Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Med ; 3(12): 860-882.e15, 2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36257298

RESUMO

BACKGROUND: The near impermeability of the blood-brain barrier (BBB) and the unique neuroimmune environment of the CNS prevents the effective use of antibodies in neurological diseases. Delivery of biotherapeutics to the brain can be enabled through receptor-mediated transcytosis via proteins such as the transferrin receptor, although limitations such as the ability to use Fc-mediated effector function to clear pathogenic targets can introduce safety liabilities. Hence, novel delivery approaches with alternative clearance mechanisms are warranted. METHODS: Binders that optimized transport across the BBB, known as transcytosis-enabling modules (TEMs), were identified using a combination of antibody discovery techniques and pharmacokinetic analyses. Functional activity of TEMs were subsequently evaluated by imaging for the ability of myeloid cells to phagocytose target proteins and cells. FINDINGS: We demonstrated significantly enhanced brain exposure of therapeutic antibodies using optimal transferrin receptor or CD98 TEMs. We found that these modules also mediated efficient clearance of tau aggregates and HER2+ tumor cells via a non-classical phagocytosis mechanism through direct engagement of myeloid cells. This mode of clearance potentially avoids the known drawbacks of FcγR-mediated antibody mechanisms in the brain such as the neurotoxic release of proinflammatory cytokines and immune cell exhaustion. CONCLUSIONS: Our study reports a new brain delivery platform that harnesses receptor-mediated transcytosis to maximize brain uptake and uses a non-classical phagocytosis mechanism to efficiently clear pathologic proteins and cells. We believe these findings will transform therapeutic approaches to treat CNS diseases. FUNDING: This research was funded by Janssen, Pharmaceutical Companies of Johnson & Johnson.


Assuntos
Barreira Hematoencefálica , Transcitose , Barreira Hematoencefálica/metabolismo , Transcitose/fisiologia , Receptores da Transferrina , Transporte Biológico/fisiologia , Anticorpos
2.
Antibodies (Basel) ; 9(4)2020 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-33217946

RESUMO

T-cell Engaging bispecific antibodies (TcEs) that can re-direct cytotoxic T-cells to kill cancer cells have been validated in clinical studies. To date, the clinical success with these agents has mainly been seen in hematologic tumor indications. However, an increasing number of TcEs are currently being developed to exploit the potent mode-of-action to treat solid tumor indications, which is more challenging in terms of tumor-cell accessibility and the complexity of the tumor microenvironment (TME). Of particular interest is the potential of TcEs as an immunotherapeutic approach for the treatment of non-immunogenic (often referred to as cold) tumors that do not respond to checkpoint inhibitors such as programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) antibodies. This has led to considerable discovery efforts for, firstly, the identification of tumor selective targeting approaches that can safely re-direct cytotoxic T-cells to cancer cells, and, secondly, bispecific antibodies and their derivatives with drug-like properties that promote a potent cytolytic synapse between T-cells and tumor cells, and in the most advanced TcEs, have IgG-like pharmacokinetics for dosing convenience. Based on encouraging pre-clinical data, a growing number of TcEs against a broad range of targets, and using an array of different molecular structures have entered clinical studies for solid tumor indications, and the first clinical data is beginning to emerge. This review outlines the different approaches that have been taken to date in addressing the challenges of exploiting the TcE mode-of-action for a broad range of solid indications, as well as opportunities for future discovery potential.

3.
Clin Cancer Res ; 26(19): 5258-5268, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32554516

RESUMO

PURPOSE: Small cell lung cancer (SCLC) is the most lethal and aggressive subtype of lung carcinoma characterized by highly chemotherapy-resistant recurrence in the majority of patients. To effectively treat SCLC, we have developed a unique and novel IgG-like T-cell engaging bispecific antibody (ITE) that potently redirects T-cells to specifically lyse SCLC cells expressing Delta-like ligand 3 (DLL3), an antigen that is frequently expressed on the cell surface of SCLC cells, with no to very little detectable expression in normal tissues. EXPERIMENTAL DESIGN: The antitumor activity and mode of action of DLL3/CD3 ITE was evaluated in vitro using SCLC cell lines and primary human effector cells and in vivo in an SCLC xenograft model reconstituted with human CD3+ T-cells. RESULTS: Selective binding of DLL3/CD3 ITE to DLL3-positive tumor cells and T-cells induces formation of an immunological synapse resulting in tumor cell lysis and activation of T-cells. In a human T-cell engrafted xenograft model, the DLL3/CD3 ITE leads to an increase in infiltration of T-cells into the tumor tissue resulting in apoptosis of the tumor cells and tumor regression. Consistent with the mode of action, the DLL3/CD3 ITE treatment led to upregulation of PD-1, PD-L1, and LAG-3. CONCLUSIONS: This study highlights the ability of the DLL3/CD3 ITE to induce strictly DLL3-dependent T-cell redirected lysis of tumor cells and recruitment of T-cells into noninflamed tumor tissues leading to tumor regression in a preclinical in vivo model. These data support clinical testing of the DLL3/CD3 ITE in patients with SCLC.


Assuntos
Complexo CD3/genética , Proliferação de Células/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Animais , Anticorpos Biespecíficos/farmacologia , Antígenos CD/genética , Apoptose/efeitos dos fármacos , Antígeno B7-H1/genética , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Xenoenxertos , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos , Receptor de Morte Celular Programada 1/genética , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/imunologia , Carcinoma de Pequenas Células do Pulmão/patologia , Linfócitos T/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
4.
Blood ; 136(11): 1298-1302, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32483610

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy that accounts for ∼20% of ALL cases. Intensive chemotherapy regimens result in cure rates >85% in children and <50% in adults, warranting a search of novel therapeutic strategies. Although immune-based therapies have tremendously improved the treatment of B-ALL and other B-cell malignancies, they are not yet available for T-ALL. We report here that humanized, non-Fcγ receptor (FcγR)-binding monoclonal antibodies (mAbs) to CD3 have antileukemic properties in xenograft (PDX) models of CD3+ T-ALL, resulting in prolonged host survival. We also report that these antibodies cooperate with chemotherapy to enhance antileukemic effects and host survival. Because these antibodies show only minor, manageable adverse effects in humans, they offer a new therapeutic option for the treatment of T-ALL. Our results also show that the antileukemic properties of anti-CD3 mAbs are largely independent of FcγR-mediated pathways in T-ALL PDXs.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Complexo CD3/imunologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Antineoplásicos Imunológicos/imunologia , Complexo CD3/antagonistas & inibidores , Terapia Combinada , Dexametasona/administração & dosagem , Relação Dose-Resposta Imunológica , Feminino , Humanos , Camundongos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/imunologia , Organismos Livres de Patógenos Específicos , Vincristina/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
5.
MAbs ; 8(8): 1487-1497, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27680183

RESUMO

Bispecific antibodies have shown promise in the clinic as medicines with novel mechanisms of action. Lack of efficient production of bispecific IgGs, however, has limited their rapid advancement. Here, we describe a single-reactor process using mammalian cell co-culture production to efficiently produce a bispecific IgG with 4 distinct polypeptide chains without the need for parallel processing of each half-antibody or additional framework mutations. This method resembles a conventional process, and the quality and yield of the monoclonal antibodies are equal to those produced using parallel processing methods. We demonstrate the application of the approach to diverse bispecific antibodies, and its suitability for production of a tissue specific molecule targeting fibroblast growth factor receptor 1 and klotho ß that is being developed for type 2 diabetes and other obesity-linked disorders.


Assuntos
Anticorpos Biespecíficos/biossíntese , Reatores Biológicos , Técnicas de Cocultura/métodos , Imunoglobulina G/biossíntese , Animais , Anticorpos Biespecíficos/imunologia , Células CHO , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/imunologia , Proteínas Klotho , Mamíferos , Proteínas de Membrana/imunologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/imunologia
6.
Mol Pharm ; 13(9): 2996-3003, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27244474

RESUMO

We have developed a tool Fab fragment of a rabbit monoclonal antibody that is useful for early evaluation in rabbit models of technologies for long acting delivery (LAD) of proteins to the eye. Using this Fab we show that vitreal clearance can be slowed through increased hydrodynamic size. Fab (G10rabFab) and Fab' (G10rabFab') fragments of a rabbit monoclonal antibody (G10rabIgG) were expressed in Chinese hamster ovary (CHO) cells and purified using antigen-based affinity chromatography. G10rabFab retains antigen-binding upon thermal stress (37 °C) for 8 weeks in phosphate-buffered saline (PBS) and can be detected in rabbit tissues using an antigen-based ELISA. Hydrodynamic radius, measured using quasi-elastic light scattering (QELS), was increased through site-specific modification of the G10rabFab' free cysteine with linear methoxy-polyethylene glycol(PEG)-maleimide of 20000 or 40000 molecular weight. Pharmacokinetic studies upon intravitreal dosing in New Zealand white rabbits were conducted on the G10rabFab and PEGylated G10rabFab'. Results of single and multidose pharmacokinetic experiments yield reproducible results and a vitreal half-life for G10rabFab of 3.2 days. Clearance from the eye is slowed through increased hydrodynamic size, with vitreal half-life showing a linear dependence on hydrodynamic radius (RH). A linear dependence of vitreal half-life on RH suggests that molecule diffusivity makes an important contribution to vitreal clearance. A method for prediction of vitreal half-life from RH measurements is proposed.


Assuntos
Anticorpos Monoclonais/farmacocinética , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Fragmentos Fab das Imunoglobulinas/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Células CHO , Cricetulus , Ensaio de Imunoadsorção Enzimática , Hidrodinâmica , Injeções Intravítreas , Cinética , Polietilenoglicóis/química , Coelhos
7.
Sci Transl Med ; 7(287): 287ra70, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25972002

RESUMO

Bispecific antibodies and antibody fragments in various formats have been explored as a means to recruit cytolytic T cells to kill tumor cells. Encouraging clinical data have been reported with molecules such as the anti-CD19/CD3 bispecific T cell engager (BiTE) blinatumomab. However, the clinical use of many reported T cell-recruiting bispecific modalities is limited by liabilities including unfavorable pharmacokinetics, potential immunogenicity, and manufacturing challenges. We describe a B cell-targeting anti-CD20/CD3 T cell-dependent bispecific antibody (CD20-TDB), which is a full-length, humanized immunoglobulin G1 molecule with near-native antibody architecture constructed using "knobs-into-holes" technology. CD20-TDB is highly active in killing CD20-expressing B cells, including primary patient leukemia and lymphoma cells both in vitro and in vivo. In cynomolgus monkeys, CD20-TDB potently depletes B cells in peripheral blood and lymphoid tissues at a single dose of 1 mg/kg while demonstrating pharmacokinetic properties similar to those of conventional monoclonal antibodies. CD20-TDB also exhibits activity in vitro and in vivo in the presence of competing CD20-targeting antibodies. These data provide rationale for the clinical testing of CD20-TDB for the treatment of CD20-expressing B cell malignancies.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antígenos CD20/imunologia , Complexo CD3/imunologia , Leucemia de Células B/terapia , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacocinética , Humanos , Leucemia de Células B/imunologia , Macaca fascicularis , Camundongos , Camundongos Transgênicos
8.
Cancer Res ; 74(19): 5561-71, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25228655

RESUMO

Clinical results from the latest strategies for T-cell activation in cancer have fired interest in combination immunotherapies that can fully engage T-cell immunity. In this study, we describe a trastuzumab-based bispecific antibody, HER2-TDB, which targets HER2 and conditionally activates T cells. HER2-TDB specifically killed HER2-expressing cancer cells at low picomolar concentrations. Because of its unique mechanism of action, which is independent of HER2 signaling or chemotherapeutic sensitivity, HER2-TDB eliminated cells refractory to currently approved HER2 therapies. HER2-TDB exhibited potent antitumor activity in four preclinical model systems, including MMTV-huHER2 and huCD3 transgenic mice. PD-L1 expression in tumors limited HER2-TDB activity, but this resistance could be reversed by anti-PD-L1 treatment. Thus, combining HER2-TDB with anti-PD-L1 yielded a combination immunotherapy that enhanced tumor growth inhibition, increasing the rates and durability of therapeutic response.


Assuntos
Anticorpos Biespecíficos/imunologia , Ativação Linfocitária , Receptor ErbB-2/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Ratos , Ratos Sprague-Dawley , Trastuzumab
9.
J Biol Chem ; 288(37): 26583-93, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23880771

RESUMO

Human bispecific antibodies have great potential for the treatment of human diseases. Although human IgG1 bispecific antibodies have been generated, few attempts have been reported in the scientific literature that extend bispecific antibodies to other human antibody isotypes. In this paper, we report our work expanding the knobs-into-holes bispecific antibody technology to the human IgG4 isotype. We apply this approach to generate a bispecific antibody that targets IL-4 and IL-13, two cytokines that play roles in type 2 inflammation. We show that IgG4 bispecific antibodies can be generated in large quantities with equivalent efficiency and quality and have comparable pharmacokinetic properties and lung partitioning, compared with the IgG1 isotype. This work broadens the range of published therapeutic bispecific antibodies with natural surface architecture and provides additional options for the generation of bispecific antibodies with differing effector functions through the use of different antibody isotypes.


Assuntos
Anticorpos Biespecíficos/imunologia , Regulação da Expressão Gênica , Imunoglobulina G/imunologia , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Animais , Anticorpos Biespecíficos/biossíntese , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Imunoglobulina G/biossíntese , Pulmão/imunologia , Pulmão/metabolismo , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/metabolismo , Engenharia de Proteínas/métodos , Ressonância de Plasmônio de Superfície
10.
Nat Biotechnol ; 31(8): 753-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23831709

RESUMO

By enabling the simultaneous engagement of two distinct targets, bispecific antibodies broaden the potential utility of antibody-based therapies. However, bispecific-antibody design and production remain challenging, owing to the need to incorporate two distinct heavy and light chain pairs while maintaining natural nonimmunogenic antibody architecture. Here we present a bispecific-antibody production strategy that relies on co-culture of two bacterial strains, each expressing a half-antibody. Using this approach, we produce 28 unique bispecific antibodies. A bispecific antibody against the receptor tyrosine kinases MET and EGFR binds both targets monovalently, inhibits their signaling, and suppresses MET and EGFR-driven cell and tumor growth. Our strategy allows rapid generation of bispecific antibodies from any two existing antibodies and yields milligram to gram quantities of bispecific antibodies sufficient for a wide range of discovery and preclinical applications.


Assuntos
Anticorpos Biespecíficos/biossíntese , Técnicas de Cocultura , Receptores ErbB/imunologia , Neoplasias/terapia , Proteínas Proto-Oncogênicas c-met/imunologia , Anticorpos Biespecíficos/imunologia , Especificidade de Anticorpos , Bactérias/imunologia , Bactérias/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/genética , Regulação Bacteriana da Expressão Gênica/imunologia , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-met/genética
11.
PLoS One ; 7(12): e51817, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284778

RESUMO

The structure of the Fab region of antibodies is critical to their function. By introducing single cysteine substitutions into various positions of the heavy and light chains of the Fab region of trastuzumab, a potent antagonist of HER2, and using thiol chemistry to link the different Fabs together, we produced a variety of monospecific F(ab')(2)-like molecules with activities spanning from activation to inhibition of breast tumor cell growth. These isomers (or bis-Fabs) of trastuzumab, with varying relative spatial arrangements between the Fv-regions, were able to either promote or inhibit cell-signaling activities through the PI3K/AKT and MAPK pathways. A quantitative phosphorylation mapping of HER2 indicated that the agonistic isomers produced a distinct phosphorylation pattern associated with activation. This study suggests that antibody geometric isomers, found both in nature and during synthetic antibody development, can have profoundly different biological activities independent of their affinities for their target molecules.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama/tratamento farmacológico , Fragmentos Fab das Imunoglobulinas/farmacologia , Receptor ErbB-2/metabolismo , Anticorpos Monoclonais Humanizados/imunologia , Apoptose , Western Blotting , Neoplasias da Mama/metabolismo , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Fragmentos Fab das Imunoglobulinas/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trastuzumab , Células Tumorais Cultivadas
12.
J Biol Chem ; 285(27): 20850-9, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20444694

RESUMO

The development of bispecific antibodies as therapeutic agents for human diseases has great clinical potential, but broad application has been hindered by the difficulty of identifying bispecific antibody formats that exhibit favorable pharmacokinetic properties and ease of large-scale manufacturing. Previously, the development of an antibody technology utilizing heavy chain knobs-into-holes mutations and a single common light chain enabled the small-scale generation of human full-length bispecific antibodies. Here we have extended the technology by developing a two-part bispecific antibody discovery strategy that facilitates proof-of-concept studies and clinical candidate antibody generation. Our scheme consists of the efficient small-scale generation of bispecific antibodies lacking a common light chain and the hinge disulfides for proof-of-concept studies coupled with the identification of a common light chain bispecific antibody for large-scale production with high purity and yield. We have applied this technology to generate a bispecific antibody suitable for development as a human therapeutic. This antibody directly inhibits the activation of the high affinity IgE receptor FcepsilonRI on mast cells and basophils by cross-linking FcepsilonRI with the inhibitory receptor FcgammaRIIb, an approach that has strong therapeutic potential for asthma and other allergic diseases. Our approach for producing human bispecific full-length antibodies enables the clinical application of bispecific antibodies to a validated therapeutic pathway in asthma.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Receptores de IgE/fisiologia , Substituição de Aminoácidos , Animais , Anticorpos Biespecíficos/genética , Especificidade de Anticorpos , Basófilos/imunologia , Linhagem Celular Tumoral , Códon/genética , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Genes , Glutationa Transferase/genética , Humanos , Imunoglobulina E/genética , Imunoglobulina E/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Leves de Imunoglobulina/genética , Camundongos , Camundongos SCID , Anafilaxia Cutânea Passiva/imunologia , Receptores de IgE/antagonistas & inibidores , Receptores de IgE/efeitos dos fármacos , Receptores de IgE/imunologia , Receptores de IgG/imunologia , Proteínas Recombinantes/uso terapêutico , Neoplasias da Retina/imunologia , Retinoblastoma/imunologia , Sensibilidade e Especificidade
13.
J Biol Chem ; 284(50): 34553-60, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19854829

RESUMO

Proapoptotic receptor agonists cause cellular demise through the activation of the extrinsic and intrinsic apoptotic pathways. Inhibitor of apoptosis (IAP) proteins block apoptosis induced by diverse stimuli. Here, we demonstrate that IAP antagonists in combination with Fas ligand (FasL) or the death receptor 5 (DR5) agonist antibody synergistically stimulate death in cancer cells and inhibit tumor growth. Single-agent activity of IAP antagonists relies on tumor necrosis factor-alpha signaling. By contrast, blockade of tumor necrosis factor-alpha does not affect the synergistic activity of IAP antagonists with FasL or DR5 agonist antibody. In most cancer cells, proapoptotic receptor agonist-induced cell death depends on amplifying the apoptotic signal via caspase-8-mediated activation of Bid and subsequent activation of the caspase-9-dependent mitochondrial apoptotic pathway. In the investigated cancer cell lines, induction of apoptosis by FasL or DR5 agonist antibody can be inhibited by knockdown of Bid. However, knockdown of X chromosome-linked IAP (XIAP) or antagonism of XIAP allows FasL or DR5 agonist antibody to induce activation of effector caspases efficiently without the need for mitochondrial amplification of the apoptotic signal and thus rescues the effect of Bid knockdown in these cells.


Assuntos
Apoptose/fisiologia , Morte Celular/fisiologia , Proteína Ligante Fas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Etanercepte , Humanos , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
14.
J Med Chem ; 52(19): 5816-25, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19736996

RESUMO

Receptor-specific proteins produced by genetic engineering are attractive as PET imaging agents, but labeling with conventional (18)F-based prosthetic groups is problematic due to long synthesis times, poor radiochemical yields, and low specific activities. Therefore, we developed a modular platform for the rapid preparation of water-soluble prosthetic groups capable of efficiently introducing (18)F into proteins. The utility of this platform is demonstrated by the thiol-specific prosthetic group, [(18)F]FPEGMA, which was used to produce site-specifically (18)F-labeled protein ((18)F-trastuzumab-ThioFab) in 82 min with a total radiochemical yield of 13 +/- 3% and a specific activity of 2.2 +/- 0.2 Ci/micromol. (18)F-trastuzumab-ThioFab retained the biological activity of native protein and was successfully validated in vivo with microPET imaging of Her2 expression in a xenograft tumor-bearing murine model modulated by the Hsp90 inhibitor, 17-(allylamino)-17-demethoxygeldanamycin.


Assuntos
Receptores ErbB/análise , Radioisótopos de Flúor , Marcação por Isótopo/métodos , Tomografia por Emissão de Pósitrons/métodos , Receptor ErbB-2/análise , Animais , Anticorpos Monoclonais , Anticorpos Monoclonais Humanizados , Benzoquinonas , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Lactamas Macrocíclicas , Camundongos , Neoplasias Experimentais/diagnóstico , Engenharia de Proteínas , Proteínas/química , Transplante Heterólogo , Trastuzumab
15.
Proc Natl Acad Sci U S A ; 103(20): 7595-600, 2006 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-16682620

RESUMO

We present a common allosteric mechanism for control of inflammatory and apoptotic caspases. Highly specific thiol-containing inhibitors of the human inflammatory caspase-1 were identified by using disulfide trapping, a method for site-directed small-molecule discovery. These compounds became trapped by forming a disulfide bond with a cysteine residue in the cavity at the dimer interface approximately 15 A away from the active site. Mutational and structural analysis uncovered a linear circuit of functional residues that runs from one active site through the allosteric cavity and into the second active site. Kinetic analysis revealed robust positive cooperativity not seen in other endopeptidases. Recently, disulfide trapping identified a similar small-molecule site and allosteric transition in the apoptotic caspase-7 that shares only a 23% sequence identity with caspase-1. Together, these studies show a general small-molecule-binding site for functionally reversing the zymogen activation of caspases and suggest a common regulatory site for the allosteric control of inflammation and apoptosis.


Assuntos
Caspases , Dissulfetos/química , Conformação Proteica , Compostos de Sulfidrila/química , Regulação Alostérica , Sequência de Aminoácidos , Apoptose/fisiologia , Sítios de Ligação , Inibidores de Caspase , Caspases/química , Caspases/genética , Caspases/metabolismo , Cristalografia por Raios X , Humanos , Inflamação/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Alinhamento de Sequência
16.
Protein Expr Purif ; 41(1): 148-53, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15802232

RESUMO

Caspases have in the past decade become some of the most intensely pursued targets for the design of small-molecule inhibitors. Two significant technological roadblocks to developing caspase-binding molecules are the poor solubility of a subset of the bacterially expressed proteins and the instability of the renatured proteins that results from rapid inactivating autolysis at high protein concentrations. In this report, we present a generalized method of renaturing human caspases and inhibiting the self-proteolytic activity of the enzymes without a need for covalent active-site inhibitors. Our method, which involves blocking the S1 region of the active site with malonate, enables one to inhibit fully the inactivating autolysis in human caspases and increases the yields of renatured active enzyme. It furthermore does not necessitate removal of malonate prior to setting up enzymatic assays since as high as 100-mM concentrations of malonate do not compete efficiently with caspase substrates or larger caspase inhibitors for binding to the active site. The method described in this report simplifies greatly caspase purification and makes it possible to stabilize the enzymes against autolysis without a need for costly, and frequently synthetically challenging, small-molecule inhibitors.


Assuntos
Caspases/isolamento & purificação , Autólise , Caspase 1/química , Caspase 1/genética , Caspase 1/isolamento & purificação , Caspase 1/metabolismo , Caspase 3 , Inibidores de Caspase , Caspases/química , Caspases/genética , Caspases/metabolismo , Domínio Catalítico , Inibidores de Cisteína Proteinase/farmacologia , Estabilidade Enzimática , Escherichia coli/genética , Humanos , Técnicas In Vitro , Malonatos/metabolismo , Malonatos/farmacologia , Modelos Moleculares , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Solubilidade
17.
Proc Natl Acad Sci U S A ; 100(17): 10114-7, 2003 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12900501

RESUMO

The tomato systemin receptor, SR160, a plasma membrane-bound, leucine-rich repeat receptor kinase that signals systemic plant defense, and the brassinolide (BL) receptor, BRI1, that regulates developmental processes, have been shown recently to have identical amino acid sequences. We report herein that tobacco, a solanaceous species that does not express a systemin precursor gene nor responds to systemin, when transformed with the SR160 receptor gene, expresses the gene in suspension-cultured cells, evidenced by mRNA and protein analyses and photoaffinity-labeling experiments. Additionally, systemin induced an alkalinization response in the transgenic tobacco cells similar to that found in tomato cells, but not in WT cells. The gain in function in tobacco cells indicates that early steps of the systemin signaling pathway found in tomato are present in tobacco cells. A tomato line, cu-3, in which a mutation in the BRI1 gene has rendered the plant nonfunctional in BL signaling, exhibits a severely reduced response to systemin. In leaves of WT tomato plants, BL strongly and reversibly antagonized systemic signaling by systemin. The results suggest that the systemin-mediated systemic defense response may have evolved in some solanaceous species by co-opting the BRI1 receptor and associated components for defense signaling.


Assuntos
Genes de Plantas , Nicotiana/genética , Nicotiana/metabolismo , Peptídeos/metabolismo , Proteínas de Plantas/metabolismo , Proteínas Quinases/genética , Solanum lycopersicum/genética , Solanum lycopersicum/metabolismo , Proteínas de Plantas/genética , Plantas Geneticamente Modificadas , Proteínas Quinases/metabolismo , Transdução de Sinais , Especificidade da Espécie , Transformação Genética
18.
Proc Natl Acad Sci U S A ; 99(14): 9585-90, 2002 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-12060717

RESUMO

The isolation to homogeneity of the 160-kDa systemin cell-surface receptor (SR160) from plasma membranes of suspension cultured cells of Lycopersicon peruvianum is reported. The purification procedure resulted in recovery of 13 microg of pure receptor protein, representing an 8,200-fold purification. Gel blot analyses using SR160-specific antibodies confirmed that a cross-reacting protein in the membranes of suspension-cultured cells comigrates with both the native and a deglycosylated form of the radiolabeled receptor. Internal amino acid sequences of the purified protein facilitated the isolation of a cDNA clone encoding the 160-kDa receptor. The identity of the encoded protein as SR160 was further confirmed by a comparison of its sequence with a mass spectral fingerprint of the SR160 protein. The deduced amino acid sequence of SR160 revealed that it is a member of the leucine-rich repeat (LRR) receptor kinase family, closely related to the brassinolide receptor kinase, BRI1.


Assuntos
Peptídeos/metabolismo , Proteínas de Plantas/metabolismo , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Solanum lycopersicum/metabolismo , Sequência de Aminoácidos , Sequência de Bases , DNA de Plantas/genética , Genes de Plantas , Proteínas de Repetições Ricas em Leucina , Solanum lycopersicum/genética , Dados de Sequência Molecular , Proteínas de Plantas/genética , Proteínas Quinases/genética , Proteínas/genética , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA