Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Med Chem ; 65(1): 485-496, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34931831

RESUMEN

Inhibitor cystine knot peptides, derived from venom, have evolved to block ion channel function but are often toxic when dosed at pharmacologically relevant levels in vivo. The article describes the design of analogues of ProTx-II that safely display systemic in vivo blocking of Nav1.7, resulting in a latency of response to thermal stimuli in rodents. The new designs achieve a better in vivo profile by improving ion channel selectivity and limiting the ability of the peptides to cause mast cell degranulation. The design rationale, structural modeling, in vitro profiles, and rat tail flick outcomes are disclosed and discussed.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Dolor/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/farmacología , Venenos de Araña/síntesis química , Animales , Degranulación de la Célula/efectos de los fármacos , Cistina/química , Diseño de Fármacos , Calor , Mastocitos/efectos de los fármacos , Modelos Moleculares , Dimensión del Dolor/efectos de los fármacos , Ratas , Venenos de Araña/farmacología
2.
Chemistry ; 26(9): 2025-2033, 2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-31769085

RESUMEN

A novel series of C12-keto-type saxitoxin (STX) derivatives bearing an unusual nonhydrated form of the ketone at C12 has been synthesized, and their NaV -inhibitory activity has been evaluated in a cell-based assay as well as whole-cell patch-clamp recording. Among these compounds, 11-benzylidene STX (3 a) showed potent inhibitory activity against neuroblastoma Neuro 2A in both cell-based and electrophysiological analyses, with EC50 and IC50 values of 8.5 and 30.7 nm, respectively. Interestingly, the compound showed potent inhibitory activity against tetrodotoxin-resistant subtype of NaV 1.5, with an IC50 value of 94.1 nm. Derivatives 3 a-d and 3 f showed low recovery rates from NaV 1.2 subtype (ca 45-79 %) compared to natural dcSTX (2), strongly suggesting an irreversible mode of interaction. We propose an interaction model for the C12-keto derivatives with NaV in which the enone moiety in the STX derivatives 3 works as Michael acceptor for the carboxylate of Asp1717 .


Asunto(s)
Saxitoxina/química , Bloqueadores de los Canales de Sodio/síntesis química , Canales de Sodio Activados por Voltaje/metabolismo , Potenciales de Acción/efectos de los fármacos , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Técnicas de Placa-Clamp , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Teoría Cuántica , Saxitoxina/metabolismo , Saxitoxina/farmacología , Bloqueadores de los Canales de Sodio/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/química , Tetrodotoxina/metabolismo , Canales de Sodio Activados por Voltaje/química , Canales de Sodio Activados por Voltaje/genética
3.
Molecules ; 23(10)2018 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-30360356

RESUMEN

µ-Conotoxins are potent and highly specific peptide blockers of voltage-gated sodium channels. In this study, the solution structure of µ-conotoxin GIIIC was determined using 2D NMR spectroscopy and simulated annealing calculations. Despite high sequence similarity, GIIIC adopts a three-dimensional structure that differs from the previously observed conformation of µ-conotoxins GIIIA and GIIIB due to the presence of a bulky, non-polar leucine residue at position 18. The side chain of L18 is oriented towards the core of the molecule and consequently the N-terminus is re-modeled and located closer to L18. The functional characterization of GIIIC defines it as a canonical µ-conotoxin that displays substantial selectivity towards skeletal muscle sodium channels (NaV), albeit with ~2.5-fold lower potency than GIIIA. GIIIC exhibited a lower potency of inhibition of NaV1.4 channels, but the same NaV selectivity profile when compared to GIIIA. These observations suggest that single amino acid differences that significantly affect the structure of the peptide do in fact alter its functional properties. Our work highlights the importance of structural factors, beyond the disulfide pattern and electrostatic interactions, in the understanding of the functional properties of bioactive peptides. The latter thus needs to be considered when designing analogues for further applications.


Asunto(s)
Conotoxinas/química , Espectroscopía de Resonancia Magnética , Secuencia de Aminoácidos , Conotoxinas/síntesis química , Conotoxinas/farmacología , Disulfuros/química , Leucina/química , Modelos Moleculares , Péptidos/síntesis química , Péptidos/química , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/química , Canales de Sodio/metabolismo , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 26(13): 3207-3211, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27038498

RESUMEN

Previously we disclosed the discovery of potent Late INa current inhibitor 2 (GS-458967, IC50 of 333nM) that has a good separation of late versus peak Nav1.5 current, but did not have a favorable CNS safety window due to high brain penetration (3-fold higher partitioning into brain vs plasma) coupled with potent inhibition of brain sodium channel isoforms (Nav1.1, 1.2, 1.3). We increased the polar surface area from 50 to 84Å(2) by adding a carbonyl to the core and an oxadiazole ring resulting in 3 GS-462808 that had lower brain penetration and serendipitously lower activity at the brain isoforms. Compound 3 has an improved CNS window (>20 rat and dog) relative to 2, and improved anti-ischemic potency relative to ranolazine. The development of 3 was not pursued due to liver lesions in 7day rat toxicology studies.


Asunto(s)
Azoles/farmacología , Descubrimiento de Drogas , Corazón/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Piridinas/farmacología , Ranolazina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Animales , Azoles/síntesis química , Azoles/química , Perros , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Haplorrinos , Humanos , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Conejos , Ranolazina/síntesis química , Ranolazina/química , Ratas , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Relación Estructura-Actividad
5.
Bioorg Med Chem Lett ; 26(13): 3202-3206, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27080178

RESUMEN

We started with a medium throughput screen of heterocyclic compounds without basic amine groups to avoid hERG and ß-blocker activity and identified [1,2,4]triazolo[4,3-a]pyridine as an early lead. Optimization of substituents for Late INa current inhibition and lack of Peak INa inhibition led to the discovery of 4h (GS-458967) with improved anti-arrhythmic activity relative to ranolazine. Unfortunately, 4h demonstrated use dependent block across the sodium isoforms including the central and peripheral nervous system isoforms that is consistent with its low therapeutic index (approximately 5-fold in rat, 3-fold in dog). Compound 4h represents our initial foray into a 2nd generation Late INa inhibitor program and is an important proof-of-concept compound. We will provide additional reports on addressing the CNS challenge in a follow-up communication.


Asunto(s)
Descubrimiento de Drogas , Corazón/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Piridinas/farmacología , Ranolazina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Triazoles/farmacología , Animales , Células CACO-2 , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Macaca fascicularis , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Conejos , Ranolazina/síntesis química , Ranolazina/química , Ratas , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/química
6.
J Biol Chem ; 291(13): 7205-20, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26817840

RESUMEN

Cone snail toxins are well known blockers of voltage-gated sodium channels, a property that is of broad interest in biology and therapeutically in treating neuropathic pain and neurological disorders. Although most conotoxin channel blockers function by direct binding to a channel and disrupting its normal ion movement, conotoxin µO§-GVIIJ channel blocking is unique, using both favorable binding interactions with the channel and a direct tether via an intermolecular disulfide bond. Disulfide exchange is possible because conotoxin µO§-GVIIJ contains anS-cysteinylated Cys-24 residue that is capable of exchanging with a free cysteine thiol on the channel surface. Here, we present the solution structure of an analog of µO§-GVIIJ (GVIIJ[C24S]) and the results of structure-activity studies with synthetic µO§-GVIIJ variants. GVIIJ[C24S] adopts an inhibitor cystine knot structure, with two antiparallel ß-strands stabilized by three disulfide bridges. The loop region linking the ß-strands (loop 4) presents residue 24 in a configuration where it could bind to the proposed free cysteine of the channel (Cys-910, rat NaV1.2 numbering; at site 8). The structure-activity study shows that three residues (Lys-12, Arg-14, and Tyr-16) located in loop 2 and spatially close to residue 24 were also important for functional activity. We propose that the interaction of µO§-GVIIJ with the channel depends on not only disulfide tethering via Cys-24 to a free cysteine at site 8 on the channel but also the participation of key residues of µO§-GVIIJ on a distinct surface of the peptide.


Asunto(s)
Conotoxinas/química , Disulfuros/química , Proteínas Musculares/química , Canal de Sodio Activado por Voltaje NAV1.2/química , Bloqueadores de los Canales de Sodio/química , Canales de Sodio/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Conotoxinas/síntesis química , Cristalografía por Rayos X , Expresión Génica , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Canal de Sodio Activado por Voltaje NAV1.2/genética , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Caracoles/química , Bloqueadores de los Canales de Sodio/síntesis química , Canales de Sodio/genética , Canales de Sodio/metabolismo , Técnicas de Síntesis en Fase Sólida , Relación Estructura-Actividad
7.
Cent Nerv Syst Agents Med Chem ; 13(2): 148-58, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24156314

RESUMEN

Semicarbazones are synthesized by the condensation of semicarbazide and aldehyde/ketone. The literature survey revealed that semicarbazones had been emerged as compounds with diverse biological activities including anticonvulsant, antitubercular, anticancer, and antimicrobial activities. The anticonvulsant activity of semicarbazones is mainly attributed due to the presence of an aryl binding site with aryl/alkyl hydrophobic group, a hydrogen bonding domain and an electron donor group and they are suggested to act by inhibiting sodium ion (Na(+)) channel. Dimmock et al., reported an extensive series of semicarbazones and reported 4-(4-fluorophenoxy) benzaldehyde semicarbazone (C0102862, V102862) as lead molecule. In MES (oral) screening C0102862 showed protective index (PI > 315) more than carbamazepine (PI 101), phenytoin (PI > 21.6) and valproate (PI > 2.17). This review briefly describes the information available about semicarbazone analogs and their anticonvulsant activity.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia Parcial Compleja/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Semicarbazonas/uso terapéutico , Animales , Anticonvulsivantes/síntesis química , Anticonvulsivantes/química , Anticonvulsivantes/toxicidad , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Electrochoque , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/toxicidad , Pentilenotetrazol/toxicidad , Ratas , Prueba de Desempeño de Rotación con Aceleración Constante , Semicarbazonas/síntesis química , Semicarbazonas/química , Semicarbazonas/toxicidad , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/uso terapéutico , Bloqueadores de los Canales de Sodio/toxicidad
8.
Toxicon ; 71: 57-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23726857

RESUMEN

Huwentoxin-IV (HWTX-IV, also named Mu-theraphotoxin-Hh2a) is a typical inhibitor cystine knot peptide isolated from the venom of Chinese tarantula Ornithoctonus huwena and is found to inhibit tetrodotoxin-sensitive (TTX-S) sodium channels from mammalian sensory neurons. This peptide binds to neurotoxin receptor site 4 located at the extracellular S3-S4 linker of domain II in neuronal sodium channels. However, the molecular surface of HWTX-IV interaction with sodium channels remains unknown. In this study, we synthesized HWTX-IV and three mutants (T28D, R29A and Q34D) and characterized their functions on TTX-S sodium channels from adult rat dorsal root ganglion (DRG) neurons. Analysis of liquid chromatography, mass spectrometry and circular dichroism spectrum indicated that all four synthetic peptides are properly folded. Synthetic HWTX-IV exhibited the same activity as native HWTX-IV, while three mutations reduced toxin binding affinities by 10-200 fold, indicating that the basic or vicinal polar residues Thr²8, Arg²9, and Gln³4 in C-terminus might play critical roles in the interaction of HWTX-IV with TTX-S sodium channels.


Asunto(s)
Neurotoxinas/química , Bloqueadores de los Canales de Sodio/farmacología , Venenos de Araña/síntesis química , Tetrodotoxina/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Femenino , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/síntesis química , Canales de Sodio/metabolismo , Venenos de Araña/farmacología , Arañas/química , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 23(14): 4230-4, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23743284

RESUMEN

A novel series of pyrrolidine derivatives as Na(+) channel blockers was synthesized and evaluated for their inhibitory effects on neuronal Na(+) channels. Structure-activity relationship (SAR) studies of a pyrrolidine analogue 2 led to the discovery of 5e as a potent Na(+) channel blocker with a low inhibitory action against human ether-a-go-go-related gene (hERG) channels. Compound 5e showed remarkably neuroprotective activity in a rat transient middle cerebral artery occlusion (MCAO) model, suggesting that 5e would act as a neuroprotectant for ischemic stroke.


Asunto(s)
Pirrolidinas/química , Bloqueadores de los Canales de Sodio/síntesis química , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , Ratas , Bloqueadores de los Canales de Sodio/farmacología , Bloqueadores de los Canales de Sodio/uso terapéutico , Relación Estructura-Actividad
10.
J Pept Sci ; 18(7): 442-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22605564

RESUMEN

Protoxin II is biologically active peptide containing the inhibitory cystine knot motif. A synthetic version of the toxin was generated with standard Fmoc solid phase peptide synthesis. If N-methylmorpholine was used as a base during synthesis of the linear protoxin II, it was found that a significant amount of racemization (approximately 50%) was observed during the process of cysteine residue coupling. This racemization could be suppressed by substituting N-methylmorpholine with 2,4,6-collidine. The crude linear toxin was then air oxidized and purified. Electrophysiological assessment of the synthesized protoxin II confirmed its previously described interactions with voltage-gated sodium channels. Eight other naturally occurring inhibitory knot peptides were also synthesized using this same methodology. The inhibitory potencies of these synthesized toxins on Nav1.7 and Nav1.2 channels are summarized.


Asunto(s)
Cisteína/química , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Péptidos/síntesis química , Péptidos/metabolismo , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/metabolismo , Técnicas de Síntesis en Fase Sólida , Venenos de Araña/síntesis química , Venenos de Araña/metabolismo , Línea Celular , Humanos , Morfolinas/química , Oxidación-Reducción , Péptidos/química , Bloqueadores de los Canales de Sodio/química , Venenos de Araña/química , Estereoisomerismo , Especificidad por Sustrato
11.
Chem Pharm Bull (Tokyo) ; 60(4): 488-98, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22466732

RESUMEN

In investigating potent sodium (Na(+)) channel blockers for the treatment of ischemic stroke, we synthesized a novel series of 3-amino-1-(5-indanyloxy)-2-propanol derivatives and evaluated their inhibitory effects on neuronal Na(+) channels. The 3-amino-1-(5-indanyloxy)-2-propanol derivatives exhibited potent blocking activity for Na(+) channels and a significantly low affinity for dopamine D(2) receptors, which demonstrates a minimal clinical risk for extrapyramidal side effects. In particular, compound 4b, a 3-amino-1-(5-indanyloxy)-2-propanol derivative bearing a benzimidazole moiety, showed desirable neuroprotective activity in a rat transient middle cerebral artery occlusion model. Furthermore, compound 4b displayed a high binding affinity for neurotoxin receptor site 2 of the Na(+) channels, which suggests that 4b would act as a use-dependent Na(+) channel blocker in sustained depolarization during ischemic stroke.


Asunto(s)
2-Propanol/química , Microsomas Hepáticos/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , 2-Propanol/farmacocinética , 2-Propanol/farmacología , 2-Propanol/uso terapéutico , Animales , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Masculino , Microsomas Hepáticos/metabolismo , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Wistar , Bloqueadores de los Canales de Sodio/farmacocinética , Bloqueadores de los Canales de Sodio/uso terapéutico
12.
Bioorg Med Chem ; 20(6): 2180-8, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22364743

RESUMEN

Voltage-gated sodium channels are known to be expressed in neurons and other excitable cells. Recently, voltage-gated sodium channels have been found to be expressed in human prostate cancer cells. α-Hydroxy-α-phenylamides are a new class of small molecules that have demonstrated potent inhibition of voltage-gated sodium channels. The hydroxyamide motif, an isostere of a hydantoin ring, provides an active scaffold from which several potent racemic sodium channel blockers have been derived. With little known about chiral preferences, the development of chiral syntheses to obtain each pure enantiomer for evaluation as sodium channel blockers is important. Using Seebach and Frater's chiral template, cyclocondensation of (R)-3-chloromandelic acid with pivaldehyde furnished both the cis- and trans-2,5-disubsituted dioxolanones. Using this chiral template, we synthesized both enantiomers of 2-(3-chlorophenyl)-2-hydroxynonanamide, and evaluated their ability to functionally inhibit hNa(v) isoforms, human prostate cancer cells and xenograft. Enantiomers of lead demonstrated significant ability to reduce prostate cancer in vivo.


Asunto(s)
Amidas/química , Amidas/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/uso terapéutico , Amidas/síntesis química , Animales , Antineoplásicos/síntesis química , Línea Celular , Línea Celular Tumoral , Técnicas de Química Sintética/métodos , Humanos , Activación del Canal Iónico/efectos de los fármacos , Isomerismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Moleculares , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Bloqueadores de los Canales de Sodio/síntesis química , Canales de Sodio/química , Canales de Sodio/metabolismo
13.
Bioorg Med Chem Lett ; 22(2): 929-32, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22197144

RESUMEN

We report the identification of a novel series of human epithelial sodium channel (ENaC) blockers that are structurally distinct from the pyrazinoyl guanidine chemotype found in prototypical ENaC blockers such as amiloride. Following a rational design hypothesis a series of quaternary amines were prepared and evaluated for their ability to block ion transport via ENaC in human bronchial epithelial cells (HBECs). Compound 11 has an IC(50) of 200nM and is efficacious in the Guinea-pig tracheal potential difference (TPD) model of ENaC blockade with an ED(50) of 44µgkg(-1) at 1h. As such, pyrazinoyl quaternary amines represent the first examples of a promising new class of human ENaC blockers.


Asunto(s)
Aminas/química , Diseño de Fármacos , Células Epiteliales/efectos de los fármacos , Bloqueadores del Canal de Sodio Epitelial , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/farmacología , Aminas/farmacología , Bronquios/citología , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Humanos , Bloqueadores de los Canales de Sodio/química , Relación Estructura-Actividad
14.
J Med Chem ; 54(13): 4427-45, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21634377

RESUMEN

Clinical human genetic studies have recently identified the tetrodotoxin (TTX) sensitive neuronal voltage gated sodium channel Nav1.7 (SCN9A) as a critical mediator of pain sensitization. Herein, we report structure-activity relationships for a novel series of 2,4-diaminotriazines that inhibit hNav1.7. Optimization efforts culminated in compound 52, which demonstrated pharmacokinetic properties appropriate for in vivo testing in rats. The binding site of compound 52 on Nav1.7 was determined to be distinct from that of local anesthetics. Compound 52 inhibited tetrodotoxin-sensitive sodium channels recorded from rat sensory neurons and exhibited modest selectivity against the hERG potassium channel and against cloned and native tetrodotoxin-resistant sodium channels. Upon oral administration to rats, compound 52 produced dose- and exposure-dependent efficacy in the formalin model of pain.


Asunto(s)
Acetamidas/síntesis química , Analgésicos/síntesis química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/síntesis química , Triazinas/síntesis química , Acetamidas/farmacocinética , Acetamidas/farmacología , Administración Oral , Analgésicos/farmacocinética , Analgésicos/farmacología , Animales , Sitios de Unión , Línea Celular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Formaldehído , Ganglios Espinales/citología , Humanos , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1 , Neuronas/efectos de los fármacos , Neuronas/fisiología , Dimensión del Dolor , Técnicas de Placa-Clamp , Ratas , Bloqueadores de los Canales de Sodio/farmacocinética , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio , Solubilidad , Relación Estructura-Actividad , Tetrodotoxina/farmacología , Triazinas/farmacocinética , Triazinas/farmacología
15.
Bioorg Med Chem ; 16(12): 6379-86, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18501613

RESUMEN

The synthesis and pharmacological characterization of a novel furan-based class of voltage-gated sodium channel blockers is reported. Compounds were evaluated for their ability to block the tetrodotoxin-resistant sodium channel Na(v)1.8 (PN3) as well as the Na(v)1.2 and Na(v)1.5 subtypes. Benchmark compounds from this series possessed enhanced potency, oral bioavailability, and robust efficacy in a rodent model of neuropathic pain, together with improved CNS and cardiovascular safety profiles compared to the clinically used sodium channel blockers mexiletine and lamotrigine.


Asunto(s)
Analgésicos no Narcóticos/química , Analgésicos no Narcóticos/farmacología , Furanos/química , Furanos/farmacología , Neuralgia/tratamiento farmacológico , Piperazinas/química , Piperazinas/farmacología , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Analgésicos no Narcóticos/síntesis química , Animales , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Furanos/síntesis química , Humanos , Masculino , Ratones , Piperazinas/síntesis química , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/síntesis química , Relación Estructura-Actividad
16.
Bioorg Med Chem Lett ; 18(6): 1963-6, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18289851
18.
Chem Biol ; 14(4): 399-407, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17462575

RESUMEN

Disulfide-rich neurotoxins from venomous animals continue to provide compounds with therapeutic potential. Minimizing neurotoxins often results in removal of disulfide bridges or critical amino acids. To address this drug-design challenge, we explored the concept of disulfide-rich scaffolds consisting of isostere polymers and peptidic pharmacophores. Flexible spacers, such as amino-3-oxapentanoic or 6-aminohexanoic acids, were used to replace conformationally constrained parts of a three-disulfide-bridged conotoxin, SIIIA. The peptide-polymer hybrids, polytides, were designed based on cladistic identification of nonconserved loci in related peptides. After oxidative folding, the polytides appeared to be better inhibitors of sodium currents in dorsal root ganglia and sciatic nerves in mice. Moreover, the polytides appeared to be significantly more potent and longer-lasting analgesics in the inflammatory pain model in mice, when compared to SIIIA. The resulting polytides provide a promising strategy for transforming disulfide-rich peptides into therapeutics.


Asunto(s)
Analgésicos no Narcóticos/síntesis química , Conotoxinas/síntesis química , Diseño de Fármacos , Péptidos/síntesis química , Secuencia de Aminoácidos , Analgésicos no Narcóticos/química , Analgésicos no Narcóticos/farmacología , Animales , Conotoxinas/química , Conotoxinas/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/tratamiento farmacológico , Dimensión del Dolor , Péptidos/química , Péptidos/farmacología , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología
19.
J Med Chem ; 49(14): 4098-115, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16821771

RESUMEN

Amiloride (1), the prototypical epithelial sodium channel (ENaC) blocker, has been administered with limited success as aerosol therapy for improving pulmonary function in patients with the genetic disorder cystic fibrosis. This study was conducted to synthesize and identify more potent, less reversible ENaC blockers, targeted for aerosol therapy and possessing minimal systemic renal activity. A series of novel 2-substituted acylguanidine analogues of amiloride were synthesized and evaluated for potency and reversibility on bronchial ENaC. All compounds tested were more potent and less reversible at blocking sodium-dependent short-circuit current than amiloride. Compounds 30-34 showed the greatest potency on ENaC with IC(50) values below 10 nM. A regioselective difference in potency was found (compounds 30, 39, and 40), whereas no stereospecific (compounds 33, 34) difference in potency on ENaC was displayed. Lead compound 32 was 102-fold more potent and 5-fold less reversible than amiloride and displayed the lowest IC(50) value ever reported for an ENaC blocker.


Asunto(s)
Bronquitis Crónica/tratamiento farmacológico , Fibrosis Quística/tratamiento farmacológico , Guanidinas/síntesis química , Pirazinas/síntesis química , Bloqueadores de los Canales de Sodio/síntesis química , Canales de Sodio/efectos de los fármacos , Animales , Bronquios/efectos de los fármacos , Bronquios/fisiología , Técnicas Químicas Combinatorias , Perros , Canales Epiteliales de Sodio , Guanidinas/química , Guanidinas/farmacología , Modelos Moleculares , Pirazinas/química , Pirazinas/farmacología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/fisiología , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/fisiología , Estereoisomerismo , Relación Estructura-Actividad , Técnicas de Cultivo de Tejidos
20.
Bioorg Med Chem Lett ; 16(5): 1358-61, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16337121

RESUMEN

Novel cyclopentane-based 3-phenyl-1-hydroxypropyl compounds were evaluated for inhibitory activity against the peripheral nerve sodium channel Na(V)1.7 and off-target activity against the cardiac potassium channel hERG. The stereochemistry of the hydroxyl group and substitution on the phenyl rings with either fluorinated O-alkyl or alkyl groups were found to be critical for conferring potency against Na(V)1.7. A benchmark compound from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Asunto(s)
Ciclopentanos/química , Ciclopentanos/farmacología , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Animales , Ciclopentanos/síntesis química , Ciclopentanos/farmacocinética , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Ratas , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacocinética , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA