustic Techniques/methods , Reactive Oxygen Species , Neoplasms/therapy , Neoplasms/drug therapy , Neoplastic Stem Cells/pathology , Oxygen , Cell Line, Tumor
3.
Redox Biol ; 67: 102913, 2023 11.
Article in English | MEDLINE | ID: mdl-37857001

ABSTRACT

Oxidative stress is a key factor leading to profound neurological deficits following spinal cord injury (SCI). In this study, we present the development and potential application of an iridium (iii) complex, (CpxbiPh) Ir (N^N) Cl, where CpxbiPh represents 1-biphenyl-2,3,4,5-tetramethyl cyclopentadienyl, and N^N denotes 2-(3-(4-nitrophenyl)-1H-1,2,4-triazol-5-yl) pyridine chelating agents, to address this challenge through a mechanism governed by the regulation of an antioxidant protein. This iridium complex, IrPHtz, can modulate the Oxidation Resistance 1 (OXR1) protein levels within spinal cord tissues, thus showcasing its antioxidative potential. By eliminating reactive oxygen species (ROS) and preventing apoptosis, the IrPHtz demonstrated neuroprotective and neural healing characteristics on injured neurons. Our molecular docking analysis unveiled the presence of π stacking within the IrPHtz-OXR1 complex, an interaction that enhanced OXR1 expression, subsequently diminishing oxidative stress, thwarting neuroinflammation, and averting neuronal apoptosis. Furthermore, in in vivo experimentation with SCI-afflicted mice, IrPHtz was efficacious in shielding spinal cord neurons, promoting their regrowth, restoring electrical signaling, and improving motor performance. Collectively, these findings underscore the potential of employing the iridium metal complex in a novel, protein-regulated antioxidant strategy, presenting a promising avenue for therapeutic intervention in SCI.


Subject(s)
Coordination Complexes , Spinal Cord Injuries , Mice , Animals , Reactive Oxygen Species/metabolism , Iridium/chemistry , Iridium/pharmacology , Iridium/therapeutic use , Antioxidants/metabolism , Molecular Docking Simulation , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Oxidative Stress , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use
4.
Acta Biomater ; 166: 524-535, 2023 08.
Article in English | MEDLINE | ID: mdl-37088161

ABSTRACT

Cerebral ischemia-reperfusion injury (CIRI) is a complex pathological condition with high mortality. In particular, reperfusion can stimulate overproduction of reactive oxygen species (ROS) and activation of inflammation, causing severe secondary injuries to the brain. Despite tremendous efforts, it remains urgent to rationally design antioxidative agents with straightforward and efficient ROS scavenging capability. Herein, a potent antioxidative agent was explored based on iridium oxide nano-agglomerates (Tf-IrO2 NAs) via the facile transferrin (Tf)-templated biomineralization approach, and innovatively applied to treat CIRI. Containing some small-size IrO2 aggregates, these NAs possess intrinsic hydroxyl radicals (•OH)-scavenging ability and multifarious enzyme activities, such as catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Moreover, they also showed improved blood-brain barrier (BBB) penetration and enhanced accumulation in the ischemic brain via Tf receptor-mediated transcytosis. Therefore, Tf-IrO2 NAs achieved robust in vitro anti-inflammatory and cytoprotection effects against oxidative stress. Importantly, mice were effectively protected against CIRI by enhanced ROS scavenging activity in vivo, and the therapeutic mechanism was systematically verified. These findings broaden the idea of expanding Ir-based NAs as potent antioxidative agents to treat CIRI and other ROS-mediated diseases. STATEMENT OF SIGNIFICANCE: (1) The ROS-scavenging activities of IrO2 are demonstrated comprehensively, which enriched the family of nano-antioxidants. (2) The engineering Tf-IrO2 nano-agglomerates present unique multifarious enzyme activities and simultaneous transferrin targeting and BBB crossing ability for cerebral ischemia-reperfusion injury therapy. (3) This work may open an avenue to enable the use of IrO2 to alleviate ROS-mediated inflammatory and brain injury diseases.


Subject(s)
Brain Ischemia , Reperfusion Injury , Mice , Animals , Reactive Oxygen Species/metabolism , Transferrin/pharmacology , Iridium/pharmacology , Iridium/therapeutic use , Brain Ischemia/drug therapy , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Oxidative Stress , Antioxidants/pharmacology , Reperfusion
5.
Respir Med ; 211: 107172, 2023 05.
Article in English | MEDLINE | ID: mdl-36906187

ABSTRACT

BACKGROUND: A novel, once-daily, fixed-dose combination of mometasone furoate/indacaterol acetate/glycopyrronium bromide (MF/IND/GLY) delivered via Breezhaler® is the first inhaled corticosteroid/long-acting ꞵ2-agonist/long-acting muscarinic antagonist (ICS/LABA/LAMA) therapy approved for the maintenance treatment of asthma in adults inadequately controlled on ICS/LABA combination. In patients with asthma and persistent airflow limitation (PAL), maximal treatment, especially with combination is suggested. This post hoc analysis of data from the IRIDIUM study assessed the efficacy of MF/IND/GLY in asthma patients with and without PAL. METHODS: Patients with post-bronchodilator FEV1 ≤80% of predicted and FEV1/FVC ratio of ≤0.7 were categorised as PAL subgroup and the remaining as the non-PAL subgroup. Lung function parameters (FEV1, PEF, and FEF25%-75%) and annualised asthma exacerbations rates were evaluated in both subgroups across the treatment arms: once-daily high-dose MF/IND/GLY (160/150/50 µg), high-dose MF/IND (320/150 µg) and twice-daily high-dose fluticasone/salmeterol (FLU/SAL; 500/50 µg). RESULTS: Of the 3092 randomised patients, 64% (n = 1981) met the criteria for PAL. Overall, there was no evidence of treatment difference between PAL and non-PAL subgroups (interaction P-value for FEV1, FEF25%-75%, PEF, moderate or severe exacerbations, severe exacerbations and all exacerbations were 0.42, 0.08, 0.43 0.29, 0.35 and 0.12, respectively). In the PAL subgroup, high-dose MF/IND/GLY versus high-dose MF/IND and high-dose FLU/SAL improved trough FEV1 (mean difference: 102 mL [P < 0.0001] and 137 mL [P < 0.0001]) and reduced moderate or severe (16% and 32%), severe (25% and 39%) and all exacerbations (19% and 38%), respectively. CONCLUSIONS: Once-daily fixed-dose MF/IND/GLY was efficacious in asthma patients with and without persistent airflow limitation.


Subject(s)
Asthma , Pulmonary Disease, Chronic Obstructive , Adult , Humans , Glycopyrrolate , Mometasone Furoate , Iridium/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Drug Combinations , Forced Expiratory Volume , Lung , Asthma/drug therapy , Indans , Nebulizers and Vaporizers , Administration, Inhalation , Bronchodilator Agents/therapeutic use
6.
Eur J Med Chem ; 233: 114250, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35276422

ABSTRACT

The increasing emergence and spread of drug resistant Candida albicans represent a serious challenge for effective treatment and call for the development of new therapeutic options. To address this need, we synthesized a series of polypyridyl iridium(III) complexes and studied their antimicrobial activities. Herein, one lead iridium(III) complex Ir-3 [(ptpy)2Ir(dppz)]PF6, with ptpy = 2-(p-tolyl)pyridine and dppz = dipyrido[3,2-a:2',3'-c]phenazine, exhibited potent and broad-spectrum antifungal activities against drug-resistant pathogens and low hemolytic activity toward mammalian cells. Furthermore, Ir-3 showed low tendency to induce resistance, displayed biofilm inhibition and eradication activities. Significantly, Ir-3 exhibited potent in vivo antifungal activity in a murine model of disseminated candidiasis. This study may pave the way for the development of novel antifungal agent based upon polypyridyl iridium(III) complex to combat antifungal resistance.


Subject(s)
Candidiasis , Iridium , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida albicans , Candidiasis/drug therapy , Candidiasis/microbiology , Drug Resistance, Fungal , Iridium/pharmacology , Iridium/therapeutic use , Mammals , Mice
7.
Adv Ther ; 39(6): 2365-2378, 2022 06.
Article in English | MEDLINE | ID: mdl-35072888

ABSTRACT

A novel, once-daily (o.d.), fixed-dose combination (FDC) of indacaterol acetate (IND), glycopyrronium bromide (GLY), and mometasone furoate (MF), delivered by the inhaler Breezhaler® device, is the first long-acting beta2-adrenergic agonist/long-acting muscarinic antagonist/inhaled corticosteroid (LABA/LAMA/ICS) therapy to be approved for maintenance treatment of asthma in adults inadequately controlled on LABA/ICS. The approval of IND/GLY/MF in the European Union (EU) also included an optional electronic sensor and smartphone (or other suitable device) application, making it the first "digital companion" that can be prescribed with an asthma medication. As a result, the European Medicines Agency included this approval as one of the "outstanding contributions to public health" (for Pneumology/Allergology) in their 2020 highlights report. Alongside IND/GLY/MF, an o.d. LABA/ICS FDC, IND/MF, was also developed and approved. This review outlines the unique strategy used in the accelerated development of IND/GLY/MF that combined various approaches: (1) selecting individual components with established efficacy/safety, (2) bridging doses to optimize efficacy/safety of IND/GLY/MF and IND/MF delivered via the Breezhaler® device, (3) developing IND/GLY/MF and IND/MF in parallel, and (4) submission for regulatory approval before formal completion of the pivotal phase III studies. IND/GLY/MF and IND/MF were combined in a single-development plan (PLATINUM program), which comprised four phase III studies: QUARTZ and PALLADIUM evaluated IND/MF while IRIDIUM and ARGON evaluated IND/GLY/MF. A unique feature was the inclusion of two LABA/ICS comparators in the pivotal IRIDIUM study-IND/MF as an internal comparator, and high-dose salmeterol xinafoate/fluticasone propionate (SAL/FLU) as a marketed comparator. In the ARGON study, IND/GLY/MF was compared against o.d. tiotropium (via Respimat®) plus twice-daily (b.i.d.) high-dose SAL/FLU (via Diskus®). As a result of this development strategy, the development and approval of IND/GLY/MF was accelerated by ca. 4 years as against what would be expected from a traditional approach, novel data were generated, and a unique optional digital companion was approved in the EU. A Video Abstract by Dr Dominic Brittain, Global Drug Development, Novartis. (MP4 228293 kb).


Subject(s)
Asthma , Pulmonary Disease, Chronic Obstructive , Acetates/therapeutic use , Administration, Inhalation , Adrenergic beta-2 Receptor Agonists/therapeutic use , Adult , Argon/therapeutic use , Asthma/drug therapy , Bronchodilator Agents/therapeutic use , Drug Combinations , Drug Development , Glycopyrrolate/therapeutic use , Humans , Indans , Iridium/therapeutic use , Mometasone Furoate/therapeutic use , Nebulizers and Vaporizers , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinolones
8.
J Asthma ; 59(8): 1627-1637, 2022 08.
Article in English | MEDLINE | ID: mdl-34346266

ABSTRACT

OBJECTIVE: PALLADIUM and IRIDIUM studies demonstrated efficacy and safety of indacaterol/mometasone (IND/MF) versus salmeterol/fluticasone (SAL/FLU). This post hoc analysis of pooled data from PALLADIUM and IRIDIUM studies evaluated efficacy and safety of IND/MF versus SAL/FLU in Asian patients with inadequately controlled asthma. METHODS: Both studies were Phase III, 52-week, randomized, double-blind, active-controlled that included patients with predicted pre-bronchodilator FEV1 (PALLADIUM, ≥50%-<85%; IRIDIUM, <80%), ACQ-7 score ≥1.5. Patients treated with IND/MF high- (150/320 µg) or medium-dose (150/160 µg) or SAL/FLU high-dose (50/500 µg) were included. Lung function, asthma control, and asthma exacerbations were evaluated. RESULTS: In total, 323 patients (IND/MF high-dose, n = 107; IND/MF medium-dose, n = 106, SAL/FLU high-dose, n = 110) were included. IND/MF high-dose showed improvement in trough FEV1 versus SAL/FLU high-dose at Weeks 26 (Δ, 42 mL; 95% CI, -35 to 120 mL), and 52 (Δ, 41 mL; 95% CI, -37 to 120 mL). IND/MF high-dose exhibited numerically greater improvement in ACQ-7 score versus SAL/FLU high-dose at Weeks 26 (Δ, -0.215; 95% CI, -0.385 to -0.044) and 52 (Δ, -0.176; 95% CI, -0.348 to -0.005). Improvements in trough FEV1 and ACQ-7 score were comparable between IND/MF medium-dose and SAL/FLU high-dose. IND/MF high- and medium-dose showed reductions in moderate or severe (45%; 30%), severe (39%; 41%), and all (9%; 25%) exacerbations, respectively, versus SAL/FLU high-dose over 52 weeks. All treatments were well tolerated. CONCLUSIONS: Once-daily, single-inhaler IND/MF high-dose improved lung function with better asthma control, reduced asthma exacerbations with comparable safety versus twice-daily SAL/FLU high-dose. IND/MF medium-dose showed comparable outcomes to SAL/FLU high-dose at a reduced steroid dose.


Subject(s)
Asthma , Palladium , Administration, Inhalation , Bronchodilator Agents , Double-Blind Method , Drug Combinations , Fluticasone-Salmeterol Drug Combination , Forced Expiratory Volume , Humans , Indans , Iridium/pharmacology , Iridium/therapeutic use , Mometasone Furoate/therapeutic use , Nebulizers and Vaporizers , Palladium/pharmacology , Palladium/therapeutic use , Quinolones , Treatment Outcome
9.
ACS Appl Mater Interfaces ; 13(24): 27934-27944, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34101408

ABSTRACT

Due to conventional photodynamic therapy encountering serious problems of phototoxicity and low tissue-penetrating depth of light, other dynamic therapy-based therapeutic methods such as sonodynamic therapy (SDT) are expected to be developed. To improve the therapeutic response to SDT, more effective sonosensitizers are imperative. In this study, a novel water-soluble iridium(III)-porphyrin sonosensitizer (IrTMPPS) was synthesized and used for SDT. IrTMPPS generated ample singlet oxygen (1O2) under US irradiation and especially showed distinguished US-activatable abilities at more than 10 cm deep-tissue depths. Interestingly, under US irradiation, IrTMPPS sonocatalytically oxidized intracellular NADH, which would enhance SDT efficiency by breaking the redox balance in the tumor. Moreover, IrTMPPS displayed great sonocytotoxicity toward various cancer cells, and in vivo experiments demonstrated efficient tumor inhibition and anti-metastasis to the lungs in the presence of IrTMPPS and US irradiation. This report gives a novel idea of metal-based sonosensitizers for sonotherapy by fully taking advantage of non-invasiveness, water solubility, and deep tumor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Porphyrins/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Iridium/chemistry , Iridium/therapeutic use , Iridium/toxicity , Mice , NAD/chemistry , NAD/metabolism , Neoplasms/pathology , Oxidation-Reduction , Porphyrins/chemical synthesis , Porphyrins/toxicity , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/toxicity , Singlet Oxygen/metabolism , Ultrasonic Waves , Zebrafish
10.
Chem Commun (Camb) ; 57(9): 1093-1096, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33434260

ABSTRACT

Modulating the hypoxic microenvironment is the priority for tumor treatment. Cytometalated iridium(iii)-metformin conjugates were synthesized for treating hypoxic cancer cells for the first time, which alleviate hypoxia via mitochondria respiration inhibition, thus displaying 10-fold higher cytotoxicity, emerging anti-metastasis and anti-inflammatory activities than a metformin-free Ir(iii) complex and cisplatin against hypoxic cancer cells.


Subject(s)
Iridium/chemistry , Iridium/therapeutic use , Metformin/chemistry , Metformin/therapeutic use , Neoplasms/drug therapy , Tumor Hypoxia/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line , Cisplatin/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Humans , Neoplasm Metastasis/drug therapy , Neoplasms/pathology , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Tumor Microenvironment
12.
Chemistry ; 25(28): 7012-7022, 2019 May 17.
Article in English | MEDLINE | ID: mdl-30913329

ABSTRACT

The chemo-anti-inflammatory strategy is attracting ever more attention for the treatment of cancer. Here, two cyclometalated IrIII complexes Ir2 and Ir3 formed by conjugation of Ir1 with two antiphlogistics (aspirin and salicylic acid) have been designed. Ir2 and Ir3 exhibit higher antitumor and anti-inflammatory potencies than a mixture of Ir1 and aspirin/salicylic acid. We show that they can be hydrolyzed, accumulate in mitochondria, and induce mitochondrial dysfunction. Due to their intense long-lived phosphorescence, Ir2 and Ir3 can track mitochondrial morphological changes. Phosphorescence lifetime imaging shows that Ir2 and Ir3 can aggregate during mitochondrial dysfunction. As expected, Ir2 and Ir3 exhibit immunomodulatory properties by regulating the activity of immune factors. Both Ir2 and Ir3 can induce caspase-dependent apoptosis and caspase-independent paraptosis and inhibit several events related to metastasis. Moreover, Ir2 and Ir3 show potent tumor growth inhibition in vivo. Our study demonstrates that the combination of mitochondrial-targeting and immunomodulatory activities is feasible to develop multifunctional metal-based anticancer agents.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Aspirin/therapeutic use , Coordination Complexes/therapeutic use , Immunomodulation/drug effects , Iridium/therapeutic use , Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aspirin/chemistry , Aspirin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Female , Humans , Iridium/chemistry , Iridium/pharmacology , Luminescent Measurements/methods , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/pathology , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging/methods
13.
Future Med Chem ; 11(2): 119-135, 2019 01.
Article in English | MEDLINE | ID: mdl-30644327

ABSTRACT

Despite improvements in the 5-year survival rate to over 80% in cancers, such as Hodgkin lymphoma and testicular cancer, more aggressive tumors including pancreatic and brain cancer still have extremely low survival rates. The establishment of chemoresistance, responsible for the reduction in treatment efficiency and cancer relapse, is one possible explanation for this setback. Metal-based compounds, a class of anticancer drugs, are largely used in the treatment of cancer. Herein, we will review the use of metal-based small molecules in chemotherapy, focusing on recent studies, and we will discuss how new nonplatinum-based agents are prompting scientists to increase drug specificity to overcome chemoresistance in cancer cells.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Metals/chemistry , Metals/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carboplatin/chemistry , Carboplatin/pharmacology , Carboplatin/therapeutic use , Cisplatin/chemistry , Cisplatin/pharmacology , Cisplatin/therapeutic use , Copper/chemistry , Copper/pharmacology , Copper/therapeutic use , Drug Discovery , Gold/chemistry , Gold/pharmacology , Gold/therapeutic use , Humans , Iridium/chemistry , Iridium/pharmacology , Iridium/therapeutic use , Iron/chemistry , Iron/pharmacology , Iron/therapeutic use , Metals/therapeutic use , Neoplasms/drug therapy , Organometallic Compounds/therapeutic use , Rhenium/chemistry , Rhenium/pharmacology , Rhenium/therapeutic use , Rhodium/chemistry , Rhodium/pharmacology , Rhodium/therapeutic use , Ruthenium/chemistry , Ruthenium/pharmacology , Ruthenium/therapeutic use
14.
Eur J Med Chem ; 163: 830-839, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30579123

ABSTRACT

A range of fluorine and naphthyridine-based half-sandwich iridium (III) and ruthenium (II) complexes were synthesized. The iridium complexes possessed excellent antiproliferative properties, a substantial improvement over cisplatin, especially the best 1C containing the fluorine atom and 2C containing the naphthyridine. On the contrary, the ruthenium complexes displayed much less antiproliferative activity. Two X-ray crystal structures were determined. The cytotoxicity of the complexes can be changed flexible by regulating the metal center and the ancillary ligands. The best complex 1C was chose to study further on the mechanism of action. The chemical reactivity such as hydrolysis, reaction with nucleobases, glutathione and catalytic conversion of NADH to NAD+, were investigated. Complex 1C can react with 9-ethylguanine (9-EtG) and catalyze oxidation of NADH. In addition, the self-luminescence of the complex 1C was also successfully used in confocal microscopy images for elucidating the subcellular localization. Complex 1C specifically targeted to lysosomes in A549 cancer cells and caused lysosomal damages and promote cathepsin B released. Flow cytometry studies confirmed that the biological effects of this type of complexes induced apoptosis, especially late apoptosis. Our results suggested that changes in the mitochondria membrane potential were responsible for apoptosis. The chemistry and biological studies has showed that this class of metal complexes are worthy of further exploration for the design of novel anticancer drugs.


Subject(s)
Coordination Complexes/therapeutic use , Fluorine/therapeutic use , Iridium/therapeutic use , Naphthyridines/therapeutic use , Ruthenium/therapeutic use , A549 Cells , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Crystallography, X-Ray , Diagnostic Imaging/methods , Fluorine/chemistry , Humans , Iridium/chemistry , Lysosomes/metabolism , Membrane Potential, Mitochondrial/drug effects , Naphthyridines/chemistry , Ruthenium/chemistry , Structure-Activity Relationship
15.
Int J Mol Sci ; 19(11)2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30463221

ABSTRACT

Platelets play a major role in hemostatic events and are associated with various pathological events, such as arterial thrombosis and atherosclerosis. Iridium (Ir) compounds are potential alternatives to platinum compounds, since they exert promising anticancer effects without cellular toxicity. Our recent studies found that Ir compounds show potent antiplatelet properties. In this study, we evaluated the in vitro antiplatelet, in vivo antithrombotic and structure⁻activity relationship (SAR) of newly synthesized Ir complexes, Ir-1, Ir-2 and Ir-4, in agonists-induced human platelets. Among the tested compounds, Ir-1 was active in inhibiting platelet aggregation induced by collagen; however, Ir-2 and Ir-4 had no effects even at their maximum concentrations of 50 µM against collagen and 500 µM against U46619-induced aggregation. Similarly, Ir-1 was potently inhibiting of adenosine triphosphate (ATP) release, calcium mobilization ([Ca2+]i) and P-selectin expression induced by collagen-induced without cytotoxicity. Likewise, Ir-1 expressively suppressed collagen-induced Akt, PKC, p38MAPKs and JNK phosphorylation. Interestingly, Ir-2 and Ir-4 had no effect on platelet function analyzer (PFA-100) collagen-adenosine diphosphate (C-ADP) and collagen-epinephrine (C-EPI) induced closure times in mice, but Ir-1 caused a significant increase when using C-ADP stimulation. Other in vivo studies revealed that Ir-1 significantly prolonged the platelet plug formation, increased tail bleeding times and reduced the mortality of adenosine diphosphate (ADP)-induced acute pulmonary thromboembolism in mice. Ir-1 has no substitution on its phenyl group, a water molecule (like cisplatin) can replace its chloride ion and, hence, the rate of hydrolysis might be tuned by the substituent on the ligand system. These features might have played a role for the observed effects of Ir-1. These results indicate that Ir-1 may be a lead compound to design new antiplatelet drugs for the treatment of thromboembolic diseases.


Subject(s)
Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Iridium/therapeutic use , Thrombosis/drug therapy , Adenosine Triphosphate/metabolism , Adult , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Movement/drug effects , Collagen/pharmacology , Coordination Complexes/pharmacology , Female , Hemorrhage/pathology , Humans , Immobilized Proteins/pharmacology , Iridium/chemistry , Iridium/pharmacology , Ligands , Male , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Embolism/drug therapy , Pulmonary Embolism/pathology , Structure-Activity Relationship , Thrombosis/pathology , Time Factors , Young Adult
16.
Int J Nanomedicine ; 13: 4417-4431, 2018.
Article in English | MEDLINE | ID: mdl-30104875

ABSTRACT

BACKGROUND AND AIM: Iridium (Ir)-based complex is a potential antitumor ingredient, but its poor physicochemical properties such as hydrophobicity and low biocompatibility hamper further application. Liposome provides a potential delivery approach for improving the poor physicochemical property and reducing the side effects of antitumor drug. In this study, we aimed at incorporating Ir ([Ir(ppy)2(BTCP)]PF6) into liposomes to enhance the biocompatibility and sustained release of Ir for intravenous administration and to elucidate the mechanism in A549 cells. MATERIALS AND METHODS: Ir-loaded PEGylated liposomes (Lipo-Ir) were formulated by thin-film dispersion and ultrasonic method. Morphology, size distribution, and zeta potential of Lipo-Ir were examined by transmission electron microscopy (TEM) and Zetasizer. The released profile and biocompatibility were investigated by dialysis method and hemolysis test, respectively. Additionally, the cytotoxic activity and mechanism of Lipo-Ir and Ir inducing apoptosis in A549 cells were evaluated. RESULTS: Lipo-Ir can keep sustained release, excellent biocompatibility, and physical stability. The average particle size, polydispersity index, zeta potential, encapsulation efficiency, and drug loading are 112.57±1.15 nm, 0.19±0.02, -10.66±0.61 mV, 94.71%±3.21%, and 4.71%±0.41%, respectively. 3-(4,5-dimethylthiazole)-2,5-diphenltetraazolium bromide (MTT) assay show that Lipo-Ir and Ir display high cytotoxicity against selected cancer cells. Furthermore, the apoptotic features of morphology, depolarization of mitochondrial membrane potential, increase in the reactive oxygen species (ROS) levels, and disorder of Ca2+ homeostasis are observed after treating A549 cells with Ir and Lipo-Ir. Besides, Lipo-Ir can arrest the cell growth in G0/G1 phase. CONCLUSION: The studies demonstrate that Lipo-Ir can trigger apoptosis in A549 cells via ROS-mediated mitochondrial dysfunctions, and the biocompatible and sustained Lipo-Ir will be a promising drug delivery system.


Subject(s)
Drug Delivery Systems , Iridium/therapeutic use , Lung Neoplasms/drug therapy , Mitochondria/metabolism , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Endocytosis/drug effects , Flow Cytometry , Humans , Inhibitory Concentration 50 , Iridium/pharmacology , Liposomes , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Rabbits , Reactive Oxygen Species/metabolism
17.
Chembiochem ; 19(15): 1574-1589, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30019476

ABSTRACT

Photodynamic therapy (PDT) is a noninvasive treatment for certain types of cancer, bacterial, fungal and viral infections, and skin diseases. In recent years, adaptation of this treatment so as to achieve more specific targeted cancer therapy in particular has attracted significant attention. We focus herein on the design of novel iridium-based photosensitizers (PSs) with tunable photophysical and photobiological properties as efficient PDT agents. We highlight the ability of some IrIII photosensitizers to target specific cellular components, including their activation by one- and two-photon irradiation.


Subject(s)
Drug Design , Iridium/chemistry , Iridium/pharmacology , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Animals , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Humans , Iridium/therapeutic use , Models, Molecular , Molecular Targeted Therapy/methods , Neoplasms/pathology , Photosensitizing Agents/therapeutic use
18.
Biomaterials ; 39: 95-104, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25477176

ABSTRACT

Mitochondria-targeted compounds represent a promising approach to target tumors selectively and overcome resistance to current anticancer therapies. In this work, three cyclometalated iridium(III) complexes (1-3) containing bis-N-heterocyclic carbene (NHC) ligands have been explored as theranostic and photodynamic agents targeting mitochondria. These complexes display rich photophysical properties, which greatly facilitates the study of their intracellular fate. All three complexes are more cytotoxic than cisplatin against the cancer cells screened. 1-3 can penetrate into human cervical carcinoma (HeLa) cells quickly and efficiently, and they can carry out theranostic functions by simultaneously inducing and monitoring the morphological changes in mitochondria. Mechanism studies show that these complexes exert their anticancer efficacy by initiating a cascade of events related to mitochondrial dysfunction. Additionally, they display up to 3 orders of magnitude higher cytotoxicity upon irradiation at 365 nm, which is so far the highest photocytotoxic responses reported for iridium complexes.


Subject(s)
Antineoplastic Agents/chemistry , Iridium/chemistry , Methane/analogs & derivatives , Mitochondria/metabolism , Organometallic Compounds/chemistry , Photochemotherapy/methods , Cell Cycle/drug effects , Cell Line, Tumor , HeLa Cells , Humans , Iridium/pharmacology , Iridium/therapeutic use , Membrane Potential, Mitochondrial/physiology , Methane/chemistry , Methane/pharmacology , Methane/therapeutic use
19.
Chemistry ; 20(49): 16242-7, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25324137

ABSTRACT

Water-soluble phosphorescent polymeric nanoparticles with an average diameter of approximately 100 nm were synthesized by a coordination cross-linking reaction. The pyridine blocks in poly(4-vinyl pyridine-b-ethylene oxide) (P4VP-b-PEO) were cross-linked by the iridium chloride-bridged dimer in DMF solution. Owing to the presence of an iridium complex with different ligands in the core of the polymeric nanoparticles, NP-1, NP-2, and NP-3 showed bright green, yellow, and red phosphorescence, respectively. PEG chains in the shell gave the polymeric nanoparticles solubility and biocompatibility, which was confirmed by an MTT assay using HeLa cells as a model cancer cell line. The flow cytometry and laser confocal fluorescence microscopy results revealed NP-2, as an example, could be effectively uptaken by HeLa cells. Therefore, these polymeric nanoparticles can be used as luminescent probes for living cells. In addition, (1) O2 could be effectively generated in the presence of NP-2 upon irradiation with visible light (λ>400 nm, 300 mW cm(-2) ), which was confirmed by a clear decrease in the fluorescence intensity of 9,10-dimethylanthracene (DMA). After incubation with NP-2 at a concentration of 200 µg mL(-1) for 6 h, approximately 90 % of HeLa cells were effectively ablated upon irradiation with visible light for only 10 min, indicating the potential for photodynamic therapy with polymeric nanoparticles.


Subject(s)
Coordination Complexes/therapeutic use , Iridium/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/diagnosis , Neoplasms/drug therapy , Coordination Complexes/chemistry , HeLa Cells , Humans , Iridium/chemistry , Luminescence , Microscopy, Confocal , Nanoparticles/chemistry , Optical Imaging , Photochemotherapy , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Vinyl Compounds/chemistry , Vinyl Compounds/therapeutic use
20.
Brachytherapy ; 11(5): 402-7, 2012.
Article in English | MEDLINE | ID: mdl-21996539

ABSTRACT

PURPOSE: Advantages for electronic brachytherapy (EBT) of the vaginal cuff include decreased physical dose to the bladder and rectum. Here we compare (192)Ir with EBT using biological effective dose (BED) to account for the different radiobiological effectiveness (RBE) predicted for low-energy x-rays. METHODS AND MATERIALS: Fifteen data sets from five consecutive postoperative endometrial cancer patients treated with EBT were analyzed. Treatment planning was performed using PLATO software. The dose was prescribed as 21Gy in three fractions to a depth of 0.5cm. Physical dose, BED(3), and BED(10) were evaluated for the mucosa, bladder, and rectum. An RBE value of 1.5 was used for BED calculations. RESULTS: Mucosal physical dose is 28.4% greater with EBT (36.6 vs. 28.5Gy, p<0.05). However, the BED(10) is increased by 79.1% (55.6 vs. 99.6Gy, p<0.05) and the BED(3) by 71.5% (118.8 vs. 203.7Gy, p<0.05). The physical dose (dose to 50% volume of the organ) to the bladder (9.3 vs. 6.6Gy, p<0.05) and rectum (7.2 vs. 4.2Gy, p<0.05) are reduced with EBT. BED(3) to the rectum and bladder are also reduced but to a lesser extent (13 vs. 8.3Gy, p<0.05; 18.9 vs. 14.7Gy, p=0.06, respectively). CONCLUSIONS: BED takes into account the higher RBE of low-energy photons generated with EBT and provides a more accurate estimate of the biological effect. When using EBT, physical dose may underestimate the biological effect on the vaginal mucosa and overestimate the benefit for the bladder and rectum. Dose adjustment for EBT based on BED should be considered.


Subject(s)
Brachytherapy/methods , Endometrial Neoplasms/radiotherapy , Iridium/therapeutic use , Radioisotopes/therapeutic use , Vagina/pathology , Female , Humans , Radiation Dosage , Rectum/pathology , Urinary Bladder/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...