Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 288(2): 1409-19, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184956

RESUMO

Protein biosynthesis and extracellular secretion are essential biological processes for therapeutic protein production in mammalian cells, which offer the capacity for correct folding and proper post-translational modifications. In this study, we have generated bispecific therapeutic fusion proteins in mammalian cells by combining a peptide and an antibody into a single open reading frame. A neutralizing peptide directed against interleukin-17A (IL17A) was genetically fused to the N termini of an anti-IL22 antibody, through either the light chain, the heavy chain, or both chains. Although the resulting fusion proteins bound and inhibited IL22 with the same affinity and potency as the unmodified anti-IL22 antibody, the peptide modality in the fusion scaffold was not active in the cell-based assay due to the N-terminal degradation. When a glutamine residue was introduced at the N terminus, which can be cyclized to form pyroglutamate in mammalian cells, the IL17A neutralization activity of the fusion protein was restored. Interestingly, the mass spectroscopic analysis of the purified fusion protein revealed an unexpected O-linked glycosylation modification at threonine 5 of the anti-IL17A peptide. The subsequent removal of this post-translational modification by site-directed mutagenesis drastically enhanced the IL17A binding affinity and neutralization potency for the resulting fusion protein. These results provide direct experimental evidence that post-translational modifications during protein biosynthesis along secretory pathways play critical roles in determining the structure and function of therapeutic proteins produced by mammalian cells. The newly engineered peptide-antibody genetic fusion is promising for therapeutically targeting multiple antigens in a single antibody-like molecule.


Assuntos
Anticorpos Biespecíficos/genética , Interleucina-17/imunologia , Interleucinas/imunologia , Polissacarídeos/química , Ácido Pirrolidonocarboxílico/química , Sequência de Aminoácidos , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Processamento de Proteína Pós-Traducional , Interleucina 22
2.
Proc Natl Acad Sci U S A ; 107(52): 22611-6, 2010 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-21149738

RESUMO

Bispecific antibodies (BsAbs) are regarded as promising therapeutic agents due to their ability to simultaneously bind two different antigens. Several bispecific modalities have been developed, but their utility is limited due to problems with stability and manufacturing complexity. Here we report a versatile technology, based on a scaffold antibody and pharmacophore peptide heterodimers, that enables rapid generation and chemical optimization of bispecific antibodies, which are termed bispecific CovX-Bodies. Two different peptides are joined together using a branched azetidinone linker and fused to the scaffold antibody under mild conditions in a site-specific manner. Whereas the pharmacophores are responsible for functional activities, the antibody scaffold imparts long half-life and Ig-like distribution. The pharmacophores can be chemically optimized or replaced with other pharmacophores to generate optimized or unique bispecific antibodies. As a prototype, we developed a bispecific antibody that binds both vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang2) simultaneously, inhibits their function, shows efficacy in tumor xenograft studies, and greatly augments the antitumor effects of standard chemotherapy. This unique antiangiogenic bispecific antibody is in phase-1 clinical trials.


Assuntos
Angiopoietina-2/imunologia , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Sequência de Aminoácidos , Angiopoietina-2/química , Angiopoietina-2/metabolismo , Animais , Anticorpos Biespecíficos/metabolismo , Especificidade de Anticorpos , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Azetidinas/química , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacocinética , Macaca fascicularis , Masculino , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Ressonância de Plasmônio de Superfície , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Sci Rep ; 6: 26071, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27184415

RESUMO

IL-17A is a pro-inflammatory cytokine that has been implicated in autoimmune and inflammatory diseases. Monoclonal antibodies inhibiting IL-17A signaling have demonstrated remarkable efficacy, but an oral therapy is still lacking. A high affinity IL-17A peptide antagonist (HAP) of 15 residues was identified through phage-display screening followed by saturation mutagenesis optimization and amino acid substitutions. HAP binds specifically to IL-17A and inhibits the interaction of the cytokine with its receptor, IL-17RA. Tested in primary human cells, HAP blocked the production of multiple inflammatory cytokines. Crystal structure studies revealed that two HAP molecules bind to one IL-17A dimer symmetrically. The N-terminal portions of HAP form a ß-strand that inserts between two IL-17A monomers while the C-terminal section forms an α helix that directly blocks IL-17RA from binding to the same region of IL-17A. This mode of inhibition suggests opportunities for developing peptide antagonists against this challenging target.


Assuntos
Inibidores Enzimáticos/metabolismo , Interleucina-17/antagonistas & inibidores , Peptídeos/metabolismo , Receptores de Interleucina-17/metabolismo , Substituição de Aminoácidos , Células Cultivadas , Cristalografia por Raios X , Inibidores Enzimáticos/isolamento & purificação , Humanos , Interleucina-17/química , Programas de Rastreamento , Modelos Moleculares , Mutagênese , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/isolamento & purificação , Ligação Proteica , Conformação Proteica
4.
Angew Chem Int Ed Engl ; 42(35): 4138-76, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-14502729

RESUMO

The high-throughput synthesis and screening of compound libraries hold tremendous promise for drug discovery and powerful methods for both solid-phase and solution-phase library preparation have been introduced. The question of which approach (solution-phase versus solid-phase) is best for the preparation of chemical libraries has been replaced by which approach is most appropriate for a particular target or screen. Herein we highlight distinctions in the two approaches that might serve as useful considerations at the onset of new programs. This is followed by a more personal account of our own focus on solution-phase techniques for the preparation of libraries designed to modulate cellular signaling by targeting protein-protein or protein-DNA interactions. The screening of our libraries against a prototypical set of extracellular and intracellular targets, using a wide range of assay formats, provided the first small-molecule modulators of the protein-protein interactions studied, and a generalized approach for conducting such studies.


Assuntos
Técnicas de Química Combinatória , DNA/metabolismo , Proteínas/metabolismo , Transdução de Sinais , Técnicas de Química Combinatória/métodos , DNA/química , Biblioteca de Peptídeos , Proteínas/química , Soluções
5.
Bioorg Med Chem Lett ; 16(19): 5194-8, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16870428

RESUMO

In the present work, we explore the possibility of introducing selectivity to existing chemotherapeutics via the design of non-pro-drug, bi-functional molecules comprising a microtubule-binding agent and a substrate for a disease-associated kinase. The design, synthesis, and in vitro biological evaluation of paclitaxel-thymidine and vinblastine-thymidine bi-functional conjugates are reported here. This work provides the first account of 'kinase-mediated trapping' of cancer therapeutics.


Assuntos
Antineoplásicos/síntese química , Sistemas de Liberação de Medicamentos/métodos , Proteínas de Neoplasias/metabolismo , Neoplasias/patologia , Paclitaxel/administração & dosagem , Proteínas Quinases/metabolismo , Timidina/administração & dosagem , Vimblastina/administração & dosagem , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Microtúbulos/metabolismo , Neoplasias/tratamento farmacológico , Relação Estrutura-Atividade , Especificidade por Substrato
6.
Bioorg Med Chem Lett ; 15(10): 2477-80, 2005 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-15863300

RESUMO

A general protocol for the synthesis of paclitaxel C-10 carbamates is described. The method entails MeI-mediated activation of 2'-O-TBS-7-O-TES-10-O-deacetyl-paclitaxel-10-O-carbonylimidazole prior to reaction with amines. This method is effective for the synthesis of paclitaxel C-10 derivatives, including bifunctional molecules.


Assuntos
Carbamatos/química , Paclitaxel/síntese química
7.
J Am Chem Soc ; 124(4): 544-55, 2002 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11804483

RESUMO

The erythropoietin receptor (EPOr) is activated by ligand-induced homodimerization, which leads to the proliferation and differentiation of erythroid progenitors. Through the screening of combinatorial libraries of dimeric iminodiacetic acid diamides, novel small molecule binders of EPOr were identified in a protein binding assay. Evaluation of a series of analogues led to optimization of binding subunits, and these were utilized in the synthesis of higher order dimer, trimer, and tetramer libraries. Several of the most active EPOr binders were found to be partial agonists and induced concentration-dependent proliferation of an EPO-dependent cell line (UT-7/EPO) while having no effect on a cell line lacking the EPOr (FDC-P1). An additional compound library, based on a symmetrical isoindoline-5,6-dicarboxylic acid template and including the optimized binding subunits, was synthesized and screened leading to the identification of additional EPO mimetics.


Assuntos
Ácidos Dicarboxílicos/química , Ácidos Dicarboxílicos/metabolismo , Eritropoetina/metabolismo , Iminoácidos/química , Iminoácidos/metabolismo , Indóis/química , Indóis/metabolismo , Mimetismo Molecular , Receptores da Eritropoetina/metabolismo , Divisão Celular/fisiologia , Técnicas de Química Combinatória , Eritropoetina/fisiologia , Humanos , Receptores da Eritropoetina/fisiologia , Soluções , Relação Estrutura-Atividade , Células Tumorais Cultivadas
8.
Biochemistry ; 41(48): 14206-15, 2002 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-12450384

RESUMO

Glycinamide ribonucleotide transformylase (GAR Tfase) is a key folate-dependent enzyme in the de novo purine biosynthesis pathway and, as such, has been the target for antitumor drug design. Here, we describe the crystal structures of the human GAR Tfase (purN) component of the human trifunctional protein (purD-purM-purN) at various pH values and in complex with its substrate. Human GAR Tfase exhibits pH-dependent enzyme activity with its maximum around pH 7.5-8. Comparison of unliganded human GAR Tfase structures at pH 4.2 and pH 8.5 reveals conformational differences in the substrate binding loop, which at pH 4.2 occupies the binding cleft and prohibits substrate binding, while at pH 8.5 is permissive for substrate binding. The crystal structure of GAR Tfase with its natural substrate, beta-glycinamide ribonucleotide (beta-GAR), at pH 8.5 confirms this conformational isomerism. Surprisingly, several important structural differences are found between human GAR Tfase and previously reported E. coli GAR Tfase structures, which have been used as the primary template for drug design studies. While the E. coli structure gave valuable insights into the active site and formyl transfer mechanism, differences in structure and inhibition between the bacterial and mammalian enzymes suggest that the human GAR Tfase structure is now the appropriate template for the design of anti-cancer agents.


Assuntos
Glicina/análogos & derivados , Glicina/química , Hidroximetil e Formil Transferases/química , Ribonucleotídeos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Carbono-Nitrogênio Ligases , Cristalografia por Raios X , Dimerização , Ativação Enzimática , Escherichia coli/enzimologia , Ácido Fólico/química , Ácido Fólico/metabolismo , Glicina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Hidroximetil e Formil Transferases/metabolismo , Cinética , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforribosilglicinamido Formiltransferase , Ribonucleotídeos/metabolismo , Especificidade por Substrato
9.
Proc Natl Acad Sci U S A ; 99(6): 3830-5, 2002 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11891322

RESUMO

Myc is a transcriptional regulator of the basic helix-loop-helix leucine zipper protein family. It has strong oncogenic potential, mutated or virally transduced forms of Myc induce lymphoid tumors in animals, and deregulated expression of Myc is associated with numerous types of human cancers. For its oncogenic activity, Myc must dimerize with the ubiquitously expressed basic helix-loop-helix leucine zipper protein Max. This requirement for dimerization may allow control of Myc activity with small molecules that interfere with Myc/Max dimerization. We have measured Myc/Max dimerization with fluorescence resonance energy transfer and have screened combinatorial chemical libraries for inhibitors of dimerization. Candidate inhibitors were isolated from a peptidomimetics library. Inhibition of Myc/Max interaction was validated by ELISA and electrophoretic mobility-shift assay. Two of the candidate inhibitors also interfere with Myc-induced oncogenic transformation in chicken embryo fibroblast cultures. Our work provides proof of principle for the identification of small molecule inhibitors of protein-protein interactions by using high-throughput screens of combinatorial chemical libraries.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/antagonistas & inibidores , Fibroblastos/patologia , Preparações Farmacêuticas , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Fatores de Transcrição , Animais , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina Básica , Transformação Celular Neoplásica/efeitos dos fármacos , Embrião de Galinha , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética , Transferência de Energia , Ensaio de Imunoadsorção Enzimática , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fluorescência , Concentração Inibidora 50 , Preparações Farmacêuticas/química , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo
11.
Bioorg Med Chem ; 11(20): 4503-9, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-13129586

RESUMO

The design and synthesis of 10-(2-benzoxazolcarbonyl)-DDACTHF (1) as an inhibitor of glycinamide ribonucleotide transformylase (GAR Tfase) and aminoimidazole carboxamide transformylase (AICAR Tfase) are reported. Ketone 1 and the corresponding alcohol 13 were evaluated for inhibition of GAR Tfase and AICAR Tfase and the former was found to be a potent inhibitor of recombinant human (rh) GAR Tfase (Ki=600 nM).


Assuntos
Hidroximetil e Formil Transferases/antagonistas & inibidores , Purinas/biossíntese , Tetra-Hidrofolatos/química , Sítios de Ligação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Concentração Inibidora 50 , Fosforribosilaminoimidazolcarboxamida Formiltransferase , Fosforribosilglicinamido Formiltransferase , Purinas/antagonistas & inibidores , Relação Estrutura-Atividade , Tetra-Hidrofolatos/farmacologia
12.
Bioorg Med Chem ; 11(20): 4511-21, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-13129587

RESUMO

The synthesis and evaluation of analogues and key derivatives of 10-CF3CO-DDACTHF as inhibitors of glycinamide ribonucleotide transformylase (GAR Tfase) and aminoimidazole carboxamide transformylase (AICAR Tfase) are reported. Polyglutamate analogues of 1 were evaluated as inhibitors of Escherichia coli and recombinant human (rh) GAR Tfase, and AICAR Tfase. Although the pentaglutamate 6 was found to be the most active inhibitor of the series tested against rhGAR Tfase (Ki=0.004 microM), little distinction between the mono-pentaglutamate derivatives was observed (Ki=0.02-0.004 microM), suggesting that the principal role of the required polyglutamation of 1 is intracellular retention. In contrast, 1 and its defined polyglutamates 3-6 were much less inactive when tested against rhAICAR Tfase (Ki=65-0.120 microM) and very selective (> or =100-fold) for rh versus E. coli GAR Tfase. Additional key analogues of 1 were examined (7 and 8) and found to be much less active (1000-fold) highlighting the exceptional characteristics of 1.


Assuntos
Ácido Fólico/análogos & derivados , Hidroximetil e Formil Transferases/antagonistas & inibidores , Purinas/biossíntese , Tetra-Hidrofolatos/química , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Fosforribosilaminoimidazolcarboxamida Formiltransferase , Fosforribosilglicinamido Formiltransferase , Purinas/antagonistas & inibidores , Relação Estrutura-Atividade , Tetra-Hidrofolatos/farmacologia
13.
Biochemistry ; 42(20): 6043-56, 2003 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-12755606

RESUMO

Glycinamide ribonucleotide transformylase (GAR Tfase) has been the target of anti-neoplastic intervention for almost two decades. Here, we use a structure-based approach to design a novel folate analogue, 10-(trifluoroacetyl)-5,10-dideazaacyclic-5,6,7,8-tetrahydrofolic acid (10-CF(3)CO-DDACTHF, 1), which specifically inhibits recombinant human GAR Tfase (K(i) = 15 nM), but is inactive (K(i) > 100 microM) against other folate-dependent enzymes that have been examined. Moreover, compound 1 is a potent inhibitor of tumor cell proliferation (IC(50) = 16 nM, CCRF-CEM), which represents a 10-fold improvement over Lometrexol, a GAR Tfase inhibitor that has been in clinical trials. Thus, this folate analogue 1 is among the most potent and selective inhibitors known toward GAR Tfase. Contributing to its efficacious activity, compound 1 is effectively transported into the cell by the reduced folate carrier and intracellularly sequestered by polyglutamation. The crystal structure of human GAR Tfase with folate analogue 1 at 1.98 A resolution represents the first structure of any GAR Tfase to be determined with a cofactor or cofactor analogue without the presence of substrate. The folate-binding loop of residues 141-146, which is highly flexible in both Escherichia coli and unliganded human GAR Tfase structures, becomes highly ordered upon binding 1 in the folate-binding site. Computational docking of the natural cofactor into this and other apo or complexed structures provides a rational basis for modeling how the natural cofactor 10-formyltetrahydrofolic acid interacts with GAR Tfase, and suggests that this folate analogue-bound conformation represents the best template to date for inhibitor design.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Hidroximetil e Formil Transferases/antagonistas & inibidores , Tetra-Hidrofolatos/química , Tetra-Hidrofolatos/farmacologia , Sítios de Ligação , Linhagem Celular , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Escherichia coli/enzimologia , Humanos , Hidroximetil e Formil Transferases/química , Técnicas In Vitro , Cinética , Substâncias Macromoleculares , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Fosforribosilglicinamido Formiltransferase , Conformação Proteica , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Eletricidade Estática , Tetra-Hidrofolatos/síntese química
14.
Bioorg Med Chem ; 10(8): 2739-49, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12057663

RESUMO

The synthesis of 10-formyl-DDACTHF (3) as a potential inhibitor of glycinamide ribonucleotide transformylase (GAR Tfase) and aminoimidazole carboxamide ribonucleotide transformylase (AICAR Tfase) is reported. Aldehyde 3, the corresponding gamma- and alpha-pentaglutamates 21 and 25 and related agents were evaluated for inhibition of folate-dependent enzymes including GAR Tfase and AICAR Tfase. The inhibitors were found to exhibit potent cytotoxic activity (CCRF-CEM IC(50) for 3=60nM) that exceeded their enzyme inhibition potency [K(i) (3)=6 and 1 microM for Escherichia coli GAR and human AICAR Tfase, respectively]. Cytotoxicity rescue by medium purines, but not pyrimidines, indicated that the potent cytotoxic activity is derived from selective purine biosynthesis inhibition and rescue by AICAR monophosphate established that the activity is derived preferentially from GAR versus AICAR Tfase inhibition. The potent cytotoxic compounds including aldehyde 3 lost activity against CCRF-CEM cell lines deficient in the reduced folate carrier (CCRF-CEM/MTX) or folylpolyglutamate synthase (CCRF-CEM/FPGS(-)) establishing that their potent activity requires both reduced folate carrier transport and polyglutamation. Unexpectedly, the pentaglutamates displayed surprisingly similar K(i)'s versus E. coli GAR Tfase and only modestly enhanced K(i)'s versus human AICAR Tfase. On the surface this initially suggested that the potent cytotoxic activity of 3 and related compounds might be due simply to preferential intracellular accumulation of the inhibitors derived from effective transport and polyglutamation (i.e., ca. 100-fold higher intracellular concentrations). However, a subsequent examination of the inhibitors against recombinant human GAR Tfase revealed they and the corresponding gamma-pentaglutamates were unexpectedly much more potent against the human versus E. coli enzyme (K(i) for 3, 14nM against rhGAR Tfase versus 6 microM against E. coli GAR Tfase) which also accounts for their exceptional cytotoxic potency.


Assuntos
Antineoplásicos/síntese química , Hidroximetil e Formil Transferases/antagonistas & inibidores , Purinas/biossíntese , Receptores de Superfície Celular , Tetra-Hidrofolatos/síntese química , Antineoplásicos/farmacologia , Proteínas de Transporte/fisiologia , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Receptores de Folato com Âncoras de GPI , Humanos , Peptídeo Sintases/fisiologia , Fosforribosilaminoimidazolcarboxamida Formiltransferase , Fosforribosilglicinamido Formiltransferase , Purinas/antagonistas & inibidores , Relação Estrutura-Atividade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa