Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.486
Filtrar
1.
J Drugs Dermatol ; 23(6): 466-471, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38834224

RESUMEN

Xerosis is experienced by almost everyone at some time in their lives and the foundation of management of dry skin (both consumer- and healthcare professional--directed) rests on the use of moisturizers. Given the wide range of available moisturizers, counseling patients about selecting the optimum moisturizer for their individual situation relies on knowledge of ingredients and formulations. Traditionally, the main focus for many moisturizers centered on the core functional and structural role of ceramides within the epidermal barrier.  However, while a key aspect of transepidermal water loss and other skin barrier functions, components other than ceramides are equally essential in increasing moisturization. The skin's natural moisturizing factors (NMFs) are a complex mixture of water-attracting compounds such as amino acids, urea, lactate, pyrrolidone carboxylic acid (PCA), and electrolytes which play a fundamental role in preserving physiologic function by regulating the water content of the stratum corneum. By facilitating water retention, NMFs contribute significantly to the suppleness, elasticity, normal desquamation, and overall integrity of the skin barrier. Incorporation of NMFs into moisturizers addresses critical deficiencies in the skin's moisture balance that exist in xerotic and atopic skin, and in many skin disorders, mitigating signs and symptoms associated with xerosis and promoting optimal skin health. The biochemical composition of NMFs and the intricate interplay with epidermal homeostasis translate to a central role in moisturizers used for prophylactic and therapeutic management of various dry skin conditions, beyond ceramides alone. J Drugs Dermatol. 2024;23(6):466-471.     doi:10.36849/JDD.8358.


Asunto(s)
Ceramidas , Emolientes , Pérdida Insensible de Agua , Humanos , Ceramidas/administración & dosificación , Pérdida Insensible de Agua/efectos de los fármacos , Emolientes/administración & dosificación , Crema para la Piel/administración & dosificación , Administración Cutánea , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Epidermis/fisiología , Urea/administración & dosificación
3.
Clin Genitourin Cancer ; 22(3): 102073, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38626661

RESUMEN

INTRODUCTION: Hand foot skin reaction (HFSR) is a common dose-limiting adverse effect of multi kinase inhibitors (MKI) whose mechanism is not fully understood, and the prophylaxis is inadequate. OBJECTIVE: In this pilot study, a double-blind, randomized placebo-controlled trial was conducted to evaluate the effect of topical urea in secondary prevention of sunitinib-induced HFSR in renal cell cancer patients. METHODS: Out of 55 screened patients, 14 were randomized to receive topical urea or placebo for four weeks. The association of HFSR with drug levels of sunitinib and its metabolite (n-desethyl sunitinib), genetic polymorphism of VEGFR2 gene, quality of life (QOL) and biochemical markers was also assessed. RESULTS: The results showed that urea-based cream was not superior to placebo (P = .075). There was no change in the QOL in both the groups. Single nucleotide polymorphism was checked for two nucleotides rs1870377 and rs2305948 located in VEGFR2 gene on chromosome 4. SNP (variant T > A) at rs1870377 was associated with appearance of new HFSR as compared to the wild type, although the association was not statistically significant (OR 0.714). There was no statistically significant difference between mean plasma levels of sunitinib and N-desethyl sunitinib in urea arm as compared to placebo arm as compared to placebo. The best fit population pharmacokinetic model for sunitinib was one compartment model with first order absorption and linear elimination. The median (IQR) of population parameters calculated from the population pharmacokinetics model for Ka, V and Cl was 0.22 (0.21-0.24) h-1, 4.4 (4.09-4.47) L, 0.049 (0.042-0.12) L/hr, respectively. CONCLUSION: The study suggested that the urea-based cream was not superior to placebo in decreasing the appearance of new HFSR in renal cancer patients receiving 4:2 regimen of sunitinib.


Asunto(s)
Carcinoma de Células Renales , Síndrome Mano-Pie , Neoplasias Renales , Sunitinib , Urea , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Humanos , Sunitinib/administración & dosificación , Sunitinib/farmacocinética , Sunitinib/efectos adversos , Método Doble Ciego , Carcinoma de Células Renales/tratamiento farmacológico , Masculino , Femenino , Persona de Mediana Edad , Urea/análogos & derivados , Urea/farmacocinética , Urea/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Síndrome Mano-Pie/etiología , Síndrome Mano-Pie/prevención & control , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Proyectos Piloto , Anciano , Polimorfismo de Nucleótido Simple , Antineoplásicos/efectos adversos , Antineoplásicos/administración & dosificación , Calidad de Vida , Resultado del Tratamiento , Administración Tópica , Adulto , Indoles/administración & dosificación , Indoles/farmacocinética , Indoles/efectos adversos
4.
Biochem Pharmacol ; 196: 114731, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34407453

RESUMEN

Cholesterol esterification proteins Sterol-O acyltransferases (SOAT) 1 and 2 are emerging prognostic markers in many cancers. These enzymes utilise fatty acids conjugated to coenzyme A to esterify cholesterol. Cholesterol esterification is tightly regulated and enables formation of lipid droplets that act as storage organelles for lipid soluble vitamins and minerals, and as cholesterol reservoirs. In cancer, this provides rapid access to cholesterol to maintain continual synthesis of the plasma membrane. In this systematic review and meta-analysis, we summarise the current depth of understanding of the role of this metabolic pathway in pan-cancer development. A systematic search of PubMed, Scopus, Web of Science, and Cochrane Library for preclinical studies identified eight studies where cholesteryl ester concentrations were compared between tumour and adjacent-normal tissue, and 24 studies where cholesterol esterification was blocked by pharmacological or genetic approaches. Tumour tissue had a significantly greater concentration of cholesteryl esters than non-tumour tissue (p < 0.0001). Pharmacological or genetic inhibition of SOAT was associated with significantly smaller tumours of all types (p ≤ 0.002). SOAT inhibition increased tumour apoptosis (p = 0.007), CD8 + lymphocyte infiltration and cytotoxicity (p ≤ 0.05), and reduced proliferation (p = 0.0003) and metastasis (p < 0.0001). Significant risk of publication bias was found and may have contributed to a 32% overestimation of the meta-analysed effect size. Avasimibe, the most frequently used SOAT inhibitor, was effective at doses equivalent to those previously reported to be safe and tolerable in humans. This work indicates that SOAT inhibition should be explored in clinical trials as an adjunct to existing anti-neoplastic agents.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Colesterol/genética , Colesterol/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Carga Tumoral/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Ensayos Clínicos como Asunto/métodos , Esterificación/efectos de los fármacos , Esterificación/fisiología , Humanos , Transportadores de Anión Orgánico/antagonistas & inhibidores , Carga Tumoral/fisiología , Urea/administración & dosificación , Urea/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Lancet Psychiatry ; 9(1): 46-58, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34861170

RESUMEN

BACKGROUND: Negative symptoms of schizophrenia are associated with adverse clinical outcomes, but there are few effective treatments. We aimed to assess the effects of pimavanserin, a selective 5-HT2A inverse agonist and antagonist, on negative symptoms of schizophrenia. METHODS: The ADVANCE study was a phase 2, 26-week, randomised, double-blind, placebo-controlled study of pimavanserin in stable outpatients with schizophrenia aged 18-55 years with predominant negative symptoms. Patients were randomly assigned (1:1) across 83 sites (18 in North America and 65 in Europe) to receive pimavanserin or placebo daily, added to an ongoing antipsychotic medication, per a computer-generated schedule (stratification by geographical region). Eligible patients had a score of at least 20 on the sum of seven Positive and Negative Syndrome Scale (PANSS) Marder negative factor items (and scores of ≥4 on at least three or ≥5 on at least two of negative symptom items). The starting dosage of 20 mg of pimavanserin or placebo could be adjusted to 34 mg or 10 mg within the first 8 weeks of the study, after which dosage remained stable until the end of the study. Both pimavanserin and placebo were administered orally once daily as two individual tablets (pimavanserin tablets were either 10 mg or 17 mg). The primary endpoint was change in total score using the 16-item Negative Symptom Assessment (NSA-16) from baseline to week 26. Primary outcomes were analysed in patients who received at least one dose of the study drug and had NSA-16 assessments at baseline and at least once post-baseline (full analysis set). Safety outcomes were analysed in patients who had received at least one dose of the study drug. This trial is registered with ClinicalTrials.gov, NCT02970305, and is complete. FINDINGS: Between Nov 4, 2016, and April 16, 2019, we randomly assigned 403 patients to pimavanserin (n=201; 131 [65%] male; 187 [93%] White) or placebo (n=202; 137 [68%] male, 186 (92%) White), of whom 400 were included in the efficacy analysis (199 in the pimavanserin group, 201 in the placebo group). Mean age was 37·7 years (SD 9·4) in the pimavanserin group and 36·7 (9·2) years in the placebo group. The change in total NSA-16 score from baseline to week 26 was significantly improved with pimavanserin (least squares mean -10·4 [SE 0·67]) versus placebo (least squares mean -8·5 [0·67]; p=0·043; effect size: 0·211). The number of patients with treatment-emergent adverse events (TEAEs) was similar between groups: 80 (40%) patients experienced TEAEs in the pimavanserin group and 71 (35%) in the placebo group. Most TEAEs were headache (6% [n=13] vs 5% [n=10]) and somnolence (5% [n=11] vs 5% [n=10]). One patient from the placebo group reported severe headache (0·5%), rhinorrhoea (0·5%), cough (0·5%), and influenza (0·5%). In the pimavanserin group, one patient reported severe toothache (0·5%), and two patients had worsening of schizophrenia (1%). Mean change in QTcF interval was higher with pimavanerin (4·5 ms [SD 18·0]) than with placebo (0·0 ms [16·0]). INTERPRETATION: Stable patients with predominant negative symptoms of schizophrenia showed a reduction in negative symptoms after treatment with pimavanserin. However, given the small effect size, further investigation with optimised dosing is warranted to determine the clinical significance of this effect. FUNDING: Acadia Pharmaceuticals.


Asunto(s)
Piperidinas/farmacología , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Urea/análogos & derivados , Adolescente , Adulto , Método Doble Ciego , Europa (Continente) , Femenino , Humanos , Masculino , Persona de Mediana Edad , América del Norte , Evaluación de Resultado en la Atención de Salud , Piperidinas/administración & dosificación , Piperidinas/efectos adversos , Antagonistas del Receptor de Serotonina 5-HT2/administración & dosificación , Antagonistas del Receptor de Serotonina 5-HT2/efectos adversos , Urea/administración & dosificación , Urea/efectos adversos , Urea/farmacología , Adulto Joven
6.
Int J Neurosci ; 132(1): 67-76, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32746675

RESUMEN

AIM OF THE STUDY: In this study, we investigated the effect of long-term administration of orexin receptor 1 (OXR1) antagonist on naloxone-precipitated morphine withdrawal symptoms and nociceptive behaviors in morphine-dependent rats. MATERIALS AND METHODS: Wistar rats received subcutaneous (s.c.) injections of morphine (6, 16, 26, 36, 46, 56, and 66 mg/kg, 2 ml/kg) at an interval of 24 h for 7 days. In chronic groups, the OXR1 antagonist, SB-334867 (20 mg/kg, i.p.), or its vehicle, was injected repetitively from postnatal day 1 (PND1)-PND23 and then for the following seven days before each morphine injection. Meanwhile, in acute groups, SB-334867, or its vehicle, was administered before each morphine injection. In groups of rats that were designated for withdrawal experiments, naloxone (2.5 mg/kg, i.p.) was administered after the last injection of morphine. In the formalin-induced pain, the effect of OXR1 inhibition on the antinociceptive effects of morphine was measured by injecting formalin after the final morphine injection. RESULTS: Animals that received long-term SB-334867 administration before morphine injection demonstrated a significant reduction in chewing, defecation, diarrhea, grooming, teeth chattering, wet-dog shake, and writhing. Inhibiting OXR1 for a long time increased formalin-induced nociceptive behaviors in interphase and phase II of the formalin-induced pain. CONCLUSIONS: Our results indicated that the inhibition of OXR1 significantly reduces the development of morphine dependence and behavioral signs elicited by the administration of naloxone in morphine-dependent rats. Furthermore, the prolonged blockade of OXR1 might be involved in formalin-induced nociceptive behaviors.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzoxazoles/farmacología , Dependencia de Morfina/tratamiento farmacológico , Naftiridinas/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Antagonistas de los Receptores de Orexina/farmacología , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Urea/análogos & derivados , Animales , Benzoxazoles/administración & dosificación , Modelos Animales de Enfermedad , Morfina/administración & dosificación , Naloxona/farmacología , Naftiridinas/administración & dosificación , Antagonistas de Narcóticos/farmacología , Narcóticos/administración & dosificación , Antagonistas de los Receptores de Orexina/administración & dosificación , Ratas , Ratas Wistar , Urea/administración & dosificación , Urea/farmacología
7.
Peptides ; 147: 170679, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34718063

RESUMEN

Stress activates multiple neural pathways and neurotransmitters that often suppress pain perception, the phenomenon called stress-induced analgesia (SIA). Orexin neurons from the lateral hypothalamus project to entire brain structures such as the hippocampus. The present study examined this hypothesis that orexinergic receptors in the CA1 region of the hippocampus may play a modulatory role in the development of SIA in formalin test as an animal model of persistent inflammatory pain. One hundred-two adult male Wistar rats were administered with intra-CA1 orexin-1 receptor (OX1r) antagonist, SB334867, at the doses of 3, 10, 30, and 100 nmol or TCS OX2 29 as orexin-2 receptor (OX2r) antagonist at the doses of 1, 3, 10, and 30 nmol. Five min later, rats were exposed to forced swim stress (FSS) for a 6-min period. Then, pain-related behaviors induced by formalin injection were measured at the 5-min blocks during a 60-min period of formalin test. The current study indicated that solely stress exposure elicits antinociception in the early and late phases of the formalin test. The FSS-induced analgesia was prevented by intra-CA1 administration of SB334867 or TCS OX2 29 during either phase of the formalin test. Moreover, the contribution of the OX2r in the mediation of analgesic effect of stress was more prominent than that of the OX1r during both phases of the formalin test. It is suggested that OX1r and OX2r in the CA1 region of the hippocampus are involved in stress-induced analgesia in the animal model of persistent inflammatory pain.


Asunto(s)
Región CA1 Hipocampal/fisiología , Receptores de Orexina/metabolismo , Dolor/etiología , Estrés Psicológico/etiología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Benzoxazoles/administración & dosificación , Benzoxazoles/farmacología , Región CA1 Hipocampal/efectos de los fármacos , Ciclofosfamida , Modelos Animales de Enfermedad , Doxorrubicina , Etopósido , Inflamación/etiología , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacología , Masculino , Microinyecciones , Naftiridinas/administración & dosificación , Naftiridinas/farmacología , Antagonistas de los Receptores de Orexina/administración & dosificación , Antagonistas de los Receptores de Orexina/farmacología , Dolor/tratamiento farmacológico , Dimensión del Dolor , Prednisona , Piridinas/administración & dosificación , Piridinas/farmacología , Ratas Wistar , Urea/administración & dosificación , Urea/análogos & derivados , Urea/farmacología , Vincristina
8.
Behav Brain Res ; 422: 113710, 2022 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-34906610

RESUMEN

Negative symptoms are a core, pervasive, and often treatment-refractory phenotype of schizophrenia, one which contributes to poor functional outcome, ability to work, pursue educational goals, and quality of life, as well as caretaker burden. Improvement of negative symptoms in some patients with schizophrenia has been reported with some atypical antipsychotic drugs [AAPDs], but improvement is absent in many patients and partial in others. Therefore, more effective treatments are needed, and better preclinical models of negative symptoms are needed to identify them. Sub-chronic [sc] treatment of rodents with phencyclidine [PCP], a noncompetitive N-methyl-d-aspartate [NMDAR] antagonist, produces deficits in social interactions [SI] that have been widely studied as a model of negative symptoms in schizophrenia. Acute restraint stress [ARS] also provides a model of treatment-refractory negative symptoms [TRS] to AAPDs. By themselves, in sc-PCP mice, the AAPDs, risperidone, olanzapine, and aripiprazole, but not the selective 5-HT2AR inverse agonist, pimavanserin [PIM], rescued the SI deficit in sc-PCP mice, as did the combination of PIM with sub-effective doses of each of these AAPDs. These three AAPDs alone did not rescue SI deficit in sc-PCP+ 2 h-ARS mice, indicating these mice were treatment refractory. However, co-administration of PIM with any of the AAPDs significantly restored SI in these mice. PIM may be an effective adjunctive therapy for treating negative symptoms of schizophrenia in some patients who have failed to respond to AAPDs, but further studies are needed.


Asunto(s)
Antipsicóticos/farmacología , Piperidinas/farmacología , Esquizofrenia Resistente al Tratamiento/tratamiento farmacológico , Urea/análogos & derivados , Animales , Antipsicóticos/administración & dosificación , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos C57BL , Piperidinas/administración & dosificación , Urea/administración & dosificación , Urea/farmacología
9.
Chem Pharm Bull (Tokyo) ; 69(11): 1110-1122, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34719594

RESUMEN

Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step of the nicotinamide adenine dinucleotide (NAD+) salvage pathway. Because NAD+ plays a pivotal role in energy metabolism and boosting NAD+ has positive effects on metabolic regulation, activation of NAMPT is an attractive therapeutic approach for the treatment of various diseases, including type 2 diabetes and obesity. Herein we report the discovery of 1-(2-phenyl-1,3-benzoxazol-6-yl)-3-(pyridin-4-ylmethyl)urea 12c (DS68702229), which was identified as a potent NAMPT activator. Compound 12c activated NAMPT, increased cellular NAD+ levels, and exhibited an excellent pharmacokinetic profile in mice after oral administration. Oral administration of compound 12c to high-fat diet-induced obese mice decreased body weight. These observations indicate that compound 12c is a promising anti-obesity drug candidate.


Asunto(s)
Fármacos Antiobesidad/síntesis química , Nicotinamida Fosforribosiltransferasa/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Urea/síntesis química , Animales , Fármacos Antiobesidad/administración & dosificación , Fármacos Antiobesidad/farmacocinética , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Masculino , Ratones Obesos , NAD/metabolismo , Obesidad/metabolismo , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacocinética , Relación Estructura-Actividad , Urea/administración & dosificación , Urea/farmacocinética
10.
Physiol Rep ; 9(16): e14988, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34405966

RESUMEN

In patients with chronic aortic regurgitation (AR), excessive preload and afterload increase left ventricle wall stress, leading to left ventricular systolic dysfunction. Thus, the objective of the present study was to evaluate the effects of the myosin activator omecamtiv mecarbil (OM) on left ventricle wall stress in an experimental rat model of severe chronic AR. Forty adult male Wistar rats were randomized into two experimental groups: induction of AR (acute phase) by retrograde puncture (n = 34) or a sham intervention (n = 6). Rats that survived the acute phase (n = 18) were randomized into an OM group (n = 8) or a placebo group (n = 10). Equal volumes of OM (1.2 mg/kg/h) or placebo (0.9% NaCl) were continuously infused into the femoral vein over 30 min. OM significantly decreased end-systolic and end-diastolic and maximum wall stress in this experimental rat model of chronic severe AR (p < 0.001) and increased systolic performance assessed by fractional shortening and left ventricle end-systolic diameter; both p < 0.05). These effects were correlated with decreased indices of global cardiac function (cardiac output and stroke volume; p < 0.05) but were not inferior to baseline pump indices. Infusion with placebo did not affect global cardiac function but decreased end-systolic wall stress (p < 0.05) and increased systolic performance (all p < 0.001). In the sham-operated (control) group, OM decreased diastolic wall stress (p < 0.05). Based on these results, OM had a favorable effect on left ventricle wall stress in an experimental rat model of severe chronic AR.


Asunto(s)
Insuficiencia de la Válvula Aórtica/tratamiento farmacológico , Cardiotónicos/uso terapéutico , Urea/análogos & derivados , Animales , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Infusiones Intravenosas , Masculino , Ratas , Ratas Wistar , Volumen Sistólico , Sístole , Urea/administración & dosificación , Urea/farmacología , Urea/uso terapéutico
11.
Clin Drug Investig ; 41(7): 647-652, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34097256

RESUMEN

BACKGROUND AND OBJECTIVE: Omecamtiv mecarbil (OM) is a novel cardiac myosin activator in development for the treatment of heart failure with reduced ejection fraction. The objective of this study was to evaluate the potential for OM to affect the pharmacokinetics of metformin. METHODS: This was an open-label, fixed-sequence study in 14 healthy subjects. On Day 1, subjects received an 850 mg oral dose of metformin. From Days 4 to 9, subjects received twice-daily 25 mg oral doses of OM tablets. On Day 10, subjects received an 850 mg oral dose of metformin and a single 25 mg tablet of OM. Blood and urine samples were collected up to 36 h post-dose following administration of metformin on Days 1 and 10 to characterize concentrations of metformin in plasma and urine. RESULTS: The ratios of the geometric least square means of metformin coadministered with OM compared to metformin alone were 98.7%, 99.3%, and 110.2% for AUCinf, AUClast, and Cmax, respectively. The mean renal clearance of metformin was similar following metformin administered alone (34.2 L/h) compared to metformin coadministered with OM (32.9 L/h). All adverse events were mild in severity and resolved prior to the end of the study. No serious adverse events or treatment-emergent adverse events led to discontinuation from the study. CONCLUSIONS: There was no clinically relevant effect of OM on the pharmacokinetics of metformin in healthy subjects.


Asunto(s)
Metformina/farmacocinética , Proteínas de Transporte de Catión Orgánico/metabolismo , Urea/análogos & derivados , Administración Oral , Adulto , Área Bajo la Curva , Diarrea/etiología , Interacciones Farmacológicas/fisiología , Femenino , Semivida , Voluntarios Sanos , Humanos , Análisis de los Mínimos Cuadrados , Masculino , Metformina/administración & dosificación , Metformina/efectos adversos , Metformina/química , Persona de Mediana Edad , Curva ROC , Especificidad por Sustrato , Comprimidos/química , Urea/administración & dosificación , Urea/efectos adversos , Urea/química
12.
Biopharm Drug Dispos ; 42(7): 319-328, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34087948

RESUMEN

Omecamtiv mecarbil (OM) is a cardiac myosin activator in clinical development for the treatment of heart failure. The effect of food on the pharmacokinetics (PK) of 25, 37.5, and 50 mg strength modified release (MR) tablets and the bioequivalence of two 25 mg tablets versus one 50 mg MR tablet were evaluated in two open-label, randomized, cross-over studies in healthy subjects. Subjects received two 25 mg tablets or one 50 mg OM MR tablet under fed or fasted states in Study 1 (n = 39), and single oral doses of 25 and 37.5 mg OM MR tablets and to assess its relative bioavailability to the 25 mg MR tablet, a 25 mg oral solution under fed or fasted states in Study 2 (n = 34). The area under the concentration-time curve (AUC) and the maximum observed concentration (Cmax ) of 25, 37.5, or 50 mg OM MR tablets were approximately 13%-22% higher and 31%-40% higher, respectively, when taken with food. The two 25 mg and one 50 mg OM MR tablets were bioequivalent (90% confidence intervals) of the geometric mean ratios for Cmax and AUC of OM were within 0.8-1.25 under the fasted or fed state. OM was well tolerated and all treatment-emergent events were mild in severity and resolved by the end of the study. In conclusion, these studies demonstrated that the effect of food on the PK of OM was minimal at all three studied strengths of the MR tablets, and two 25 mg MR tablets may be switched for one 50 mg MR tablet (EudraCT Number: 2019-003683-44).


Asunto(s)
Urea/análogos & derivados , Administración Oral , Adulto , Área Bajo la Curva , Miosinas Cardíacas , Estudios Cruzados , Preparaciones de Acción Retardada , Sustitución de Medicamentos , Ayuno/metabolismo , Femenino , Interacciones Alimento-Droga , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Comprimidos , Equivalencia Terapéutica , Urea/administración & dosificación , Urea/efectos adversos , Urea/sangre , Urea/farmacocinética
13.
Acta Med Okayama ; 75(3): 289-297, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34176932

RESUMEN

Early treatment with an oral ß-blocker is recommended in patients with a ST-segment-elevation myocardial infarction (STEMI). In this multicenter study, we evaluated the effects of a continuous administration of landiolol, an ultrashort-acting ß-blocker, before primary percutaneous coronary intervention (PCI) on myocardial salvage and its safety in STEMI patients. A total of 47 Japanese patients with anterior or lateral STEMI undergoing a primary PCI within 12 h of symptom onset were randomized to receive intravenous landiolol (started at 3 µg/min/kg dose and continued to a total of 50 mg; n=23) or not (control; n=24). Patients with Killip class III or more were excluded. The primary outcome was the myocardial salvage index on cardiac magnetic resonance imaging (MRI) performed 5-7 days after the PCI. Cardiac MRI was performed in 35 patients (74%). The myocardial salvage index in the landiolol group was significantly greater than that in the control group (44.4±14.6% vs. 31.7±18.9%, respectively; p=0.04). There were no significant differences in adverse events at 24 h between the landiolol and control groups. A continuous administration of landiolol before a primary PCI may increase the degree of myocardial salvage without additional hemodynamic adverse effects within the first 24 h after STEMI.


Asunto(s)
Antagonistas Adrenérgicos beta/administración & dosificación , Morfolinas/administración & dosificación , Daño por Reperfusión Miocárdica/prevención & control , Infarto del Miocardio con Elevación del ST/tratamiento farmacológico , Urea/análogos & derivados , Administración Intravenosa , Anciano , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/patología , Intervención Coronaria Percutánea/métodos , Estudios Prospectivos , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Urea/administración & dosificación
14.
Clin Transl Sci ; 14(4): 1600-1610, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33955165

RESUMEN

Omecamtiv mecarbil (OM) is a myosin activator (myotrope), developed as a potential therapeutic agent for heart failure with reduced ejection fraction. To characterize the potential pro-arrhythmic risk of this novel sarcomere activator, we evaluated OM in a series of International Conference on Harmonization S7B core and follow-up assays, including an in silico action potential (AP) model. OM was tested in: (i) hERG, Nav1.5 peak, and Cav1.2 channel assays; (ii) in silico computation in a human ventricular AP (hVAP) population model; (iii) AP recordings in canine cardiac Purkinje fibers (PF); and (iv) electrocardiography analysis in isolated rabbit hearts (IRHs). OM had low potency in the hERG (half-maximal inhibitory concentration [IC50 ] = 125.5 µM) and Nav1.5 and Cav1.2 assays (IC50  > 300 µM). These potency values were used as inputs to investigate the occurrence of repolarization abnormalities (biomarkers of pro-arrhythmia) in an hVAP model over a wide range of OM concentrations. The outcome of hVAP analysis indicated low pro-arrhythmia risk at OM concentration up to 30 µM (100-fold the effective free therapeutic plasma concentration). In the isolated canine PF assay, OM shortened AP duration (APD)60 and APD90 significantly from 3 to 30 µM. In perfused IRH, ventricular repolarization (corrected QT and corrected JT intervals) was decreased significantly at greater than or equal to 1 µM OM. In summary, the comprehensive proarrhythmic assessment in human and non-rodent cardiac models provided data indicative that OM did not delay ventricular repolarization at therapeutic relevant concentrations, consistent with clinical findings.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Insuficiencia Cardíaca/tratamiento farmacológico , Urea/análogos & derivados , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/inducido químicamente , Simulación por Computador , Perros , Evaluación Preclínica de Medicamentos , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Preparación de Corazón Aislado , Miocitos Cardíacos/efectos de los fármacos , Cultivo Primario de Células , Ramos Subendocárdicos , Conejos , Urea/administración & dosificación , Urea/efectos adversos
15.
Parkinsonism Relat Disord ; 87: 25-31, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33933853

RESUMEN

INTRODUCTION: Pimavanserin, a selective 5-HT2A inverse agonist/antagonist, was approved for hallucinations and delusions associated with Parkinson's disease psychosis (PDP). We present durability of response with pimavanserin in patients with PDP for an additional 4 weeks of treatment. METHODS: This was an open-label extension (OLE) study in patients previously completing one of three double-blind, placebo-controlled (Core) studies. All patients received pimavanserin 34 mg once daily. Efficacy assessments included the Scale for the Assessment of Positive Symptoms (SAPS) PD and H + D scales, Clinical Global Impression (CGI) Improvement and Severity scales and Caregiver Burden Scale (CBS), through 4 weeks in the OLE. Safety assessments were conducted at each visit. RESULTS: Of 459 patients, 424 (92.4%) had a Week 4 efficacy assessment. At Week 4 (10 weeks total treatment), SAPS-PD mean (standard deviation) change from OLE baseline was -1.8 (5.5) and for SAPS-H + D was -2.1 (6.2) with pimavanserin 34 mg. Patients receiving placebo during the Core studies had greater improvements (SAPS-PD -2.9 [5.6]; SAPS-H + D -3.5 [6.3]) during the OLE. For participants treated with pimavanserin 8.5 or 17 mg during the Core studies, further improvement was observed during the OLE with pimavanserin 34 mg. The mean change from Core Study baseline for SAPS-PD score was similar among prior pimavanserin 34 mg and prior placebo-treated participants (-7.1 vs. -7.0). The CGI-I response rate (score of 1 or 2) at Week 4 was 51.4%. Adverse events were reported by 215 (46.8%) patients during the first 4 weeks of OLE. The most common AEs were fall (5.9%), hallucination (3.7%), urinary tract infection (2.8%), insomnia (2.4%), and peripheral edema (2.2%) CONCLUSIONS: Patients previously on pimavanserin 34 mg during three blinded core studies had durability of efficacy during the subsequent 4 week OLE SAPS-PD assessment. Patients previously on blinded placebo improved after 4 weeks of OL pimavanserin treatment. These results in over 400 patients from 14 countries support the efficacy of pimavanserin for treating PDP.


Asunto(s)
Antipsicóticos/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Piperidinas/farmacología , Trastornos Psicóticos/tratamiento farmacológico , Urea/análogos & derivados , Anciano , Anciano de 80 o más Años , Antipsicóticos/administración & dosificación , Antipsicóticos/efectos adversos , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Enfermedad de Parkinson/complicaciones , Piperidinas/administración & dosificación , Piperidinas/efectos adversos , Trastornos Psicóticos/etiología , Urea/administración & dosificación , Urea/efectos adversos , Urea/farmacología
16.
Drug Metab Dispos ; 49(8): 619-628, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34011533

RESUMEN

Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that is currently in clinical development for the treatment of heart failure. The absorption and disposition of [14C]OM (60 µCi) were studied after a single intravenous infusion (35 mg over 1 hour) or oral solution dose (35 mg) in 14 healthy male subjects. Mean recovery of the administered [14C]OM dose was 85.1% and 86.5% over 336 hours for the intravenous and oral routes, respectively. After intravenous dosing, 47.8% and 37.3% of the dose was recovered in urine and feces, respectively; after oral dosing, 48.6% and 38.0% was recovered in urine and feces, respectively. Unchanged OM accounted for a minor percentage of radioactivity in urine (mean 7.7% of dose) and feces (mean 4.1% of dose) across all subjects. The major metabolites recovered in urine and feces were M3 (decarbamoylation product) and sequential metabolite M4 (lactam of M3), which accounted for means of 26.5% and 11.6% of the administered dose, respectively. The CYP4 family of enzymes was primarily responsible for the formation of M3 based on in vitro studies. Other metabolic pathways accounted for 14.9% of the administered dose. In pooled plasma, OM, M3, and M4 accounted for 83.8%, 6.0%, and 3.3% of the total [14C]OM-related materials. No other plasma metabolites constituted more than 3% of the administered dose. The bioavailability for OM solution was 93.5% after rapid and extensive absorption. SIGNIFICANCE STATEMENT: This study characterized the absorption and disposition of OM, a novel small molecule being developed for the treatment of heart failure. OM was primarily cleared through metabolism by the CYP4 family through oxidative cleavage of a terminal carbamate moiety that resembles hydrolysis.


Asunto(s)
Familia 4 del Citocromo P450/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Eliminación Hepatobiliar/fisiología , Absorción Intestinal/fisiología , Eliminación Renal/fisiología , Urea/análogos & derivados , Administración Intravenosa , Administración Oral , Adulto , Disponibilidad Biológica , Biotransformación , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/farmacocinética , Voluntarios Sanos , Humanos , Infusiones Intravenosas , Masculino , Urea/administración & dosificación , Urea/farmacocinética
18.
Neurosci Lett ; 755: 135909, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33892002

RESUMEN

Orexin neuropeptides are implicated in the expression of morphine dependence. Locus coeruleus (LC) nucleus is an important brain area involving in the development of withdrawal signs of morphine and contains high expression of orexin type 1 receptors (OX1Rs). Despite extensive considerations, effects of immediate inhibition of OX1Rs by a single dose administration of SB-334867 prior to the naloxone-induced activation of LC neurons remains unknown. Therefore, we examined the direct effects of OX1Rs acute blockade on the neuronal activity of the morphine-dependent rats which underwent naloxone administration. Adult male rats underwent subcutaneous administration of 10 mg/kg morphine (two times/day) for a ten-day period. On the last day of experiment, intra-cerebroventricular administration of 10 µg/µl antagonist of OX1Rs, SB-334867, was performed just before intra-peritoneal injection of 2 mg/kg naloxone. Thereafter, in vivo extracellular single unit recording was employed to evaluate the electrical activity of LC neuronal cells. The outcomes demonstrated that morphine tolerance developed following ten-day of injection. Then, naloxone administration causes hyperactivity of LC neuronal cells, whereas a single dose administration of SB-334867 prior to naloxone prevented the enhanced activity of neurons upon morphine withdrawal. Our findings indicate that increased response of LC neuronal cells to applied naloxone could be prevented by the acute inhibition of the OX1Rs just before the naloxone treatment.


Asunto(s)
Locus Coeruleus/fisiología , Dependencia de Morfina/fisiopatología , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de los Receptores de Orexina/administración & dosificación , Receptores de Orexina/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Animales , Benzoxazoles/administración & dosificación , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Locus Coeruleus/efectos de los fármacos , Masculino , Morfina/administración & dosificación , Morfina/efectos adversos , Dependencia de Morfina/tratamiento farmacológico , Naftiridinas/administración & dosificación , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Wistar , Urea/administración & dosificación , Urea/análogos & derivados
19.
Mol Imaging Biol ; 23(5): 745-755, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33891265

RESUMEN

PURPOSE: PSMA overexpression has been associated with aggressive prostate cancer (PCa). However, PSMA PET imaging has revealed highly variable changes in PSMA expression in response to ADT treatment ranging from increases to moderate decreases. To better understand these PSMA responses and potential relationship to progressive PCa, the PET imaging agent, [18F]DCFPyL, was used to assess changes in PSMA expression in response to ADT using genomically characterized LuCaP patient-derived xenograft mouse models (LuCaP-PDXs) which were found to be sensitive to ADT (LuCaP73 and LuCaP136;CS) or resistant (LuCaP167;CR). METHODS: [18F]DCFPyL (2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid) was used to assess PSMA in vitro (saturation assays) in LuCaP tumor membrane homogenates and in vivo (imaging/biodistribution) in LuCaP-PDXs. Control and ADT-treated LuCaPs were imaged before ADT (0 days) and 2-, 7-, 14-, and 21-days post-ADT from which tumor:muscle ratios (T:Ms) were determined and concurrently tumor volumes were measured (caliper). After the 21-day imaging, biodistributions and histologic/genomic (PSMA, AR) analysis were done. RESULTS: [18F]DCFPyL exhibited high affinity for PSMA and distinguished different levels of PSMA in LuCaP tumors. Post-ADT CS LuCaP73 and LuCaP136 tumor volumes significantly decreased at day 7 or 14 respectively vs controls, whereas the CR LuCaP167 tumor volumes were minimally changed. [18F]DCFPyL imaging T:Ms were increased 3-5-fold in treated LuCaP73 tumors vs controls, while treated LuCaP136 T:Ms remained unchanged which was confirmed by day 21 biodistribution results. For treated LuCaP167, T:Ms were decreased (~ 45 %) vs controls but due to low T:M values (<2) may not be indicative of PSMA level changes. LuCaP73 tumor PSMA histologic/genomic results were comparable to imaging/biodistribution results, whereas the results for other tumor types varied. CONCLUSION: Tumor responses to ADT varied from sensitive to resistant among these LuCaP PDXs, while only the high PSMA expressing LuCaP model exhibited an increase in PSMA levels in response to ADT. These models may be useful in understanding the clinical relevance of PSMA PET responses to ADT and potentially the relationship to disease progression as it may relate to the genomic signature.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Lisina/análogos & derivados , Tomografía de Emisión de Positrones/métodos , Antígeno Prostático Específico , Neoplasias de la Próstata , Urea/análogos & derivados , Animales , Antineoplásicos Hormonales/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Lisina/administración & dosificación , Lisina/metabolismo , Lisina/farmacocinética , Masculino , Ratones , Antígeno Prostático Específico/análisis , Antígeno Prostático Específico/química , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Urea/administración & dosificación , Urea/metabolismo , Urea/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Methods Mol Biol ; 2283: 15-20, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33765304

RESUMEN

The concept of the urea breath test (UBT), as a method for H. pylori detection, is based on the ability of the H. pylori urease enzyme to break down an isotope-labelled urea solution ingested by the patient into carbon dioxide (CO2) and ammonia. This chapter summarizes the current use of the UBT and the utility of the "UBT and Treat" strategy compared to other strategies for the management of H. pylori infection . Different UBT methods are described as well as factors affecting the accuracy of the test.


Asunto(s)
Infecciones por Helicobacter/diagnóstico , Helicobacter pylori/enzimología , Urea/administración & dosificación , Ureasa/metabolismo , Amoníaco/química , Proteínas Bacterianas/metabolismo , Pruebas Respiratorias , Dióxido de Carbono/química , Femenino , Helicobacter pylori/aislamiento & purificación , Humanos , Masculino , Sensibilidad y Especificidad , Urea/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...