Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(16): 7926-7931, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30926668

RESUMEN

Dysregulation of the alternative complement pathway (AP) predisposes individuals to a number of diseases including paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, and C3 glomerulopathy. Moreover, glomerular Ig deposits can lead to complement-driven nephropathies. Here we describe the discovery of a highly potent, reversible, and selective small-molecule inhibitor of factor B, a serine protease that drives the central amplification loop of the AP. Oral administration of the inhibitor prevents KRN-induced arthritis in mice and is effective upon prophylactic and therapeutic dosing in an experimental model of membranous nephropathy in rats. In addition, inhibition of factor B prevents complement activation in sera from C3 glomerulopathy patients and the hemolysis of human PNH erythrocytes. These data demonstrate the potential therapeutic value of using a factor B inhibitor for systemic treatment of complement-mediated diseases and provide a basis for its clinical development.


Asunto(s)
Factor B del Complemento/antagonistas & inhibidores , Vía Alternativa del Complemento/efectos de los fármacos , Descubrimiento de Drogas/métodos , Factores Inmunológicos/farmacología , Animales , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas Sprague-Dawley
2.
Chimia (Aarau) ; 73(7): 581-590, 2019 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-31431218

RESUMEN

The macrolide rapamycin ( 1 ) was first described as an antifungal agent in 1975. Even though its biological target and the molecular details were yet to be discovered, rapamycin attracted our interest in the early 90s based on its reported immunosuppressive activity in transplantation models and based on findings that its mechanism of action was different from those of the known immunosuppressive agents ciclosporin and FK506. In this review we describe our efforts to chemically modify this complex and chemically very sensitive natural product. Despite the limitations regarding the reaction conditions compatible with rapamycin we discovered ways of selectively modifying specific functional groups. This allowed us, among others, to improve the stability of the parent molecule towards ring-opening. Our efforts culminated in the discovery and development of the 40-O-alkylated derivative everolimus 2 which became a useful drug in solid organ transplantation, in various cancer indications and as the active principle of the market leading drug-eluting stent.


Asunto(s)
Sirolimus/química , Productos Biológicos , Stents Liberadores de Fármacos , Everolimus , Inmunosupresores
3.
Bioorg Med Chem Lett ; 24(3): 731-6, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24439847

RESUMEN

The successful launches of dipeptidyl peptidase IV (DPP IV) inhibitors as oral anti-diabetics warrant and spur the further quest for additional chemical entities in this promising class of therapeutics. Numerous pharmaceutical companies have pursued their proprietary candidates towards the clinic, resulting in a large body of published chemical structures associated with DPP IV. Herein, we report the discovery of a novel chemotype for DPP IV inhibition based on the C-(1-aryl-cyclohexyl)-methylamine scaffold and its optimization to compounds which selectively inhibit DPP IV at low-nM potency and exhibit an excellent oral pharmacokinetic profile in the rat.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/síntesis química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Descubrimiento de Drogas , Metilaminas/síntesis química , Metilaminas/farmacocinética , Adamantano/análogos & derivados , Adamantano/química , Adamantano/farmacología , Administración Oral , Animales , Células CACO-2 , Cristalografía por Rayos X , Ciclización , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Metilaminas/química , Metilaminas/farmacología , Estructura Molecular , Nitrilos/química , Nitrilos/farmacología , Pirazinas/química , Pirazinas/farmacología , Pirrolidinas/química , Pirrolidinas/farmacología , Ratas , Fosfato de Sitagliptina , Triazoles/química , Triazoles/farmacología , Vildagliptina
4.
Bioorg Med Chem Lett ; 22(3): 1464-8, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22177783

RESUMEN

Novel deazaxanthine-based DPP-4 inhibitors have been identified that are potent (IC(50) <10nM) and highly selective versus other dipeptidyl peptidases. Their synthesis and SAR are reported, along with initial efforts to improve the PK profile through decoration of the deazaxanthine core. Optimisation of compound 3a resulted in the identification of compound (S)-4i, which displayed an improved in vitro and ADME profile. Further enhancements to the PK profile were possible by changing from the deazahypoxanthine to the deazaxanthine template, culminating in compound 12g, which displayed good ex vivo DPP-4 inhibition and a superior PK profile in rat, suggestive of once daily dosing in man.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Animales , Células CACO-2 , Cristalografía por Rayos X , Inhibidores de la Dipeptidil-Peptidasa IV/síntesis química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Activación Enzimática/efectos de los fármacos , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacología , Compuestos Heterocíclicos/uso terapéutico , Humanos , Concentración 50 Inhibidora , Masculino , Modelos Moleculares , Estructura Molecular , Ratas , Relación Estructura-Actividad
5.
Cancer Discov ; 12(6): 1500-1517, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35404998

RESUMEN

Covalent inhibitors of KRASG12C have shown antitumor activity against advanced/metastatic KRASG12C-mutated cancers, though resistance emerges and additional strategies are needed to improve outcomes. JDQ443 is a structurally unique covalent inhibitor of GDP-bound KRASG12C that forms novel interactions with the switch II pocket. JDQ443 potently inhibits KRASG12C-driven cellular signaling and demonstrates selective antiproliferative activity in KRASG12C-mutated cell lines, including those with G12C/H95 double mutations. In vivo, JDQ443 induces AUC exposure-driven antitumor efficacy in KRASG12C-mutated cell-derived (CDX) and patient-derived (PDX) tumor xenografts. In PDX models, single-agent JDQ443 activity is enhanced by combination with inhibitors of SHP2, MEK, or CDK4/6. Notably, the benefit of JDQ443 plus the SHP2 inhibitor TNO155 is maintained at reduced doses of either agent in CDX models, consistent with mechanistic synergy. JDQ443 is in clinical development as monotherapy and in combination with TNO155, with both strategies showing antitumor activity in patients with KRASG12C-mutated tumors. SIGNIFICANCE: JDQ443 is a structurally novel covalent KRASG12C inhibitor with a unique binding mode that demonstrates potent and selective antitumor activity in cell lines and in vivo models. In preclinical models and patients with KRASG12C-mutated malignancies, JDQ443 shows potent antitumor activity as monotherapy and in combination with the SHP2 inhibitor TNO155. This article is highlighted in the In This Issue feature, p. 1397.


Asunto(s)
Inhibidores Enzimáticos , Indazoles , Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Indazoles/química , Indazoles/farmacología , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
6.
J Med Chem ; 65(24): 16173-16203, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36399068

RESUMEN

Rapid emergence of tumor resistance via RAS pathway reactivation has been reported from clinical studies of covalent KRASG12C inhibitors. Thus, inhibitors with broad potential for combination treatment and distinct binding modes to overcome resistance mutations may prove beneficial. JDQ443 is an investigational covalent KRASG12C inhibitor derived from structure-based drug design followed by extensive optimization of two dissimilar prototypes. JDQ443 is a stable atropisomer containing a unique 5-methylpyrazole core and a spiro-azetidine linker designed to position the electrophilic acrylamide for optimal engagement with KRASG12C C12. A substituted indazole at pyrazole position 3 results in novel interactions with the binding pocket that do not involve residue H95. JDQ443 showed PK/PD activity in vivo and dose-dependent antitumor activity in mouse xenograft models. JDQ443 is now in clinical development, with encouraging early phase data reported from an ongoing Phase Ib/II clinical trial (NCT04699188).


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Diseño de Fármacos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Pirazoles/farmacología , Pirazoles/uso terapéutico
7.
J Med Chem ; 63(11): 5697-5722, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32073845

RESUMEN

The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.


Asunto(s)
Ácido Benzoico/química , Factor B del Complemento/antagonistas & inhibidores , Indoles/química , Síndrome Hemolítico Urémico Atípico/metabolismo , Síndrome Hemolítico Urémico Atípico/patología , Ácido Benzoico/metabolismo , Ácido Benzoico/farmacocinética , Sitios de Unión , Dominio Catalítico , Factor B del Complemento/metabolismo , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Indoles/metabolismo , Indoles/farmacocinética , Concentración 50 Inhibidora , Degeneración Macular/metabolismo , Degeneración Macular/patología , Simulación de Dinámica Molecular , Relación Estructura-Actividad
8.
Drug Metab Dispos ; 36(8): 1457-60, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18448571

RESUMEN

The immunosuppressant macrolide everolimus was found to be metabolized in animals and humans to a phosphocholine ester (ATG181), a hitherto unknown type of conjugate in xenobiotic metabolism. The structure of ATG181 was elucidated by mass spectrometry and confirmed by synthesis. ATG181 was among the most prominent metabolites of everolimus in rat, monkey, and human blood and was found also in various tissues of the rat, whereas no ATG181 was identified in the urine and feces of the species investigated. The metabolite showed binding to FK506 binding protein with a 2- to 3-fold higher affinity than everolimus. However, ATG181 exhibited only marginal in vitro immunosuppressive activity and is therefore very unlikely to contribute in a relevant manner to the immunosuppressive effect of everolimus.


Asunto(s)
Inmunosupresores/farmacología , Fosforilcolina/metabolismo , Sirolimus/análogos & derivados , Animales , Cromatografía Liquida , Ésteres , Everolimus , Humanos , Inmunosupresores/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Masculino , Espectrometría de Masas , Ratas , Ratas Wistar , Sirolimus/metabolismo , Sirolimus/farmacología
9.
Nat Commun ; 7: 13166, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27774986

RESUMEN

The COP9 signalosome (CSN) is a central component of the activation and remodelling cycle of cullin-RING E3 ubiquitin ligases (CRLs), the largest enzyme family of the ubiquitin-proteasome system in humans. CRLs are implicated in the regulation of numerous cellular processes, including cell cycle progression and apoptosis, and aberrant CRL activity is frequently associated with cancer. Remodelling of CRLs is initiated by CSN-catalysed cleavage of the ubiquitin-like activator NEDD8 from CRLs. Here we describe CSN5i-3, a potent, selective and orally available inhibitor of CSN5, the proteolytic subunit of CSN. The compound traps CRLs in the neddylated state, which leads to inactivation of a subset of CRLs by inducing degradation of their substrate recognition module. CSN5i-3 differentially affects the viability of tumour cell lines and suppresses growth of a human xenograft in mice. Our results provide insights into how CSN regulates CRLs and suggest that CSN5 inhibition has potential for anti-tumour therapy.


Asunto(s)
Antineoplásicos/farmacología , Azepinas/farmacología , Complejo del Señalosoma COP9/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Pirazoles/farmacología , Ubiquitina-Proteína Ligasas/genética , Animales , Antineoplásicos/síntesis química , Azepinas/síntesis química , Complejo del Señalosoma COP9/genética , Complejo del Señalosoma COP9/metabolismo , Femenino , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/metabolismo , Linfoma Anaplásico de Células Grandes/patología , Ratones , Ratones SCID , Terapia Molecular Dirigida , Proteína NEDD8/genética , Proteína NEDD8/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis/efectos de los fármacos , Pirazoles/síntesis química , Células THP-1 , Carga Tumoral/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Nat Rev Drug Discov ; 13(8): 577-87, 2014 08.
Artículo en Inglés | MEDLINE | ID: mdl-25033734

RESUMEN

Analysis of the origins of new drugs approved by the US Food and Drug Administration (FDA) from 1999 to 2008 suggested that phenotypic screening strategies had been more productive than target-based approaches in the discovery of first-in-class small-molecule drugs. However, given the relatively recent introduction of target-based approaches in the context of the long time frames of drug development, their full impact might not yet have become apparent. Here, we present an analysis of the origins of all 113 first-in-class drugs approved by the FDA from 1999 to 2013, which shows that the majority (78) were discovered through target-based approaches (45 small-molecule drugs and 33 biologics). In addition, of 33 drugs identified in the absence of a target hypothesis, 25 were found through a chemocentric approach in which compounds with known pharmacology served as the starting point, with only eight coming from what we define here as phenotypic screening: testing a large number of compounds in a target-agnostic assay that monitors phenotypic changes. We also discuss the implications for drug discovery strategies, including viewing phenotypic screening as a novel discipline rather than as a neoclassical approach.


Asunto(s)
Descubrimiento de Drogas/métodos , Aprobación de Drogas/historia , Descubrimiento de Drogas/historia , Evaluación Preclínica de Medicamentos/historia , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/métodos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Biología de Sistemas/métodos
11.
J Med Chem ; 56(6): 2196-206, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23360239

RESUMEN

A small library of fragments comprising putative recognition motifs for the catalytic dyad of aspartic proteases was generated by in silico similarity searches within the corporate compound deck based on rh-renin active site docking and scoring filters. Subsequent screening by NMR identified the low-affinity hits 3 and 4 as competitive active site binders, which could be shown by X-ray crystallography to bind to the hydrophobic S3-S1 pocket of rh-renin. As part of a parallel multiple hit-finding approach, the 3,5-disubstituted piperidine (rac)-5 was discovered by HTS using a enzymatic assay. X-ray crystallography demonstrated the eutomer (3S,5R)-5 to be a peptidomimetic inhibitor binding to a nonsubstrate topography of the rh-renin prime site. The design of the potent and selective (3S,5R)-12 bearing a P3(sp)-tethered tricyclic P3-P1 pharmacophore derived from 3 is described. (3S,5R)-12 showed oral bioavailability in rats and demonstrated blood pressure lowering activity in the double-transgenic rat model.


Asunto(s)
Diseño de Fármacos , Piperidinas/química , Piperidinas/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Renina/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Concentración 50 Inhibidora , Modelos Moleculares , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacocinética , Conformación Proteica , Ratas , Renina/química
12.
J Med Chem ; 54(17): 6028-39, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21797275

RESUMEN

Protein kinase C (PKC) isotypes have emerged as key targets for the blockade of early T-cell activation. Herein, we report on the structure-activity relationship and the detailed physicochemical and in vivo pharmacokinetic properties of sotrastaurin (AEB071, 1), a novel maleimide-based PKC inhibitor currently in phase II clinical trials. Most notably, the preferred uptake of sotrastaurin into lymphoid tissues is an important feature, which is likely to contribute to its in vivo efficacy.


Asunto(s)
Rechazo de Injerto/prevención & control , Activación de Linfocitos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Psoriasis/tratamiento farmacológico , Pirroles/uso terapéutico , Quinazolinas/uso terapéutico , Animales , Células Cultivadas , Humanos , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirroles/química , Pirroles/farmacocinética , Quinazolinas/química , Quinazolinas/farmacocinética , Ratas , Relación Estructura-Actividad , Distribución Tisular
13.
J Med Chem ; 52(20): 6193-6, 2009 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-19827831

RESUMEN

A series of novel maleimide-based inhibitors of protein kinase C (PKC) were designed, synthesized, and evaluated. AEB071 (1) was found to be a potent, selective inhibitor of classical and novel PKC isotypes. 1 is a highly efficient immunomodulator, acting via inhibition of early T cell activation. The binding mode of maleimides to PKCs, proposed by molecular modeling, was confirmed by X-ray analysis of 1 bound in the active site of PKCalpha.


Asunto(s)
Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Quinazolinas/farmacología , Animales , Ensayos Clínicos como Asunto , Descubrimiento de Drogas , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Maleimidas/química , Maleimidas/metabolismo , Ratones , Modelos Moleculares , Conformación Molecular , Peso Molecular , Proteína Quinasa C/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirroles/química , Pirroles/farmacocinética , Quinazolinas/química , Quinazolinas/farmacocinética , Ratas , Especificidad por Sustrato , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Tolerancia al Trasplante
15.
J Biol Chem ; 280(23): 21965-71, 2005 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-15772070

RESUMEN

Sanglifehrin A (SFA) is a novel immunosuppressant isolated from Streptomyces sp. that binds strongly to the human immunophilin cyclophilin A (CypA). SFA exerts its immunosuppressive activity through a mode of action different from that of all other known immunophilin-binding substances, namely cyclosporine A (CsA), FK506, and rapamycin. We have determined the crystal structure of human CypA in complex with SFA at 1.6 A resolution. The high resolution of the structure revealed the absolute configuration at all 17 chiral centers of SFA as well as the details of the CypA/SFA interactions. In particular, it was shown that the 22-membered macrocycle of SFA is deeply embedded in the same binding site as CsA and forms six direct hydrogen bonds with CypA. The effector domain of SFA, on the other hand, has a chemical and three-dimensional structure very different from CsA, already strongly suggesting different immunosuppressive mechanisms. Furthermore, two CypA.SFA complexes form a dimer in the crystal as well as in solution as shown by light scattering and size exclusion chromatography experiments. This observation raises the possibility that the dimer of CypA.SFA complexes is the molecular species mediating the immunosuppressive effect.


Asunto(s)
Ciclofilina A/química , Inmunosupresores/farmacología , Lactonas/química , Compuestos de Espiro/química , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Humanos , Ligandos , Luz , Modelos Químicos , Modelos Moleculares , Péptidos/química , Unión Proteica , Conformación Proteica , Dispersión de Radiación , Estereoisomerismo
16.
J Am Chem Soc ; 125(13): 3849-59, 2003 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-12656618

RESUMEN

Sanglifehrin A (SFA) is a novel immunosuppressive natural product isolated from Streptomyces sp. A92-308110. SFA has a very strong affinity for cyclophilin A (IC(50) = 6.9 +/- 0.9 nM) but is structurally different from cyclosporin A (CsA) and exerts its immunosuppressive activity via a novel mechanism. SFA has a complex molecular structure consisting of a 22-membered macrocycle, bearing in position 23 a nine-carbon tether terminated by a highly substituted spirobicyclic moiety. Selective oxidative cleavage of the C(26)=C(27) exocyclic double bond affords the spirolactam containing fragment 1 and macrolide 2. The affinity of 2 for cyclophilin (IC(50) = 29 +/- 2.1 nM) is essentially identical to SFA, which indicates that the interaction between SFA and cyclophilin A is mediated exclusively by the macrocyclic portion of the molecule. This observation was confirmed by the X-ray crystal structure resolved at 2.1 A of cyclophilin A complexed to macrolide 16, a close analogue of 2. The X-ray crystal structure showed that macrolide 16 binds to the same deep hydrophobic pocket of cyclophilin A as CsA. Additional valuable details of the structure-activity relationship were obtained by two different chemical approaches: (1) degradation work on macrolide 2 or (2) synthesis of a library of macrolide analogues using the ring-closing metathesis reaction as the key step. Altogether, it appears that the complex macrocyclic fragment of SFA is a highly optimized combination of multiple functionalities including an (E,E)-diene, a short polypropionate fragment, and an unusual tripeptide unit, which together provide an extremely strong affinity for cyclophilin A.


Asunto(s)
Ciclofilina A/química , Inmunosupresores/química , Lactonas/química , Compuestos de Espiro/química , Unión Competitiva , Cristalografía por Rayos X , Ciclofilina A/metabolismo , Inmunosupresores/síntesis química , Inmunosupresores/metabolismo , Inmunosupresores/farmacología , Cinética , Lactonas/síntesis química , Lactonas/metabolismo , Lactonas/farmacología , Modelos Moleculares , Estructura Molecular , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/síntesis química , Compuestos de Espiro/metabolismo , Compuestos de Espiro/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA