Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Am Soc Mass Spectrom ; 32(7): 1567-1574, 2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-33415981

RESUMEN

NK group 2 member A (NKG2A), an immune checkpoint inhibitor, is an emerging therapeutic target in immuno-oncology. NKG2A forms a heterodimer with CD94 on the cell surface of NK and a subset of T cells and recognizes the nonclassical human leukocyte antigen (HLA-E) in humans. Therapeutic blocking antibodies that block the ligation between HLA-E and NKG2A/CD94 have been shown to enhance antitumor immunity in mice and humans. In this study, we illustrate the practical utilities of mass spectrometry (MS)-based protein footprinting in areas from reagent characterization to antibody epitope mapping. Hydrogen/deuterium exchange mass spectrometry (HDX-MS) in the higher-order structure characterization of NKG2A in complex with CD94 provides novel insights into the conformational dynamics of NKG2A/CD94 heterodimer. To fully understand antibody/target interactions, we employed complementary protein footprinting methods, including HDX-MS and fast photochemical oxidation of proteins (FPOP)-MS, to determine the binding epitopes of therapeutic monoclonal antibodies targeting NKG2A. Such a combination approach provides molecular insights into the binding mechanisms of antibodies to NKG2A with high specificity, demonstrating the blockade of NKG2A/HLA-E interaction.


Asunto(s)
Anticuerpos , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio/métodos , Subfamília C de Receptores Similares a Lectina de Células NK , Subfamília D de Receptores Similares a Lectina de las Células NK , Huella de Proteína/métodos , Anticuerpos/química , Anticuerpos/metabolismo , Mapeo Epitopo , Epítopos , Humanos , Subfamília C de Receptores Similares a Lectina de Células NK/química , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Subfamília D de Receptores Similares a Lectina de las Células NK/química , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo
2.
J Med Chem ; 63(22): 13913-13950, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33155811

RESUMEN

A series of tetrahydroisoquinoline-based benzodiazepine dimers were synthesized and tested for in vitro cytotoxicity against a panel of cancer cell lines. Structure-activity relationship investigation of various spacers guided by molecular modeling studies helped to identify compounds with picomolar activity. Payload 17 was conjugated to anti-mesothelin and anti-fucosylated monosialotetrahexosylganglioside (FucGM1) antibodies using lysosome-cleavable valine-citrulline dipeptide linkers via heterogeneous lysine conjugation and bacterial transglutaminase-mediated site-specific conjugation. In vitro, these antibody drug conjugates (ADCs) exhibited significant cytotoxic and target-mediated selectivity on human cancer cell lines. The pharmacokinetics and efficacy of these ADCs were further evaluated in gastric and lung cancer xenograft models in mice. Consistent pharmacokinetic profiles, high target specificity, and robust antitumor activity were observed in these models after a single dose of the ADC-46 (0.02 µmol/kg).


Asunto(s)
Anticuerpos Monoclonales/química , Antineoplásicos/farmacología , Benzodiazepinas/química , Diseño de Fármacos , Inmunoconjugados/farmacología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Tetrahidroisoquinolinas/química , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antineoplásicos/química , Apoptosis , Benzodiazepinas/metabolismo , Proliferación Celular , Femenino , Gangliósido G(M1)/análogos & derivados , Gangliósido G(M1)/inmunología , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunoconjugados/química , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Mesotelina , Ratones , Ratones SCID , Carcinoma Pulmonar de Células Pequeñas/patología , Neoplasias Gástricas/patología , Relación Estructura-Actividad , Tetrahidroisoquinolinas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Anal Chem ; 92(2): 2065-2073, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31860282

RESUMEN

Antibody drug conjugates (ADCs) can undergo in vivo biotransformation (e.g., payload metabolism, deconjugation) leading to reduced or complete loss of activity. The location/site of conjugation of payload-linker can have an effect on ADC stability and hence needs to be carefully optimized. Affinity capture LC-MS of intact ADCs or ADC subfragments has been extensively used to evaluate ADC biotransformation. However, the current methods have certain limitations such as the requirement of specific capture reagents, limited mass resolution of low mass change metabolites, low sensitivity, and use of capillary or nanoflow LC-MS. To address these challenges, we developed a generic affinity capture LC-MS assay that can be utilized to evaluate the biotransformation of any site-specific ADC independent of antibody type and site of conjugation (Fab and Fc) in preclinical studies. The method involves a combination of some or all of these steps: (1) "mono capture" or "dual capture" of ADCs from serum with streptavidin magnetic beads coated with a generic biotinylated antihuman capture reagent, (2) "on-bead" digestion with IdeS and/or PNGase F, and (3) reduction of interchain disulfide bonds to generate ∼25 kDa ADC subfragments, which are finally analyzed by LC-HRMS on a TOF mass spectrometer. The advantages of this method are that it can be performed using commercially available generic reagents and requires sample preparation time of less than 7 h. Furthermore, by reducing the size of intact ADC (∼150 kDa) to subfragments (∼25 kDa), the identification of conjugated payload and its metabolites can be achieved with excellent sensitivity and resolution (hydrolysis and other small mass change metabolites). This method was successfully applied to evaluate the in vitro and in vivo biotransformation of ADCs conjugated at different sites (LC, HC-Fab, and HC-Fc) with various classes of payload-linkers.


Asunto(s)
Biotransformación , Inmunoconjugados/sangre , Inmunoconjugados/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Cromatografía Liquida , Humanos , Espectrometría de Masas
4.
J Clin Invest ; 129(12): 5553-5567, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31710313

RESUMEN

Immune checkpoint inhibitors (ICIs), although promising, have variable benefit in head and neck cancer (HNC). We noted that tumor galectin-1 (Gal1) levels were inversely correlated with treatment response and survival in patients with HNC who were treated with ICIs. Using multiple HNC mouse models, we show that tumor-secreted Gal1 mediates immune evasion by preventing T cell migration into the tumor. Mechanistically, Gal1 reprograms the tumor endothelium to upregulate cell-surface programmed death ligand 1 (PD-L1) and galectin-9. Using genetic and pharmacological approaches, we show that Gal1 blockade increases intratumoral T cell infiltration, leading to a better response to anti-PD1 therapy with or without radiotherapy. Our study reveals the function of Gal1 in transforming the tumor endothelium into an immune-suppressive barrier and that its inhibition synergizes with ICIs.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Endotelio/fisiología , Galectina 1/fisiología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígeno B7-H1/fisiología , Femenino , Galectina 1/antagonistas & inhibidores , Galectinas/fisiología , Neoplasias de Cabeza y Cuello/inmunología , Humanos , Tolerancia Inmunológica , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Factor de Transcripción STAT1/fisiología
5.
Bioorg Med Chem Lett ; 29(3): 466-470, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30579797

RESUMEN

Uncialamycin analogs were evaluated as potential cytotoxic agents in an antibody-drug conjugate (ADC) approach to treating human cancer. These analogs were synthesized using Hauser annulations of substituted phthalides as a key step. A highly potent uncialamycin analog 3c with a valine-citrulline dipeptide linker was conjugated to an anti-mesothelin monoclonal antibody (mAb) through lysines to generate a meso-13 conjugate. This conjugate demonstrated subnanomolar potency (IC50 = 0.88 nM, H226 cell line) in in vitro cytotoxicity experiments with good immunological specificity to mesothelin-positive lung cancer cell lines. The potency and mechanism of action of this uncialamycin class of enediyne antitumor antibiotics make them attractive payloads in ADC-based cancer therapy.


Asunto(s)
Antraquinonas/farmacología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Inmunoconjugados/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Antraquinonas/química , Anticuerpos Monoclonales/química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inmunoconjugados/química , Neoplasias Pulmonares/patología , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
6.
Clin Cancer Res ; 24(20): 5178-5189, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30021910

RESUMEN

Purpose: The ganglioside fucosyl-GM1 (FucGM1) is a tumor-associated antigen expressed in a large percentage of human small cell lung cancer (SCLC) tumors, but absent in most normal adult tissues, making it a promising target in immuno-oncology. This study was undertaken to evaluate the preclinical efficacy of BMS-986012, a novel, nonfucosylated, fully human IgG1 antibody that binds specifically to FucGM1.Experimental Design: The antitumor activity of BMS-986012 was evaluated in in vitro assays using SCLC cells and in mouse xenograft and syngeneic tumor models, with and without chemotherapeutic agents and checkpoint inhibitors.Results: BMS-986012 showed a high binding affinity for FcγRIIIa (CD16), which resulted in enhanced antibody-dependent cellular cytotoxicity (ADCC) against FucGM1-expressing tumor cell lines. BMS-986012-mediated tumor cell killing was also observed in complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) assays. In several mouse SCLC models, BMS-986012 demonstrated efficacy and was well tolerated. In the DMS79 xenograft model, tumor regression was achieved with BMS-986012 doses of 0.3 mg/kg and greater; antitumor activity was enhanced when BMS-986012 was combined with standard-of-care cisplatin or etoposide. In a syngeneic model, tumors derived from a genetically engineered model of SCLC were treated with BMS-986012 or anti-FucGM1 with a mouse IgG2a Fc and their responses evaluated; when BMS-986012 was combined with anti-PD-1 or anti-CD137 antibody, therapeutic responses significantly improved.Conclusions: Single-agent BMS-986012 demonstrated robust antitumor activity, with the addition of chemotherapeutic or immunomodulatory agents further inhibiting SCLC growth in the same models. These preclinical data supported evaluation of BMS-986012 in a phase I clinical trial of patients with relapsed, refractory SCLC. Clin Cancer Res; 24(20); 5178-89. ©2018 AACR.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Gangliósido G(M1)/análogos & derivados , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos de Neoplasias/inmunología , Carcinoma de Células Pequeñas/tratamiento farmacológico , Carcinoma de Células Pequeñas/metabolismo , Carcinoma de Células Pequeñas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Gangliósido G(M1)/antagonistas & inhibidores , Gangliósido G(M1)/inmunología , Gangliósido G(M1)/metabolismo , Humanos , Inmunohistoquímica , Inmunomodulación/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Unión Proteica , Receptores de IgG/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Proc Natl Acad Sci U S A ; 114(21): E4223-E4232, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28484017

RESUMEN

Rational modulation of the immune response with biologics represents one of the most promising and active areas for the realization of new therapeutic strategies. In particular, the use of function blocking monoclonal antibodies targeting checkpoint inhibitors such as CTLA-4 and PD-1 have proven to be highly effective for the systemic activation of the human immune system to treat a wide range of cancers. Ipilimumab is a fully human antibody targeting CTLA-4 that received FDA approval for the treatment of metastatic melanoma in 2011. Ipilimumab is the first-in-class immunotherapeutic for blockade of CTLA-4 and significantly benefits overall survival of patients with metastatic melanoma. Understanding the chemical and physical determinants recognized by these mAbs provides direct insight into the mechanisms of pathway blockade, the organization of the antigen-antibody complexes at the cell surface, and opportunities to further engineer affinity and selectivity. Here, we report the 3.0 Å resolution X-ray crystal structure of the complex formed by ipilimumab with its human CTLA-4 target. This structure reveals that ipilimumab contacts the front ß-sheet of CTLA-4 and intersects with the CTLA-4:Β7 recognition surface, indicating that direct steric overlap between ipilimumab and the B7 ligands is a major mechanistic contributor to ipilimumab function. The crystallographically observed binding interface was confirmed by a comprehensive cell-based binding assay against a library of CTLA-4 mutants and by direct biochemical approaches. This structure also highlights determinants responsible for the selectivity exhibited by ipilimumab toward CTLA-4 relative to the homologous and functionally related CD28.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Antineoplásicos Inmunológicos/farmacología , Sitios de Unión de Anticuerpos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Ipilimumab/farmacología , Melanoma/tratamiento farmacológico , Factores Biológicos/farmacología , Antígeno CTLA-4/inmunología , Línea Celular , Cristalografía por Rayos X , Células HEK293 , Humanos , Inmunoterapia/métodos , Unión Proteica , Estructura Terciaria de Proteína
8.
Bioanalysis ; 8(6): 519-31, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26915587

RESUMEN

BACKGROUND: The bioanalytical strategy for antibody-drug conjugates (ADC) includes multiple integrated measurements of pharmacologically relevant ADC. METHODS & RESULTS: Three ligand-binding assays were validated for the measurement of total antibody, active ADC and total ADC. Accuracy and precision demonstrate %bias from -8 to 14%, %CV from 3 to 11% and total error from 3 to 21%, with >98% samples meeting incurred sample reanalysis criteria. Each assay met stability, selectivity, dilutional integrity, carry over and specificity criteria with no interference from associated metabolite/impurity. Given the active ADC assay sensitivity to payload, active ADC was used to assess drug to antibody ratio. DISCUSSION & CONCLUSION: Implementation of a microfluidic automated platform enabled high throughput sample analysis of multiple analytes with minimal sample processing.


Asunto(s)
Inmunoensayo , Inmunoconjugados/análisis , Anticuerpos Monoclonales/química , Semivida , Inmunoensayo/normas , Inmunoconjugados/farmacocinética , Lignanos , Preparaciones Farmacéuticas/química , Control de Calidad
9.
Biopharm Drug Dispos ; 37(2): 93-106, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25869904

RESUMEN

CD70 is a tumor necrosis factor (TNF)-like type II integral membrane protein that is transiently expressed on activated T- and B-lymphocytes. Aberrant expression of CD70 was identified in both solid tumors and haematologic malignancies. BMS-936561 (αCD70_MED-A) is an antibody-drug conjugate composed of a fully human anti-CD70 monoclonal antibody (αCD70) conjugated with a duocarmycin derivative, MED-A, through a maleimide-containing citrulline-valine dipeptide linker. MED-A is a carbamate prodrug that is activated by carboxylesterase to its active form, MED-B, to exert its DNA alkylation activity. In vitro serum stability studies suggested the efficiencies of hydrolyzing the carbamate-protecting group in αCD70_MED-A followed a rank order of mouse>rat > >monkey>dog~human. Pharmacokinetics of αCD70_MED-A was evaluated in mice, monkeys, and dogs after single intravenous doses. In mice, αCD70_MED-A was cleared rapidly, with no detectable exposures after 15 min following dosing. In contrast, αCD70_MED-A was much more stable in monkeys and dogs. The clearance of αCD70_MED-A in monkeys was 58 mL/d/kg, ~2-fold faster than that in dogs (31 mL/d/kg). The human PK profiles of the total αCD70 and αCD70_MED-A were predicted using allometrically scaled monkeys PK parameters of αCD70 and the carbamate hydrolysis rate constant estimated in dogs. Comparing the predicted and observed human PK from the phase I study, the dose-normalized concentration-time profiles of αCD70_MED-A and the total αCD70 were largely within the 5(th)-95(th) percentile of the predicted profiles.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos Alquilantes/farmacocinética , Ligando CD27/antagonistas & inhibidores , Inmunoconjugados/farmacocinética , Indoles/farmacocinética , Profármacos/farmacocinética , Animales , Anticuerpos Monoclonales/sangre , Antineoplásicos Alquilantes/sangre , Ligando CD27/inmunología , Perros , Humanos , Inmunoconjugados/sangre , Indoles/sangre , Macaca fascicularis , Ratones Endogámicos BALB C , Modelos Biológicos
10.
Artículo en Inglés | MEDLINE | ID: mdl-26310897

RESUMEN

Antibody drug conjugates (ADCs) are complex molecules composed of two pharmacologically distinct components, the cytotoxic payload and the antibody. The measurement of the payload molecules that are attached to the antibody in vivo is important for the evaluation of the safety and efficacy of ADCs, and can also provide distinct information compared to the antibody-related analytes. However, analyzing the antibody-conjugated payload is challenging and in some cases may not be feasible. The in vivo change in drug antibody ratio (DAR), due to deconjugation, biotransformation or other clearance phenomena, generates unique and additional challenges for ADC analysis in biological samples. Here, we report a novel hybrid approach with immuno-capture of the ADC, payload cleavage by specific enzyme, and LC-MS/MS of the cleaved payload to quantitatively measure the concentration of payload molecules still attached to the antibody via linker in plasma. The ADC reference material used for the calibration curve is not likely to be identical to the ADC measured in study samples due to the change in DAR distribution over the PK time course. The assay clearly demonstrated that there was no bias in the measurement of antibody-conjugated payload for ADC with varying DAR, which thus allowed accurate quantification even when the DAR distribution dynamically changes in vivo. This hybrid assay was fully validated based on a combination of requirements for both chromatographic and ligand binding methods, and was successfully applied to support a GLP safety study in monkeys.


Asunto(s)
Cromatografía Liquida/métodos , Haplorrinos/sangre , Inmunoconjugados/sangre , Espectrometría de Masas en Tándem/métodos , Animales
12.
Bioanalysis ; 7(13): 1569-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26226308

RESUMEN

BACKGROUND: The bioanalytical strategy for antibody-drug conjugates (ADC) includes numerous measurements integrally designed to provide comprehensive characterization of PK, PD and immunogenicity. This manuscript describes the utilization of reagents specifically tailored to an ADC with a microtubule polymerization inhibitor payload and cathepsin B cleavable linker. METHODS: The PK strategy includes the evaluation of physiological levels of total antibody, active ADC, total ADC, antibody-conjugated payload and unconjugated payload. These data are evaluated in the context of target and antidrug antibody levels to elucidate bioactive ADC. RESULTS & CONCLUSION: Herein, we discuss how this strategy has been applied and present our preliminary observations. Continuously evolving to meet pipeline demands, the integrated bioanalytical data will provide critical insights into the exposure-response relationship.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoconjugados/inmunología , Anticuerpos Monoclonales/química , Humanos , Inmunoconjugados/química
14.
PLoS One ; 10(5): e0125614, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25933020

RESUMEN

In Alzheimer's disease (AD), an extensive accumulation of extracellular amyloid plaques and intraneuronal tau tangles, along with neuronal loss, is evident in distinct brain regions. Staging of tau pathology by postmortem analysis of AD subjects suggests a sequence of initiation and subsequent spread of neurofibrillary tau tangles along defined brain anatomical pathways. Further, the severity of cognitive deficits correlates with the degree and extent of tau pathology. In this study, we demonstrate that phospho-tau (p-tau) antibodies, PHF6 and PHF13, can prevent the induction of tau pathology in primary neuron cultures. The impact of passive immunotherapy on the formation and spread of tau pathology, as well as functional deficits, was subsequently evaluated with these antibodies in two distinct transgenic mouse tauopathy models. The rTg4510 transgenic mouse is characterized by inducible over-expression of P301L mutant tau, and exhibits robust age-dependent brain tau pathology. Systemic treatment with PHF6 and PHF13 from 3 to 6 months of age led to a significant decline in brain and CSF p-tau levels. In a second model, injection of preformed tau fibrils (PFFs) comprised of recombinant tau protein encompassing the microtubule-repeat domains into the cortex and hippocampus of young P301S mutant tau over-expressing mice (PS19) led to robust tau pathology on the ipsilateral side with evidence of spread to distant sites, including the contralateral hippocampus and bilateral entorhinal cortex 4 weeks post-injection. Systemic treatment with PHF13 led to a significant decline in the spread of tau pathology in this model. The reduction in tau species after p-tau antibody treatment was associated with an improvement in novel-object recognition memory test in both models. These studies provide evidence supporting the use of tau immunotherapy as a potential treatment option for AD and other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Anticuerpos Monoclonales/farmacología , Trastornos del Conocimiento/terapia , Inmunización Pasiva , Fosfoproteínas/farmacología , Proteínas tau/antagonistas & inhibidores , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/inmunología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Regulación de la Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/patología , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/patología , Cultivo Primario de Células , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Transducción de Señal , Resultado del Tratamiento , Proteínas tau/genética , Proteínas tau/inmunología
15.
Biotechnol Appl Biochem ; 53(Pt 1): 51-61, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18680480

RESUMEN

Biologically active membrane proteins are difficult to isolate. Very often the isolated membrane proteins have low binding affinity or no biological integrity at all. Despite some success in isolation, one has to overcome the hurdles of obtaining sufficient quantity of the proteins and maintaining biological activity upon coating them on surfaces for developing an ELISA. Thus an alternative approach may be useful. The present study describes a quantification assay method for a therapeutic hmAb-1 (human monoclonal antibody-1) that recognizes a cell-surface protein employing an anti-ID (anti-idiotypic antibody) to hmAb-1 as a surrogate antigen in an immunoassay format using surface- plasmon-resonance technology. This assay is applicable for quantification of hmAb-1 in process streams, final drug-product quality control, as well as low-concentration drug substances in intravenous-solution bags. The surrogate nature of the anti-ID was confirmed by demonstrating that the anti-ID displaced the interaction between the hmAb-1 and its membrane antigen in a FACS titration test. The assay format involves first capturing hmAb-1 on the flow-cell surface, which then binds quantitatively to anti-ID in a mixture of increasing quantity of hmAb-1 in solution. An inverse dose-response relation between this anti-ID bound signal (or resonance units) and hmAb-1 concentration was established. The dose-response range of the calibration curve for hmAb-1 was between 20 and 300 ng/ml. The precision, accuracy and specificity of the assay are reported in the present paper.


Asunto(s)
Anticuerpos Antiidiotipos/inmunología , Anticuerpos Monoclonales/análisis , Inmunoensayo/métodos , Proteínas de la Membrana/inmunología , Resonancia por Plasmón de Superficie , Animales , Anticuerpos Antiidiotipos/análisis , Anticuerpos Monoclonales/inmunología , Células CHO , Calibración , Cricetinae , Cricetulus , Relación Dosis-Respuesta Inmunológica , Citometría de Flujo , Humanos , Proteínas Inmovilizadas/inmunología , Inmunoglobulina G/inmunología , Proteínas de la Membrana/análisis , Sensibilidad y Especificidad
16.
Nat Biotechnol ; 23(9): 1159-69, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16127450

RESUMEN

The tubular gland of the chicken oviduct is an attractive system for protein expression as large quantities of proteins are deposited in the egg, the production of eggs is easily scalable and good manufacturing practices for therapeutics from eggs have been established. Here we examined the ability of upstream and downstream DNA sequences of ovalbumin, a protein produced exclusively in very high quantities in chicken egg white, to drive tissue-specific expression of human mAb in chicken eggs. To accommodate these large regulatory regions, we established and transfected lines of chicken embryonic stem (cES) cells and formed chimeras that express mAb from cES cell-derived tubular gland cells. Eggs from high-grade chimeras contained up to 3 mg of mAb that possesses enhanced antibody-dependent cellular cytotoxicity (ADCC), nonantigenic glycosylation, acceptable half-life, excellent antigen recognition and good rates of internalization.


Asunto(s)
Anticuerpos Monoclonales/química , Animales , Southern Blotting , Western Blotting , Células CHO , Rastreo Diferencial de Calorimetría , Carbohidratos/química , Pollos , Cricetinae , ADN/metabolismo , Clara de Huevo , Embrión de Mamíferos/citología , Embrión no Mamífero , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos , Genoma , Glicosilación , Humanos , Inmunoglobulina G , Inmunohistoquímica , Focalización Isoeléctrica , Ratones , Ratones Endogámicos BALB C , Modelos Genéticos , Monosacáridos/química , Oligosacáridos/química , Ovalbúmina/genética , Ovalbúmina/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Ionización de Electrospray , Células Madre/citología
17.
J Biol Chem ; 278(35): 33142-9, 2003 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12815048

RESUMEN

A single candidate 4'-phosphopantetheine transferase, identified by BLAST searches of the human genome sequence data base, has been cloned, expressed, and characterized. The human enzyme, which is expressed mainly in the cytosolic compartment in a wide range of tissues, is a 329-residue, monomeric protein. The enzyme is capable of transferring the 4'-phosphopantetheine moiety of coenzyme A to a conserved serine residue in both the acyl carrier protein domain of the human cytosolic multifunctional fatty acid synthase and the acyl carrier protein associated independently with human mitochondria. The human 4'-phosphopantetheine transferase is also capable of phosphopantetheinylation of peptidyl carrier and acyl carrier proteins from prokaryotes. The same human protein also has recently been implicated in phosphopantetheinylation of the alpha-aminoadipate semialdehyde dehydrogenase involved in lysine catabolism (Praphanphoj, V., Sacksteder, K. A., Gould, S. J., Thomas, G. H., and Geraghty, M. T. (2001) Mol. Genet. Metab. 72, 336-342). Thus, in contrast to yeast, which utilizes separate 4'-phosphopantetheine transferases to service each of three different carrier protein substrates, humans appear to utilize a single, broad specificity enzyme for all posttranslational 4'-phosphopantetheinylation reactions.


Asunto(s)
Transferasas (Grupos de Otros Fosfatos Sustitutos)/química , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatografía en Gel , Clonación Molecular , Coenzima A/metabolismo , Citosol/enzimología , Citosol/metabolismo , Cartilla de ADN/química , ADN Complementario/metabolismo , Bases de Datos Genéticas , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Ácido Graso Sintasas/metabolismo , Humanos , Insectos , Cinética , Lisina/metabolismo , Mitocondrias/metabolismo , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Serina/química , Especificidad por Sustrato , Factores de Tiempo , Distribución Tisular , Transferasas (Grupos de Otros Fosfatos Sustitutos)/biosíntesis
18.
Chem Biol ; 10(2): 169-73, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12618189

RESUMEN

Animal fatty acid synthases are large polypeptides containing seven functional domains that are active only in the dimeric form. Inactivity of the monomeric form has long been attributed to the obligatory participation of domains from both subunits in catalysis of substrate loading and condensation reactions. However, we have engineered a fatty acid synthase containing one wild-type subunit and one subunit compromised by mutations in all seven functional domains that is active in fatty acid synthesis. This finding indicates that a single subunit, in the context of a dimer, is able to catalyze the entire biosynthetic pathway and suggests that, in the natural complex, each of the two subunits forms a scaffold that optimizes the conformation of the companion subunit.


Asunto(s)
Acetona/análogos & derivados , Ácido Graso Sintasas/síntesis química , Acetona/química , Animales , Western Blotting , Catálisis , Reactivos de Enlaces Cruzados , ADN Complementario/biosíntesis , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Ácido Graso Sintasas/química , Ácido Graso Sintasas/genética , Calor , Indicadores y Reactivos , Conformación Molecular , Mutación/genética , Ingeniería de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA