Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 754
Filter
1.
Int J Nanomedicine ; 19: 7997-8014, 2024.
Article in English | MEDLINE | ID: mdl-39130683

ABSTRACT

Purpose: Mitochondrial damage may lead to uncontrolled oxidative stress and massive apoptosis, and thus plays a pivotal role in the pathological processes of myocardial ischemia-reperfusion (I/R) injury. However, it is difficult for the drugs such as puerarin (PUE) to reach the mitochondrial lesion due to lack of targeting ability, which seriously affects the expected efficacy of drug therapy for myocardial I/R injury. Methods: We prepared triphenylphosphonium (TPP) cations and ischemic myocardium-targeting peptide (IMTP) co-modified puerarin-loaded liposomes (PUE@T/I-L), which effectively deliver the drug to mitochondria and improve the effectiveness of PUE in reducing myocardial I/R injury. Results: In vitro test results showed that PUE@T/I-L had sustained release and excellent hemocompatibility. Fluorescence test results showed that TPP cations and IMTP double-modified liposomes (T/I-L) enhanced the intracellular uptake, escaped lysosomal capture and promoted drug targeting into the mitochondria. Notably, PUE@T/I-L inhibited the opening of the mitochondrial permeability transition pore, reduced intracellular reactive oxygen species (ROS) levels and increased superoxide dismutase (SOD) levels, thereby decreasing the percentage of Hoechst-positive cells and improving the survival of hypoxia-reoxygenated (H/R)-injured H9c2 cells. In a mouse myocardial I/R injury model, PUE@T/I-L showed a significant myocardial protective effect against myocardial I/R injury by protecting mitochondrial integrity, reducing myocardial apoptosis and decreasing infarct size. Conclusion: This drug delivery system exhibited excellent mitochondrial targeting and reduction of myocardial apoptosis, which endowed it with good potential extension value in the precise treatment of myocardial I/R injury.


Subject(s)
Isoflavones , Liposomes , Myocardial Reperfusion Injury , Organophosphorus Compounds , Animals , Liposomes/chemistry , Myocardial Reperfusion Injury/drug therapy , Isoflavones/chemistry , Isoflavones/pharmacology , Isoflavones/administration & dosage , Isoflavones/pharmacokinetics , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacokinetics , Male , Mice , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Cations/chemistry , Myocardium/pathology , Myocardium/metabolism , Oxidative Stress/drug effects , Peptides/chemistry , Peptides/pharmacology , Peptides/administration & dosage , Drug Delivery Systems/methods
2.
Carbohydr Polym ; 282: 119087, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35123755

ABSTRACT

The efficient triggering of prodrug release has become a challengeable task for stimuli-responsive nanomedicine utilized in cancer therapy due to the subtle differences between normal and tumor tissues and heterogeneity. In this work, a dual ROS-responsive nanocarriers with the ability to self-regulate the ROS level was constructed, which could gradually respond to the endogenous ROS to achieve effective, hierarchical and specific drug release in cancer cells. In brief, DOX was conjugated with MSNs via thioketal bonds and loaded with ß-Lapachone. TPP modified chitosan was then coated to fabricate nanocarriers for mitochondria-specific delivery. The resultant nanocarriers respond to the endogenous ROS and release Lap specifically in cancer cells. Subsequently, the released Lap self-regulated the ROS level, resulting in the specific DOX release and mitochondrial damage in situ, enhancing synergistic oxidation-chemotherapy. The tumor inhibition Ratio was achieved to 78.49%. The multi-functional platform provides a novel remote drug delivery system in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Naphthoquinones/administration & dosage , Neoplasms/drug therapy , Oxidative Stress , Prodrugs/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Chitosan/administration & dosage , Chitosan/chemistry , Chitosan/pharmacokinetics , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Mitochondria/physiology , Nanoparticles/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacokinetics , Neoplasms/metabolism , Neoplasms/pathology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Oxidation-Reduction , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Reactive Oxygen Species/metabolism , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Tumor Burden/drug effects
3.
J Ocul Pharmacol Ther ; 37(6): 321-330, 2021.
Article in English | MEDLINE | ID: mdl-34152861

ABSTRACT

Purpose: Inflammation of the ocular surface is central to dry eye disease (DED). The anti-inflammatory agent phospho-sulindac (PS) at a high dose was efficacious against DED in a rabbit model. We assessed the dose, formulation and structure dependence of PS's effect. Methods: In rabbits with concanavalin A-induced DED we evaluated a range of PS concentrations (0.05%-1.6%) and dosing frequencies, assessed the duration of its effect with PS in 2 solution formulations and one emulsion formulation, and compared the efficacy of PS to that of sulindac, and of the structurally similar phospho-ibuprofen amide. We determined tear breakup time (TBUT) (tear stability), Schirmer's tear test (tear production), and by esthesiometry corneal sensitivity (symptoms). We also determined the biodistribution in the eye of topically applied PS. Results: PS in a solution formulation, given as eye drops q.i.d. was efficacious starting at a dose of 0.1%. The effect was apparent after 2 days of treatment and lasted at least 8 days after the last dose. Both signs (evidenced by TBUT and Schirmer's test) and symptoms (measured by corneal sensitivity) improved significantly. The best formulation was the solution formulation; a cyclodextrin-based formulation was also successful but the emulsion formulation was not. PS and its metabolites were essentially restricted to the anterior chamber of the eye. Sulindac and phospho-ibuprofen amide had no efficacy on DED. Conclusions: PS is efficacious against DED. Its effect, encompassing signs, and symptoms, are dose, formulation, and structure dependent. PS has therapeutic promise and merits further development.


Subject(s)
Drug Compounding , Dry Eye Syndromes/drug therapy , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Sulindac/analogs & derivatives , Administration, Topical , Animals , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Male , Organophosphorus Compounds/pharmacokinetics , Rabbits , Sulindac/administration & dosage , Sulindac/chemistry , Sulindac/pharmacokinetics , Tissue Distribution
4.
Methods Mol Biol ; 2275: 49-64, 2021.
Article in English | MEDLINE | ID: mdl-34118031

ABSTRACT

We have previously reported that radiolabeled phosphonium cations accumulate in the mitochondria down a transmembrane potential gradient. We present an optimized procedure for synthesis of three [18F]-labeled fluoroalkyl triphenylphosphonium salts ([18F]FATPs) via two-step simple nucleophilic substitution reactions to develop new myocardial imaging agents for positron emission tomography (PET) . The total reaction time of [18F]FATPs was within 60 min, and the overall decay-corrected radiochemical yield was approximately 15-30% (decay corrected). Radiochemical purity was >98% according to analytical high-performance liquid chromatography (HPLC) . The specific activity of [18F]FATPs was >6.1 TBq/µmol. The [18F]FATPs exhibited higher first-pass extraction fraction values in isolated heart, higher uptake in the myocardium, and a more rapid clearance from the liver and lung than [13N]NH3 in normal rats. The images from rats with an occluded left coronary artery demonstrated sharply defined myocardial defects in the corresponding area of the myocardium. This imaging technology may enable high-throughput, multiple studies daily and wide distribution of PET myocardial studies in clinic.


Subject(s)
Arterial Occlusive Diseases/diagnostic imaging , Coronary Vessels/diagnostic imaging , Fluorine Radioisotopes/chemistry , Heart/diagnostic imaging , Organophosphorus Compounds/chemical synthesis , Radiopharmaceuticals/chemistry , Animals , Chromatography, High Pressure Liquid , Coronary Vessels/pathology , Disease Models, Animal , Drug Elimination Routes , Male , Mitochondria, Heart/metabolism , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Positron-Emission Tomography , Rats , Salts/administration & dosage , Salts/chemical synthesis , Salts/chemistry
5.
Methods Mol Biol ; 2275: 65-85, 2021.
Article in English | MEDLINE | ID: mdl-34118032

ABSTRACT

The mitochondrion can be considered as the metabolic powerhouse of the cell, having a key impact on energy production, cell respiration, and intrinsic cell death. Mitochondria are also the main source of endogenous reactive oxygen species , including free radicals (FR), which are physiologically involved in signaling pathways but may promote cell damage when unregulated or excessively formed in inappropriate locations. A variety of chronic pathologies have been associated with FR-induced mitochondrial dysfunctions , such as cancer, age-related neurodegenerative diseases, and metabolic syndrome.In recent years drug design based on specific mitochondria-targeted antioxidants has become a very attractive therapeutic strategy and, among target compounds, nitrones have received growing attention because of their specific affinity toward FR. Here, we describe protocols dealing with the preparation, mitochondria permeation assessment, electron paramagnetic resonance (EPR) spin trapping setting, and antiapoptotic properties evaluation of a series of new linear nitrones vectorized by a triphenylphosphonium cation and labeled with a diethoxyphosphoryl moiety as 31P nuclear magnetic resonance (NMR) reporter with antioxidant property.


Subject(s)
Antioxidants/chemical synthesis , Mitochondria/chemistry , Nitrogen Oxides/chemistry , Organophosphorus Compounds/chemical synthesis , 3T3 Cells , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Electron Spin Resonance Spectroscopy , Humans , Magnetic Resonance Spectroscopy , Male , Mice , Molecular Structure , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Phosphorus Isotopes/chemistry , Phosphorylation , Rats , Spin Trapping
6.
J Clin Pharm Ther ; 46(5): 1480-1483, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33754343

ABSTRACT

WHAT IS KNOWN AND OBJECTIVES: Poor image quality was randomly seen in [99m Tc]Tc-tetrofosmin myocardial perfusion scintigraphic imaging. The interference hampered or even precluded medical interpretation. Our objective was to identify the cause of the random interferences. METHODS: Out of 40 patients planned for [99m Tc]Tc-tetrofosmin MPS, 36 presented normal tracer uptake and 4 exhibited subdiaphragmatic artefacts. Pharmaceutical interviews (P.I.) were set up to formally identify aetiologies of subdiaphragmatic uptake of [99m Tc]Tc-tetrofosmin. Patients were questioned about their diet and current drug treatments. RESULTS AND DISCUSSION: P.I. led to identification of dipyridamole as the cause of the artefacts. The systematic ingestion of a solid 25-gram high-fat snack bar and a glass of fresh water was introduced immediately after the injection of dipyridamole in 12 other patients undergoing [99m Tc]Tc-tetrofosmin MPS. None of the 12 patients presented subdiaphragmatic artefacts. WHAT IS NEW AND CONCLUSION: P.I. identified the cause of poor scintigraphic images to allow improved diagnoses.


Subject(s)
Dipyridamole/pharmacology , Myocardial Perfusion Imaging/methods , Organophosphorus Compounds/pharmacokinetics , Organotechnetium Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Vasodilator Agents/pharmacology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Tomography, Emission-Computed, Single-Photon
7.
Invest New Drugs ; 39(5): 1306-1314, 2021 10.
Article in English | MEDLINE | ID: mdl-33742299

ABSTRACT

Background Brigatinib, a next-generation anaplastic lymphoma kinase (ALK) inhibitor, targets activated, mutant forms of ALK and overcomes mechanisms of resistance to the ALK inhibitors crizotinib, ceritinib, and alectinib. Brigatinib is approved in multiple countries for treatment of patients with ALK-positive non-small cell lung cancer. Based on population pharmacokinetic (PK) analyses, no dosage adjustment is required for patients with mild or moderate renal impairment. Methods An open-label, single-dose study was conducted to evaluate the PK of brigatinib (90 mg) in patients with severe renal impairment (estimated glomerular filtration rate < 30 mL/min/1.73 m2; n = 8) and matched healthy volunteers with normal renal function (estimated glomerular filtration rate ≥ 90 mL/min/1.73 m2; n = 8). Plasma and urine were collected for the determination of plasma protein binding and estimation of plasma and urine PK parameters. Results Plasma protein binding of brigatinib was similar between patients with severe renal impairment (92 % bound) and matched healthy volunteers with normal renal function (91 % bound). Unbound brigatinib exposure (area under the plasma concentration-time curve from time zero to infinity) was approximately 92 % higher in patients with severe renal impairment compared with healthy volunteers with normal renal function. The renal clearance of brigatinib in patients with severe renal impairment was approximately 20 % of that observed in volunteers with normal renal function. Conclusions These findings support a brigatinib dosage reduction of approximately 50 % in patients with severe renal impairment.Trial registry: Not applicable.


Subject(s)
Organophosphorus Compounds/pharmacokinetics , Pyrimidines/pharmacokinetics , Renal Insufficiency/metabolism , Aged , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Area Under Curve , Female , Glomerular Filtration Rate , Half-Life , Humans , Male , Metabolic Clearance Rate , Middle Aged , Organophosphorus Compounds/blood , Organophosphorus Compounds/urine , Patient Acuity , Protein Binding/physiology , Pyrimidines/blood , Pyrimidines/urine
8.
J Mater Chem B ; 9(7): 1877-1887, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33533366

ABSTRACT

Acute organophosphorus pesticide poisoning (AOPP) is a worldwide health concern that has threatened human lives for decades, which attacks acetylcholinesterase (AChE) and causes nervous system disorders. Classical treatment options are associated with short in vivo half-life and side effects. As a potential alternative, delivery of mammalian-derived butyrylcholinesterase (BChE) offers a cost-effective way to block organophosphorus attack on acetylcholinesterase, a key enzyme in the neurotransmitter cycle. Yet the use of exotic BChE as a prophylactic or therapeutic agent is compromised by short plasma residence, immune response and unfavorable biodistribution. To overcome these obstacles, BChE nanodepots (nBChE) composed of a BChE core/polymorpholine shell structure were prepared via in situ polymerization, which showed enhanced stability, prolonged plasma circulation, attenuated antigenicity and reduced accumulation in non-targeted tissues. In vivo administration of nBChE pre- or post-organophosphorus exposure in a BALB/C mouse model resulted in potent prophylactic and therapeutic efficiency. To our knowledge, this is the first systematic delivery of non-human BChE to tackle AOPP. In addition, this work also opens up a new avenue for real applications in both research and clinical settings to cope with acute intoxication-related diseases.


Subject(s)
Butyrylcholinesterase/metabolism , Nanoparticles/metabolism , Organophosphate Poisoning/metabolism , Organophosphorus Compounds/metabolism , 3T3-L1 Cells , Animals , Butyrylcholinesterase/administration & dosage , Butyrylcholinesterase/chemistry , Cells, Cultured , Female , HEK293 Cells , Humans , Injections, Intravenous , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Particle Size , Surface Properties , Tissue Distribution
9.
Sci Rep ; 11(1): 598, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436690

ABSTRACT

The synthesis of a new series of Gd(III)-arylphosphonium complexes is described and the solution stability of selected compounds is reported. Their lipophilicity and uptake in human glial (SVG p12) and human glioblastoma multiforme (T98G) cell lines are presented. The in vitro cytotoxicity of all complexes was determined to be low at therapeutically-relevant concentrations. Selected Gd(III) complexes are potential candidates for further investigation as theranostic agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Gadolinium/chemistry , Glioblastoma/drug therapy , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacology , Antineoplastic Agents/pharmacokinetics , Cell Proliferation , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Organophosphorus Compounds/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
10.
Clin Pharmacokinet ; 60(2): 235-247, 2021 02.
Article in English | MEDLINE | ID: mdl-32816246

ABSTRACT

BACKGROUND AND OBJECTIVES: Brigatinib is an oral tyrosine kinase inhibitor approved in multiple countries for the treatment of patients with anaplastic lymphoma kinase-positive metastatic non-small cell lung cancer who have progressed on or are intolerant to crizotinib. We report a population pharmacokinetic model-based analysis for brigatinib. METHODS: Plasma concentration-time data were collected from 442 participants (105 healthy volunteers and 337 patients with cancer) who received single or multiple doses of oral brigatinib in one of five trials. Data were analyzed using non-linear mixed-effects modeling (NONMEM software version 7.3). RESULTS: Brigatinib plasma concentrations were best described by a three-compartment model with a transit compartment input (number of transit compartments = 2.35; mean transit time = 0.9 h). The final model included albumin as a covariate on apparent clearance. None of the additional covariates examined, including sex, age, race, body weight, mild or moderate renal impairment, total bilirubin, aspartate aminotransferase, and alanine aminotransferase, were found to meaningfully explain variability in apparent clearance, suggesting that no dose adjustment is required based on these covariates. CONCLUSIONS: Results from these population pharmacokinetic analyses informed the prescribing guidance for patients with mild or moderate renal impairment in the US Prescribing Information and the European Summary of Product Characteristics for brigatinib.


Subject(s)
Neoplasms , Organophosphorus Compounds , Pyrimidines , Adolescent , Adult , Aged , Aged, 80 and over , Clinical Trials as Topic , Female , Healthy Volunteers , Humans , Male , Middle Aged , Neoplasms/metabolism , Organophosphorus Compounds/pharmacokinetics , Pyrimidines/pharmacokinetics , Young Adult
11.
Bioorg Chem ; 104: 104185, 2020 11.
Article in English | MEDLINE | ID: mdl-32911200

ABSTRACT

Phosphonates-based agents are well-known bone-seeking radiopharmaceuticals with application in detection and therapy. With higher sensitivity and resolution offered by Positron Emission Tomography (PET), tracers based on this technique are gaining huge attention. 68Ga-based generator and radiotracers render independence from the on-site cyclotron. We report the development of 68Ga-labeled DOTA-based bismacrocyclic phosphonate derivative, for bone PET imaging. The synthesis and characterization of 68Ga- DO3P-AME-DO3P was carried out in > 95% purity. The radiotracer displayed high stability and low binding affinity (<3%) to blood serum. High in vitro binding affinity were observed for synthetic hydroxyapatite, SAOS-2, osteoclast and osteoblast cells. In vivo pharmacokinetics revealed fast washout with biphasic release pattern. The deposition of radiotracer in osseous tissues was high (Bone/Muscle ratio:18), as studied from the biodistribution studies. In vivo PET/CT and biodistribution analyses revealed the ability of 68Ga-DO3P-AME-DO3P to target and accumulate in bone, thus displaying its potential as a PET bone imaging agent.


Subject(s)
Acetamides/chemistry , Bone and Bones/diagnostic imaging , Macrocyclic Compounds/chemistry , Organophosphorus Compounds/chemistry , Positron-Emission Tomography , Radiopharmaceuticals/chemistry , Acetamides/blood , Acetamides/pharmacokinetics , Gallium Radioisotopes , Humans , Macrocyclic Compounds/blood , Macrocyclic Compounds/pharmacokinetics , Molecular Structure , Organophosphorus Compounds/blood , Organophosphorus Compounds/pharmacokinetics , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
12.
J Clin Oncol ; 38(31): 3592-3603, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32780660

ABSTRACT

PURPOSE: Brigatinib, a next-generation anaplastic lymphoma kinase (ALK) inhibitor, demonstrated superior progression-free survival (PFS) and improved health-related quality of life (QoL) versus crizotinib in advanced ALK inhibitor-naive ALK-positive non-small cell lung cancer (NSCLC) at first interim analysis (99 events; median brigatinib follow-up, 11.0 months) in the open-label, phase III ALTA-1L trial (ClinicalTrials.gov identifier: NCT02737501). We report results of the second prespecified interim analysis (150 events). METHODS: Patients with ALK inhibitor-naive advanced ALK-positive NSCLC were randomly assigned 1:1 to brigatinib 180 mg once daily (7-day lead-in at 90 mg once daily) or crizotinib 250 mg twice daily. The primary end point was PFS as assessed by blinded independent review committee (BIRC). Investigator-assessed efficacy, blood samples for pharmacokinetic assessments, and patient-reported outcomes were also collected. RESULTS: Two hundred seventy-five patients were randomly assigned (brigatinib, n = 137; crizotinib, n = 138). With median follow-up of 24.9 months for brigatinib (150 PFS events), brigatinib showed consistent superiority in BIRC-assessed PFS versus crizotinib (hazard ratio [HR], 0.49 [95% CI, 0.35 to 0.68]; log-rank P < .0001; median, 24.0 v 11.0 months). Investigator-assessed PFS HR was 0.43 (95% CI, 0.31 to 0.61; median, 29.4 v 9.2 months). No new safety concerns emerged. Brigatinib delayed median time to worsening of global health status/QoL scores compared with crizotinib (HR, 0.70 [95% CI, 0.49 to 1.00]; log-rank P = .049). Brigatinib daily area under the plasma concentration-time curve was not a predictor of PFS (HR, 1.005 [95% CI, 0.98 to 1.031]; P = .69). CONCLUSION: Brigatinib represents a once-daily ALK inhibitor with superior efficacy, tolerability, and QoL over crizotinib, making it a promising first-line treatment of ALK-positive NSCLC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Crizotinib/therapeutic use , Lung Neoplasms/drug therapy , Organophosphorus Compounds/therapeutic use , Pyrimidines/therapeutic use , Adolescent , Adult , Aged , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Crizotinib/adverse effects , Crizotinib/blood , Crizotinib/pharmacokinetics , Female , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/blood , Organophosphorus Compounds/pharmacokinetics , Progression-Free Survival , Pyrimidines/adverse effects , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Quality of Life , Survival Rate , Young Adult
13.
Arch Toxicol ; 94(11): 3751-3757, 2020 11.
Article in English | MEDLINE | ID: mdl-32720193

ABSTRACT

We present a simple method for chiral separation and analysis of organophosphorus nerve agents and apply it to monitor the enantioselective blood elimination kinetics of sarin in-vitro. The method is implemented in standard reverse phase LC-MS operating conditions, relieving the user of the dedicated operating conditions frequently demanded in chiral LC-MS analysis. The method consists of formation of diastereomers by a rapid derivatization with (R)-2-(1 aminoethyl) phenol, followed by LC-MS/MS analysis. Derivatization enantioselectivity was studied by comparing the reaction of optically pure sarin and racemic sarin, proving no substantial enantiomeric preference in the reaction and demonstrating the enantiomeric discrimination abilities of the technique. Enantioselective sarin elimination pathways were probed in-vitro by following the fast elimination kinetics of the two sarin enantiomers as well as its hydrolysis metabolite (isopropyl methyl-phosphonic acid, IMPA) in whole blood and plasma compared to water. Sarin enantiomers showed the known marked differences in elimination kinetics with rapid elimination of the (+) enantiomer and slower elimination of the (-) enantiomer in whole blood and plasma as well as dose-dependent kinetics (faster elimination at lower concentrations). We found that small amounts of acetonitrile in plasma prevent the rapid elimination of the (+) enantiomer, resulting in similar, slower elimination kinetics for both enantiomers.


Subject(s)
Sarin/metabolism , Sarin/pharmacokinetics , Blood/metabolism , Chemical Warfare Agents/metabolism , Chemical Warfare Agents/pharmacokinetics , Chromatography, Liquid , Humans , Hydrolysis , Nerve Agents/metabolism , Nerve Agents/pharmacokinetics , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacokinetics , Stereoisomerism , Tandem Mass Spectrometry , Water/chemistry
14.
AAPS PharmSciTech ; 21(5): 160, 2020 May 31.
Article in English | MEDLINE | ID: mdl-32476084

ABSTRACT

The aim of this present study was to investigate the ability of different dissolution methods to predict the in vivo performance of efonidipine hydrochloride (EFH). The solid dispersions of EFH were prepared by solvent evaporation method with HPMC-AS as matrix and urea as a pH adjusting agent. The paddle method, the open-loop, and the closed-loop flow-through cell methods were studied. In the study, Weibull's model was the best fit to explain release profiles. The pharmacokinetics behaviors of two kinds of solid dispersions with different release rate were investigated in comparison to the EFH after oral administration in rats. In vivo absorption was calculated by a numerical deconvolution method. In the study, the level A in vivo and in vitro correlation (IVIVC) was utilized. The correlation coefficient was calculated and interpreted by means of linear regression analysis (Origin.Pro.8.5 software). As a result, excellent IVIVC for solid dispersions and crude drug (r2 = 0.9352-0.9916) was obtained for the dissolution rate determined with flow-through cell open-loop system in phosphate buffer solution with 0.1% (w/v) polysorbate 80 at pH 6.5, the flow-rate of 4 mL/min. In addition, the self-assembled flow cell system had good repeatability and accuracy. The dissolution rate of the solid dispersion could be slowed down by the flow-through method, and the difference caused by preparation was significantly distinguished. The study demonstrated that flow-through cell method of the open-loop, compared with paddle method, was suitable for predicting in vivo performance of EFH solid dispersions.


Subject(s)
Calcium Channel Blockers/chemistry , Dihydropyridines/chemistry , Nitrophenols/chemistry , Animals , Calcium Channel Blockers/pharmacokinetics , Dihydropyridines/pharmacokinetics , In Vitro Techniques , Male , Nitrophenols/pharmacokinetics , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Pharmaceutical Preparations , Rats , Rats, Sprague-Dawley , Solubility , Solvents , Water
15.
Mol Pharm ; 17(6): 2044-2053, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32383887

ABSTRACT

The strategy of using radioligands for targeting the prostate-specific membrane antigen (PSMA) revealed to be promising for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Recently developed albumin-binding PSMA radioligands showed a remarkably increased tumor uptake because of the enhanced blood circulation, but higher accumulation of activity was also observed in off-target organs and tissues. The aim of this study was to investigate the option of using fast-cleared, small-molecular-weight PSMA inhibitors (PSMA-11, 2-PMPA, and ZJ-43) to reduce the kidney uptake of [177Lu]Lu-PSMA-ALB-56, a previously developed albumin-binding PSMA radioligand. Dual-isotope SPECT/CT imaging was performed with tumor-bearing mice coinjected with [177Lu]Lu-PSMA-ALB-56 and a 2.5-fold molar excess of [67Ga]Ga-PSMA-11. At early timepoints after injection, the high renal uptake of [67Ga]Ga-PSMA-11 reduced the accumulation of [177Lu]Lu-PSMA-ALB-56 in the kidneys substantially, whereas the tumor uptake of [177Lu]Lu-PSMA-ALB-56 was only slightly affected. These findings were confirmed in biodistribution studies, which revealed reduced uptake of [177Lu]Lu-PSMA-ALB-56 in the kidneys due to coadministered unlabeled PSMA-11 (9.1 ± 0.8% IA/g vs 46 ± 11% IA/g; 1 h p.i.). The tumor uptake of [177Lu]Lu-PSMA-ALB-56 was almost the same at 1 h p.i., irrespective of whether or not PSMA-11 was coinjected (24 ± 6% IA/g vs 27 ± 7% IA/g). The application of [177Lu]Lu-PSMA-ALB-56 with 2-PMPA or ZJ-43, respectively, showed similar results in biodistribution studies. Among all three tested PSMA inhibitors, 2-PMPA, applied at a 2.5-fold molar excess relative to [177Lu]Lu-PSMA-ALB-56, was most effective to improve the tumor-to-kidney ratios over the first hours after injection of [177Lu]Lu-PSMA-ALB-56. The concept of using a PSMA inhibitor together with [177Lu]Lu-PSMA-ALB-56 appears promising in view of a clinical translation of this and possibly other long-circulating PSMA radioligands.


Subject(s)
Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Humans , Kidney/metabolism , Male , Single Photon Emission Computed Tomography Computed Tomography
16.
Drug Deliv ; 27(1): 502-518, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32228100

ABSTRACT

Reactive oxygen species (ROS)-induced neuronal mitochondrial dysfunction is a key pathologic factor in sporadic Alzheimer's disease (AD). Neuronal mitochondria have been proposed to be a promising therapeutic target for AD, especially for the failures of phase III clinical trials on conventional amyloid-ß (Aß) targeted therapy. However, the efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge due to the complicated physiological environment. Recently, biomaterials-based nanomedicine has been widely investigated for the treatment of AD. Herein, we devised a strategy for functional antioxidant delivery to neuronal mitochondria by loading antioxidants into red blood cell (RBC) membrane-coated nanostructured lipid carriers (NLC) bearing rabies virus glycoprotein (RVG29) and triphenylphosphine cation (TPP) molecules attached to the RBC membrane surface (RVG/TPP NPs@RBCm). With the advantage of suitable physicochemical properties of NLC and unique biological functions of the RBC membrane, RVG/TPP NPs@RBCm are stabilized and enabled sustained drug release, providing improved biocompatibility and long-term circulation. Under the synergistic effects of RVG29 and TPP, RVG/TPP NPs@RBCm can not only penetrate the blood-brain barrier (BBB) but also target neuron cells and further localize in the mitochondria. After encapsulating Resveratrol (RSV) as the model antioxidant, the data demonstrated that RVG/TPP-RSV NPs@RBCm can relieve AD symptoms by mitigating Aß-related mitochondrial oxidative stress both in vitro and in vivo. The memory impairment in APP/PS1 mice is significantly improved following the systemic administration of RVG/TPP-RSV NPs@RBCm. In conclusion, intravenous neuronal mitochondria-targeted dual-modified novel biomimetic nanosystems are a promising therapeutic candidate for ROS-induced mitochondrial dysfunction in AD.


Subject(s)
Alzheimer Disease/drug therapy , Antioxidants/administration & dosage , Biomimetic Materials/chemistry , Mitochondria/drug effects , Nanoparticles/chemistry , Neurons/drug effects , Resveratrol/administration & dosage , Administration, Intravenous , Alzheimer Disease/metabolism , Animals , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Biological Transport/drug effects , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/therapeutic use , Erythrocyte Membrane/chemistry , Male , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Nanoparticles/metabolism , Nanoparticles/therapeutic use , Neurons/metabolism , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/therapeutic use , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resveratrol/pharmacokinetics , Resveratrol/therapeutic use , Tissue Distribution
17.
Bioorg Med Chem Lett ; 30(7): 126986, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32046903

ABSTRACT

Our HCV research program investigated novel 2'-dihalogenated nucleoside HCV polymerase inhibitors and identified compound 1, a 5'-phosphoramidate prodrug of 2'-deoxy-2'-α-bromo-ß-chloro uridine. Although 1 had a favorable in vitro activity profile in HCV replicons, oral dosing in dog resulted in low levels of the active 5'-triphosphate (TP) in liver. Metabolism studies using human hepatocytes provided a simple assay for screening alternative phosphoramidate prodrug analogs. Compounds that produced high TP concentrations in hepatocytes were tested in dog liver biopsy studies. This method identified 2-aminoisobutyric acid ethyl ester (AIBEE) phosphoramidate prodrug 14, which provided 100-fold higher TP concentrations in dog liver in comparison to 1 (4 and 24 h after 5 mg/kg oral dose).


Subject(s)
Antiviral Agents/pharmacology , Deoxyuridine/analogs & derivatives , Deoxyuridine/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Prodrugs/pharmacology , Aminoisobutyric Acids/metabolism , Aminoisobutyric Acids/pharmacokinetics , Aminoisobutyric Acids/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Deoxyuridine/metabolism , Deoxyuridine/pharmacokinetics , Dogs , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Hepacivirus/enzymology , Hepatocytes/metabolism , Humans , Liver/metabolism , Microbial Sensitivity Tests , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/pharmacology , Prodrugs/metabolism , Prodrugs/pharmacokinetics , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects
18.
Neurobiol Dis ; 133: 104455, 2020 01.
Article in English | MEDLINE | ID: mdl-31022458

ABSTRACT

There is a unique in vivo interplay involving the mechanism of inactivation of acetylcholinesterase (AChE) by toxic organophosphorus (OP) compounds and the restoration of AChE activity by oxime antidotes. OP compounds form covalent adducts to this critical enzyme target and oximes are introduced to directly displace the OP from AChE. For the most part, the in vivo inactivation of AChE leading to neurotoxicity and antidote-based therapeutic reversal of this mechanism are well understood, however, these molecular-level events have not been evaluated by dynamic imaging in living systems at millimeter resolution. A deeper understanding of these critically, time-dependent mechanisms is needed to develop new countermeasures. To address this void and to help accelerate the development of new countermeasures, positron-emission tomography (PET) has been investigated as a unique opportunity to create platform technologies to directly examine the interdependent toxicokinetic/pharmacokinetic and toxicodynamic/pharmacodynamic features of OPs and oximes in real time within live animals. This review will cover two first-in-class PET tracers representing an OP and an oxime antidote, including their preparation, requisite pharmacologic investigations, mechanistic interpretations, biodistribution and imaging.


Subject(s)
Cholinesterase Reactivators/pharmacokinetics , Nerve Agents , Organophosphorus Compounds , Positron-Emission Tomography/methods , Radiopharmaceuticals , Animals , Antidotes/pharmacokinetics , Humans , Nerve Agents/pharmacokinetics , Nerve Agents/toxicity , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/toxicity , Oximes/pharmacokinetics
19.
Clin Pharmacol Drug Dev ; 9(2): 214-223, 2020 02.
Article in English | MEDLINE | ID: mdl-31287236

ABSTRACT

In vitro data support involvement of cytochrome P450 (CYP)2C8 and CYP3A4 in the metabolism of the anaplastic lymphoma kinase inhibitor brigatinib. A 3-arm, open-label, randomized, single-dose, fixed-sequence crossover study was conducted to characterize the effects of the strong inhibitors gemfibrozil (of CYP2C8) and itraconazole (of CYP3A) and the strong inducer rifampin (of CYP3A) on the single-dose pharmacokinetics of brigatinib. Healthy subjects (n = 20 per arm) were administered a single dose of brigatinib (90 mg, arms 1 and 2; 180 mg, arm 3) alone in treatment period 1 and coadministered with multiple doses of gemfibrozil 600 mg twice daily (BID; arm 1), itraconazole 200 mg BID (arm 2), or rifampin 600 mg daily (QD; arm 3) in period 2. Compared with brigatinib alone, coadministration of gemfibrozil with brigatinib did not meaningfully affect brigatinib area under the plasma concentration-time curve (AUC0-inf ; geometric least-squares mean [LSM] ratio [90%CI], 0.88 [0.83-0.94]). Coadministration of itraconazole with brigatinib increased AUC0-inf (geometric LSM ratio [90%CI], 2.01 [1.84-2.20]). Coadministration of rifampin with brigatinib substantially reduced AUC0-inf (geometric LSM ratio [90%CI], 0.20 [0.18-0.21]) compared with brigatinib alone. The treatments were generally tolerated. Based on these results, strong CYP3A inhibitors and inducers should be avoided during brigatinib treatment. If concomitant use of a strong CYP3A inhibitor is unavoidable, the results of this study support a dose reduction of brigatinib by approximately 50%. Furthermore, CYP2C8 is not a meaningful determinant of brigatinib clearance, and no dose modifications are needed during coadministration of brigatinib with CYP2C8 inhibitors.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Organophosphorus Compounds/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Administration, Oral , Adult , Aged , Anaplastic Lymphoma Kinase/metabolism , Area Under Curve , Cross-Over Studies , Cytochrome P-450 CYP2B6 Inducers/administration & dosage , Cytochrome P-450 CYP2B6 Inducers/pharmacokinetics , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C8 Inhibitors/administration & dosage , Cytochrome P-450 CYP2C8 Inhibitors/pharmacokinetics , Drug Interactions , Drug Therapy, Combination , Female , Gemfibrozil/administration & dosage , Gemfibrozil/pharmacokinetics , Healthy Volunteers , Humans , Itraconazole/administration & dosage , Itraconazole/pharmacokinetics , Lung Neoplasms/pathology , Male , Middle Aged , Organophosphorus Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Rifampin/administration & dosage , Rifampin/pharmacokinetics
20.
Anal Chem ; 92(1): 740-748, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31750649

ABSTRACT

The dynamics of mitochondria in live cells play a pivotal role in biological events such as cell metabolism, early stage apoptosis, and cell differentiation. Triphenylphosphonium (TPP) is a commonly used mitochondria-targeting agent for mitochondrial studies. However, there has been a lack of understanding in intracellular behaviors of TPP in the course of targeting mitochondria due to the difficulty in tracking and quantifying small molecules in a biological environment. Here, we report the utility of hyperspectral stimulated Raman scattering (SRS) microscopy associated with a Raman tag synthesized for real-time visualization and quantitation of TPP dynamics within live cells at the subcellular level. With the myriad of merits offered by a synthesized aryl-diyne-based Raman tag such as excellent photostability, negligible background interferences, and a linear dependence of the SRS signal on the TPP concentration, we successfully establish a quantitative model to associate the mitochondrial membrane potential with the key pharmacokinetic parameters of TPP inside the live cells. The model reveals that reduction in the mitochondrial membrane potential leads to significant decreases in both the uptake rate and intracellular concentrations of TPP. Further, on the basis of the multiplexed SRS images concurrently highlighting the cellular proteins and lipids without further labeling, we find that the TPP uptake causes little cytotoxicity to the host cells. The bioorthogonal hyperspectral SRS microscopy imaging reveals that TPP can maintain stable affinity to mitochondria during the restructuring of mitochondrial networking, demonstrating its great potential for real-time monitoring of pharmacokinetics of small molecules associated with live biological hosts, thereby promoting the development of mitochondria-targeting imaging probes and therapies in the near future.


Subject(s)
Mitochondria/metabolism , Nonlinear Optical Microscopy/methods , Organophosphorus Compounds/pharmacokinetics , Cell Survival , Equipment Design , HeLa Cells , Humans , Indicators and Reagents , Nonlinear Optical Microscopy/instrumentation , Organophosphorus Compounds/analysis
SELECTION OF CITATIONS
SEARCH DETAIL