Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
2.
Pediatr Diabetes ; 22(2): 192-201, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33244872

RESUMEN

BACKGROUND: While circulating levels of alpha1 -proteinase inhibitor (alpha1 -PI) are typically normal, antiprotease activity appears to be compromised in patients with Type 1 diabetes mellitus (T1DM). Because alpha1 -PI [human] (alpha1 -PI[h]) therapy can inhibit pro-inflammatory mediators associated with ß-cell destruction and reduced insulin production, it has been proposed for T1DM disease prevention. The aim of this study was to evaluate safety, tolerability, and efficacy of intravenous (IV) alpha1 -PI[h] in preserving C-peptide production in newly diagnosed T1DM patients. PARTICIPANTS: Seventy-six participants (aged 6-35 years) were randomized at 25 centers within 3 months of T1DM diagnosis. METHODS: A Phase II, multicenter, partially blinded, placebo-controlled, proof-of-concept study evaluating four dosing regimens of alpha1 -PI[h] (NCT02093221, GTI1302): weekly IV infusions of either 90 or 180 mg/kg, each for either 13 or 26 weeks. Safety and efficacy were monitored over 52 weeks with an efficacy evaluation planned at 104 weeks. The primary efficacy endpoint was change from baseline in the 2-h area-under-the-curve C-peptide level from a mixed-meal tolerance test at 52 weeks. A battery of laboratory tests, including inflammatory biomarkers, constituted exploratory efficacy variables. RESULTS: Infusions were well tolerated with no new safety signals. All groups exhibited highly variable declines in the primary outcome measure at 52 weeks with no statistically significant difference from placebo. Interleukin-6 (IL-6) was reduced from baseline in all alpha1 -PI treatment groups but not the placebo group. CONCLUSION: Pharmacologic therapy with alpha1 -PI[h] is safe, well tolerated, and able to reduce IL-6 levels; however, due to variability in the efficacy endpoint, its effects on preservation of C-peptide production were inconclusive.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Inhibidores de Serina Proteinasa/administración & dosificación , alfa 1-Antitripsina/administración & dosificación , Adolescente , Adulto , Péptido C/sangre , Niño , Diabetes Mellitus Tipo 1/sangre , Esquema de Medicación , Femenino , Humanos , Infusiones Intravenosas , Interleucina-6/sangre , Masculino , Prueba de Estudio Conceptual , Inhibidores de Serina Proteinasa/farmacocinética , Resultado del Tratamiento , Adulto Joven , alfa 1-Antitripsina/farmacocinética
3.
J Med Chem ; 63(23): 14740-14760, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33226226

RESUMEN

The discovery of a pan-genotypic hepatitis C virus (HCV) NS3/4A protease inhibitor based on a P1-P3 macrocyclic tripeptide motif is described. The all-carbon tether linking the P1-P3 subsites of 21 is functionalized with alkyl substituents, which are shown to effectively modulate both potency and absorption, distribution, metabolism, and excretion (ADME) properties. The CF3Boc-group that caps the P3 amino moiety was discovered to be an essential contributor to metabolic stability, while positioning a methyl group at the C1 position of the P1' cyclopropyl ring enhanced plasma trough values following oral administration to rats. The C7-fluoro, C6-CD3O substitution pattern of the P2* isoquinoline heterocycle of 21 was essential to securing the targeted potency, pharmacokinetic (PK), and toxicological profiles. The C6-CD3O redirected metabolism away from a problematic pathway, thereby circumventing the time-dependent cytochrome P (CYP) 450 inhibition observed with the C6-CH3O prototype.


Asunto(s)
Antivirales/farmacología , Péptidos Cíclicos/farmacología , Inhibidores de Serina Proteinasa/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Antivirales/farmacocinética , Células CHO , Cricetulus , Descubrimiento de Drogas , Estabilidad de Medicamentos , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Pruebas de Sensibilidad Microbiana , Microsomas Hepáticos/metabolismo , Estructura Molecular , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacocinética , Ratas , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/metabolismo , Inhibidores de Serina Proteinasa/farmacocinética , Relación Estructura-Actividad
4.
J Nucl Med ; 61(12): 1806-1813, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32332144

RESUMEN

Fibroblast activation protein (FAP) has emerged as an interesting molecular target used in the imaging and therapy of various types of cancers. 68Ga-labeled chelator-linked FAP inhibitors (FAPIs) have been successfully applied to PET imaging of various tumor types. To broaden the spectrum of applicable PET tracers for extended imaging studies of FAP-dependent diseases, we herein report the radiosynthesis and preclinical evaluation of an 18F-labeled glycosylated FAPI ([18F]FGlc-FAPI). Methods: An alkyne-bearing precursor was synthesized and subjected to click chemistry-based radiosynthesis of [18F]FGlc-FAPI by 2-step 18F-fluoroglycosylation. FAP-expressing HT1080hFAP cells were used to study competitive binding to FAP, cellular uptake, internalization, and efflux of [18F]FGlc-FAPI in vitro. Biodistribution studies and in vivo small-animal PET studies of [18F]FGlc-FAPI compared with [68Ga]Ga-FAPI-04 were conducted in nude mice bearing HT1080hFAP tumors or U87MG xenografts. Results: [18F]FGlc-FAPI was synthesized with a 15% radioactivity yield and a high radiochemical purity of more than 99%. In HT1080hFAP cells, [18F]FGlc-FAPI showed specific uptake, a high internalized fraction, and low cellular efflux. Compared with FAPI-04 (half maximal inhibitory concentration [IC50] = 32 nM), the glycoconjugate, FGlc-FAPI (IC50 = 167 nM), showed slightly lower affinity for FAP in vitro, whereas plasma protein binding was higher for [18F]FGlc-FAPI. Biodistribution studies revealed significant hepatobiliary excretion of [18F]FGlc-FAPI; however, small-animal PET studies in HT1080hFAP xenografts showed higher specific tumor uptake of [18F]FGlc-FAPI (4.5 percentage injected dose per gram of tissue [%ID/g]) than of [68Ga]Ga-FAPI-04 (2 %ID/g). In U87MG tumor-bearing mice, both tracers showed similar tumor uptake, but [18F]FGlc-FAPI showed a higher tumor retention. Interestingly, [18F]FGlc-FAPI demonstrated high specific uptake in bone structures and joints. Conclusion: [18F]FGlc-FAPI is an interesting candidate for translation to the clinic, taking advantage of the longer half-life and physical imaging properties of 18F. The availability of [18F]FGlc-FAPI may allow extended PET studies of FAP-related diseases, such as cancer, but also arthritis, heart diseases, or pulmonary fibrosis.


Asunto(s)
Radioisótopos de Flúor/química , Gelatinasas/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/síntesis química , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Técnicas de Química Sintética , Endopeptidasas , Femenino , Humanos , Marcaje Isotópico , Ratones , Terapia Molecular Dirigida , Tomografía de Emisión de Positrones , Radioquímica , Serina Endopeptidasas , Inhibidores de Serina Proteinasa/farmacocinética , Inhibidores de Serina Proteinasa/farmacología , Distribución Tisular
5.
Bioorg Med Chem Lett ; 30(4): 126949, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31932224

RESUMEN

The discovery of orally bioavailable FXIa inhibitors has been a challenge. Herein, we describe our efforts to address this challenge by optimization of our imidazole-based macrocyclic series. Our optimization strategy focused on modifications to the P2 prime, macrocyclic amide linker, and the imidazole scaffold. Replacing the amide of the macrocyclic linker with amide isosteres led to the discovery of substituted amine linkers which not only maintained FXIa binding affinity but also improved oral exposure in rats. Combining the optimized macrocyclic amine linker with a pyridine scaffold afforded compounds 23 and 24 that were orally bioavailable, single-digit nanomolar FXIa inhibitors with excellent selectivity against relevant blood coagulation enzymes.


Asunto(s)
Aminas/química , Factor XIa/antagonistas & inhibidores , Compuestos Macrocíclicos/química , Inhibidores de Serina Proteinasa/síntesis química , Administración Oral , Animales , Sitios de Unión , Diseño de Fármacos , Factor XIa/metabolismo , Semivida , Compuestos Macrocíclicos/metabolismo , Compuestos Macrocíclicos/farmacocinética , Simulación de Dinámica Molecular , Estructura Terciaria de Proteína , Piridinas/química , Ratas , Inhibidores de Serina Proteinasa/metabolismo , Inhibidores de Serina Proteinasa/farmacocinética , Relación Estructura-Actividad
6.
Respir Investig ; 57(1): 89-96, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30416054

RESUMEN

BACKGROUND: Alpha1-Proteinase Inhibitor, Modified Process (Alpha-1 MP) is used for augmentation therapy in alpha1-antitrypsin deficiency (AATD), an extremely rare disease in Japan. Weekly doses of 60 mg/kg Alpha-1 MP have been shown to be safe and well tolerated in non-Japanese subjects, but the safety and pharmacokinetics (PK) have not been evaluated in Japanese subjects. The objectives of this study were to evaluate the safety and PK of 60 mg/kg Alpha-1 MP administered by weekly IV infusions over 8 weeks in Japanese subjects with AATD. METHODS: This was a multicenter, open-label trial in Japanese adults aged ≥20 years with AATD. Samples for evaluation of serum alpha1-PI concentration and PK parameters were collected at 10 time points until the seventh day after the last dose at Week 8: immediately before dosing, immediately after dosing (time 0), and 0.25, 2, 4, 8, 24, 48, 120, and 168 hours after dosing. RESULTS: Four subjects were analyzed. The median tmax was 0.534 h. Mean ± SD values for t½, Cmax, and AUC0-7days were 150.4 ± 36.18 h, 174.2 ± 30.51 mg/dL, and 14,913.2 ± 1633.45 mg*h/dL, respectively. Mean trough concentration at week 8 was 55.4 ± 7.23 mg/dL. Alpha-1 MP therapy was safe, with no serious adverse events or deaths reported. Two treatment-emergent adverse events of fatigue in one subject were considered to be possibly related. CONCLUSIONS: The PK and safety of Alpha-1 MP in Japanese subjects with AATD were consistent with the Alpha-1 MP profile in non-Japanese subjects (ClinicalTrials.gov: NCT02870309; JAPIC CTI: JapicCTI-163160).


Asunto(s)
Inhibidores de Serina Proteinasa/administración & dosificación , Inhibidores de Serina Proteinasa/farmacocinética , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , alfa 1-Antitripsina/administración & dosificación , alfa 1-Antitripsina/farmacocinética , Anciano , Pueblo Asiatico , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Seguridad , Factores de Tiempo , Deficiencia de alfa 1-Antitripsina/metabolismo
7.
J Nucl Med ; 60(3): 386-392, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30072500

RESUMEN

Fibroblast activation protein (FAP) is overexpressed in cancer-associated fibroblasts of several tumor entities. The recent development of quinoline-based PET tracers that act as FAP inhibitors (FAPIs) demonstrated promising results preclinically and already in a few clinical cases. Consequently, these tracers are now applied in our hospital to amend the diagnostics of cancer patients facing the limitations of standard examinations. Here, we analyze the tissue biodistribution and preliminary dosimetry of 2 members of this new class of PET radiopharmaceutical. Methods: A preliminary dosimetry estimate for 68Ga-FAPI-2 and 68Ga-FAPI-4 was based on 2 patients examined at 0.2, 1, and 3 h after tracer injection using the QDOSE dosimetry software suit. Further PET/CT scans of tumor patients were acquired 1 h after injection of either 68Ga-FAPI-2 (n = 25) or 68Ga-FAPI-4 (n = 25); for 6 patients an intraindividual related 18F-FDG scan (also acquired 1 h after injection) was available. For the normal tissue of 16 organs, a 2-cm spheric volume of interest was placed in the parenchyma; for tumor lesions, a threshold-segmented volume of interest was used to quantify SUVmean and SUVmaxResults: Similar to literature values for 18F-FDG, 68Ga-DOTATATE, and 68Ga-PSMA-11, an examination with 200 MBq of 68Ga-FAPI-2 or 68Ga-FAPI-4 corresponds to an equivalent dose of approximately 3-4 mSv. After a fast clearance via the kidneys, the normal organs showed a low tracer uptake with only minimal changes between 10 min and 3 h after injection. In 68Ga-FAPI-2, the tumor uptake from 1 to 3 h after injection decreased by 75%, whereas the tumor retention was prolonged with 68Ga-FAPI-4 (25% washout). Regarding tumor-to-background ratios, at 1 h after injection both 68Ga-FAPI tracers performed equally. In comparison to 18F-FDG, the tumor uptake was almost equal (average SUVmax, 7.41 for 18F-FDG and 7.37 for 68Ga-FAPI-2; not statistically significant); the background uptake in brain (11.01 vs. 0.32), liver (2.77 vs. 1.69), and oral/pharyngeal mucosa (4.88 vs. 2.57) was significantly lower with 68Ga-FAPI. Other organs did not relevantly differ between 18F-FDG and 68Ga-FAPI. Conclusion: FAPI PET/CT is a new diagnostic method in imaging cancer patients. In contrast to 18F-FDG, no diet or fasting in preparation for the examination is necessary, and image acquisition can potentially be started a few minutes after tracer application. Tumor-to-background contrast ratios were equal to or even better than those of 18F-FDG.


Asunto(s)
Radioisótopos de Galio , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacocinética , Adulto , Anciano , Anciano de 80 o más Años , Endopeptidasas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Radiometría , Radiofármacos/química , Radiofármacos/metabolismo , Radiofármacos/farmacocinética , Inhibidores de Serina Proteinasa/metabolismo , Distribución Tisular
8.
Rev Cardiovasc Med ; 19(S1): S31-S46, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30207556

RESUMEN

Low-density lipoprotein (LDL) receptors on the surface of liver hepatocytes are the primary way that humans regulate serum LDL cholesterol levels. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a proteolytic enzyme that indirectly regulates serum LDL cholesterol (LDL-C) by causing the destruction of LDL receptors. Less LDL receptors result in increased LDL-C in the bloodstream but inhibiting or binding the circulating PCSK9 results in increased LDL receptors with the resultant decrease in serum LDL-C. Two PCSK9 inhibitors are currently approved for use: alirocumab and evolocumab. Both are fully human monoclonal antibodies that bind free PCSK9. Herein we discuss the mechanism of action, efficacy, and safety of PCSK9 inhibitors. clinical problem.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticolesterolemiantes/uso terapéutico , LDL-Colesterol/sangre , Dislipidemias/tratamiento farmacológico , Inhibidores de PCSK9 , Inhibidores de Serina Proteinasa/uso terapéutico , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Anticolesterolemiantes/efectos adversos , Anticolesterolemiantes/farmacocinética , Biomarcadores/sangre , Dislipidemias/sangre , Dislipidemias/diagnóstico , Dislipidemias/enzimología , Humanos , Proproteína Convertasa 9/metabolismo , Inhibidores de Serina Proteinasa/efectos adversos , Inhibidores de Serina Proteinasa/farmacocinética , Resultado del Tratamiento
9.
Bioorg Med Chem Lett ; 28(20): 3372-3375, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201291

RESUMEN

We report here the design and synthesis of a novel series of benzylamines that are potent and selective inhibitors of uPA with promising oral availability in rat. Further evaluation of one representative (ZK824859) of the new structural class showed that this compound lowered clinical scores when dosed in either acute or chronic mouse EAE models, suggesting that uPA inhibitors of this type could be useful for the treatment of multiple sclerosis.


Asunto(s)
Bencilaminas/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Inhibidores de Serina Proteinasa/uso terapéutico , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Animales , Bencilaminas/síntesis química , Bencilaminas/química , Bencilaminas/farmacocinética , Sitios de Unión , Femenino , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Ratas , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacocinética , Relación Estructura-Actividad , Activador de Plasminógeno de Tipo Uroquinasa/química
10.
Acta Pharmacol Sin ; 39(11): 1746-1752, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29930277

RESUMEN

NS3/4A serine protease is a prime target for direct-acting antiviral therapies against hepatitis C virus (HCV) infection. Several NS3/4A inhibitors have been widely used in clinic, while new inhibitors with better characteristics are still urgently needed. GP205 is a new macrocyclic inhibitor of NS3/4A with low nanomolar activities against HCV replicons of genotypes 1b, 2a, 4a, and 5a, with EC50 values ranging from 1.5 to 12.8 nmol/L. In resistance selection study in vitro, we found resistance-associated substitutions on D168: The activity of GP205 was significantly attenuated against 1b replicon with D168V or D168A mutation, similar as simeprevir. No cross resistance of GP205 with NS5B or NS5A inhibitor was observed. Combination of GP205 with sofosbuvir or daclatasvir displayed additive or synergistic efficacy. The pharmacokinetic profile of GP205 was characterized in rats and dogs after oral administration, which revealed good drug exposure both in plasma and in liver and long plasma half-life. The in vitro stability test showed ideal microsomal and hepatic cells stability of GP205. The preclinical profiles of GP205 support further research on this NS3/4A inhibitor to expand the existing HCV infection therapies.


Asunto(s)
Antivirales/uso terapéutico , Hepatitis C/tratamiento farmacológico , Compuestos Macrocíclicos/uso terapéutico , Inhibidores de Serina Proteinasa/uso terapéutico , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/farmacocinética , Carbamatos , Línea Celular Tumoral , Perros , Combinación de Medicamentos , Estabilidad de Medicamentos , Sinergismo Farmacológico , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Humanos , Imidazoles/farmacocinética , Imidazoles/uso terapéutico , Compuestos Macrocíclicos/farmacocinética , Masculino , Microsomas Hepáticos/metabolismo , Mutación , Pirrolidinas , Ratas Sprague-Dawley , Serina Proteasas/genética , Inhibidores de Serina Proteinasa/farmacocinética , Sofosbuvir/farmacocinética , Sofosbuvir/uso terapéutico , Valina/análogos & derivados , Proteínas no Estructurales Virales/genética
11.
Cell Chem Biol ; 25(8): 1031-1037.e4, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-29779956

RESUMEN

Prolyl oligopeptidase (POP), a serine protease highly expressed in the brain, has recently emerged as an enticing therapeutic target for the treatment of cognitive and neurodegenerative disorders. However, most reported inhibitors suffer from short duration of action, poor protease selectivity, and low blood-brain barrier (BBB) permeability, which altogether limit their potential as drugs. Here, we describe the structure-based design of the first irreversible, selective, and brain-permeable POP inhibitors. At low-nanomolar concentrations, these covalent peptidomimetics produce a fast, specific, and sustained inactivation of POP, both in vitro and in human cells. More importantly, they are >1,000-fold selective against two family-related proteases (DPPIV and FAP) and display high BBB permeability, as shown in both lipid membranes and MDCK cells.


Asunto(s)
Fluoruros/química , Fluoruros/farmacología , Peptidomiméticos/química , Peptidomiméticos/farmacología , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Línea Celular , Perros , Descubrimiento de Drogas , Fluoruros/farmacocinética , Humanos , Células de Riñón Canino Madin Darby , Modelos Moleculares , Peptidomiméticos/farmacocinética , Permeabilidad , Prolil Oligopeptidasas , Inhibidores de Serina Proteinasa/farmacocinética
12.
Cardiovasc Drugs Ther ; 32(2): 175-180, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29627892

RESUMEN

PURPOSE: Clinical trials of statins and other lipid-lowering therapies (LLTs) often report large inter-individual variations in their effects on low-density lipoprotein cholesterol (LDL-C). We evaluated apparent hyporesponsiveness to the proprotein convertase subtilisin/kexin type 9 inhibitor alirocumab (defined as < 15% LDL-C reduction from baseline at all timepoints) using data from 10 Phase 3 trials (3120 hypercholesterolemic patients). METHODS: This report assessed the LDL-C percent reduction from baseline at weeks 4-104 (depending on study), and alirocumab serum levels and antidrug antibodies, in patients with apparent hyporesponsiveness. RESULTS: Among the 3120 patients evaluated, 98.9% responded to alirocumab, and 33 (1.1%) had < 15% LDL C reduction at all measured timepoints. Pharmacokinetics data indicated that 13/33 apparent hyporesponders had not received alirocumab; no pharmacokinetics data were available for 14/33, and 6/33 had detectable alirocumab. For the six patients with confirmed alirocumab receipt, the degree of adherence to pre-study concurrent LLTs could not be determined after study start; one of these patients had persistent antidrug antibodies. CONCLUSIONS: Apparent hyporesponsiveness to alirocumab appeared to be due to lack of receipt of alirocumab determined by serum alirocumab levels, possible lack of adherence to concurrent LLTs, a theoretical and rare possibility of biological non-responsiveness due to persistent antidrug antibodies, or other causes, as yet unidentified.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticolesterolemiantes/uso terapéutico , LDL-Colesterol/sangre , Hipercolesterolemia/tratamiento farmacológico , Inhibidores de PCSK9 , Inhibidores de Serina Proteinasa/uso terapéutico , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Anticolesterolemiantes/farmacocinética , Biomarcadores/sangre , Ensayos Clínicos Fase III como Asunto , Regulación hacia Abajo , Resistencia a Medicamentos , Quimioterapia Combinada , Humanos , Hipercolesterolemia/sangre , Hipercolesterolemia/diagnóstico , Cumplimiento de la Medicación , Proproteína Convertasa 9/inmunología , Proproteína Convertasa 9/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Inhibidores de Serina Proteinasa/farmacocinética , Resultado del Tratamiento
13.
Clin Pharmacol Ther ; 104(6): 1155-1164, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29484635

RESUMEN

Neutrophil serine proteases (NSPs), such as neutrophil elastase (NE), are activated by dipeptidyl peptidase 1 (DPP1) during neutrophil maturation. High NSP levels can be detrimental, particularly in lung tissue, and inhibition of NSPs is therefore an interesting therapeutic opportunity in multiple lung diseases, including chronic obstructive pulmonary disease (COPD) and bronchiectasis. We conducted a randomized, placebo-controlled, first-in-human study to assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of single and multiple oral doses of the DPP1 inhibitor AZD7986 in healthy subjects. Pharmacokinetic and pharmacodynamic data were analyzed using nonlinear mixed effects modeling and showed that AZD7986 inhibits whole blood NE activity in an exposure-dependent, indirect manner-consistent with in vitro and preclinical predictions. Several dose-dependent, possibly DPP1-related, nonserious skin findings were observed, but these were not considered to prevent further clinical development. Overall, the study results provided confidence to progress AZD7986 to phase II and supported selection of a clinically relevant dose.


Asunto(s)
Benzoxazoles/administración & dosificación , Catepsina C/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/administración & dosificación , Elastasa de Leucocito/antagonistas & inhibidores , Neutrófilos/efectos de los fármacos , Oxazepinas/administración & dosificación , Inhibidores de Serina Proteinasa/administración & dosificación , Administración Oral , Benzoxazoles/efectos adversos , Benzoxazoles/farmacocinética , Inhibidores de Cisteína Proteinasa/efectos adversos , Inhibidores de Cisteína Proteinasa/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Voluntarios Sanos , Humanos , Elastasa de Leucocito/sangre , Masculino , Modelos Biológicos , Neutrófilos/enzimología , Dinámicas no Lineales , Oxazepinas/efectos adversos , Oxazepinas/farmacocinética , Inhibidores de Serina Proteinasa/farmacocinética
14.
Cardiovasc Ther ; 36(1)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29057618

RESUMEN

AIMS: Two multiple-dose phase II studies were conducted in subjects with primary hypercholesterolemia to evaluate the LDL-C lowering efficacy, safety, and tolerability of bococizumab, a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor. METHODS: The results from the two phase II, double-blinded, randomized, placebo-controlled, multicenter studies conducted in the USA and Canada were combined. In Study 1, 90 subjects with LDL-C ≥100 mg/dL received intravenous (IV) placebo or bococizumab 0.25, 1, 3, or 6 mg/kg. In Study 2, 45 subjects with LDL-C ≥80 mg/dL received IV placebo or bococizumab 1 or 3 mg/kg. Subjects were treated every 4 weeks for 12 weeks. Dosing was interrupted if LDL-C dipped to ≤25 mg/dL and resumed if LDL-C returned to ≥40 mg/dL. The primary endpoint was percent LDL-C reduction from baseline at Week 12. RESULTS: At Week 12, the reductions from baseline in LDL-C vs placebo in the bococizumab 0.25, 1, 3, and 6 mg/kg groups were 9.3%, 10.2%, 41.6%, and 52.0%, respectively (P < .001 vs placebo for all). LDL-C reductions were greater (69.9%) in subjects who received all three doses of bococizumab 6 mg/kg (P < .001 vs placebo). Pharmacogenomic analysis revealed that 15% of hyperlipidemic subjects carried polymorphisms associated with familial hypercholesterolemia, with maximal LDL-C reductions being similar between carriers and noncarriers. Adverse events were mild, unrelated to bococizumab, and resolved by Week 12. CONCLUSIONS: These studies demonstrated that bococizumab safely and effectively lowered LDL-C in hypercholesterolemic subjects on high doses of statin.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticolesterolemiantes/administración & dosificación , LDL-Colesterol/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Hipercolesterolemia/tratamiento farmacológico , Inhibidores de PCSK9 , Inhibidores de Serina Proteinasa/administración & dosificación , Administración Intravenosa , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticolesterolemiantes/efectos adversos , Anticolesterolemiantes/farmacocinética , Biomarcadores/sangre , Canadá , Método Doble Ciego , Regulación hacia Abajo , Quimioterapia Combinada , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Hipercolesterolemia/sangre , Hipercolesterolemia/enzimología , Hipercolesterolemia/genética , Masculino , Persona de Mediana Edad , Farmacogenética , Proproteína Convertasa 9/metabolismo , Inhibidores de Serina Proteinasa/efectos adversos , Inhibidores de Serina Proteinasa/farmacocinética , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos
15.
Cardiovasc Ther ; 36(1)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29078037

RESUMEN

AIMS: Three single-dose and one multiple-dose phase I studies were conducted in subjects with primary hypercholesterolemia to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of bococizumab, a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor. METHODS: The dosing schedules for hypercholesterolemic subjects randomized in the four phase I studies were (1) ascending, single, intravenous (IV) bococizumab (0.3, 1, 3, 6, 12, or 18 mg/kg), or placebo (N = 48; baseline low-density lipoprotein cholesterol [LDL-C] ≥130 mg/dL); (2) single, IV bococizumab (0.5 or 4 mg/kg; no placebo) added to ongoing atorvastatin 40 mg/day (N = 24); (3) single, fixed, subcutaneous (SC) bococizumab (100 or 200 mg), or IV bococizumab (200 mg; no placebo; N = 49; baseline LDL-C ≥130 mg/dL); and (4) weekly IV bococizumab (0.25, 0.5, 1, or 1.5 mg/kg) or placebo for 4 weeks (N = 67; baseline LDL-C ≥130 mg/dL). RESULTS: Bococizumab pharmacokinetics were well characterized following single IV or SC doses and following multiple IV doses. Exposure to single-dose bococizumab increased slightly greater than dose-proportionally and clearance decreased with increasing dose. In the single-dose studies, maximal mean percent reductions from baseline in LDL-C ranged from 43% (0.3 mg/kg) to 84% (18 mg/kg) in bococizumab-treated subjects, compared with 2% for placebo. For the multiple-dose study, maximal reductions in LDL-C ranged from 55% (0.25 mg/kg) to 66% (1 mg/kg) in bococizumab-treated subjects, compared with 9% for placebo. In all studies, adverse events were infrequent, transient, and not dose-related. CONCLUSIONS: Bococizumab was generally safe and well tolerated. Bococizumab lowered LDL-C levels substantially in all four studies.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticolesterolemiantes/administración & dosificación , Atorvastatina/administración & dosificación , LDL-Colesterol/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Hipercolesterolemia/tratamiento farmacológico , Inhibidores de PCSK9 , Inhibidores de Serina Proteinasa/administración & dosificación , Administración Intravenosa , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticolesterolemiantes/efectos adversos , Anticolesterolemiantes/farmacocinética , Atorvastatina/efectos adversos , Biomarcadores/sangre , Regulación hacia Abajo , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Hipercolesterolemia/sangre , Hipercolesterolemia/enzimología , Hipercolesterolemia/genética , Masculino , Persona de Mediana Edad , Proproteína Convertasa 9/metabolismo , Inhibidores de Serina Proteinasa/efectos adversos , Inhibidores de Serina Proteinasa/farmacocinética , Factores de Tiempo , Resultado del Tratamiento
16.
J Med Chem ; 60(3): 1060-1075, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28085275

RESUMEN

A novel series of macrocyclic FXIa inhibitors was designed based on our lead acyclic phenyl imidazole chemotype. Our initial macrocycles, which were double-digit nanomolar FXIa inhibitors, were further optimized with assistance from utilization of structure-based drug design and ligand bound X-ray crystal structures. This effort resulted in the discovery of a macrocyclic amide linker which was found to form a key hydrogen bond with the carbonyl of Leu41 in the FXIa active site, resulting in potent FXIa inhibitors. The macrocyclic FXIa series, exemplified by compound 16, had a FXIa Ki = 0.16 nM with potent anticoagulant activity in an in vitro clotting assay (aPTT EC1.5x = 0.27 µM) and excellent selectivity against the relevant blood coagulation enzymes.


Asunto(s)
Amidas/química , Factor XIa/antagonistas & inhibidores , Compuestos Macrocíclicos/farmacología , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Descubrimiento de Drogas , Enlace de Hidrógeno , Ligandos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacocinética , Estructura Molecular , Inhibidores de Serina Proteinasa/farmacocinética
17.
Eur Neuropsychopharmacol ; 27(2): 180-191, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27986355

RESUMEN

Cognitive deficits are considered a key feature of schizophrenia, and they usually precede the onset of the illness and continue after psychotic symptoms appear. Current antipsychotic drugs have little or no effect on the cognitive deficits of this disorder. Prolyl oligopeptidase (POP) is an 81-kDa monomeric serine protease that is expressed in brain and other tissues. POP inhibitors have shown neuroprotective, anti-amnesic and cognition-enhancing properties. Here we studied the potential of IPR19, a new POP inhibitor, for the treatment of the cognitive symptoms related to schizophrenia. The efficacy of the inhibitor was evaluated in mouse models based on subchronic phencyclidine and acute dizocilpine administration, and in adult offspring from mothers with immune reaction induced by polyinosinic:polycytidylic acid administration during pregnancy. Acute IPR19 administration (5mg/kg, i.p.) reversed the cognitive performance deficits of the three mouse models in the novel object recognition test, T-maze, and eight-arm radial maze. The compound also ameliorates deficits of the prepulse inhibition response. The in vitro inhibitory efficacy and selectivity, brain penetration and exposure time after injection of IPR19 were also addressed. Our results indicate that the inhibition of POP using IPR19 may offer a promising strategy to develop drugs to ameliorate the cognitive deficits of schizophrenia.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Prolina/análogos & derivados , Psicotrópicos/farmacología , Esquizofrenia/tratamiento farmacológico , Psicología del Esquizofrénico , Animales , Línea Celular Tumoral , Cognición/efectos de los fármacos , Cognición/fisiología , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Poli I-C , Inhibición Prepulso/efectos de los fármacos , Inhibición Prepulso/fisiología , Prolina/química , Prolina/farmacocinética , Prolina/farmacología , Prolina/toxicidad , Prolil Oligopeptidasas , Psicotrópicos/química , Psicotrópicos/farmacocinética , Psicotrópicos/toxicidad , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Esquizofrenia/complicaciones , Esquizofrenia/enzimología , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacocinética , Inhibidores de Serina Proteinasa/farmacología , Inhibidores de Serina Proteinasa/toxicidad
18.
Vasc Health Risk Manag ; 12: 163-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27143910

RESUMEN

Multiple categories of medications have been developed to manage lipid profiles and reduce the risk of cardiovascular events in patients with heart disease. However, currently marketed medications have not solved the problems associated with preventing and treating cardiovascular diseases completely. A substantial population of patients cannot take advantage of statin therapy due to statin intolerance, heart failure, or kidney hemodialysis, suggesting a need for additional effective agents to reduce low-density lipoprotein cholesterol (LDL-C) levels. Proprotein convertase subtilisin/kexin type 9 (PCSK9) was discovered in 2003 and subsequently emerged as a novel target for LDL-C-lowering therapy. Evolocumab is a fully human monoclonal immunoglobulin G2 (IgG2) directed against human PCSK9. By inactivating PCSK9, evolocumab upregulates LDL receptors causing increased catabolism of LDL-C and the consequent reduction of LDL-C levels in blood. Overall, evolocumab has had notable efficacy, with LDL-C reduction ranging from 53% to 75% in monotherapy and combination therapies, and is associated with minor adverse effects. However, studies regarding the ability of evolocumab to reduce mortality as well as long-term safety concerns are limited. The fact that the drug was introduced at a cost much higher than the existing medications and shows a low incremental mortality benefit suggests that many payers will consider evolocumab to have an unfavorable cost-benefit ratio.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticolesterolemiantes/uso terapéutico , LDL-Colesterol/sangre , Hipercolesterolemia/tratamiento farmacológico , Proproteína Convertasas/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/uso terapéutico , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/economía , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Anticolesterolemiantes/efectos adversos , Anticolesterolemiantes/economía , Anticolesterolemiantes/farmacocinética , Biomarcadores/sangre , Análisis Costo-Beneficio , Regulación hacia Abajo , Costos de los Medicamentos , Quimioterapia Combinada , Humanos , Hipercolesterolemia/sangre , Hipercolesterolemia/diagnóstico , Hipercolesterolemia/economía , Hipercolesterolemia/mortalidad , Proproteína Convertasa 9 , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/efectos adversos , Inhibidores de Serina Proteinasa/economía , Inhibidores de Serina Proteinasa/farmacocinética , Resultado del Tratamiento
19.
Curr Opin Endocrinol Diabetes Obes ; 23(2): 97-105, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26859553

RESUMEN

PURPOSE OF REVIEW: This article summarizes the role that the recently approved proprotein convertase subtilisin/kexin type 9 (PCSK9) monoclonal antibodies (mAbs) will play in LDL cholesterol management. RECENT FINDINGS: The discovery of PCSK9 in 2003 and its association with a rare form of autosomal-dominant hypercholesterolemia quickly led to the understanding of the role of circulating PCSK9 in regulating the recycling of the LDL receptor and LDL cholesterol (LDL-C) and impact on cardiovascular disease. Development of monoclonal antibodies that bind and inhibit PCSK9 entered clinical development in late 2009 and demonstrated substantial LDL-C reductions as monotherapy, added to statins, in heterozygous familial hypercholesterolemia and patients intolerant of statins. Large and comprehensive trials over the last 5 years also indicated good tolerability and safety and resulted in the 2015 regulatory approval in the USA and Europe for the marketing of two mAbs, evolocumab and alirocumab, for the treatment of LDL-C. SUMMARY: The background, clinical trials and approved indications for the current PCSK9 inhibitors are reviewed along with their likely role in the management of LDL-C and cardiovascular disease prevention.


Asunto(s)
Hiperlipidemias/tratamiento farmacológico , Hipolipemiantes/uso terapéutico , Lípidos/sangre , Inhibidores de PCSK9 , Inhibidores de Serina Proteinasa/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Biomarcadores/sangre , Descubrimiento de Drogas , Humanos , Hiperlipidemias/sangre , Hiperlipidemias/diagnóstico , Hiperlipidemias/enzimología , Masculino , Terapia Molecular Dirigida , Proproteína Convertasa 9/metabolismo , Inhibidores de Serina Proteinasa/efectos adversos , Inhibidores de Serina Proteinasa/farmacocinética , Resultado del Tratamiento
20.
J Cyst Fibros ; 15(2): 227-33, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26321218

RESUMEN

BACKGROUND: Inhaled alpha1-proteinase inhibitor (PI) is known to reduce neutrophil elastase burden in some patients with CF. This phase 2a study was designed to test inhaled Alpha-1 HC, a new aerosolized alpha1-PI formulation, in CF patients. METHODS: We performed a randomized, double-blind, placebo-controlled study and evaluated the safety of 100 or 200mg of inhaled Alpha-1 HC once daily for 3 weeks in subjects with CF. Thirty adult subjects were randomized in a 2:1 ratio to receive Alpha-1 HC or placebo. RESULTS: Drug delivery was confirmed by a dose-dependent increase in the sputum alpha1-PI. Seven (20.0%) of the 35 adverse events in the 100-mg dose group, 3 (13.0%) of 23 in the 200-mg dose group, and 4 (14.3%) of 28 in the placebo group were drug-related in these subjects. One serious adverse event occurred in 1 subject within each group. CONCLUSIONS: Alpha-1 HC inhalation was safe and well tolerated.


Asunto(s)
Fibrosis Quística/tratamiento farmacológico , alfa 1-Antitripsina/administración & dosificación , Administración por Inhalación , Adulto , Fibrosis Quística/metabolismo , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Elastasa de Leucocito/sangre , Masculino , Inhibidores de Serina Proteinasa/administración & dosificación , Inhibidores de Serina Proteinasa/farmacocinética , Esputo/metabolismo , Factores de Tiempo , Resultado del Tratamiento , alfa 1-Antitripsina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...