Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Therapeutic Methods and Therapies TCIM
Database
Language
Publication year range
1.
Mol Pharm ; 18(2): 593-609, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32926630

ABSTRACT

Deferoxamine mesylate (DFO) is an FDA-approved, hexadentate iron chelator routinely used to alleviate systemic iron burden in thalassemia major and sickle cell patients. Iron accumulation in these disease states results from the repeated blood transfusions required to manage these conditions. Iron accumulation has also been implicated in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), and secondary injury following intracerebral hemorrhage (ICH). Chelation of brain iron is thus a promising therapeutic strategy for improving behavioral outcomes and slowing neurodegeneration in the aforementioned disease states, though the effectiveness of DFO treatment is limited on several accounts. Systemically administered DFO results in nonspecific toxicity at high doses, and the drug's short half-life leads to low patient compliance. Mixed reports of DFO's ability to cross the blood-brain barrier (BBB) also appear in literature. These limitations necessitate novel DFO formulations prior to the drug's widespread use in managing neurodegeneration. Herein, we discuss the various dosing regimens and formulations employed in intranasal (IN) or systemic DFO treatment, as well as the physiological and behavioral outcomes observed in animal models of AD, PD, and ICH. The clinical progress of chelation therapy with DFO in managing neurodegeneration is also evaluated. Finally, the elimination of intranasally administered particles via the glymphatic system and efflux transporters is discussed. Abundant preclinical evidence suggests that intranasal DFO treatment improves memory retention and behavioral outcome in rodent models of AD, PD, and ICH. Several other biochemical and physiological metrics, such as tau phosphorylation, the survival of tyrosine hydroxylase-positive neurons, and infarct volume, are also positively affected by intranasal DFO treatment. However, dosing regimens are inconsistent across studies, and little is known about brain DFO concentration following treatment. Systemic DFO treatment yields similar results, and some complex formulations have been developed to improve permeability across the BBB. However, despite the success in preclinical models, clinical translation is limited with most clinical evidence investigating DFO treatment in ICH patients, where high-dose treatment has proven dangerous and dosing regimens are not consistent across studies. DFO is a strong drug candidate for managing neurodegeneration in the aging population, but before it can be routinely implemented as a therapeutic agent, dosing regimens must be standardized, and brain DFO content following drug administration must be understood and controlled via novel formulations.


Subject(s)
Alzheimer Disease/drug therapy , Cerebral Hemorrhage/drug therapy , Deferoxamine/administration & dosage , Drug Carriers/chemistry , Parkinson Disease/drug therapy , Siderophores/administration & dosage , Administration, Intranasal , Alzheimer Disease/pathology , Animals , Biological Availability , Blood-Brain Barrier/metabolism , Brain/cytology , Brain/drug effects , Brain/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Deferoxamine/pharmacokinetics , Disease Models, Animal , Half-Life , Humans , Injections, Intramuscular , Injections, Intraventricular , Injections, Spinal , Injections, Subcutaneous , Iron/metabolism , Medication Adherence , Nanoparticles/chemistry , Nasal Mucosa/metabolism , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/pathology , Permeability , Siderophores/pharmacokinetics , Tissue Distribution
2.
Article in English | MEDLINE | ID: mdl-31262762

ABSTRACT

The pharmacokinetic (PK) and pharmacodynamic (PD) parameters which correlated with the in vivo efficacy of cefiderocol were evaluated using neutropenic murine thigh and lung infection models in which the infections were caused by a variety of Gram-negative bacilli. The dose fractionation study using the thigh infection model in which the infection was caused by Pseudomonas aeruginosa showed that the cumulative percentage of a 24-h period that the free drug concentration in plasma exceeds the MIC (%fT>MIC) rather than the free peak level divided by the MIC (fCmax/MIC) and the area under the free concentration-time curve over 24 h divided by the MIC (fAUC/MIC) was the PK/PD parameter that best correlated with efficacy. The study with multiple carbapenem-resistant strains revealed that the %fT>MIC determined in iron-depleted cation-adjusted Mueller-Hinton broth (ID-CAMHB) better reflected the in vivo efficacy of cefiderocol than the %fT>MIC determined in cation-adjusted Mueller-Hinton broth (CAMHB). The mean %fT>MIC of cefiderocol required for a 1-log10 reduction against 10 strains of Enterobacteriaceae and 3 strains of Pseudomonas aeruginosa in the thigh infection models were 73.3% and 77.2%, respectively. The mean %fT>MIC for Enterobacteriaceae, P. aeruginosa, Acinetobacter baumannii, and Stenotrophomonas maltophilia in the lung infection model were 64.4%, 70.3%, 88.1%, and 53.9%, respectively. These results indicate that cefiderocol has potent efficacy against Gram-negative bacilli, including carbapenem-resistant strains, irrespective of the bacterial species, in neutropenic thigh and lung infection models and that the in vivo efficacy correlated with the in vitro MIC under iron-deficient conditions.


Subject(s)
Cephalosporins/pharmacokinetics , Cephalosporins/therapeutic use , Lung/microbiology , Siderophores/therapeutic use , Thigh/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Carbapenems/pharmacokinetics , Carbapenems/therapeutic use , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae/drug effects , Enterobacteriaceae/pathogenicity , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/pathogenicity , Male , Mice , Microbial Sensitivity Tests , Protein Binding , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Siderophores/pharmacokinetics , Stenotrophomonas maltophilia/drug effects , Stenotrophomonas maltophilia/pathogenicity , Cefiderocol
3.
ACS Nano ; 12(5): 4123-4139, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29617109

ABSTRACT

Excess iron deposition in the brain often causes oxidative stress-related damage and necrosis of dopaminergic neurons in the substantia nigra and has been reported to be one of the major vulnerability factors in Parkinson's disease (PD). Iron chelation therapy using deferoxamine (DFO) may inhibit this nigrostriatal degeneration and prevent the progress of PD. However, DFO shows very short half-life in vivo and hardly penetrates the blood brain barrier (BBB). Hence, it is of great interest to develop DFO formulations for safe and efficient intracerebral drug delivery. Herein, we report a polymeric nanoparticle system modified with brain-targeting peptide rabies virus glycoprotein (RVG) 29 that can intracerebrally deliver DFO. The nanoparticle system penetrates the BBB possibly through specific receptor-mediated endocytosis triggered by the RVG29 peptide. Administration of these nanoparticles significantly decreased iron content and oxidative stress levels in the substantia nigra and striatum of PD mice and effectively reduced their dopaminergic neuron damage and as reversed their neurobehavioral deficits, without causing any overt adverse effects in the brain or other organs. This DFO-based nanoformulation holds great promise for delivery of DFO into the brain and for realizing iron chelation therapy in PD treatment.


Subject(s)
Brain/metabolism , Deferoxamine/administration & dosage , Drug Delivery Systems , Glycoproteins/chemistry , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Parkinson Disease/drug therapy , Peptide Fragments/chemistry , Viral Proteins/chemistry , Animals , Brain/drug effects , Deferoxamine/pharmacokinetics , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Glycoproteins/administration & dosage , Male , Mice , Mice, Inbred C57BL , Parkinson Disease/metabolism , Peptide Fragments/administration & dosage , Siderophores/administration & dosage , Siderophores/pharmacokinetics , Siderophores/pharmacology , Siderophores/therapeutic use , Viral Proteins/administration & dosage
4.
Int J Hematol ; 91(3): 401-12, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20213118

ABSTRACT

Iron chelators are effective at removing iron from the body in iron overload, but little is known about the handling of iron chelates by the kidney. We studied the transport of deferoxamine, deferasirox, and three hydroxypyridones, and their iron chelates, in polarized renal epithelial MDCK cells growing on Transwell inserts. Directional iron efflux was also studied in (59)Fe-loaded cells. The chelators were transported at comparable rates in the apical and basolateral directions and moved faster than their corresponding chelates, except for deferoxamine, which did not move from the basolateral to the apical side. In contrast, the chelates were transported faster in the apical-to-basolateral direction. More permeable chelators were more efficient at removing iron from iron-loaded cells compared with deferoxamine. Iron is preferentially removed from the basolateral side, and kinetic modeling suggests facilitated diffusion of chelates in some cases. Basolateral iron efflux is temperature-dependent and partially sensitive to ATP depletion. Polarized transport of chelates suggests the kidney may be involved in reabsorption of iron bound to chelators, with a temperature-sensitive facilitated removal of some iron complexes from the basolateral side. Further studies are warranted to determine if these processes may contribute to the observed nephrotoxicity of some iron chelators.


Subject(s)
Benzoates/pharmacokinetics , Deferoxamine/pharmacokinetics , Epithelial Cells/metabolism , Iron Overload/metabolism , Pyridones/pharmacokinetics , Triazoles/pharmacokinetics , Adenosine Triphosphate/metabolism , Animals , Benzoates/chemistry , Biological Transport/physiology , Cell Line , Cell Polarity/physiology , Deferasirox , Deferoxamine/chemistry , Dogs , Epithelial Cells/cytology , Epithelial Cells/drug effects , Exocytosis/physiology , Iron/pharmacokinetics , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacokinetics , Iron Radioisotopes , Kidney/cytology , Models, Biological , Pyridones/chemistry , Siderophores/chemistry , Siderophores/pharmacokinetics , Temperature , Triazoles/chemistry
5.
Biochem Pharmacol ; 65(8): 1351-60, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12694876

ABSTRACT

Our study demonstrates the capacity of FR160, a catechol iron chelator, to reach and accumulate into infected Plasmodium falciparum erythrocytes and parasites (cellular accumulation ratio between 12 and 43). Steady-state FR160 accumulation is obtained after 2 hr of exposure. After 2 hr exposure, it reaches intracellular levels that are 4- to 10-fold higher in infected red blood cells than those attained in normal erythrocytes. There is quite a good correlation between the accumulation of chloroquine and FR160 in the different strains (r=0.939) and in the IC(50) values (r=0.719). In contrast, the accumulation of FR160 and its activity is poorly correlated (r=0.500), suggesting that activity of FR160 may be independent of its penetration into infected erythrocytes. The mechanism of accumulation is yet unknown but based on inhibitor studies, the uptake of FR160 seems to be not associated with the calcium pump or channel, the potassium channel or the Na(+)/H(+) exchanger. Combinations of FR160 with verapamil, diltiazem, clotrimazole, amiloride, diazoxide, 4-aminopyridine, and picrotoxin should be avoided (antagonistic effects). The potent in vitro activity of FR160 on chloroquine-resistant strains or isolates, its lower toxicity against Vero cells, its mechanisms of action, its capacity to reach rapidly and accumulate into infected erythrocytes suggest that FR160 holds much promise as a new structural lead and effective antimalarial agent or at least a promising adjuvant in treatment of malaria.


Subject(s)
Chloroquine/blood , Erythrocytes/metabolism , Erythrocytes/parasitology , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Spermidine/analogs & derivatives , Spermidine/blood , Animals , Biological Transport , Chloroquine/pharmacokinetics , Humans , Iron/metabolism , Kinetics , Siderophores/blood , Siderophores/pharmacokinetics , Spermidine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL