Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 190
Filtrar
1.
Drug Deliv ; 28(1): 1822-1835, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34515590

RESUMEN

Brain-targeting delivery of 1,1'-methylenebis[4-[(hydroxyimino)methyl]-pyridinium] dimethanesulfonate (MMB4 DMS) is limited by its hydrophilic property and chemical instability. In order to solve this problem, herein, we develop a facile protocol through combining antisolvent precipitation and emulsion-solvent evaporation method to synthesize midazolam (MDZ) coated MMB4 DMS (MMB4@MDZ) nanoparticles. The as-prepared MMB4@MDZ had a MMB4 DMS nanocrystal (MMB4-NC) core and a MDZ shell. The MDZ shell prevented the MMB4-NC core from contacting the aqueous environment, and thus, guaranteed the chemical stability of MMB4 DMS. Most charmingly, the iron mimic cyclic peptide CRTIGPSVC (CRT) was modified on MMB4@MDZ surfaces to produce CRT-MMB4@MDZ which was endowed with ability to absorb transferrin (Tf)-abundant corona. Taking advantages of the Tf-abundant corona, CRT-MMB4@MDZ achieved transferrin receptor (TfR)-mediated brain-targeting delivery. With the fascinating chemical stability and brain-targeting delivery effect, CRT-MMB4@MDZ showed great clinical transform prospect as a brand-new nanomedicine. Of particular importance, this work promised not only a core-shell carrier-free nanomedicine platform for effective delivery of unstable water-soluble drug, but also a protein corona-manipulating strategy for targeting delivery.


Asunto(s)
Antídotos/farmacocinética , Encéfalo/metabolismo , Midazolam/farmacocinética , Nanopartículas/química , Oximas/química , Animales , Antídotos/administración & dosificación , Línea Celular , Relación Dosis-Respuesta a Droga , Liberación de Fármacos , Estabilidad de Medicamentos , Masculino , Ratones , Ratones Endogámicos C57BL , Midazolam/administración & dosificación , Tamaño de la Partícula , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores de Transferrina/metabolismo , Propiedades de Superficie
2.
Drug Deliv ; 28(1): 1455-1465, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34236248

RESUMEN

Chromium poisoning has become one of the most common heavy metal poisoning occupational diseases with high morbidity and mortality. However, most antidotes detoxify the whole body and are highly toxic. To achieve hepato-targeted chromium poisoning detoxification, a novel hepato-targeted strategy was developed using aging erythrocyte membranes (AEMs) as biomimetic material coated with a dimercaptosuccinic acid (DMSA) nanostructured lipid carrier to construct a biomimetic nano-drug delivery system. The particle size, potential, drug loading, encapsulation rate, in vitro release, and stability of the nanoparticles (NPs) were characterized. Confocal microscopy and flow cytometry showed that the prepared NPs could be phagocytized by RAW264.7 macrophage cells. The efficacy of AEM-DMSA-NPs for targeted liver detoxification was evaluated by in vitro MTT analysis and an in vivo model of chromium poisoning. The results showed that the NPs could safely and efficiently achieve targeted liver chromium poisoning detoxification. All the results indicated that the biomimetic nano-drug delivery system mediated by aging erythrocyte membranes and containing DMSA nanoparticles could be used as a novel therapeutic drug delivery system potentially targeting liver detoxification.


Asunto(s)
Antídotos/farmacología , Materiales Biomiméticos/metabolismo , Cromo/envenenamiento , Membrana Eritrocítica/metabolismo , Nanopartículas/química , Succímero/farmacología , Animales , Antídotos/administración & dosificación , Antídotos/farmacocinética , Química Farmacéutica , Portadores de Fármacos , Liberación de Fármacos , Hígado/efectos de los fármacos , Masculino , Ratones , Tamaño de la Partícula , Células RAW 264.7 , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Succímero/administración & dosificación , Succímero/farmacocinética
3.
Biochem Biophys Res Commun ; 555: 32-39, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33812056

RESUMEN

Protein-protein (e.g., antibody-antigen) interactions comprise multiple weak interactions. We have previously reported that lipid nanoparticles (LNPs) bind to and neutralize target toxic peptides after multifunctionalization of the LNP surface (MF-LNPs) with amino acid derivatives that induce weak interactions; however, the MF-LNPs aggregated after target capture and showed short blood circulation times. Here we optimized polyethylene glycol (PEG)-modified MF-LNPs (PEG-MF-LNPs) to inhibit the aggregation and increase the blood circulation time. Melittin was used as a target toxin, and MF-LNPs were prepared with negatively charged, hydrophobic, and neutral amino-acid-derivative-conjugated functional lipids. In this study, MF-LNPs modified with only PEG5k (PEG5k-MF-LNPs) and with both PEG5k and PEG2k (PEGmix-MF-LNPs) were prepared, where PEG5k and PEG2k represent PEG with a molecular weight of 5000 and 2000, respectively. PEGylation of the MF-LNPs did not decrease the melittin neutralization ability of nonPEGylated MF-LNPs, as tested by hemolysis assay. The PEGmix-MF-LNPs showed better blood circulation characteristics than the PEG5k-MF-LNPs. Although the nonPEGylated MF-LNPs immediately aggregated when mixed with melittin, the PEGmix-MF-LNPs did not aggregate. The PEGmix-MF-LNPs dramatically increased the survival rate of melittin-treated mice, whereas the nonPEGylated MF-LNPs increased slightly. These results provide a fundamental strategy to improve the in vivo toxin neutralization ability of MF-LNPs.


Asunto(s)
Antídotos/farmacología , Meliteno/toxicidad , Nanopartículas Multifuncionales/química , Polietilenglicoles/química , Animales , Antídotos/química , Antídotos/farmacocinética , Bovinos , Línea Celular , Hemólisis/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Lípidos/química , Masculino , Meliteno/sangre , Meliteno/metabolismo , Meliteno/farmacocinética , Ratones Endogámicos BALB C , Nanopartículas Multifuncionales/administración & dosificación , Nanopartículas Multifuncionales/metabolismo , Distribución Tisular
4.
Toxins (Basel) ; 13(2)2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33573016

RESUMEN

Ricin, a highly toxic protein from Ricinus communis, is considered a potential biowarfare agent. Despite the many data available, no specific treatment has yet been approved. Due to their ability to provide immediate protection, antibodies (Abs) are an approach of choice. However, their high specificity might compromise their capacity to protect against the different ricin isoforms (D and E) found in the different cultivars. In previous work, we have shown the neutralizing potential of different Abs (43RCA-G1 (anti ricin A-chain) and RB34 and RB37 (anti ricin B-chain)) against ricin D. In this study, we evaluated their protective capacity against both ricin isoforms. We show that: (i) RB34 and RB37 recognize exclusively ricin D, whereas 43RCA-G1 recognizes both isoforms, (ii) their neutralizing capacity in vitro varies depending on the cultivar, and (iii) there is a synergistic effect when combining RB34 and 43RCA-G1. This effect is also demonstrated in vivo in a mouse model of intranasal intoxication with ricin D/E (1:1), where approximately 60% and 40% of mice treated 0 and 6 h after intoxication, respectively, are protected. Our results highlight the importance of evaluating the effectiveness of the Abs against different ricin isoforms to identify the treatment with the broadest spectrum neutralizing effect.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Antídotos/farmacología , Intoxicación/prevención & control , Ricina/antagonistas & inhibidores , Ricinus/metabolismo , Animales , Especificidad de Anticuerpos , Antídotos/farmacocinética , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada , Femenino , Humanos , Células Jurkat , Dosificación Letal Mediana , Ratones Endogámicos BALB C , Intoxicación/inmunología , Isoformas de Proteínas , Ricina/inmunología , Ricina/aislamiento & purificación , Ricina/envenenamiento , Ricinus/crecimiento & desarrollo
5.
Clin Biochem ; 91: 31-38, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33444605

RESUMEN

OBJECTIVES: Hydroxocobalamin (OHCob) is an antidote for cyanide poisoning in patients rescued from house fires and is known to cause interference with certain laboratory tests. Consensus is lacking on the extent of this interference and on how to handle these samples. The objectives of this study were to characterize OHCob interference across a wide range of laboratory tests and to develop protocols for identifying and reporting these samples. DESIGNS & METHODS: Patient plasma samples (n = 5) were spiked with OHCob (1.5 mg/mL) and compared to controls without this drug. A series of analytes were measured using chemistry, urinalysis, coagulation, hematology, and blood gas instruments. Dose-response testing was performed on a subset of assays that showed interferences ≥10%. RESULTS: Of the 77 analytes evaluated, 27 (35%) showed interference from OHCob, with chemistry and coagulation analytes showing the greatest effects. Of those affected, 22 analytes had a positive interference, whereas 5 analytes had negative interference. Dose-response studies showed dose-dependent increases and/or decreases consistent with initial spiking studies. Although red in colour, plasma samples with OHCob did not trigger hemolysis index flags, necessitating a special sample identification and reporting protocol. CONCLUSION: OHCob had significant effects on several analytes across different instruments. These findings led to the development of special sample handling and reporting protocols to identify OHCob samples and ensure only accurate results are released. It is vital for emergency departments to document and notify their laboratories whenever blood samples from these patients are drawn.


Asunto(s)
Antídotos/farmacocinética , Análisis Químico de la Sangre , Hidroxocobalamina/farmacocinética , Intoxicación , Cianuro de Potasio , Antídotos/administración & dosificación , Femenino , Humanos , Hidroxocobalamina/administración & dosificación , Masculino , Intoxicación/sangre , Intoxicación/tratamiento farmacológico
6.
Toxicology ; 444: 152578, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32898602

RESUMEN

The nanotechnological approach is an innovative strategy of high potential to achieve reactivation of organophosphorus-inhibited acetylcholinesterase in central nervous system. It was previously shown that pralidoxime chloride-loaded solid lipid nanoparticles (2-PAM-SLNs) are able to protect the brain against pesticide (paraoxon) central toxicity. In the present work, we increased brain AChE reactivation efficacy by PEGylation of 2-PAM-SLNs using PEG-lipid N-(carbonyl-methoxypolyethylene glycol-2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine, sodium salt) (DSPE-PEG2000) as a surface-modifier of SLNs. To perform pharmacokinetic study, a simple, sensitive (LLOQ 1.0 ng/mL) high-performance liquid chromatography tandem mass spectrometry with atmospheric pressure chemical ionization by multiple reaction monitoring mode (HPLC-APCI-MS) was developed. The method was compared to mass spectrometry with electrospray ionization. The method was validated for linearity, accuracy, precision, extraction recovery, matrix effect and stability. Acetophenone oxime was used as the internal standard for the quantification of 2-PAM in rat plasma and brain tissue after intravenous administration. 2-PAM-DSPE-PEG2000-SLNs of mean size about 80 nm (PDI = 0.26), zeta-potential of -55 mV and of high in vitro stability, prolonged the elimination phase of 2-PAM from the bloodstream more than 3 times compared to free 2-PAM. An increase in reactivation of POX-inhibited human brain acetylcholinesterase up to 36.08 ± 4.3 % after intravenous administration of 2-PAM-DSPE-PEG2000-SLNs (dose of 2-PAM is 5 mg/kg) was achieved. The result is one of the first examples where this level of brain acetylcholinesterase reactivation was achieved. Thus, the implementation of different approaches for targeting and modifying nanoparticles' surface gives hope for improving the antidotal treatment of organophosphorus poisoning by marketed reactivators.


Asunto(s)
Antídotos/administración & dosificación , Inhibidores de la Colinesterasa/toxicidad , Reactivadores de la Colinesterasa/administración & dosificación , Nanopartículas/administración & dosificación , Compuestos de Pralidoxima/administración & dosificación , Acetilcolinesterasa/metabolismo , Animales , Antídotos/química , Antídotos/farmacocinética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Reactivadores de la Colinesterasa/sangre , Reactivadores de la Colinesterasa/química , Reactivadores de la Colinesterasa/farmacocinética , Liberación de Fármacos , Femenino , Humanos , Lípidos/administración & dosificación , Lípidos/química , Lípidos/farmacocinética , Masculino , Nanopartículas/química , Compuestos Organofosforados/toxicidad , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Compuestos de Pralidoxima/sangre , Compuestos de Pralidoxima/química , Compuestos de Pralidoxima/farmacocinética , Ratas Wistar , Propiedades de Superficie
7.
Neurobiol Dis ; 133: 104455, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31022458

RESUMEN

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.


Asunto(s)
Reactivadores de la Colinesterasa/farmacocinética , Agentes Nerviosos , Compuestos Organofosforados , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Animales , Antídotos/farmacocinética , Humanos , Agentes Nerviosos/farmacocinética , Agentes Nerviosos/toxicidad , Compuestos Organofosforados/farmacocinética , Compuestos Organofosforados/toxicidad , Oximas/farmacocinética
8.
Am J Cardiovasc Drugs ; 20(3): 295-299, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31709499

RESUMEN

BACKGROUND: Clinical experience with using activated prothrombin complex concentrates (aPCCs) to reverse the effects of factor Xa inhibitors is limited. OBJECTIVES: Our objective was to assess the achievement of effective clinical hemostasis using aPCC in patients on chronic apixaban or rivaroxaban therapy presenting with major bleeding in whom a reversal agent is warranted. We also assessed the safety of the drug. METHODS: A retrospective medical records review was conducted at a tertiary referral medical center in the USA. Patients presenting with major bleeding while receiving apixaban or rivaroxaban and treated with aPCC were included. Clinical hemostasis was assessed using International Society of Thrombosis and Hemostasis Scientific and Standardization Subcommittee criteria. RESULTS: A total of 35 patients were included in the study. The most common site of bleeding was intracerebral hemorrhage (ICH) (n = 18 [51.4%]), followed by gastrointestinal bleed (n = 10 [28.6%]). Clinical hemostasis was achieved in 24 (68.6%) patients; 11 patients (31.4%) did not achieve clinical hemostasis; nine of these patients had ICH. Seven of the patients who did not achieve hemostasis died during hospitalization. Three (8.6%) patients experienced thromboembolic events during hospitalization. In total, 21 (60%) patients were receiving concomitant medications that interact with anti-factor Xa inhibitors and can increase the risk of bleeding. CONCLUSIONS: Our study suggests that aPCC could be an option in patients with major bleeding associated with apixaban or rivaroxaban. It may be an alternative for patients who need anticoagulation reversal if the specific antidote, andexanet alfa, is unavailable.


Asunto(s)
Factores de Coagulación Sanguínea , Coagulación Sanguínea/efectos de los fármacos , Hemorragia Gastrointestinal , Hemorragias Intracraneales , Pirazoles/efectos adversos , Piridonas/efectos adversos , Rivaroxabán/efectos adversos , Anciano , Antídotos/administración & dosificación , Antídotos/efectos adversos , Antídotos/farmacocinética , Factores de Coagulación Sanguínea/administración & dosificación , Factores de Coagulación Sanguínea/efectos adversos , Factores de Coagulación Sanguínea/farmacocinética , Inhibidores del Factor Xa/efectos adversos , Femenino , Hemorragia Gastrointestinal/sangre , Hemorragia Gastrointestinal/inducido químicamente , Hemorragia Gastrointestinal/tratamiento farmacológico , Hemorragia Gastrointestinal/mortalidad , Hemostasis/efectos de los fármacos , Humanos , Hemorragias Intracraneales/sangre , Hemorragias Intracraneales/inducido químicamente , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/mortalidad , Masculino , Evaluación de Procesos y Resultados en Atención de Salud , Estudios Retrospectivos , Análisis de Supervivencia , Trombosis/diagnóstico , Trombosis/etiología , Estados Unidos/epidemiología
9.
Tidsskr Nor Laegeforen ; 139(13)2019 Sep 24.
Artículo en Noruego, Inglés | MEDLINE | ID: mdl-31556537

RESUMEN

BACKGROUND: Bystander administration with naloxone nasal spray can prevent deaths from opioid overdose. To achieve optimal nasal absorption of naloxone, the spray must be administered at low volume with high concentration of the drug. The study aimed to investigate the bioavailability and absorption pattern for a new naloxone nasal spray. MATERIAL AND METHOD: In an open, randomised, two-way crossover study undertaken in five healthy men, naloxone 2 mg (20 mg/ml) in nasal spray was compared with 1 mg intravenously administered naloxone. A total of 15 blood samples were taken over a period of six hours after administration. The drug concentration was determined using liquid chromatography tandem-mass spectrometry. Pharmacokinetic variables were calculated using non-compartmental analysis. RESULTS: Bioavailability for intranasal naloxone was 47 % (minimum-maximum values 24-66 %). Maximum concentration (Cmax) was 4.2 (1.5-7.1) ng/ml, and this was achieved (Tmax ) after 16 (5-25) minutes. INTERPRETATION: The nasal spray resulted in a rapid systemic absorption with higher serum concentrations than intravenous naloxone 10-240 minutes after intake. The pilot study indicated that the highly concentrated nasal spray may provide a therapeutic dose of naloxone with a single spray actuation. The findings led to further commercial development of the medication.


Asunto(s)
Antídotos , Naloxona , Rociadores Nasales , Administración Intravenosa , Adulto , Analgésicos Opioides/envenenamiento , Antídotos/administración & dosificación , Antídotos/farmacocinética , Disponibilidad Biológica , Estudios Cruzados , Sobredosis de Droga/tratamiento farmacológico , Cromatografía de Gases y Espectrometría de Masas , Humanos , Masculino , Naloxona/administración & dosificación , Naloxona/farmacocinética , Proyectos Piloto , Espectrometría de Masas en Tándem , Adulto Joven
10.
J Pharmacokinet Pharmacodyn ; 46(6): 513-529, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31396799

RESUMEN

The primary goal of this work was to develop a computational tool to enable personalized prediction of pharmacological disposition and associated responses for opioids and antidotes. Here we present a computational framework for physiologically-based pharmacokinetic (PBPK) modeling of an opioid (morphine) and an antidote (naloxone). At present, the model is solely personalized according to an individual's mass. These PK models are integrated with a minimal pharmacodynamic model of respiratory depression induction (associated with opioid administration) and reversal (associated with antidote administration). The model was developed and validated on human data for IV administration of morphine and naloxone. The model can be further extended to consider different routes of administration, as well as to study different combinations of opioid receptor agonists and antagonists. This work provides the framework for a tool that could be used in model-based management of pain, pharmacological treatment of opioid addiction, appropriate use of antidotes for opioid overdose and evaluation of abuse deterrent formulations.


Asunto(s)
Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacocinética , Antídotos/efectos adversos , Antídotos/farmacocinética , Analgésicos Opioides/administración & dosificación , Antídotos/administración & dosificación , Humanos , Masculino , Morfina/efectos adversos , Morfina/farmacocinética , Naloxona/administración & dosificación , Naloxona/efectos adversos , Naloxona/farmacocinética , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/efectos adversos , Antagonistas de Narcóticos/farmacocinética , Trastornos Relacionados con Opioides/tratamiento farmacológico , Dolor/tratamiento farmacológico , Receptores Opioides/metabolismo
12.
Chem Biol Interact ; 310: 108737, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31279792

RESUMEN

AIMS: K117 and K127 are bis-pyridinium aldoximes but K117 is a bis-pyridinium bis-aldoxime while K127 has only one single aldoxime in addition to its amide substituent. Is there any difference in pharmacokinetics in these compounds that otherwise have the same chemical structure? Both K117 and K127 are developed as antidotes in acetylcholinesterase and butyrylcholinesterase poisoning in terrorist attacks or intoxication with other organophosphorous compounds. Their distributions have been scouted in the bodies of rats. MAIN METHODS: White male Wistar rats were intramuscularly injected. The animals were sacrificed, tissue samples were homogenized, and either K117 or K127 concentrations were determined using reversed-phase high-performance liquid chromatography. KEY FINDINGS: Both K117 and K127 were present in all tissues that were analyzed including blood (serum), the brains, cerebrospinal fluid, the eyes, livers, kidneys, lungs and testes. Their pharmacokinetics and body distributions are similar. SIGNIFICANCE: Either K117 or K127 meets the essential requirements for antidotes. Dose dependence and kinetics of their distribution were compared to that of other pyridinium aldoximes.


Asunto(s)
Antídotos/farmacocinética , Organofosfatos/antagonistas & inhibidores , Oximas/farmacocinética , Compuestos de Piridinio/farmacocinética , Acetilcolinesterasa/química , Animales , Butirilcolinesterasa/química , Sustancias para la Guerra Química/farmacocinética , Inhibidores de la Colinesterasa/farmacocinética , Reactivadores de la Colinesterasa/farmacocinética , Oximas/análisis , Compuestos de Piridinio/análisis , Ratas , Ratas Wistar , Distribución Tisular
13.
AAPS PharmSciTech ; 20(6): 221, 2019 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-31214899

RESUMEN

Fomepizole is used as an antidote to treat methanol poisoning due to its selectivity towards alcohol dehydrogenase. In the present study, the goal is to develop a method to predict the fomepizole human plasma concentration versus time profile based on the preclinical pharmacokinetics using the assumption of superimposability on simulated time course profiles of animals and humans. Standard allometric equations with/without correction factors were also assimilated in the prediction. The volume of distribution at steady state (Vss) predicted by simple allometry (57.55 L) was very close to the reported value (42.17 L). However, clearance (CL) prediction by simple allometry was at least 3-fold higher to the reported value (33.86 mL/min); hence, multiple correction factors were used to predict the clearance. Both brain weight and maximum life span potential could predict the CL with 1.22- and 1.01-fold difference. Specifically, the predicted Vss and CL values via interspecies scaling were used in the prediction of series of human intravenous pharmacokinetic parameters, while the simulation of human oral profile was done by the use of absorption rate constant (Ka) from dog following the applicability of human bioavailability value scaled from dog data. In summary, the findings indicate that the utility of diverse allometry approaches to derive the human pharmacokinetics of fomepizole after intravenous/oral dosing.


Asunto(s)
Antídotos/farmacocinética , Fomepizol/farmacocinética , Administración Intravenosa , Animales , Antídotos/administración & dosificación , Disponibilidad Biológica , Fomepizol/administración & dosificación , Fomepizol/sangre , Humanos , Masculino , Ratones , Modelos Biológicos , Conejos , Ratas
14.
Ann Emerg Med ; 74(3): 423-429, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31080026

RESUMEN

STUDY OBJECTIVE: Cyanide is a deadly poison, particularly with oral exposure, in which larger doses can occur before any symptoms develop. Multiple governmental agencies highlight oral cyanide as an agent that can be used in a terrorist attack because it can be easily weaponized and is readily available. Currently, there are no Food and Drug Administration-approved antidotes specifically for oral cyanide. An oral countermeasure that can neutralize and prevent absorption of cyanide from the gastrointestinal tract after oral exposure is needed. The objective of this study is to determine if the combination of glycine and sodium thiosulfate administered orally is effective in reducing mortality in a large, swine model of oral cyanide toxicity. METHODS: Nine swine (45 to 55 kg) were instrumented, sedated, and stabilized. Potassium cyanide (at 8 mg/kg) in saline solution was delivered as a onetime bolus through an orogastric tube. Three minutes after cyanide administration, animals that were randomized to the treatment group received sodium thiosulfate (508.2 mg/kg, 3.25-M solution) and glycine (30 mg/kg, 3.5-M solution) through an orogastric tube. Survival at 60 minutes was the primary outcome. We compared survival between groups by log-rank Mantel-Cox analysis and trended laboratory results and vital signs. RESULTS: At baseline and treatment, all animals were similar. Survival at 60 minutes was 100% in treated animals compared with 0% in the control group (P=.003). By the study end, defined as death or 60 minutes after cyanide administration, there was a significant difference in the lactate concentration between the treatment and control groups (control 9.43 mmol/L [SD 4.08]; treatment 1.66 mmol/L [SD 0.82]; difference between means 7.69 mmol/L [SD 2.07]; 95% confidence interval difference -14.05 to -1.32). Mean arterial pressure was significantly different between the treatment and control groups at study end (control 26 mm Hg [SD 6.7]; treatment 81 mm Hg [SD 14]; difference between means 55.2 mm Hg [SD 7.1]; 95% confidence interval difference 37.8 to 72.6). pH and oxygen saturation were also significantly different between the treatment and control groups at study end. CONCLUSION: The combination of oral sodium thiosulfate and glycine significantly improved survival and physiologic parameters in a large-animal model of oral cyanide toxicity.


Asunto(s)
Antídotos/administración & dosificación , Glicina/administración & dosificación , Cianuro de Potasio/envenenamiento , Tiosulfatos/administración & dosificación , Administración Oral , Animales , Antídotos/farmacocinética , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Glicina/farmacología , Humanos , Venenos , Distribución Aleatoria , Porcinos , Tiosulfatos/farmacología , Factores de Tiempo
15.
Theranostics ; 9(3): 633-645, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809298

RESUMEN

Accidental or suicidal ingestion of the world's most widely used herbicide, paraquat (PQ), may result in rapid multi-organ failure with a 60% fatality rate due to the absence of an effective detoxification solution. Effective, specific antidotes to PQ poisoning have been highly desired. Methods: The binding constant of PQ and a synthetic receptor, cucurbit[7]uril (CB[7]), was first determined in various pH environments. The antidotal effects of CB[7] on PQ toxicity were firstly evaluated with in-vitro cell lines. With in-vivo mice models, the pharmacokinetics and the biodistribution of PQ in major organs were determined to evaluate the influence of CB[7] on the oral bioavailability of PQ. Major organs' injuries and overall survival rates of the mice were systemically examined to evaluate the therapeutic efficacy of CB[7] on PQ poisoning. Results: We demonstrate that CB[7] may complex PQ strongly under various conditions and significantly reduce its toxicity in vitro and in vivo. Oral administration of PQ in the presence of CB[7] in a mouse model significantly decreased PQ levels in the plasma and major organs and alleviated major organs' injuries, when compared to those of mice administered with PQ alone. Further studies indicated that oral administration of CB[7] within 2 h post PQ ingestion significantly increased the survival rates and extended the survival time of the mice, in contrast to the ineffective treatment by activated charcoal, which is commonly recommended for PQ decontamination. Conclusion: CB[7] may be used as a specific oral antidote for PQ poisoning by strongly binding with PQ and inhibiting its absorption in the gastrointestinal tracts.


Asunto(s)
Antídotos/administración & dosificación , Hidrocarburos Aromáticos con Puentes/administración & dosificación , Herbicidas/antagonistas & inhibidores , Imidazoles/administración & dosificación , Paraquat/antagonistas & inhibidores , Intoxicación/terapia , Receptores Artificiales/administración & dosificación , Administración Oral , Estructuras Animales/patología , Animales , Antídotos/farmacocinética , Antídotos/farmacología , Hidrocarburos Aromáticos con Puentes/farmacocinética , Hidrocarburos Aromáticos con Puentes/farmacología , Línea Celular , Herbicidas/toxicidad , Imidazoles/farmacocinética , Imidazoles/farmacología , Ratones , Paraquat/toxicidad , Análisis de Supervivencia
16.
PLoS One ; 14(1): e0209350, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30615630

RESUMEN

BACKGROUND: Idarucizumab is a humanized Fab fragment that specifically reverses dabigatran anticoagulation. In trauma, volume expanders are used for resuscitation to compensate for blood loss and hemorrhagic shock, but it is unknown whether volume expanders influence the binding of dabigatran to its antidote. Using a porcine dilutional coagulopathy model, this study investigated whether volume replacement strategies affect binding of dabigatran to idarucizumab. METHODS: Twenty-five male pigs were treated orally with dabigatran etexilate (30 mg/kg bid) for 3 days. The following day, animals were anesthetized, infused with dabigatran (total dose 0.645 mg/kg) to achieve supratherapeutic concentrations, and randomized 1:1:1:1:1 (n = 5 per group) to control (no hemodilution) or hemodilution where ~50% of blood volume was substituted with Ringer's solution, 6% hydroxyethyl starch 130/0.4, 6% hydroxyethyl starch 200/0.5 or 4% gelatin. Idarucizumab was then administered intravenously (30 mg/kg) and serial blood samples were taken for up to 24 hours to measure diluted thrombin time (corresponding with dabigatran activity), total dabigatran (bound to antidote and free drug) and a panel of coagulation parameters. RESULTS: Mean plasma dabigatran levels were 617 ± 16 ng/mL after infusion and 600 ± 114 ng/mL after ~50% hemodilution with no significant differences between groups. Following treatment with idarucizumab, plasma concentrations of unbound dabigatran decreased markedly, with similar reductions in all groups. Dabigatran-induced prolongation of coagulation parameters was rapidly reversed in all groups. CONCLUSION: This study indicates that several volume expanders used for resuscitation in trauma do not interfere with the binding of idarucizumab to dabigatran.


Asunto(s)
Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacocinética , Antídotos/farmacocinética , Antitrombinas/sangre , Antitrombinas/farmacocinética , Dabigatrán/antagonistas & inhibidores , Dabigatrán/farmacocinética , Hemodilución , Animales , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/terapia , Volumen Sanguíneo/fisiología , Dabigatrán/sangre , Masculino , Modelos Animales , Sustitutos del Plasma/administración & dosificación , Sustitutos del Plasma/metabolismo , Sus scrofa
17.
J Pharmacol Exp Ther ; 367(2): 363-372, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30190337

RESUMEN

In the development of antidotal therapy for treatment of organophosphate exposure from pesticides used in agriculture and nerve agents insidiously employed in terrorism, the alkylpyridinium aldoximes have received primary attention since their early development by I. B. Wilson in the 1950s. Yet these agents, by virtue of their quaternary structure, are limited in rates of crossing the blood-brain barrier, and they require administration parenterally to achieve full distribution in the body. Oximes lacking cationic charges or presenting a tertiary amine have been considered as alternatives. Herein, we examine the pharmacokinetic properties of a lead ionizable, zwitterionic hydroxyiminoacetamido alkylamine in mice to develop a framework for studying these agents in vivo and generate sufficient data for their consideration as appropriate antidotes for humans. Consequently, in vitro and in vivo efficacies of immediate structural congeners were explored as leads or backups for animal studies. We compared oral and parenteral dosing, and we developed an intramuscular loading and oral maintenance dosing scheme in mice. Steady-state plasma and brain levels of the antidote were achieved with sequential administrations out to 10 hours, with brain levels exceeding plasma levels shortly after administration. Moreover, the zwitterionic oxime showed substantial protection after gavage, whereas the classic methylpyridinium aldoxime (2-pyridinealdoxime methiodide) was without evident protection. Although further studies in other animal species are necessary, ionizing zwitterionic aldoximes present viable alternatives to existing antidotes for prophylaxis and treatment of large numbers of individuals in terrorist-led events with nerve agent organophosphates, such as sarin, and in organophosphate pesticide exposure.


Asunto(s)
Antídotos/farmacología , Antídotos/farmacocinética , Intoxicación por Organofosfatos/tratamiento farmacológico , Organofosfatos/efectos adversos , Administración Oral , Animales , Encéfalo/efectos de los fármacos , Femenino , Plomo/efectos adversos , Masculino , Ratones , Agentes Nerviosos/efectos adversos , Compuestos Organofosforados/efectos adversos , Oximas/farmacocinética , Oximas/farmacología , Plaguicidas/efectos adversos , Distribución Tisular
18.
ACS Chem Neurosci ; 9(12): 3007-3014, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30071719

RESUMEN

2-Pyridinealdoxime methiodide (2-PAM) is a widely used antidote for the treatment of organophosphorus (OP) exposure that reactivates the target protein acetylcholinesterase. Carbon-11 2-PAM was prepared to more fully understand the in vivo mode of action, distribution, and dynamic qualities of this important countermeasure. Alkylation of 2-pyridinealdoxime with [11C]CH3I provided the first-in-class [11C]2-PAM tracer in 3.5% decay corrected radiochemical yield from [11C]CH3I, >99% radiochemical purity, and 4831 Ci/mmol molar activity. [11C]2-PAM tracer distribution was evaluated by ex vivo biodistribution and in vivo dynamic positron emission tomography (PET) imaging in naïve (OP exposure deficient) rats. Tracer alone and tracer coinjected with a body mass-scaled human therapeutic dose of 30 mg/kg nonradioactive 2-PAM demonstrated statistically similar tissue and blood distribution profiles with the greatest uptake in kidney and significantly lower levels in liver, heart, and lung with lesser amounts in blood and brain. The imaging and biodistribution data show that radioactivity uptake in brain and peripheral organs is rapid and characterized by differential tissue radioactivity washout profiles. Analysis of arterial blood samples taken 5 min after injection showed ∼82% parent [11C]2-PAM tracer. The imaging and biodistribution data are now established, enabling future comparisons to outcomes acquired in OP intoxicated rodent models.


Asunto(s)
Antídotos/farmacocinética , Radioisótopos de Carbono/farmacocinética , Intoxicación por Organofosfatos , Compuestos de Pralidoxima/farmacocinética , Radiofármacos/farmacocinética , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Radioisótopos de Carbono/química , Corazón/diagnóstico por imagen , Riñón/diagnóstico por imagen , Riñón/metabolismo , Hígado/diagnóstico por imagen , Hígado/metabolismo , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Miocardio/metabolismo , Tomografía de Emisión de Positrones , Compuestos de Pralidoxima/síntesis química , Trazadores Radiactivos , Radiofármacos/síntesis química , Ratas , Distribución Tisular
19.
Curr Opin Hematol ; 25(5): 382-388, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30015643

RESUMEN

PURPOSE OF REVIEW: Since the selection of the first thrombin-binding aptamer in 1992, the use of nucleic acid aptamers to target specific coagulation factors has emerged as a valuable approach for generating novel anticoagulant and procoagulant therapeutics. Herein, we highlight the most recent discoveries involving application of aptamers for those purposes. RECENT FINDINGS: Learning from the successes and pitfalls of the FIXa-targeting aptamer pegnivacogin in preclinical and clinical studies, the latest efforts to develop antidote-controllable anticoagulation strategies for cardiopulmonary bypass that avoid unfractionated heparin involve potentiation of the exosite-binding factor X (FX)a aptamer 11F7t by combination with either a small molecule FXa catalytic site inhibitor or a thrombin aptamer. Recent work has also focused on identifying aptamer inhibitors of contact pathway factors such as FXIa and kallikrein, which may prove to be well tolerated and effective antithrombotic agents in certain clinical settings. Finally, new approaches to develop procoagulant aptamers to control bleeding associated with hemophilia and other coagulopathies involve targeting activated protein C and tissue plasminogen activator. SUMMARY: Overall, these recent findings exemplify the versatility of aptamers to modulate a variety of procoagulant and anticoagulant factors, along with their capacity to be used complementarily with other aptamers or drugs for wide-ranging applications.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Factor IXa , Inhibidores del Factor Xa/uso terapéutico , Hemostasis , Animales , Antídotos/farmacocinética , Antídotos/uso terapéutico , Aptámeros de Nucleótidos/efectos adversos , Aptámeros de Nucleótidos/farmacocinética , Puente Cardiopulmonar , Dominio Catalítico , Factor IXa/antagonistas & inhibidores , Factor IXa/metabolismo , Inhibidores del Factor Xa/efectos adversos , Inhibidores del Factor Xa/farmacocinética , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Calicreínas/metabolismo
20.
Int J Toxicol ; 37(5): 352-363, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29879849

RESUMEN

Oxime reactivators are critical antidotes after organophosphate pesticide or nerve agent poisoning, directly restoring the function of inhibited acetylcholinesterase. In the continuing search for more broad-spectrum acetylcholinesterase reactivators, this study evaluated one of the leading next-generation oxime reactivators: methoxime, (1,1'-trimethylene bis[4-(hydroxyimino)methyl]pyridinium dichloride (MMB-4). The pharmacokinetics of both salts of MMB-4 (dichloride [2Cl] and dimethanesulphonate [DMS]) were characterized across a range of relevant doses (19, 58, and 116 µmol/kg, intramuscular) in a nonhuman primate model (male African green monkeys), and only subtle differences were observed between the salts. Additionally, the behavioral and cardiovascular safety of these MMB-4 salts was compared directly to other available oximes (HI-6 2Cl, HI-6 DMS, and pyridine-2-aldoxime chloride (2-PAM Cl)) at comparable projected doses. Automated operant behavioral tests were used to examine attention, motivation, visual discrimination, concept execution, and fine motor coordination after high doses of all oxime salts, and of all oximes studied, only the highest dose of 2-PAM Cl (447 µmol/kg) disrupted behavioral performance. Likewise, the effects of a range of doses of MMB-4 2Cl or DMS, HI-6 2Cl or DMS, or 2-PAM Cl on cardiovascular parameters were measured in African green monkeys implanted with telemetry devices. Only a small transient decrease in pulse pressure was observed following administration of the highest dose of MMB-4 DMS (116 µmol/kg). Thus, MMB-4 salts, up to the 9× equivalent of a projected autoinjector dose in humans, did not produce behavioral or cardiovascular toxicity in African green monkeys in the current study, and the pharmacokinetic parameters were orderly and predictable.


Asunto(s)
Antídotos , Reactivadores de la Colinesterasa , Oximas , Animales , Antídotos/farmacocinética , Antídotos/toxicidad , Conducta Animal/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Chlorocebus aethiops , Conducta de Elección/efectos de los fármacos , Reactivadores de la Colinesterasa/sangre , Reactivadores de la Colinesterasa/farmacocinética , Reactivadores de la Colinesterasa/toxicidad , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Oximas/sangre , Oximas/farmacocinética , Oximas/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...