Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Res ; 10(10): 1175-1189, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-35981087

RESUMEN

Novel therapeutic approaches combining immune-checkpoint inhibitors are needed to improve clinical outcomes for patients with cancer. Lymphocyte-activation gene 3 (LAG-3) is an immune-checkpoint molecule that inhibits T-cell activity and antitumor immune responses, acting through an independent mechanism from that of programmed death-1 (PD-1) and cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4). Here, we describe the development and preclinical characterization of relatlimab, a human antibody that binds to human LAG-3 with high affinity and specificity to block the interaction of LAG-3 with the ligands MHC II and fibrinogen-like protein-1, and to reverse LAG-3-mediated inhibition of T-cell function in vitro. Consistent with previous reports, in mouse models, the combined blockade of LAG-3 and PD-1 with surrogate antibodies resulted in enhanced antitumor activity greater than the individual blockade of either receptor. In toxicity studies in cynomolgus monkeys, relatlimab was generally well tolerated when combined with nivolumab. These results are consistent with findings from the RELATIVITY-047 phase II/III trial showing that relatlimab combined with nivolumab is a well-tolerated regimen that demonstrates superior progression-free survival compared with nivolumab monotherapy in patients with unresectable or metastatic melanoma.


Asunto(s)
Melanoma , Nivolumab , Animales , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4 , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Fibrinógeno/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico , Macaca fascicularis , Melanoma/patología , Ratones , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1
2.
Sci Rep ; 12(1): 3530, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35241687

RESUMEN

T-cell engagers (TCEs) are a growing class of biotherapeutics being investigated in the clinic for treatment of a variety of hematological and solid tumor indications. However, preclinical evaluation of TCEs in vivo has been mostly limited to xenograft tumor models in human T-cell reconstituted immunodeficient mice, which have a number of limitations. To explore the efficacy of human TCEs in fully immunocompetent hosts, we developed a knock-in mouse model (hCD3E-epi) in which a 5-residue N-terminal fragment of murine CD3-epsilon was replaced with an 11-residue stretch from the human sequence that encodes for a common epitope recognized by anti-human CD3E antibodies in the clinic. T cells from hCD3E-epi mice underwent normal thymic development and could be efficiently activated upon crosslinking of the T-cell receptor with anti-human CD3E antibodies in vitro. Furthermore, a TCE targeting human CD3E and murine CD20 induced robust T-cell redirected killing of murine CD20-positive B cells in ex vivo hCD3E-epi splenocyte cultures, and also depleted nearly 100% of peripheral B cells for up to 7 days following in vivo administration. These results highlight the utility of this novel mouse model for exploring the efficacy of human TCEs in vivo, and suggest a useful tool for evaluating TCEs in combination with immuno-oncology/non-immuno-oncology agents against heme and solid tumor targets in hosts with a fully intact immune system.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Antígenos CD20 , Complejo CD3 , Epítopos , Humanos , Ratones , Linfocitos T
3.
MAbs ; 12(1): 1818436, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32936727

RESUMEN

Treatment of ocular disease is hindered by the presence of the blood-retinal barrier, which restricts access of systemic drugs to the eye. Intravitreal injections bypass this barrier, delivering high concentrations of drug to the targeted tissue. However, the recommended dosing interval for approved biologics is typically 6-12 weeks, and frequent travel to the physician's office poses a substantial burden for elderly patients with poor vision. Real-world data suggest that many patients are under-treated. Here, we investigate IgMs as a novel platform for treating ocular disease. We show that IgMs are well-suited to ocular administration due to moderate viscosity, long ocular exposure, and rapid systemic clearance. The complement-dependent cytotoxicity of IgMs can be readily removed with a P436G mutation, reducing safety liabilities. Furthermore, dodecavalent binding of IgM hexamers can potently activate pathways implicated in the treatment of progressive blindness, including the Tie2 receptor tyrosine kinase signaling pathway for the treatment of diabetic macular edema, or the death receptor 4 tumor necrosis family receptor pathway for the treatment of wet age-related macular degeneration. Collectively, these data demonstrate the promise of IgMs as therapeutic agonists for treating progressive blindness.


Asunto(s)
Sistemas de Liberación de Medicamentos , Inmunoglobulina M/farmacología , Degeneración Macular , Cuerpo Vítreo/metabolismo , Animales , Células CHO , Cricetulus , Humanos , Inyecciones Intravítreas , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Ratas
4.
MAbs ; 11(6): 1122-1138, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31122132

RESUMEN

IgA antibodies have broad potential as a novel therapeutic platform based on their superior receptor-mediated cytotoxic activity, potent neutralization of pathogens, and ability to transcytose across mucosal barriers via polymeric immunoglobulin receptor (pIgR)-mediated transport, compared to traditional IgG-based drugs. However, the transition of IgA into clinical development has been challenged by complex expression and characterization, as well as rapid serum clearance that is thought to be mediated by glycan receptor scavenging of recombinantly produced IgA monomer bearing incompletely sialylated N-linked glycans. Here, we present a comprehensive biochemical, biophysical, and structural characterization of recombinantly produced monomeric, dimeric and polymeric human IgA. We further explore two strategies to overcome the rapid serum clearance of polymeric IgA: removal of all N-linked glycosylation sites creating an aglycosylated polymeric IgA and engineering in FcRn binding with the generation of a polymeric IgG-IgA Fc fusion. While previous reports and the results presented in this study indicate that glycan-mediated clearance plays a major role for monomeric IgA, systemic clearance of polymeric IgA in mice is predominantly controlled by mechanisms other than glycan receptor clearance, such as pIgR-mediated transcytosis. The developed IgA platform now provides the potential to specifically target pIgR expressing tissues, while maintaining low systemic exposure.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/genética , Perros , Femenino , Glicosilación , Semivida , Humanos , Inmunoglobulina A/genética , Inmunoglobulina G/genética , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética
5.
PLoS One ; 10(4): e0121177, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25837374

RESUMEN

More than 30% of acute myeloid leukemia (AML) patients possess activating mutations in the receptor tyrosine kinase FMS-like tyrosine kinase 3 or FLT3. A small-molecule inhibitor of FLT3 (known as quizartinib or AC220) that is currently in clinical trials appears promising for the treatment of AML. Here, we report the co-crystal structure of the kinase domain of FLT3 in complex with quizartinib. FLT3 with quizartinib bound adopts an "Abl-like" inactive conformation with the activation loop stabilized in the "DFG-out" orientation and folded back onto the kinase domain. This conformation is similar to that observed for the uncomplexed intracellular domain of FLT3 as well as for related receptor tyrosine kinases, except for a localized induced fit in the activation loop. The co-crystal structure reveals the interactions between quizartinib and the active site of FLT3 that are key for achieving its high potency against both wild-type FLT3 as well as a FLT3 variant observed in many AML patients. This co-complex further provides a structural rationale for quizartinib-resistance mutations.


Asunto(s)
Antineoplásicos/química , Benzotiazoles/química , Proteínas de Neoplasias/química , Compuestos de Fenilurea/química , Inhibidores de Proteínas Quinasas/química , Proteínas Recombinantes de Fusión/química , Tirosina Quinasa 3 Similar a fms/química , Animales , Baculoviridae/genética , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Células Sf9 , Spodoptera/genética , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética
6.
Elife ; 42015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25699547

RESUMEN

Bruton's tyrosine kinase (Btk), a Tec-family tyrosine kinase, is essential for B-cell function. We present crystallographic and biochemical analyses of Btk, which together reveal molecular details of its autoinhibition and activation. Autoinhibited Btk adopts a compact conformation like that of inactive c-Src and c-Abl. A lipid-binding PH-TH module, unique to Tec kinases, acts in conjunction with the SH2 and SH3 domains to stabilize the inactive conformation. In addition to the expected activation of Btk by membranes containing phosphatidylinositol triphosphate (PIP3), we found that inositol hexakisphosphate (IP6), a soluble signaling molecule found in both animal and plant cells, also activates Btk. This activation is a consequence of a transient PH-TH dimerization induced by IP6, which promotes transphosphorylation of the kinase domains. Sequence comparisons with other Tec-family kinases suggest that activation by IP6 is unique to Btk.


Asunto(s)
Ácido Fítico/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Regulación Alostérica/efectos de los fármacos , Animales , Sitios de Unión , Biocatálisis/efectos de los fármacos , Bovinos , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Modelos Moleculares , Fosforilación/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Proteínas Tirosina Quinasas/química , Proteínas Proto-Oncogénicas c-abl/metabolismo , Solubilidad , Soluciones , Electricidad Estática , Termodinámica , Dominios Homologos src
7.
Methods Enzymol ; 548: 23-67, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25399641

RESUMEN

The aberrant activation of protein kinases is associated with many human diseases, most notably cancer. Due to this link between kinase deregulation and disease progression, kinases are one of the most targeted protein families for small-molecule inhibition. Within the last 15 years, the U.S. Food and Drug Administration has approved over 20 small-molecule inhibitors of protein kinases for use in the clinic. These inhibitors target the kinase active site and represent the successful hurdling by medicinal chemists of the formidable challenge posed by the high similarity among the active sites of the approximately 500 human kinases. We review the conserved structural features of kinases that are important for inhibitor binding as well as for catalysis. Many clinically approved drugs elicit selectivity by exploiting subtle variation within the kinase active site. We highlight some of the crystallographic studies on the kinase-inhibitor complexes that have provided valuable guidance for the development of these drugs as well as for future drug design efforts.


Asunto(s)
Modelos Moleculares , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Biocatálisis/efectos de los fármacos , Dominio Catalítico , Secuencia Conservada , Humanos , Mutación , Fosforilación/efectos de los fármacos , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos
8.
Nat Chem Biol ; 10(11): 969-76, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25262416

RESUMEN

We previously discovered a small-molecule inducer of cell death, named 1541, that noncovalently self-assembles into chemical fibrils ('chemi-fibrils') and activates procaspase-3 in vitro. We report here that 1541-induced cell death is caused by the fibrillar rather than the soluble form of the drug. A short hairpin RNA screen reveals that knockdown of genes involved in endocytosis, vesicle trafficking and lysosomal acidification causes partial 1541 resistance. We confirm the role of these pathways using pharmacological inhibitors. Microscopy shows that the fluorescent chemi-fibrils accumulate in punctae inside cells that partially colocalize with lysosomes. Notably, the chemi-fibrils bind and induce liposome leakage in vitro, suggesting they may do the same in cells. The chemi-fibrils induce extensive proteolysis including caspase substrates, yet modulatory profiling reveals that chemi-fibrils form a distinct class from existing inducers of cell death. The chemi-fibrils share similarities with proteinaceous fibrils and may provide insight into their mechanism of cellular toxicity.


Asunto(s)
Benzamidas/química , Benzamidas/farmacología , Caspasa 3/metabolismo , Cumarinas/química , Cumarinas/farmacología , Secuencia de Aminoácidos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células K562 , Lisosomas/química , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Modelos Biológicos , Estructura Molecular , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
J Biol Chem ; 287(40): 33781-95, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22872644

RESUMEN

Most proteases are expressed as inactive precursors, or zymogens, that become activated by limited proteolysis. We previously identified a small molecule, termed 1541, that dramatically promotes the maturation of the zymogen, procaspase-3, to its mature form, caspase-3. Surprisingly, compound 1541 self-assembles into nanofibrils, and localization of procaspase-3 to the fibrils promotes activation. Here, we interrogate the biochemical mechanism of procaspase-3 activation on 1541 fibrils in addition to proteogenic amyloid-ß(1-40) fibrils. In contrast to previous reports, we find no evidence that procaspase-3 alone is capable of self-activation, consistent with its fate-determining role in executing apoptosis. In fact, mature caspase-3 is >10(7)-fold more active than procaspase-3, making this proenzyme a remarkably inactive zymogen. However, we also show that fibril-induced colocalization of trace amounts of caspase-3 or other initiator proteases with procaspase-3 dramatically stimulates maturation of the proenzyme in vitro. Thus, similar to known cellular signaling complexes, these synthetic or natural fibrils can serve as platforms to concentrate procaspase-3 for trans-activation by upstream proteases.


Asunto(s)
Apoptosis , Caspasa 3/química , Péptidos beta-Amiloides/química , Catálisis , Dimerización , Sistemas de Liberación de Medicamentos , Activación Enzimática , Precursores Enzimáticos/química , Humanos , Cinética , Modelos Biológicos , Modelos Químicos , Péptido Hidrolasas/química , Transducción de Señal , Activación Transcripcional
10.
J Am Chem Soc ; 133(49): 19630-3, 2011 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22066605

RESUMEN

Modulating enzyme function with small-molecule activators, as opposed to inhibitors, offers new opportunities for drug discovery and allosteric regulation. We previously identified a compound, called 1541, from a high-throughput screen (HTS) that stimulates activation of a proenzyme, procaspase-3, to generate mature caspase-3. Here we further investigate the mechanism of activation and report the surprising finding that 1541 self-assembles into nanofibrils exceeding 1 µm in length. These particles are an unanticipated outcome from an HTS that have properties distinct from standard globular protein aggregators. Moreover, 1541 nanofibrils function as a unique biocatalytic material that activates procaspase-3 via induced proximity. These studies demonstrate a novel approach for proenzyme activation through binding to fibrils, which may mimic how procaspases are naturally processed on protein scaffolds.


Asunto(s)
Caspasa 3/metabolismo , Activación Enzimática/efectos de los fármacos , Precursores Enzimáticos/metabolismo , Nanofibras/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Nanofibras/ultraestructura
11.
Nat Chem Biol ; 6(3): 179-188, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20154666

RESUMEN

Drug discovery and chemical genetic efforts typically focus on the identification and design of inhibitors or loss-of-function probes as a means to perturb enzyme function. These tools are effective in determining the physiological consequence of ablating the activity of a specific enzyme. Remarkably, nearly a dozen examples of non-natural small molecules that activate enzyme catalysis have been identified within the past decade. In aggregate, these studies delineate four unique activation mechanisms that the small molecules exploit. These complementary gain-of-function probes offer a way to address the sufficiency of an enzyme to drive a particular cellular phenotype, and they also provide new opportunities for drug discovery. This review covers the identification and characterization of these unique small-molecule activators.

12.
Science ; 326(5954): 853-8, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19892984

RESUMEN

Virtually all of the 560 human proteases are stored as inactive proenyzmes and are strictly regulated. We report the identification and characterization of the first small molecules that directly activate proenzymes, the apoptotic procaspases-3 and -6. It is surprising that these compounds induce autoproteolytic activation by stabilizing a conformation that is both more active and more susceptible to intermolecular proteolysis. These procaspase activators bypass the normal upstream proapoptotic signaling cascades and induce rapid apoptosis in a variety of cell lines. Systematic biochemical and biophysical analyses identified a cluster of mutations in procaspase-3 that resist small-molecule activation both in vitro and in cells. Compounds that induce gain of function are rare, and the activators reported here will enable direct control of the executioner caspases in apoptosis and in cellular differentiation. More generally, these studies presage the discovery of other proenzyme activators to explore fundamental processes of proenzyme activation and their fate-determining roles in biology.


Asunto(s)
Benzopiranos/metabolismo , Caspasa 3/metabolismo , Caspasa 6/metabolismo , Activadores de Enzimas/metabolismo , Precursores Enzimáticos/metabolismo , Imidazoles/metabolismo , Piridinas/metabolismo , Animales , Apoptosis , Benzopiranos/química , Benzopiranos/farmacología , Biocatálisis , Caspasa 3/química , Caspasa 3/genética , Caspasa 6/química , Caspasa 6/genética , Inhibidores de Caspasas , Dominio Catalítico , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Activación Enzimática , Activadores de Enzimas/química , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Precursores Enzimáticos/antagonistas & inhibidores , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Granzimas/metabolismo , Humanos , Imidazoles/química , Imidazoles/farmacología , Cinética , Ratones , Estructura Molecular , Mutagénesis , Piridinas/química , Piridinas/farmacología , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo
13.
Nat Neurosci ; 11(7): 772-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18568022

RESUMEN

In traditional folk medicine, Xanthoxylum plants are referred to as 'toothache trees' because their anesthetic or counter-irritant properties render them useful in the treatment of pain. Psychophysical studies have identified hydroxy-alpha-sanshool as the compound most responsible for the unique tingling and buzzing sensations produced by Szechuan peppercorns or other Xanthoxylum preparations. Although it is generally agreed that sanshool elicits its effects by activating somatosensory neurons, the underlying cellular and molecular mechanisms remain a matter of debate. Here we show that hydroxy-alpha-sanshool excites two types of sensory neurons, including small-diameter unmyelinated cells that respond to capsaicin (but not mustard oil) as well as large-diameter myelinated neurons that express the neurotrophin receptor TrkC. We found that hydroxy-alpha-sanshool excites neurons through a unique mechanism involving inhibition of pH- and anesthetic-sensitive two-pore potassium channels (KCNK3, KCNK9 and KCNK18), providing a framework for understanding the unique and complex psychophysical sensations associated with the Szechuan pepper experience.


Asunto(s)
Amidas/farmacología , Neuronas Aferentes/efectos de los fármacos , Piper nigrum/química , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Capsaicina/farmacología , Células Cultivadas , Estimulación Eléctrica/métodos , Ganglios Sensoriales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Proteínas de Neurofilamentos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio de Dominio Poro en Tándem/efectos de los fármacos , Cloruro de Potasio/farmacología , Receptor trkC/metabolismo , Canal Catiónico TRPA1 , Canales Catiónicos TRPV/deficiencia , Canales de Potencial de Receptor Transitorio/deficiencia
14.
J Gen Virol ; 88(Pt 4): 1392-1401, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17374787

RESUMEN

Quinacrine and related 9-aminoacridine compounds are effective in eliminating the alternatively folded prion protein, termed PrP(Sc), from scrapie-infected cultured cells. Clinical evaluations of quinacrine for the treatment of human prion diseases are progressing in the absence of a clear understanding of the molecular mechanism by which prion replication is blocked. Here, insight into the mode of action of 9-aminoacridine compounds was sought by using a chemical proteomics approach to target identification. Cellular macromolecules that bind 9-aminoacridine ligands were affinity-purified from tissue lysates by using a 9-aminoacridine-functionalized solid-phase matrix. Although the 9-aminoacridine matrix was conformationally selective for PrP(Sc), it was inefficient: approximately 5 % of PrP(Sc) was bound under conditions that did not support binding of the cellular isoform, PrP(C). Our findings suggest that 9-aminoacridine compounds may reduce the PrP(Sc) burden either by occluding epitopes necessary for templating on the surface of PrP(Sc) or by altering the stability of PrP(Sc) oligomers, where a one-to-one stoichiometry is not necessary.


Asunto(s)
Aminacrina/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Priones/química , Priones/metabolismo , Animales , Western Blotting , Línea Celular , Cricetinae , Mesocricetus , Ratones , Ratones Transgénicos , Proteínas PrPC/química , Proteínas PrPC/aislamiento & purificación , Proteínas PrPSc/química , Proteínas PrPSc/aislamiento & purificación , Enfermedades por Prión/metabolismo , Unión Proteica , Pliegue de Proteína
15.
Antimicrob Agents Chemother ; 51(6): 2164-72, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17371810

RESUMEN

Parasitic diseases are of enormous public health significance in developing countries-a situation compounded by the toxicity of and resistance to many current chemotherapeutics. We investigated a focused library of 18 structurally diverse bis-acridine compounds for in vitro bioactivity against seven protozoan and one helminth parasite species and compared the bioactivities and the cytotoxicities of these compounds toward various mammalian cell lines. Structure-activity relationships demonstrated the influence of both the bis-acridine linker structure and the terminal acridine heterocycle on potency and cytotoxicity. The bioactivity of polyamine-linked acridines required a minimum linker length of approximately 10 A. Increasing linker length resulted in bioactivity against most parasites but also cytotoxicity toward mammalian cells. N alkylation, but less so N acylation, of the polyamine linker ameliorated cytotoxicity while retaining bioactivity with 50% effective concentration (EC(50)) values similar to or better than those measured for standard drugs. Substitution of the polyamine for either an alkyl or a polyether linker maintained bioactivity and further alleviated cytotoxicity. Polyamine-linked compounds in which the terminal acridine heterocycle had been replaced with an aza-acridine also maintained acceptable therapeutic indices. The most potent compounds recorded low- to mid-nanomolar EC(50) values against Plasmodium falciparum and Trypanosoma brucei; otherwise, low-micromolar potencies were measured. Importantly, the bioactivity of the library was independent of P. falciparum resistance to chloroquine. Compound bioactivity was a function of neither the potential to bis-intercalate DNA nor the inhibition of trypanothione reductase, an important drug target in trypanosomatid parasites. Our approach illustrates the usefulness of screening focused compound libraries against multiple parasite targets. Some of the bis-acridines identified here may represent useful starting points for further lead optimization.


Asunto(s)
Acridinas , Antiparasitarios , Técnicas Químicas Combinatorias/métodos , Eucariontes/efectos de los fármacos , Schistosoma mansoni/efectos de los fármacos , Acridinas/síntesis química , Acridinas/química , Acridinas/farmacología , Acridinas/toxicidad , Animales , Antiparasitarios/síntesis química , Antiparasitarios/química , Antiparasitarios/farmacología , Antiparasitarios/toxicidad , Eucariontes/clasificación , Eucariontes/crecimiento & desarrollo , Células HL-60 , Humanos , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/efectos de los fármacos , Poliaminas/química , Schistosoma mansoni/crecimiento & desarrollo , Relación Estructura-Actividad , Trypanosoma brucei brucei/efectos de los fármacos
16.
J Med Chem ; 50(1): 65-73, 2007 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-17201410

RESUMEN

2-Aminopyridine-3,5-dicarbonitrile compounds were previously identified as mimetics of dominant-negative prion protein mutants and inhibit prion replication in cultured cells. Here, we report findings from a comprehensive structure-activity relationship study of the 6-aminopyridine-3,5-dicarbonitrile scaffold. We identify compounds with significantly improved bioactivity (approximately 40-fold) against replication of the infectious prion isoform (PrPSc) and suitable pharmacokinetic profiles to warrant evaluation in animal models of prion disease.


Asunto(s)
Aminopiridinas/síntesis química , Nitrilos/síntesis química , Proteínas PrPSc/antagonistas & inhibidores , Aminopiridinas/química , Aminopiridinas/farmacología , Animales , Línea Celular Tumoral , Técnicas Químicas Combinatorias , Membranas Artificiales , Ratones , Modelos Moleculares , Nitrilos/química , Nitrilos/farmacología , Permeabilidad , Solubilidad , Relación Estructura-Actividad
17.
Bioorg Med Chem Lett ; 16(18): 4913-6, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16860557

RESUMEN

A focused library of variously substituted 9-aminoacridine compounds was screened for bioactivity against accumulation of the infectious prion protein isoform, denoted PrP(Sc), in a cell model of prion replication. The efficacy of compounds against PrP(Sc) accumulation was influenced by both substituents of the distal tertiary amine and acridine heterocycle, while cellular cytotoxicity was encoded in the acridine heterocycle substituents.


Asunto(s)
Aminacrina/química , Aminacrina/farmacología , Neuroblastoma/patología , Scrapie , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Estructura Molecular , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...