Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Anticancer Drugs ; 32(4): 365-375, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33595947

RESUMEN

Patients with advanced breast cancer often develop bone metastases. Treatment is limited to palliative care. Parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) antagonists for bone metastases failed clinically due to short half-life and inadequate concentration in bone. We synthesized two novel PTHrP antagonists fused to an inert bacterial collagen binding domain (CBD) that directs drugs to bone. PTH(7-33)-CBD is an N-terminal truncated PTHrP antagonist. [W2]PTH(1-33)-CBD is an PTHrP inverse-agonist. The aim of this study was to assess PTH(7-33)-CBD to reduce breast cancer bone metastases and prevent osteolytic destruction in mice and to assess both drugs for apoptosis of breast cancer cells in vitro and inhibition of PTH receptor (PTHR1). PTH(7-33)-CBD (1000 µg/kg, subcutaneous) or vehicle was administered 24 h prior to MDA-MB-231 breast cancer cell inoculation into the tibia marrow. Weekly tumor burden and bone density were measured. Pharmacokinetic analysis of PTH(7-33)-CBD in rat serum was evaluated. Drug effect on cAMP accumulation in SaOS-2 osteosarcoma cells and apoptosis of MDA-MB-231 cells was assessed. PTH(7-33)-CBD reduced MDA-MB-231 tumor burden and osteolytic destruction in mice 4-5 weeks post-treatment. PTH(7-33)-CBD (1000 µg/kg i.v. and subcutaneous) in rats was rapidly absorbed with peak concentration 5-min and terminal half-life 3-h. Bioavailability by the subcutaneous route was 43% relative to the i.v. route. PTH(7-33)-CBD was detected only on rat periosteal bone surfaces that stained positive for collagen-1. PTH(7-33)-CBD and [W2]PTH(1-33)-CBD (10-8M) blocked basal and PTH agonist-induced cAMP accumulation in SaOS-2 osteosarcoma cells. Both drugs induced PTHR1-dependent apoptosis of MDA-MB-231 cells in vitro. Novel bone-targeted PTHrP antagonists represent a new paradigm for treatment of breast cancer bone metastases.


Asunto(s)
Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Hormona Paratiroidea/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Relacionada con la Hormona Paratiroidea/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Pharm Res ; 38(10): 1677-1695, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34671921

RESUMEN

PURPOSE: We developed an accessible method for labeling small extracellular vesicles (sEVs) without disrupting endogenous ligands. Using labeled sEVs administered to conscious rats, we developed a multiple compartment pharmacokinetic model to identify potential differences in the disposition of sEVs from three different cell types. METHODS: Crude sEVs were labeled with a non-homologous oligonucleotide and isolated from cell culture media using a commercial reagent. Jugular vein catheters were used to introduce EVs to conscious rats (n = 30) and to collect blood samples. Digital PCR was leveraged to allow for quantification over a wide dynamic range. Non-linear mixed effects analysis with first order conditional estimation - extended least squares (FOCE ELS) was used to estimate population-level parameters with associated intra-animal variability. RESULTS: 86.5% ± 1.5% (mean ± S.E.) of EV particles were in the 45-195 nm size range and demonstrated protein and lipid markers of endosomal origin. Incorporated oligonucleotide was stable in blood and detectable over five half-lives. Data were best described by a three-compartment model with one elimination from the central compartment. We performed an observation-based simulated posterior predictive evaluation with prediction-corrected visual predictive check. Covariate and bootstrap analyses identified cell type having an influence on peripheral volumes (V2 and V3) and clearance (Cl3). CONCLUSIONS: Our method relies upon established laboratory techniques, can be tailored to a variety of biological questions regarding the pharmacokinetic disposition of extracellular vesicles, and will provide a complementary approach for the of study EV ligand-receptor interactions in the context of EV uptake and targeted therapeutics.


Asunto(s)
Vesículas Extracelulares/metabolismo , Nanopartículas/metabolismo , Oligonucleótidos/farmacocinética , Animales , Secuencia de Bases , Transporte Biológico , Caenorhabditis elegans/genética , Humanos , Ligandos , Lípidos/química , Masculino , MicroARNs , Modelos Biológicos , Oligonucleótidos/metabolismo , Ratas Sprague-Dawley , Imagen Individual de Molécula
3.
Pharm Res ; 37(12): 235, 2020 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-33140122

RESUMEN

PURPOSE: The pharmacokinetic properties of plasma NO3- and its reduced metabolite, NO2-, have been separately described, but there has been no reported attempt to simultaneously model their pharmacokinetics following NO3- ingestion. This report describes development of such a model from retrospective analyses of concentrations largely obtained from primary endpoint efficacy trials. METHODS: Linear and non-linear mixed effects analyses were used to statistically define concentration dependency on time, dose, as well as patient and study variables, and to integrate NO3- and NO2- concentrations from studies conducted at different times, locations, patient groups, and several studies in which sample range was limited to a few hours. Published pharmacokinetic studies for both substances were used to supplement model development. RESULTS: A population pharmacokinetic model relating NO3- and NO2- concentrations was developed. The model incorporated endogenous levels of the two entities, and determined these were not influenced by exogenous NO3- delivery. Covariate analysis revealed intersubject variability in NO3- exposure was partially described by body weight differences influencing volume of distribution. The model was applied to visualize exposure versus response (muscle contraction performance) in individual patients. CONCLUSIONS: Extension of the present first-generation model, to ultimately optimize NO3- dose versus pharmacological effects, is warranted.


Asunto(s)
Suplementos Dietéticos , Modelos Biológicos , Nitratos/farmacocinética , Nitritos/farmacocinética , Administración Oral , Anciano , Envejecimiento/metabolismo , Disponibilidad Biológica , Peso Corporal , Estudios Cruzados , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/dietoterapia , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Nitratos/administración & dosificación , Nitratos/metabolismo , Nitritos/metabolismo , Estudios Retrospectivos , Sarcopenia/sangre , Sarcopenia/dietoterapia , Sarcopenia/metabolismo
4.
Mol Pharmacol ; 96(2): 272-296, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31221824

RESUMEN

Tamoxifen is used to prevent and treat estrogen receptor-positive (ER+) breast cancer (BC); however, its chronic use can increase uterine cancer risk and induce tamoxifen resistance. Novel melatonin-tamoxifen drug conjugates may be promising to treat BC and may help offset the adverse effects of tamoxifen usage alone due to the presence of melatonin. We synthesized and screened five drug conjugates (C2, C4, C5, C9, and C15 linked) for their effects on BC cell (MCF-7, tamoxifen-resistant MCF-7, mouse mammary carcinoma, MDA-MB-231, and BT-549) viability, migration, and binding affinity to melatonin receptor 1 (MT1R) and estrogen receptor 1 (ESR1). C4 and C5 demonstrated the most favorable pharmacological characteristics with respect to binding profiles (affinity for ESR1 and MT1R) and their potency/efficacy to inhibit BC cell viability and migration in four phenotypically diverse invasive ductal BC cell lines. C4 and C5 were further assessed for their actions against tamoxifen-resistant MCF-7 cells and a patient-derived xenograft triple-negative BC cell line (TU-BcX-4IC) and for their mechanisms of action using selective mitogen-activated protein kinase kinase MEK1/2, MEK5, and phosphoinositide 3-kinase (PI3K) inhibitors. C4 and C5 inhibited tamoxifen-resistant MCF-7 cells with equal potency (IC50 = 4-8 µM) and efficacy (∼90% inhibition of viability and migration) but demonstrated increased potency (IC50 = 80-211 µM) and efficacy (∼140% inhibition) to inhibit migration versus cell viability (IC50 = 181-304 mM; efficacy ∼80% inhibition) in TU-BcX-4IC cells. Unique pharmacokinetic profiles were observed, with C4 having greater bioavailability than C5. Further assessment of C4 and C5 demonstrates that they create novel pharmacophores within each BC cell that is context specific and involves MEK1/2/pERK1/2, MEK5/pERK5, PI3K, and nuclear factor κB. These melatonin-tamoxifen drug conjugates show promise as novel anticancer drugs and further preclinical and clinical evaluation is warranted.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/metabolismo , Melatonina/administración & dosificación , Receptor de Melatonina MT1/metabolismo , Tamoxifeno/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Disponibilidad Biológica , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células MCF-7 , Melatonina/farmacocinética , Melatonina/farmacología , Ratones , Tamoxifeno/farmacocinética , Tamoxifeno/farmacología
5.
J Pharmacol Exp Ther ; 369(1): 107-120, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30733244

RESUMEN

Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. A semimechanistic pharmacokinetic and pharmacodynamic (PK/PD) model with consideration of these multiple effects as a basis was developed. Integrated pharmacokinetics of d-amphetamine in plasma, brain extracellular fluid (ECF) via microdialysis, and cerebrospinal fluid were characterized using a population approach. This PK model was then linked to an indirect-response pharmacodynamic model using as a basis the measurement of extracellular striatal dopamine, also via microdialysis. In both rats and nonhuman primates (NHPs), d-amphetamine stimulation of dopamine outflow (reverse transport) through DAT was primarily responsible for the dose-linear increase in dopamine. As well, in both species a moderator function was needed to account for loss of the dopamine response in the presence of a relatively sustained d-amphetamine ECF exposure, presumptive of an acute tolerance response. PK/PD model structure was consistent between species; however, there was a 10-fold faster return to baseline dopamine in NHPs in response to an acute d-amphetamine challenge. These results suggest preservation from rodents to NHPs regarding the mechanism by which amphetamine increases extracellular dopamine, but a faster system response in NHPs to tolerate this increase. This microdialysis-based PK/PD model suggests greater value in directing preclinical discovery of novel approaches that modify reverse transport stimulation to treat amphetamine abuse. General value regarding insertion of an NHP model in paradigm rodent-to-human translational research is also suggested.


Asunto(s)
Dextroanfetamina/farmacología , Dextroanfetamina/farmacocinética , Dopamina/metabolismo , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Animales , Dextroanfetamina/efectos adversos , Cinética , Macaca fascicularis , Masculino , Ratas , Seguridad
6.
Drug Metab Dispos ; 44(5): 624-33, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26916207

RESUMEN

Administration of bupropion [(±)-2-(tert-butylamino)-1-(3-chlorophenyl)propan-1-one] and its preformed active metabolite, hydroxybupropion [(±)-1-(3-chlorophenyl)-2-[(1-hydroxy-2-methyl-2-propanyl)amino]-1-propanone], to rats with measurement of unbound concentrations by quantitative microdialysis sampling of plasma and brain extracellular fluid was used to develop a compartmental pharmacokinetics model to describe the blood-brain barrier transport of both substances. The population model revealed rapid equilibration of both entities across the blood-brain barrier, with resultant steady-state brain extracellular fluid/plasma unbound concentration ratio estimates of 1.9 and 1.7 for bupropion and hydroxybupropion, respectively, which is thus indicative of a net uptake asymmetry. An overshoot of the brain extracellular fluid/plasma unbound concentration ratio at early time points was observed with bupropion; this was modeled as a time-dependent uptake clearance of the drug across the blood-brain barrier. Translation of the model was used to predict bupropion and hydroxybupropion exposure in human brain extracellular fluid after twice-daily administration of 150 mg bupropion. Predicted concentrations indicate that preferential inhibition of the dopamine and norepinephrine transporters by the metabolite, with little to no contribution by bupropion, would be expected at this therapeutic dose. Therefore, these results extend nuclear imaging studies on dopamine transporter occupancy and suggest that inhibition of both transporters contributes significantly to bupropion's therapeutic efficacy.


Asunto(s)
Encéfalo/metabolismo , Bupropión/análogos & derivados , Bupropión/farmacocinética , Líquido Extracelular/metabolismo , Plasma/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Humanos , Masculino , Microdiálisis/métodos , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Bioorg Med Chem Lett ; 25(19): 4158-63, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26299349

RESUMEN

The observation that cholinergic deafferentation of circuits projecting from forebrain basal nuclei to frontal and hippocampal circuits occurs in Alzheimer's disease has led to drug-targeting of muscarinic M1 receptors to alleviate cognitive symptoms. The high homology within the acetylcholine binding domain of this family however has made receptor-selective ligand development challenging. This work presents the synthesis scheme, pharmacokinetic and structure-activity-relationship study findings for M1-selective ligand, LY593093. Pharmacologically the compound acts as an orthosteric ligand. The homology modeling work presented however will illustrate that compound binding spans from the acetylcholine pocket to the extracellular loops of the receptor, a common allosteric vestibule for the muscarinic protein family. Altogether LY593093 represents a growing class of multi-topic ligands which interact with the receptors in both the ortho- and allosteric binding sites, but which exert their activation mechanism as an orthosteric ligand.


Asunto(s)
Amidas/química , Amidas/farmacología , Diseño de Fármacos , Receptor Muscarínico M1/agonistas , Amidas/síntesis química , Animales , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Ratas , Relación Estructura-Actividad
8.
J Transl Med ; 12: 203, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25142323

RESUMEN

BACKGROUND: Clozapine is highly effective in treatment-resistant schizophrenia, although, there remains significant variability in the response to this drug. To better understand this variability, the objective of this study was to predict brain extracellular fluid (ECF) concentrations and receptor occupancy of clozapine and norclozapine in human central nervous system by translating plasma and brain ECF pharmacokinetic (PK) relationships in the rat and coupling these with known human disposition of clozapine in the plasma. METHODS: Unbound concentrations of clozapine and norclozapine were measured in rat brain ECF using quantitative microdialysis after subcutaneous administration of a 10 mg/kg single dose of clozapine or norclozapine. These data were linked with plasma concentrations obtained in the same rats to develop a plasma-brain ECF compartmental model. Parameters describing brain ECF disposition were then allometrically scaled and linked with published human plasma PK to predict human ECF concentrations. Subsequently, prediction of human receptor occupancy at several CNS receptors was based on an effect model that related the predicted ECF concentrations to published concentration-driven receptor occupancy parameters. RESULTS: A one compartment model with first order absorption and elimination best described clozapine and norclozapine plasma concentrations in rats. A delay in the transfer of clozapine and norclozapine from plasma to the brain ECF compartment was captured using a transit compartment model approach. Human clozapine and norclozapine concentrations in brain ECF were simulated, and from these the median percentage of receptor occupancy of dopamine-2, serotonin-2A, muscarinic-1, alpha-1 adrenergic, alpha-2 adrenergic and histamine-1 for clozapine, and dopamine-2 for norclozapine were consistent with values reported in the literature. CONCLUSIONS: A PK model that relates clozapine and norclozapine disposition in rat plasma and brain, including blood-brain barrier transport, was developed. Using allometry and published human plasma PK, the model was successfully translated to predict clozapine and norclozapine concentrations and accordant receptor occupancy of both agents in human brain. These predicted exposure and occupancy measures at several receptors that bind clozapine may be employed to extend our understanding of clozapine's complex behavioral effects in humans.


Asunto(s)
Química Encefálica , Clozapina/análogos & derivados , Clozapina/análisis , Clozapina/farmacocinética , Animales , Disponibilidad Biológica , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Clozapina/sangre , Humanos , Masculino , Modelos Animales , Ratas , Ratas Wistar , Estadística como Asunto , Investigación Biomédica Traslacional
9.
CPT Pharmacometrics Syst Pharmacol ; 13(1): 106-117, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37884051

RESUMEN

APX3330 ((2E)-2-[(4,5-dimethoxy-2-methyl-3,6-dioxo-1,4-cyclohexadien-1-yl)methylene]-undecanoic acid), a selective inhibitor of APE1/Ref-1, has been investigated in treatment of hepatitis, cancer, diabetic retinopathy, and macular edema. APX3330 is administered orally as a quinone but is rapidly converted to the hydroquinone form. This study describes the pharmacokinetics of APX3330 and explores effect of food on absorption. Total plasma quinone concentrations of APX3330 were obtained following oral administration from studies in healthy Japanese male subjects (single dose-escalation; multiple-dose; food-effect) and patients with cancer patients. Nonlinear mixed effects modeling was performed using Monolix to estimate pharmacokinetic parameters and assess covariate effects. To further evaluate the effect of food on absorption, a semi-physiologic pharmacokinetic model was developed in Gastroplus to delineate effects of food on dissolution and absorption. A two-compartment, first order absorption model with lag time best described plasma concentration-time profiles from 49 healthy Japanese males. Weight was positively correlated with apparent clearance (CL/F) and volume. Administration with food led to an 80% higher lag time. CL/F was 41% higher in the cancer population. The semi-physiologic model indicates a switch from dissolution-rate control of absorption in the fasted-state to gastric emptying rate determining absorption rate in the fed-state. Oral clearance of APX3330 is higher in patients with cancer than healthy Japanese males, possibly due to reduced serum albumin in patients with cancer. Delayed APX3330 absorption with food may be related to higher conversion to the more soluble but less permeable hydroquinone form in the gastrointestinal tract. Future work should address pharmacokinetic differences between APX3330 quinone and hydroquinone forms.


Asunto(s)
Hidroquinonas , Neoplasias , Humanos , Masculino , Administración Oral , Inhibidores de la Angiogénesis , Quinonas
10.
Anal Chem ; 85(3): 1727-33, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23294002

RESUMEN

A method has been developed for screening glyceollins and their metabolites based on precursor ion scanning. Under higher-energy collision conditions with the employment of a triple quadrupole mass spectrometer in the negative ion mode, deprotonated glyceollin precursors yield a diagnostic radical product ion at m/z 148. We propose this resonance-stabilized radical anion, formed in violation of the even-electron rule, to be diagnostic of glyceollins and glyceollin metabolites. Liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) established that scanning for precursors of m/z 148 can identify glyceollins and their metabolites from plasma samples originating from rats dosed with glyceollins. Precursor peaks of interest were found at m/z 337, 353, 355, 417, and 433. The peak at m/z 337 corresponds to deprotonated glyceollins, whereas the others represent metabolites of glyceollins. Accurate mass measurement confirmed m/z 417 to be a sulfated metabolite of glyceollins. The peak at m/z 433 is also sulfated, but it contains an additional oxygen, as confirmed by accurate mass measurement. The latter metabolite differs from the former likely by the replacement of a hydrogen with a hydroxyl moiety. The peaks at m/z 353 and 355 are proposed to correspond to hydroxylated metabolites of glyceollins, wherein the latter additionally undergoes a double bond reduction.


Asunto(s)
Pterocarpanos/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masas en Tándem/métodos , Animales , Extractos Vegetales/análisis , Extractos Vegetales/química , Extractos Vegetales/metabolismo , Pterocarpanos/análisis , Pterocarpanos/química , Ratas , Semillas
11.
Bioorg Med Chem Lett ; 23(23): 6459-62, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24119554

RESUMEN

The synthesis and structure-activity relationship of decahydroisoquinoline derivatives with various benzoic acid substitutions as GluK1 antagonists are described. Potent and selective antagonists were selected for a tailored prodrug approach in order to facilitate the evaluation of the new compounds in pain models after oral administration. Several diester prodrugs allowed for acceptable amino acid exposure and moderate efficacy in vivo.


Asunto(s)
Isoquinolinas/farmacología , Dolor/tratamiento farmacológico , Profármacos/farmacología , Receptores de Ácido Kaínico/antagonistas & inhibidores , Administración Oral , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Haplorrinos , Isoquinolinas/química , Datos de Secuencia Molecular , Profármacos/química , Receptores de Ácido Kaínico/química , Relación Estructura-Actividad
12.
Bioorg Med Chem Lett ; 23(23): 6463-6, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24140446

RESUMEN

We have explored the decahydroisoquinoline scaffold, bearing a phenyl tetrazole, as GluK1 antagonists with potential as oral analgesics. We have established the optimal linker atom between decahydroisoquinoline and phenyl rings and demonstrated an improvement of both the affinity for the GluK1 receptor and the selectivity against the related GluA2 receptor with proper phenyl substitution. In this Letter, we also disclose in vivo data that led to the discovery of LY545694·HCl, a compound with oral efficacy in two persistent pain models.


Asunto(s)
Isoquinolinas/farmacología , Dolor/tratamiento farmacológico , Profármacos/farmacología , Receptores de Ácido Kaínico/antagonistas & inhibidores , Tetrazoles/farmacología , Administración Oral , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Isoquinolinas/química , Masculino , Datos de Secuencia Molecular , Profármacos/química , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/química , Relación Estructura-Actividad , Tetrazoles/química
13.
Eur J Drug Metab Pharmacokinet ; 48(2): 171-187, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36823342

RESUMEN

BACKGROUND AND OBJECTIVES: Bupropion is an atypical antidepressant and smoking cessation aid; its use is associated with wide intersubject variability in efficacy and safety. Knowledge of the brain pharmacokinetics of bupropion and its pharmacologically active metabolites is considered important for understanding the cause-effect relationships driving this variability. METHODS: Brain concentrations from rats administered a 10 mg/kg subcutaneous dose of racemic bupropion were analyzed using a stereoselective LC/MS-MS method. A 2 mg/kg dose of (S,S)-hydroxybupropion, which has comparable pharmacologic potency to bupropion, was administered to a separate group of rats. Plasma exposure and unbound concentrations in both matrices from companion equilibrium dialysis experiments were determined to assess potential carrier-mediated transport at the blood-brain barrier. RESULTS: Exposures to unbound forms of bupropion enantiomers were similar in plasma; this was also true in brain. This trend held for reductive diastereomer metabolite pairs in the two matrices. Unbound (R,R)-hydroxybupropion exposure was 1.5-fold higher than (S,S)-hydroxybupropion exposure in plasma and brain following bupropion administration. Unbound concentration ratios (Kp,uu) of a given molecular form decreased over time: between 4 and 6 h, these were < 1 for the two bupropion enantiomers, and they were ~ 1 for metabolites that formed. Administration of preformed (S,S)-hydroxybupropion also demonstrated a declining Kp,uu. CONCLUSIONS: The temporal shift in Kp,uu among the different molecular forms provides evidence regarding the operation of carrier-mediated transport and/or within-brain metabolism of bupropion, and, thereby, fresh insight regarding the causes of intersubject variability in the safety and efficacy of bupropion therapy.


Asunto(s)
Antidepresivos de Segunda Generación , Bupropión , Ratas , Animales , Bupropión/farmacocinética , Encéfalo/metabolismo , Espectrometría de Masas en Tándem/métodos
14.
Drug Metab Dispos ; 40(10): 1909-16, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22736307

RESUMEN

A significant barrier to realization of the full potential of clozapine as a therapeutic agent in the treatment of schizophrenia is the substantial interpatient variability that exists along the therapeutic continuum of no response-efficacious response-adverse response. Genetic polymorphisms that manifest as highly variable pharmacodynamic and pharmacokinetic measures are its expected causes. To support investigations that seek to understand these causes, the plasma and central nervous system pharmacokinetics of clozapine were determined in rats, the latter using microdialysis sampling. Results obtained with clozapine and N-desmethylclozapine, a pharmacologically active human metabolite that was administered to a separate group of animals, support a conclusion of net carrier-mediated efflux of both compounds across the blood-brain barrier. These results are supported by the replication of published findings regarding the passive transport and net efflux transport of two model compounds, escitalopram and risperidone, respectively. The results obtained with clozapine and N-desmethylclozapine are considered a first step in the development of preclinical pharmacokinetic-pharmacodynamic models that will support deeper mechanistic studies of clozapine in in vivo pharmacology, as well as the development of translational models that augment pharmacogenetic investigations that seek to improve the safety and efficacy of clozapine therapeutic intervention in the treatment of schizophrenia.


Asunto(s)
Antipsicóticos/farmacocinética , Encéfalo/metabolismo , Clozapina/análogos & derivados , Microdiálisis , Animales , Antipsicóticos/administración & dosificación , Biotransformación , Barrera Hematoencefálica/metabolismo , Clozapina/administración & dosificación , Clozapina/farmacocinética , Inyecciones Subcutáneas , Masculino , Modelos Biológicos , Permeabilidad , Ratas , Ratas Wistar
15.
Viruses ; 14(3)2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35337012

RESUMEN

We extend our established agent-based multiscale computational model of infection of lung tissue by SARS-CoV-2 to include pharmacokinetic and pharmacodynamic models of remdesivir. We model remdesivir treatment for COVID-19; however, our methods are general to other viral infections and antiviral therapies. We investigate the effects of drug potency, drug dosing frequency, treatment initiation delay, antiviral half-life, and variability in cellular uptake and metabolism of remdesivir and its active metabolite on treatment outcomes in a simulated patch of infected epithelial tissue. Non-spatial deterministic population models which treat all cells of a given class as identical can clarify how treatment dosage and timing influence treatment efficacy. However, they do not reveal how cell-to-cell variability affects treatment outcomes. Our simulations suggest that for a given treatment regime, including cell-to-cell variation in drug uptake, permeability and metabolism increase the likelihood of uncontrolled infection as the cells with the lowest internal levels of antiviral act as super-spreaders within the tissue. The model predicts substantial variability in infection outcomes between similar tissue patches for different treatment options. In models with cellular metabolic variability, antiviral doses have to be increased significantly (>50% depending on simulation parameters) to achieve the same treatment results as with the homogeneous cellular metabolism.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Antivirales/farmacología , Antivirales/uso terapéutico , Epitelio , Humanos , SARS-CoV-2 , Replicación Viral
16.
J Pharm Biomed Anal ; 195: 113872, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33388643

RESUMEN

Pharmacologic effects elicited by drugs most directly relate to their unbound concentrations. Measurement of binding in blood, plasma and target tissues are used to estimate these concentrations by determining the fraction of total concentration in a biological matrix that is not bound. In the case of attempting to estimate R- and S-bupropion concentrations in plasma and brain following racemic bupropion administration, reversible chiral inversion and irreversible degradation of the enantiomers were hypothesized to confound attempts at unbound fraction estimation. To address this possibility, a kinetic modeling approach was used to quantify inversion and degradation specific processes for each enantiomer from separate incubations of each enantiomer in the two matrices, and in pH 7.4 buffer, which is also used in binding experiments based on equilibrium dialysis. Modeling analyses indicated that chiral inversion kinetics were two to four-fold faster in plasma and brain than degradation, with only inversion observed in buffer. Inversion rate was faster for S-bupropion in the three media; whereas, degradation rates were similar for the two enantiomers in plasma and brain, with overall degradation in plasma approximately 2-fold higher than in brain homogenate. Incorporation of degradation and chiral inversion kinetic terms into a model to predict enantiomer-specific binding in plasma and brain revealed that, despite existence of these two processes, empirically derived estimates of fraction unbound were similar to model-derived values, leading to a firm conclusion that observed extent of plasma and brain binding are accurate largely because binding kinetics are faster than parallel degradation and chiral inversion processes.


Asunto(s)
Bupropión , Preparaciones Farmacéuticas , Animales , Encéfalo , Cinética , Ratas , Estereoisomerismo
17.
Clin Transl Sci ; 14(5): 1864-1874, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33939284

RESUMEN

Clinical trial efficiency, defined as facilitating patient enrollment, and reducing the time to reach safety and efficacy decision points, is a critical driving factor for making improvements in therapeutic development. The present work evaluated a machine learning (ML) approach to improve phase II or proof-of-concept trials designed to address unmet medical needs in treating schizophrenia. Diagnostic data from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) trial were used to develop a binary classification ML model predicting individual patient response as either "improvement," defined as greater than 20% reduction in total Positive and Negative Syndrome Scale (PANSS) score, or "no improvement," defined as an inadequate treatment response (<20% reduction in total PANSS). A random forest algorithm performed best relative to other tree-based approaches in model ability to classify patients after 6 months of treatment. Although model ability to identify true positives, a measure of model sensitivity, was poor (<0.2), its specificity, true negative rate, was high (0.948). A second model, adapted from the first, was subsequently applied as a proof-of-concept for the ML approach to supplement trial enrollment by identifying patients not expected to improve based on their baseline diagnostic scores. In three virtual trials applying this screening approach, the percentage of patients predicted to improve ranged from 46% to 48%, consistently approximately double the CATIE response rate of 22%. These results show the promising application of ML to improve clinical trial efficiency and, as such, ML models merit further consideration and development.


Asunto(s)
Antipsicóticos/uso terapéutico , Aprendizaje Automático , Selección de Paciente , Esquizofrenia/tratamiento farmacológico , Adolescente , Adulto , Anciano , Ensayos Clínicos Fase II como Asunto/estadística & datos numéricos , Conjuntos de Datos como Asunto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prueba de Estudio Conceptual , Esquizofrenia/diagnóstico , Resultado del Tratamiento , Adulto Joven
18.
Clin Cancer Res ; 27(21): 5810-5817, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34400413

RESUMEN

PURPOSE: Fatty acid synthase (FASN) is overexpressed in 70% of operable triple-negative breast cancer (TNBC) and is associated with poor prognosis. Proton pump inhibitors selectively inhibit FASN activity and induce apoptosis in TNBC cell lines. PATIENTS AND METHODS: Patients with operable TNBC were enrolled in this single-arm phase II study. Patients began omeprazole 80 mg orally twice daily for 4-7 days prior to neoadjuvant anthracycline-taxane-based chemotherapy (AC-T) and continued until surgery. The primary endpoint was pathologic complete response (pCR) in patients with baseline FASN overexpression (FASN+). Secondary endpoints included pCR in all surgery patients, change in FASN expression, enzyme activity, and downstream protein expression after omeprazole monotherapy, safety, and limited omeprazole pharmacokinetics. RESULTS: Forty-two patients were recruited with a median age of 51 years (28-72). Most patients had ≥cT2 (33, 79%) and ≥N1 (22, 52%) disease. FASN overexpression prior to AC-T was identified in 29 of 34 (85%) evaluable samples. The pCR rate was 72.4% [95% confidence interval (CI), 52.8-87.3] in FASN+ patients and 74.4% (95% CI, 57.9-87.0) in all surgery patients. Peak omeprazole concentration was significantly higher than the IC50 for FASN inhibition observed in preclinical testing; FASN expression significantly decreased with omeprazole monotherapy [mean change 0.12 (SD, 0.25); P = 0.02]. Omeprazole was well tolerated with no grade ≥ 3 toxicities. CONCLUSIONS: FASN is commonly expressed in early TNBC. Omeprazole can be safely administered in doses that inhibit FASN. The addition of omeprazole to neoadjuvant AC-T yields a promising pCR rate that needs further confirmation in randomized studies.


Asunto(s)
Ácido Graso Sintasas/antagonistas & inhibidores , Terapia Neoadyuvante , Omeprazol/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto , Anciano , Femenino , Humanos , Persona de Mediana Edad , Omeprazol/farmacología , Resultado del Tratamiento
19.
Eur J Drug Metab Pharmacokinet ; 44(2): 261-274, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30298475

RESUMEN

BACKGROUND AND OBJECTIVES: Bupropion is an atypical antidepressant and smoking cessation aid associated with wide intersubject variability. This study compared the formation kinetics of three phase I metabolites (hydroxybupropion, threohydrobupropion, and erythrohydrobupropion) in human, marmoset, rat, and mouse liver microsomes. The objective was to establish suitability and limitations  for subsequent use of nonclinical species to model bupropion central nervous system (CNS) disposition in humans. METHODS: Hepatic microsomal incubations were conducted separately for the R- and S-bupropion enantiomers, and the formation of enantiomer-specific metabolites was determined using LC-MS/MS. Intrinsic formation clearance (CLint) of metabolites across the four species was determined from the formation rate versus substrate concentration relationship. RESULTS: The total clearance of S-bupropion was higher than that of R-bupropion in monkey and human liver microsomes. The contribution of hydroxybupropion to the total racemic bupropion clearance was 38%, 62%, 17%, and 96% in human, monkey, rat, and mouse, respectively.  In the same species order, threohydrobupropion contributed 53%, 23%, 17%, and 3%, and erythrohydrobupropion contributed 9%, 14%, 66%, and 1.3%, respectively, to racemic bupropion clearance. CONCLUSION: The results demonstrate that phase I metabolism in monkeys best approximates that observed in humans, and support the preferred use of this species to investigate possible pharmacokinetic factors that influence the CNS disposition of bupropion and contribute to its high intersubject variability.


Asunto(s)
Antidepresivos de Segunda Generación/química , Antidepresivos de Segunda Generación/metabolismo , Bupropión/química , Bupropión/metabolismo , Microsomas Hepáticos/metabolismo , Animales , Antidepresivos de Segunda Generación/farmacología , Bupropión/farmacología , Callithrix , Relación Dosis-Respuesta a Droga , Femenino , Haplorrinos , Humanos , Masculino , Ratones , Microsomas Hepáticos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
20.
J Neurosci Methods ; 317: 71-81, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30768951

RESUMEN

BACKGROUND: Challenges specific to the discovery and development of candidate CNS drugs have led to implementation of various in silico, in vitro and in vivo approaches to improve the odds for commercialization of novel treatments. NEW METHOD: Advances in analytical methodology and microdialysis probe design have enabled development of a non-human primate model capable of measuring concentrations of drugs or endogenous chemicals in brain extracellular fluid (ECF) and cerebrospinal fluid (CSF). Linking these to population modeling reduces animal numbers to support predictive translational sciences in primates. Application to measure D-amphetamine exposure and dopamine response in ECF and CSF demonstrate the approach. RESULTS: Following a 0.1 mg/kg intravenous bolus dose of D-amphetamine, a population approach was used to build a plasma compartmental-based and brain physiologic-based pharmacokinetic (PK) model linking drug concentrations in plasma to brain ECF and CSF concentrations. Dopamine was also measured in brain ECF. The PK model was used to simulate the relationship between D-amphetamine exposure and dopamine response in ECF over a wide dose range. COMPARISONS WITH EXISTING METHODS: Ability to co-sample and measure drug and endogenous substances in blood, brain ECF and/or CSF, coupled with population modeling, provides an in vivo approach to evaluate CNS drug penetration and effect in non-human primates. CONCLUSIONS: A method to measure drug and endogenous neurochemicals in non-human primate brain fluids is demonstrated. Its basis in non-human primates merits improved confidence regarding predictions of drug exposure and target engagement in human CNS.


Asunto(s)
Química Encefálica , Líquido Cefalorraquídeo/química , Dextroanfetamina/análisis , Dextroanfetamina/farmacología , Dopamina/análisis , Líquido Extracelular/química , Microdiálisis/métodos , Animales , Biomarcadores/análisis , Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Macaca fascicularis , Masculino , Ratas Wistar , Investigación Biomédica Traslacional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA