Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Bioorg Med Chem Lett ; 27(8): 1670-1680, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28302397

RESUMEN

The emergence and spread of multidrug-resistant (MDR) Gram negative bacteria presents a serious threat for public health. Novel antimicrobials that could overcome the resistance problems are urgently needed. UDP-3-O-(R-3-hydroxymyristol)-N-acetylglucosamine deacetylase (LpxC) is a cytosolic zinc-based deacetylase that catalyzes the first committed step in the biosynthesis of lipid A, which is essential for the survival of Gram-negative bacteria. Our efforts toward the discovery of novel LpxC inhibitors are presented herein.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Antibacterianos/química , Antibacterianos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Amidohidrolasas/metabolismo , Descubrimiento de Drogas , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Humanos , Simulación del Acoplamiento Molecular
2.
Bioorg Med Chem Lett ; 22(19): 6248-51, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22951041

RESUMEN

Daptomycin was shown to interact in vitro with pulmonary surfactant leading to reduction of its antibacterial activity. We report herein the preparation and anti-staphylococcal activity of a series of daptomycin analogs with reduced pulmonary surfactant interaction by replacing tryptophan with various amino acids.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Daptomicina/análogos & derivados , Daptomicina/farmacología , Surfactantes Pulmonares/química , Staphylococcus aureus/efectos de los fármacos , Triptófano/metabolismo , Daptomicina/química , Pruebas de Sensibilidad Microbiana , Conformación Molecular
3.
Bioorg Med Chem Lett ; 21(12): 3743-8, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21561767

RESUMEN

Ponatinib (AP24534) was previously identified as a pan-BCR-ABL inhibitor that potently inhibits the T315I gatekeeper mutant, and has advanced into clinical development for the treatment of refractory or resistant CML. In this study, we explored a novel series of five and six membered monocycles as alternate hinge-binding templates to replace the 6,5-fused imidazopyridazine core of ponatinib. Like ponatinib, these monocycles are tethered to pendant toluanilides via an ethynyl linker. Several compounds in this series displayed excellent in vitro potency against both native BCR-ABL and the T315I mutant. Notably, a subset of inhibitors exhibited desirable PK and were orally active in a mouse model of T315I-driven CML.


Asunto(s)
Alquinos/síntesis química , Alquinos/farmacología , Compuestos de Anilina/síntesis química , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Tolueno/síntesis química , Administración Oral , Alquinos/química , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Animales , Ciclización , Modelos Animales de Enfermedad , Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Ratones , Modelos Moleculares , Estructura Molecular , Mutación , Ratas , Relación Estructura-Actividad , Tolueno/química , Tolueno/farmacología
4.
J Med Chem ; 62(21): 9541-9559, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31593466

RESUMEN

Novel nanoparticle-drug conjugates (NDCs) containing diverse, clinically relevant anticancer drug payloads (docetaxel, cabazitaxel, and gemcitabine) were successfully generated and tested in drug discovery studies. The NDCs utilized structurally varied linkers that attached the drug payloads to a ß-cyclodextrin-PEG copolymer to form self-assembled nanoparticles. In vitro release studies revealed a diversity of release rates driven by linker structure-activity relationships (SARs). Improved in vivo pharmacokinetics (PK) for the cabazitaxel (CBTX) NDCs with glycinate-containing (1c) and hexanoate-containing linkers (2c) were demonstrated, along with high and sustained tumor levels (>168 h of released drug in tumor tissues). This led to potent efficacy and survival in both taxane- and docetaxel-resistant in vivo anticancer mouse efficacy models. Overall, the CBTX-hexanoate NDC 2c (CRLX522), demonstrated optimal and improved in vivo PK (plasma and tumor) and efficacy profile versus those of the parent drug, and the results support the potential therapeutic use of CRLX522 as a new anticancer agent.


Asunto(s)
Portadores de Fármacos/química , Diseño de Fármacos , Nanopartículas/química , Polietilenglicoles/química , Taxoides/química , Taxoides/farmacología , beta-Ciclodextrinas/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Masculino , Melanoma Experimental/patología , Ratones , Taxoides/farmacocinética , Distribución Tisular
5.
Clin Cancer Res ; 12(20 Pt 2): 6291s-6295s, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17062716

RESUMEN

Src tyrosine kinase was the first gene product shown to have an essential function in bone using recombinant DNA technology after its expression was knocked out in mice approximately 15 years ago. Since then, our understanding of the regulation of bone catabolism has advanced significantly with the identification of other key enzymes that regulate osteoclast formation, activation, and survival after their knockout in mice or recognition of mutations in them in humans. This led to the discovery or development of specific inhibitors of some of these key enzymes, including Src, as proof-of-concept lead compounds or potential clinical candidates for the prevention of diseases associated with increased bone resorption, such as osteoporosis and metastatic bone disease. Although bisphosphonates have been prescribed with proven and improving efficacy for the prevention of bone loss for >30 years, adverse effects, such as upper gastrointestinal tract symptoms, and the requirement to take them at least 2 hours before food have limited patient compliance. Thus, with growing knowledge of the pathways regulating osteoclast function and the appreciation that some of these are active also in tumor cells, drug companies have made efforts to identify small-molecular lead compounds for development into new therapeutic agents for the prevention of bone loss with efficacy that matches or supersedes that of bisphosphonates. In this article, we review our current understanding of the signaling pathways that regulate osteoclast formation, activation, and survival with specific reference to the role of Src tyrosine kinase and downstream signaling and highlight in a variety of models of increased bone resorption the effects of Src kinase inhibitors that have been targeted to bone to limit potential adverse effects on other cells.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Resorción Ósea/prevención & control , Osteoclastos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones , Osteoclastos/citología , Osteoclastos/enzimología , Inhibidores de Proteínas Quinasas/química , Transducción de Señal/fisiología , Relación Estructura-Actividad , Familia-src Quinasas/efectos de los fármacos
6.
Cancer Res ; 65(4): 1335-42, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735019

RESUMEN

Src tyrosine kinase expression and activity are elevated during colon cancer progression. How this contributes to the malignant phenotype is not fully understood. We show that in KM12C colon carcinoma cells, expression of kinase-deficient Src proteins (SrcMF and Src251) does not alter cell growth. Src kinase activity is required for turnover of cell-matrix adhesions and, in particular, the Src-dependent phosphorylation of focal adhesion kinase (FAK) is required for their disassembly. Surprisingly, we found that expression of SrcMF or Src251 resulted in increased tyrosine phosphorylation of FAK on Tyr(407), Tyr(576), Tyr(577), and Tyr(861), which are considered to be Src kinase substrates. This Src kinase-independent phosphorylation of FAK required an intact Src SH2 domain that mediates association of Src and FAK at peripheral adhesions. Use of a novel highly potent and selective Src kinase inhibitor AP23464 combined with experiments in Src/Fyn/Yes-deficient fibroblasts showed that increased phosphorylation of FAK in cells expressing SrcMF did not require Src-like kinases. However, specific phosphorylation on Tyr(925) of FAK was not evident in SrcMF- or Src251-expressing cells, and lack of Src kinase-dependent phosphorylation on this site was associated with impaired adhesion turnover. Our data show that Src kinase activity is required for adhesion turnover associated with cell migration in cancer cells and that, in addition to the catalytic activity, Src also acts as an adaptor to recruit other kinases that can phosphorylate key substrates including FAK. These studies have implications for tumor progression with respect to the use of Src kinase inhibitors.


Asunto(s)
Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Proteínas Tirosina Quinasas/metabolismo , Familia-src Quinasas/metabolismo , Animales , Sitios de Unión , Catálisis , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Femenino , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Ratones , Ratones Desnudos , Fosforilación , Transducción de Señal , Especificidad por Sustrato , Tirosina/metabolismo , Dominios Homologos src
8.
Ann N Y Acad Sci ; 1068: 447-57, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16831942

RESUMEN

Understanding of the regulation of bone catabolism has advanced significantly over the past two decades with the identification of key enzymes that regulate osteoclast formation, activation, and survival following their knockout in mice or recognition of mutations in humans. This led to the discovery of specific inhibitors of some of these key enzymes as proof-of-concept lead compounds or potential clinical candidates for the prevention of osteoporosis and other diseases associated with increased bone resorption. Bisphosphonates have been the major therapeutic agents prescribed for the prevention of bone loss in a variety of pathologic conditions for over 30 years. More potent amino bisphosphonates have increased efficacy than earlier drugs, but side effects such as upper gastrointestinal symptoms and the requirement to take them at least 2 h before food have limited patient compliance. This, coupled with the growing knowledge of the pathways regulating osteoclast function, has driven efforts to identify small molecular lead compounds that could be developed into new therapeutic agents with efficacy that matches or supersedes that of bisphosphonates for the prevention of bone loss. In this article, we review briefly the effects of specific inhibitors of bone resorption that have been developed to date and highlight in a variety of models of increased bone resorption the effects of Src kinase inhibitors that have been targeted to bone to limit potential unwanted side effects on other cells.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Huesos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Resorción Ósea , Catepsina K , Catepsinas/antagonistas & inhibidores , Humanos , Osteoclastos/fisiología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Sirolimus/antagonistas & inhibidores , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores
9.
Mol Cancer Ther ; 4(12): 1900-11, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16373705

RESUMEN

c-Src is frequently activated in human malignancies, including colon, breast, and pancreatic carcinomas. Several recent studies have shown that activation of Src family kinases leads to tumor progression and metastasis by increasing cellular migration and invasion, promoting cell growth and survival, and deregulating expression of proangiogenic molecules. Therefore, selective inhibitors of Src are being developed for cancer therapy. In this study, we characterize the biological effects of the novel ATP-based Src family kinase inhibitor, AP23846, in tumor cells with high Src activity. As a lead compound, AP23846 is a potent c-Src kinase inhibitor (IC50 approximately 0.5 nmol/L in vitro, approximately 10-fold more potent than PP2, the most widely used commercially available Src family kinase inhibitor). At concentrations of 1 micromol/L, AP23846 led to complete Src inhibition for 48 hours in cells. No cytotoxicity was observed under these conditions, although proliferation rates were slower. Therefore, this was an excellent inhibitor to examine Src-regulated signaling pathways in tumor cells. AP23846 reduced cellular migration, vascular endothelial growth factor, and interleukin-8 in a dose-dependent fashion in pancreatic adenocarcinoma cells grown in vitro. Correspondingly, cell culture supernatants from L3.6pl pancreatic adenocarcinoma cells pretreated with AP23846 failed to promote migration of hepatic endothelial cells in vitro and failed to support angiogenesis into gel foams implanted s.c. in mice in vivo. These results suggest that Src inhibitors affect biological properties of tumor progression and may be useful as cancer therapeutic agents in more advanced disease.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Inhibidores Enzimáticos/farmacología , Interleucina-8/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/prevención & control , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/farmacología , Animales , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Cartilla de ADN , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C3H , Neoplasias/irrigación sanguínea , Fosforilación , ARN Interferente Pequeño
10.
ACS Med Chem Lett ; 7(4): 374-8, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27096044

RESUMEN

The ATPase subunit of DNA gyrase B is an attractive antibacterial target due to high conservation across bacteria and the essential role it plays in DNA replication. A novel class of pyrazolopyridone inhibitors was discovered by optimizing a fragment screening hit scaffold using structure guided design. These inhibitors show potent Gram-positive antibacterial activity and low resistance incidence against clinically important pathogens.

11.
J Med Chem ; 58(12): 5137-42, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-25993059

RESUMEN

Novel cyclic lipopeptides with different acyl tails were synthesized via a semisynthetic approach. Structure-activity relationship studies revealed that lipophilicity, chain length, and the location of key aromatic functionalities of the tail modulated activity. The lead compound surotomycin exhibited significantly improved in vitro activity compared with daptomycin (MIC90 0.5 vs 2 µg/mL) against Clostridium difficile including NAP1 epidemic strains. In hamster efficacy studies, surotomycin protected animals at a dose of 0.5 mg/kg, PO.


Asunto(s)
Antibacterianos/química , Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Diarrea/tratamiento farmacológico , Enterocolitis Seudomembranosa/tratamiento farmacológico , Lipopéptidos/química , Lipopéptidos/uso terapéutico , Animales , Cricetinae , Diarrea/microbiología , Enterocolitis Seudomembranosa/complicaciones , Masculino , Pruebas de Sensibilidad Microbiana , Péptidos Cíclicos/química , Péptidos Cíclicos/uso terapéutico , Relación Estructura-Actividad
12.
ACS Med Chem Lett ; 6(10): 1080-5, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26487916

RESUMEN

Antibacterials with a novel mechanism of action offer a great opportunity to combat widespread antimicrobial resistance. Bacterial DNA Gyrase is a clinically validated target. Through physiochemical property optimization of a pyrazolopyridone hit, a novel class of GyrB inhibitors were discovered. Guided by structure-based drug design, indazole derivatives with excellent enzymatic and antibacterial activity as well as great animal efficacy were discovered.

13.
J Med Chem ; 58(21): 8503-12, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26460684

RESUMEN

The emergence and spread of multidrug resistant bacteria are widely believed to endanger human health. New drug targets and lead compounds exempt from cross-resistance with existing drugs are urgently needed. We report on the discovery of azaindole ureas as a novel class of bacterial gyrase B inhibitors and detail the story of their evolution from a de novo design hit based on structure-based drug design. These inhibitors show potent minimum inhibitory concentrations against fluoroquinolone resistant MRSA and other Gram-positive bacteria.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Girasa de ADN/metabolismo , Indoles/farmacología , Staphylococcus aureus Resistente a Meticilina/enzimología , Inhibidores de Topoisomerasa II/farmacología , Urea/farmacología , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/enzimología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Indoles/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Modelos Moleculares , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Inhibidores de Topoisomerasa II/química , Urea/análogos & derivados
14.
Curr Pharm Des ; 8(23): 2049-75, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12171518

RESUMEN

The dynamic and highly regulated processes of bone remodeling involve two major cells, osteoclasts and osteoblasts, both of which command a multitude of cellular signaling pathways involving protein kinases. Of the possible kinases in these cells, Src tyrosine kinase stands out as a promising therapeutic target for bone disease as validated by Src knockout mouse studies and in vitro cellular experiments, suggesting a regulatory role for Src in both osteoclasts (positive) and osteoblasts (negative). Advances in structural studies involving both Src and non-Src family kinases, in activated and unactivated protein states, have uncovered key binding site interactions that have led to the design of potent Src inhibitors. The lead compounds originate from a variety of synthetic templates and have demonstrated nM potency in enzymatic/binding assays and efficacy in animal models of bone disease. This review will provide a current understanding of critical Src signalling pathways in osteoclasts and osteoblasts, while detailing the structure-based design and screening-based lead discovery of Src inhibitors to be developed as therapeutic agents for bone disease.


Asunto(s)
Enfermedades Óseas/enzimología , Inhibidores Enzimáticos/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Factores Biológicos/química , Factores Biológicos/farmacología , Enfermedades Óseas/tratamiento farmacológico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Huesos/citología , Huesos/enzimología , Técnicas Químicas Combinatorias , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Imitación Molecular , Osteoclastos/citología , Osteoclastos/fisiología , Osteoporosis/tratamiento farmacológico , Osteoporosis/enzimología , Osteoporosis/patología , Péptidos/química , Unión Proteica , Dominios Homologos src , Familia-src Quinasas/química
15.
Curr Opin Drug Discov Devel ; 6(5): 729-41, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14579523

RESUMEN

Bone-targeted Src tyrosine kinase (STK) inhibitors have recently been developed for the treatment of osteoporosis and cancer-related bone diseases. The concept of bone targeting derives from bisphosphonates, and from the evolution of such molecules in terms of therapeutic efficacy for the treatment of bone disorders. Interestingly, some of the earliest bisphosphonates were recognized for their ability to inhibit calcium carbonate precipitation (scaling) by virtue of their affinity to chelate calcium. This chelating property was subsequently exploited in the development of bisphosphonate analogs as inhibitors of the bone-resorbing cells known as osteoclasts, giving rise to breakthrough medicines, such as Fosamax (for the treatment of osteoporosis) and Zometa (for the treatment of osteoporosis and bone metastases). Relative to these milestone achievements, there is a tremendous opportunity to explore beyond the limited chemical space (functional group diversity) of such bisphosphonates to design novel bone-targeting moieties, which may be used to develop other classes of promising small-molecule drugs affecting different biological pathways. Here, we review studies focused on bone-targeted inhibitors of STK, a key enzyme in osteoclast-dependent bone resorption. Two strategies are described relative to bone-targeted STK inhibitor drug discovery: (i) the development of novel Src homology (SH)-2 inhibitors incorporating non-hydrolyzable phosphotyrosine mimics and exhibiting molecular recognition and bone-targeting properties, leading to the in vivo-effective lead compound AP-22408; and (ii) the development of novel ATP-based Src kinase inhibitors incorporating bone-targeting moieties, leading to the in vivo-effective lead compound AP-23236. In summary, AP-22408 and AP-23236, which differ mechanistically by virtue of blocking Src-dependent non-catalytic or catalytic activities in osteoclasts, exemplify ARIAD Pharmaceuticals' structure-based design of novel bone-targeted lead compounds, successfully achieving in vivo proof-of-concept and providing the framework for the next-generation molecules that have further advanced, in terms of preclinical studies, for the treatment of osteoporosis and related bone diseases, including osteolytic bone metastases.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/enzimología , Difosfonatos/farmacología , Fenoles/farmacología , Purinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/enzimología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/enzimología , Difosfonatos/uso terapéutico , Diseño de Fármacos , Humanos , Modelos Moleculares , Osteoclastos/efectos de los fármacos , Osteoclastos/enzimología , Osteoporosis/tratamiento farmacológico , Osteoporosis/enzimología , Fenoles/uso terapéutico , Conformación Proteica , Purinas/uso terapéutico , Relación Estructura-Actividad , Dominios Homologos src , Familia-src Quinasas/química
16.
Mini Rev Med Chem ; 2(5): 475-88, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12370048

RESUMEN

The structural and functional characterization of Src homology-2 (SH2) domains and their relationship to catalytic proteins (e.g., kinases, phosphatases, and lipases) or non-catalytic proteins (e.g., upstream adapters, and downstream transcription factors) has significantly impacted our understanding of signal transduction pathways and the identification of promising therapeutic targets for drug discovery. Such SH2-containing proteins are known to be intimately involved in the regulation of a number of cellular processes, including growth, mitogenesis, motility, metabolism, and gene transcription. Molecular recognition and biochemical selectivity exists for various SH2 domains based on their binding to phosphotyrosine (pTyr) and contiguous C-terminal amino acids of cognate protein 'partners' in a sequence-dependent manner (i.e., -pTyr-AA(1)-AA(2)-AA(3)-) which result in the formation of signal transduction protein complexes in cells. In recent years, drug discovery efforts have advanced peptidomimetic and nonpeptide inhibitors of such protein-protein interactions based on mimicking pTyr-containing peptide ligands as well as SH2 structure-based de novo design of nonpeptide templates that can capture key binding sites on the target protein. Noteworthy are peptidomimetic and nonpeptide inhibitors of Src, Lck, Grb2, PI-3K, and Zap70 from pioneering efforts that led to the first examples of cellularly and in vivo active SH2 inhibitors. This mini-review highlights key achievements in SH2 inhibitor drug discovery with an emphasis on peptidomimetic and nonpeptide lead compounds in terms of structure-based design, key chemical and biological properties, and proof-of-concept studies relative to further defining the role(s) of SH2 domains in signal transduction processes, cellular functions, and in vivo disease models.


Asunto(s)
Imitación Molecular , Péptidos/farmacología , Dominios Homologos src/efectos de los fármacos , Conformación Molecular , Péptidos/química , Transducción de Señal , Relación Estructura-Actividad
17.
J Med Chem ; 53(12): 4701-19, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20513156

RESUMEN

In the treatment of chronic myeloid leukemia (CML) with BCR-ABL kinase inhibitors, the T315I gatekeeper mutant has emerged as resistant to all currently approved agents. This report describes the structure-guided design of a novel series of potent pan-inhibitors of BCR-ABL, including the T315I mutation. A key structural feature is the carbon-carbon triple bond linker which skirts the increased bulk of Ile315 side chain. Extensive SAR studies led to the discovery of development candidate 20g (AP24534), which inhibited the kinase activity of both native BCR-ABL and the T315I mutant with low nM IC(50)s, and potently inhibited proliferation of corresponding Ba/F3-derived cell lines. Daily oral administration of 20g significantly prolonged survival of mice injected intravenously with BCR-ABL(T315I) expressing Ba/F3 cells. These data, coupled with a favorable ADME profile, support the potential of 20g to be an effective treatment for CML, including patients refractory to all currently approved therapies.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Imidazoles/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Piridazinas/síntesis química , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Proteínas de Fusión bcr-abl/genética , Imidazoles/farmacocinética , Imidazoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Ratones , Ratones SCID , Modelos Moleculares , Mutación , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Piridazinas/farmacocinética , Piridazinas/farmacología , Ratas
19.
Cancer Cell ; 16(5): 401-12, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19878872

RESUMEN

Inhibition of BCR-ABL by imatinib induces durable responses in many patients with chronic myeloid leukemia (CML), but resistance attributable to kinase domain mutations can lead to relapse and a switch to second-line therapy with nilotinib or dasatinib. Despite three approved therapeutic options, the cross-resistant BCR-ABL(T315I) mutation and compound mutants selected on sequential inhibitor therapy remain major clinical challenges. We report design and preclinical evaluation of AP24534, a potent, orally available multitargeted kinase inhibitor active against T315I and other BCR-ABL mutants. AP24534 inhibited all tested BCR-ABL mutants in cellular and biochemical assays, suppressed BCR-ABL(T315I)-driven tumor growth in mice, and completely abrogated resistance in cell-based mutagenesis screens. Our work supports clinical evaluation of AP24534 as a pan-BCR-ABL inhibitor for treatment of CML.


Asunto(s)
Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Imidazoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piridazinas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Cristalografía por Rayos X , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Imidazoles/química , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones SCID , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Piridazinas/química , Transducción de Señal/efectos de los fármacos
20.
Proc Natl Acad Sci U S A ; 103(24): 9244-9, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16754879

RESUMEN

Mutation in the ABL kinase domain is the principal mechanism of imatinib resistance in patients with chronic myelogenous leukemia. Many mutations favor active kinase conformations that preclude imatinib binding. Because the active forms of ABL and SRC resemble one another, we tested two dual SRC-ABL kinase inhibitors, AP23464 and PD166326, against 58 imatinib-resistant (IM(R)) BCR/ABL kinase variants. Both compounds potently inhibit most IM(R) variants, and in vitro drug selection demonstrates that active (AP23464) and open (PD166326) conformation-specific compounds are less susceptible to resistance than imatinib. Combinations of inhibitors suppressed essentially all resistance mutations, with the notable exception of T315I. Guided by mutagenesis studies and molecular modeling, we designed a series of AP23464 analogues to target T315I. The analogue AP23846 inhibited both native and T315I variants of BCR/ABL with submicromolar potency but showed nonspecific cellular toxicity. Our data illustrate how conformational dynamics of the ABL kinase accounts for the activity of dual SRC-ABL inhibitors against IM(R)-mutants and provides a rationale for combining conformation specific inhibitors to suppress resistance.


Asunto(s)
Resistencia a Medicamentos/fisiología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Benzamidas , Proteínas de Fusión bcr-abl , Humanos , Mesilato de Imatinib , Modelos Moleculares , Estructura Molecular , Mutación , Piperazinas/química , Piperazinas/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-abl/genética , Piridinas/química , Piridinas/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA