Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Blood Cells Mol Dis ; 47(2): 107-16, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21641240

RESUMEN

The ß-hemoglobinopathies and thalassemias are serious genetic blood disorders affecting the ß-globin chain of hemoglobin A (α(2)ß(Α)(2)). Their clinical severity can be reduced by enhancing expression of fetal hemoglobin (γ-globin), producing HbF (α(2)γ(2,)). In studies reported here, γ-globin induction by 23 novel, structurally-unrelated compounds, which had been predicted through molecular modeling and in silico screening of a 13,000 chemical library, was evaluated in vitro in erythroid progenitors cultured from normal subjects and ß-thalassemia patients, and in vivo in transgenic mice or anemic baboons. Four predicted candidates were found to have high potency, with 4- to 8-fold induction of HbF. Two of these compounds have pharmacokinetic profiles favorable for clinical application. These studies thus effectively identified high potency γ-globin inducing candidate therapeutics and validated the utility of in silico molecular modeling.


Asunto(s)
Anemia/tratamiento farmacológico , Productos Biológicos/administración & dosificación , Diseño de Fármacos , Células Precursoras Eritroides/efectos de los fármacos , Hemoglobina Fetal/biosíntesis , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Talasemia beta/tratamiento farmacológico , gamma-Globinas/biosíntesis , Administración Oral , Anemia/genética , Anemia/metabolismo , Animales , Productos Biológicos/química , Productos Biológicos/uso terapéutico , Células Cultivadas , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Expresión Génica , Humanos , Inyecciones Intravenosas , Ratones , Ratones Transgénicos , Modelos Moleculares , Papio , Flebotomía , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Globinas beta/deficiencia , Globinas beta/genética , Talasemia beta/genética , Talasemia beta/metabolismo , gamma-Globinas/genética
2.
Ann N Y Acad Sci ; 1054: 257-65, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16339673

RESUMEN

Accelerated apoptosis of erythroid progenitors in beta-thalassemia is a significant barrier to definitive therapy because the beneficial effects of fetal globin-inducing agents on globin chain balance may not be inducible in cells in which programmed cell death is established early. Accordingly, our objectives have been to identify methods to decrease cellular apoptosis and to identify orally tolerable fetal globin gene inducers. A pilot clinical trial was conducted to determine whether combined use of a fetal globin gene inducer (butyrate) and rhu-erythropoietin (EPO), the hematopoietic growth factor that prolongs erythroid cell survival and stimulates erythroid proliferation, would produce additive hematologic responses in any thalassemia subjects. Butyrate and EPO were administered in 10 patients. Novel fetal globin gene inducers that also stimulate erythroid proliferation were evaluated for pharmacokinetic profiles. Patients with beta+-thalassemia had relatively low levels of endogenous EPO (<145 mU/mL) and had additive responses to administered EPO and butyrate. Patients with at least one beta 0-globin mutation had higher baseline HbF levels (>60%) and EPO levels (>160 mU/mL), and three-fourths of these subjects responded to the fetal globin gene inducer alone. A few select fetal globin-inducing short-chain fatty acid derivatives that stimulated cell proliferation also had favorable pharmacokinetics. These studies identify a significant subset of thalassemia patients who appear to require exogenous EPO to respond optimally to any HbF inducer, as well as new therapeutic candidates that act on both cellular and molecular pathologies of beta-thalassemia. Both approaches now offer excellent potential for tolerable, definitive treatment of beta-thalassemia.


Asunto(s)
Butiratos/uso terapéutico , Células Eritroides/efectos de los fármacos , Eritropoyetina/uso terapéutico , Hemoglobina Fetal/biosíntesis , Expresión Génica/efectos de los fármacos , Talasemia beta/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Transfusión Sanguínea , Butiratos/administración & dosificación , Células Cultivadas/efectos de los fármacos , Terapia Combinada , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Células Eritroides/metabolismo , Eritropoyetina/administración & dosificación , Ácidos Grasos Volátiles/farmacocinética , Ácidos Grasos Volátiles/farmacología , Hemoglobina Fetal/genética , Humanos , Papio , Proyectos Piloto , Proteínas Recombinantes , Resultado del Tratamiento , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/terapia
3.
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
4.
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
5.
Endocr Relat Cancer ; 18(6): 759-71, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21990324

RESUMEN

The concept of targeting cancer therapeutics toward specific mutations or abnormalities in tumor cells, which are not found in normal tissues, has the potential advantages of high selectivity for the tumor and correspondingly low secondary toxicities. Many human malignancies display activating mutations in the Ras family of signal-transducing genes or over-activity of p21(Ras)-signaling pathways. Carcinoid and other neuroendocrine tumors have been similarly demonstrated to have activation of Ras signaling directly by mutations in Ras, indirectly by loss of Ras-regulatory proteins, or via constitutive activation of upstream or downstream effector pathways of Ras, such as growth factor receptors or PI(3)-kinase and Raf/mitogen-activated protein kinases. We previously reported that aberrant activation of Ras signaling sensitizes cells to apoptosis when the activity of the PKCδ isozyme is suppressed and that PKCδ suppression is not toxic to cells with normal levels of p21(Ras) signaling. We demonstrate here that inhibition of PKCδ by a number of independent means, including genetic mechanisms (shRNA) or small-molecule inhibitors, is able to efficiently and selectively repress the growth of human neuroendocrine cell lines derived from bronchopulmonary, foregut, or hindgut tumors. PKCδ inhibition in these tumors also efficiently induced apoptosis. Exposure to small-molecule inhibitors of PKCδ over a period of 24  h is sufficient to significantly suppress cell growth and clonogenic capacity of these tumor cell lines. Neuroendocrine tumors are typically refractory to conventional therapeutic approaches. This Ras-targeted therapeutic approach, mediated through PKCδ suppression, which selectively takes advantage of the very oncogenic mutations that contribute to the malignancy of the tumor, may hold potential as a novel therapeutic modality.


Asunto(s)
Proliferación Celular , Tumores Neuroendocrinos/terapia , Proteína Quinasa C-delta/antagonistas & inhibidores , Acetofenonas/farmacología , Animales , Apoptosis , Benzopiranos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Neoplasias Gastrointestinales , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares , Ratones , Células 3T3 NIH , Tumores Neuroendocrinos/enzimología , Tumores Neuroendocrinos/genética , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo
7.
Chem Biol Drug Des ; 71(2): 97-105, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18179464

RESUMEN

Targeted disruption of the pp60(src) (Src) gene has implicated this tyrosine kinase in osteoclast-mediated bone resorption and as a therapeutic target for the treatment of osteoporosis and other bone-related diseases. Here, we describe structure activity relationships of a novel series of carbon-linked, 2-substituted purines that led to the identification of AP23451 as a potent inhibitor of Src tyrosine kinase with antiresorptive activity in vivo. AP23451 features the use of an arylphosphinylmethylphosphinic acid moiety which confers bone-targeting properties to the molecule, thereby increasing local concentrations of the inhibitor to actively resorbing osteoclasts at the bone interface. AP23451 exhibited an IC50 = 68 nm against Src kinase; an X-ray crystal structure of the molecule complexed with Src detailed the molecular interactions responsible for its Src inhibition. In vivo, AP23451 demonstrated a dose-dependent decrease in PTH-induced hypercalcemia. Moreover, AP23517, a structurally and biochemically similar molecule with comparable activity (IC50 = 73 nm) except devoid of the bone-targeting element, demonstrated significantly reduced in vivo efficacy, suggesting that Src activity was necessary but not sufficient for in vivo activity in this series of compounds.


Asunto(s)
Adenina/análogos & derivados , Resorción Ósea/tratamiento farmacológico , Organofosfonatos/farmacología , Purinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adenina/química , Adenina/farmacología , Cristalografía por Rayos X , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Hipercalcemia , Concentración 50 Inhibidora , Estructura Molecular , Organofosfonatos/química , Osteoporosis/tratamiento farmacológico , Hormona Paratiroidea/farmacología , Ácidos Fosfínicos , Purinas/síntesis química , Relación Estructura-Actividad , Familia-src Quinasas/química
8.
Chem Biol Drug Des ; 67(5): 318-28, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16784456

RESUMEN

Pharmacologic reinduction of the developmentally silenced fetal (gamma) globin genes has been achieved in hemoglobinopathy patients using short chain fatty acid derivatives, with therapeutic effects. However, higher-potency inducers than are available in currently identified short chain fatty acid derivatives are desirable for long-term use. Using several short-chain fatty acids with established gamma-globin induction activity, a pharmacophore template was constructed with the TFIT module of the flo software and used to select several new candidate compounds, three of which exhibited significant activity in a gamma-globin gene reporter transcriptional assay which detects only strong inducers. The data were used to construct a new pharmacophore and a 'pseudo' receptor around it. Six hundred and thirty low-molecular weight compounds were docked into this receptor model. Of 26 compounds selected and tested in functional assays, two compounds showed activity >500% over control levels and two had activity 200% over control range, significantly more active than previously identified short chain fatty acid derivative fetal globin gene inducers. Three compounds had less activity; the remainder showed moderate activity. These findings demonstrate the feasibility of using iterative construction of pharmacophores, pseudo-binding site modeling, and virtual screening to identify small molecules with the ability to induce transcription of specific target genes, for potential therapeutics.


Asunto(s)
Diseño de Fármacos , Ácidos Grasos/farmacología , Hemoglobina Fetal/genética , Modelos Moleculares , Activación Transcripcional , Animales , Sitios de Unión , Butiratos/química , Butiratos/farmacología , Línea Celular Tumoral , Leucemia Eritroblástica Aguda/metabolismo , Luciferasas , Ratones , Estructura Molecular , Fenilbutiratos/química , Fenilbutiratos/farmacología
9.
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
10.
Chem Biol Drug Des ; 67(1): 46-57, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16492148

RESUMEN

The tyrosine kinase pp60src (Src) is the prototypical member of a family of proteins that participate in a broad array of cellular signal transduction processes, including cell growth, differentiation, survival, adhesion, and migration. Abnormal Src family kinase (SFK) signaling has been linked to several disease states, including osteoporosis and cancer metastases. Src has thus emerged as a molecular target for the discovery of small-molecule inhibitors that regulate Src kinase activity by binding to the ATP pocket within the catalytic domain. Here, we present crystal structures of the kinase domain of Src in complex with two purine-based inhibitors: AP23451, a small-molecule inhibitor designed to inhibit Src-dependent bone resorption, and AP23464, a small-molecule inhibitor designed to inhibit the Src-dependent metastatic spread of cancer. In each case, a trisubstituted purine template core was elaborated using structure-based drug design to yield a potent Src kinase inhibitor. These structures represent early examples of high affinity purine-based Src family kinase-inhibitor complexes, and they provide a detailed view of the specific protein-ligand interactions that lead to potent inhibition of Src. In particular, the 3-hydroxyphenethyl N9 substituent of AP23464 forms unique interactions with the protein that are critical to the picomolar affinity of this compound for Src. The comparison of these new structures with two relevant kinase-inhibitor complexes provides a structural basis for the observed kinase inhibitory selectivity. Further comparisons reveal a concerted induced-fit movement between the N- and C-terminal lobes of the kinase that correlates with the affinity of the ligand. Binding of the most potent inhibitor, AP23464, results in the largest induced-fit movement, which can be directly linked to interactions of the hydrophenethyl N9 substituent with a region at the interface between the two lobes. A less pronounced induced-fit movement is also observed in the Src-AP23451 complex. These new structures illustrate how the combination of structural, computational, and medicinal chemistry can be used to rationalize the process of developing high affinity, selective tyrosine kinase inhibitors as potential therapeutic agents.


Asunto(s)
Adenina/análogos & derivados , Diseño de Fármacos , Inhibidores Enzimáticos/química , Organofosfonatos/química , Purinas/química , Relación Estructura-Actividad , Familia-src Quinasas/química , Adenina/química , Adenina/metabolismo , Adenina/farmacología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Huesos/metabolismo , Carbono/química , Dominio Catalítico/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Ligandos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/antagonistas & inhibidores , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/química , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Nitrógeno/química , Organofosfonatos/metabolismo , Organofosfonatos/farmacología , Conformación Proteica/efectos de los fármacos , Purinas/síntesis química , Purinas/farmacología , Pirimidinas/química , Especificidad por Sustrato , Familia-src Quinasas/antagonistas & inhibidores
11.
Biopolymers ; 71(6): 717-29, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14991680

RESUMEN

A series of novel nonpeptide inhibitors of the pp60(c-Src) (Src) SH2 domain is described that exploit multifunctional group replacement of the phenylphosphate moiety of phosphotyrosine (pTyr). Relative to an x-ray structure of citrate complexed to the pTyr binding site of the Src SH2 domain, these nonpeptide ligands illustrate the systematic replacement of the phosphate group by multiple nonhydrolyzable, mono- or dianionic functionalities. Specifically, several phenylalanine (Phe) analogs incorporating key 4' and 3' substituents were synthesized and incorporated into a bicyclic benzamide template previously reported (W. C. Shakespeare et al., Proceedings of the National Academy of Science USA, 2000, Vol. 97, pp. 9373-9378). These pTyr mimetics included 4',3'-diphosphono-Phe (Dpp), 4',3'-dicarboxymethyloxy-Phe (Dcp), and 4'-phosphono-3'-carboxymethyloxy-Phe (Cpp). Noteworthy were nonpeptide inhibitors 8-11 that were 5- to 10-fold more potent than the cognate tetrapeptide ligand Ac-pTyr-Glu-Glu-Ile-NH(2) in binding to the Src SH2 domain.


Asunto(s)
Péptidos/química , Fosfotirosina/química , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Dominios Homologos src , Sitios de Unión , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Modelos Moleculares , Péptidos/farmacología , Relación Estructura-Actividad
12.
Bioorg Med Chem Lett ; 13(18): 3063-6, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941334

RESUMEN

Src tyrosine kinase is a therapeutic target for bone diseases that has been validated by gene knockout studies. Furthermore, in vitro cellular studies implicate that Src has a positive regulatory role in osteoclasts and a negative regulatory role in osteoblasts. The potential use of Src inhibitors for osteoporosis therapy has been previously shown by novel bone-targeted ligands of the Src SH2 (e.g., AP22408) and non-bone-targeted, ATP-based inhibitors of Src kinase. Significant to this study, compounds 2-12 exemplify novel analogues of known pyrrolopyrimidine and pyrazolopyrimidine template-based Src kinase inhibitors that incorporate bone-targeting group modifications designed to provide tissue (bone) selectivity and diminished side effects. Accordingly, we report here the structure-based design, synthetic chemistry and biological testing of these compounds and proof-of-concept studies thereof.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Diseño de Fármacos , Osteoporosis/tratamiento farmacológico , Pirimidinas/síntesis química , Familia-src Quinasas/antagonistas & inhibidores , Animales , Enfermedades Óseas/tratamiento farmacológico , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Purinas/síntesis química , Purinas/farmacología , Pirimidinas/farmacología , Relación Estructura-Actividad
13.
Bioorg Med Chem Lett ; 13(18): 3067-70, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941335

RESUMEN

Novel bone-targeted 2,6,9-trisubstituted purine template-based inhibitors of Src tyrosine kinase are described. Drug design studies of known purine compounds revealed that both positions-2 and -6 were suitable for incorporating bone-seeking moieties. A variety of bone-targeting groups with different affinity to hydroxyapatite were utilized in the study. Compound 3d was determined to be a potent Src inhibitor and was quite selective against a panel of other protein kinases.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Purinas/síntesis química , Familia-src Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/análogos & derivados , Animales , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Durapatita/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Purinas/farmacología , Relación Estructura-Actividad
14.
Blood ; 104(8): 2532-9, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15256422

RESUMEN

The deregulated, oncogenic tyrosine kinase Bcr-Abl causes chronic myeloid leukemia (CML). Imatinib mesylate (Gleevec, STI571), a Bcr-Abl kinase inhibitor, selectively inhibits proliferation and promotes apoptosis of CML cells. Despite the success of imatinib mesylate in the treatment of CML, resistance is observed, particularly in advanced disease. The most common imatinib mesylate resistance mechanism involves Bcr-Abl kinase domain mutations that impart varying degrees of drug insensitivity. AP23464, a potent adenosine 5'-triphosphate (ATP)-based inhibitor of Src and Abl kinases, displays antiproliferative activity against a human CML cell line and Bcr-Abl-transduced Ba/F3 cells (IC(50) = 14 nM; imatinib mesylate IC(50) = 350 nM). AP23464 ablates Bcr-Abl tyrosine phosphorylation, blocks cell cycle progression, and promotes apoptosis of Bcr-Abl-expressing cells. Biochemical assays with purified glutathione S transferase (GST)-Abl kinase domain confirmed that AP23464 directly inhibits Abl activity. Importantly, the low nanomolar cellular and biochemical inhibitory properties of AP23464 extend to frequently observed imatinib mesylate-resistant Bcr-Abl mutants, including nucleotide binding P-loop mutants Q252H, Y253F, E255K, C-terminal loop mutant M351T, and activation loop mutant H396P. AP23464 was ineffective against mutant T315I, an imatinib mesylate contact residue. The potency of AP23464 against imatinib mesylate-refractory Bcr-Abl and its distinct binding mode relative to imatinib mesylate warrant further investigation of AP23464 for the treatment of CML.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Mutación/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfato/química , Aminoácidos/genética , Aminoácidos/metabolismo , Apoptosis/efectos de los fármacos , Benzamidas , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/química , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HL-60 , Humanos , Mesilato de Imatinib , Concentración 50 Inhibidora , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de la Leche/metabolismo , Modelos Moleculares , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Piperazinas/química , Piperazinas/farmacología , Estructura Terciaria de Proteína , Piridonas/química , Piridonas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Factor de Transcripción STAT5 , Transactivadores/metabolismo
15.
Bioorg Med Chem Lett ; 13(18): 3071-4, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941336

RESUMEN

The design of bone-targeted pyrido[2,3-d]pyrimidin-7-ones as Src tyrosine kinase inhibitors is described. Leveraging SAR from known compounds and using structure-based methods, we were able to rapidly incorporate bone binding components, which maintained, and even increased potency against the target enzyme. Compound 4 displayed a high affinity for hydroxyapatite, a major constituent of bone, and demonstrated antiresoprtive activity in our cell-based assay.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Resorción Ósea/prevención & control , Pirimidinonas/síntesis química , Familia-src Quinasas/antagonistas & inhibidores , Animales , Resorción Ósea/tratamiento farmacológico , Simulación por Computador , Dentina/metabolismo , Diseño de Fármacos , Durapatita/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Concentración 50 Inhibidora , Osteoclastos/efectos de los fármacos , Pirimidinonas/farmacología , Conejos , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA