Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 17(2): 507-516, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31841002

RESUMEN

Targeted strategies to deliver and retain drugs to kidneys are needed to improve drug accumulation and efficacy in a myriad of kidney diseases. These drug delivery systems show potential for improving the therapeutic windows of drugs acting in the kidney. Biodistribution of antibody-based therapeutics in vivo is governed by several factors including binding affinity, size, and valency. Investigations of how the biophysical and biochemical properties of biologics enable them to overcome biological barriers and reach kidneys are therefore of interest. Although renal accumulation of antibody fragments in cancer diagnostics and treatment has been observed, reports on effective delivery of antibody fragments to the kidneys remain scarce. Previously, we demonstrated that targeting plasmalemma vesicle-associated protein (PV1), a caveolae-associated protein, can promote accumulation of antibodies in both the lungs and the kidneys. Here, by fine-tuning the binding affinity of an antibody toward PV1, we observe that the anti-PV1 antibody with reduced binding affinity lost the capability for kidney targeting while retaining the lung targeting activity, suggesting that binding affinity is a critical factor for kidney targeting of the anti-PV1 antibody. We next use the antibody fragment F(ab')2 targeting PV1 to assess the dual effects of rapid kidney filtration and PV1 targeting on kidney-selective targeting. Ex vivo fluorescence imaging results demonstrated that after rapidly accumulating in kidneys at 4 h, PV1-targeted F(ab')2 was continually retained in the kidney at 24 h, whereas the isotype control F(ab')2 underwent urinary elimination with significantly reduced signaling in the kidney. Confocal imaging studies confirmed the localization of PV1-targeted F(ab')2 in the kidney. In addition, the monovalent antibody fragment (Fab-C4) lost the capability for kidney homing, indicating that the binding avidity of anti-PV1 F(ab')2 is important for kidney targeting. Our findings suggest that PV1-targeted F(ab')2 might be useful as a drug carrier for renal targeting and highlight the importance of affinity optimization for tissue targeting antibodies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Caveolas/metabolismo , Portadores de Fármacos/farmacocinética , Fragmentos Fab de Inmunoglobulinas/inmunología , Riñón/efectos de los fármacos , Proteínas de la Membrana/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Afinidad de Anticuerpos , Portadores de Fármacos/administración & dosificación , Femenino , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Riñón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
2.
Biomacromolecules ; 21(9): 3596-3607, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32786528

RESUMEN

Synthetic gene delivery systems employ multiple functions to enable safe and effective transport of DNA to target cells. Here, we describe metabolite-based poly(l-lysine) (PLL) modifiers that improve transfection by imparting both pH buffering and nanoparticle stabilization functions within a single molecular unit. PLL modifiers were based on morpholine (M), morpholine and niacin (MN), or thiomorpholine (TM). PLL modification with (MN) or (TM) imparted buffering function over the pH range of 5-7 both in solution and live cells and enhanced the stability of PLL DNA nanoparticles, which exhibited higher resistance to polyanion exchange and prolonged blood circulation. These properties translated into increased transfection efficiency in vitro coupled with reduced toxicity compared to unmodified PLL and PLL(M). Furthermore, PEG-PLL(MN) DNA nanoparticles transfected muscle tissue in vivo for >45 days following intramuscular injection. These polymer modifiers demonstrate the successful design of multifunctional units that improve transfection of synthetic gene delivery systems while maintaining biocompatibility.


Asunto(s)
Técnicas de Transferencia de Gen , Polilisina , ADN/genética , Terapia Genética , Polietilenglicoles , Transfección
3.
J Biol Chem ; 293(22): 8439-8448, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669810

RESUMEN

Pathways of human epidermal growth factor (EGF) receptors are activated upon ligand-dependent or -independent homo- or heterodimerization and their subsequent transphosphorylation. Overexpression of these receptors positively correlates with transphosphorylation rates and increased tumor growth rates. MEDI4276, an anti-human epidermal growth factor receptor 2 (HER2) biparatopic antibody-drug conjugate, has two paratopes within each antibody arm. One, 39S, is aiming at the HER2 site involved in receptor dimerization and the second, single chain fragment (scFv), mimicking trastuzumab. Here we present the cocrystal structure of the 39S Fab-HER2 complex and, along with biophysical and functional assays, determine the corresponding epitope of MEDI4276 and its underlying mechanism of action. Our results reveal that MEDI4276's uniqueness is based first on the ability of its 39S paratope to block HER2 homo- or heterodimerization and second on its ability to cluster the receptors on the surface of receptor-overexpressing cells.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Multimerización de Proteína , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Secuencia de Aminoácidos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cristalografía por Rayos X , Femenino , Humanos , Modelos Moleculares , Fosforilación , Conformación Proteica , Homología de Secuencia , Células Tumorales Cultivadas
4.
Bioconjug Chem ; 30(9): 2340-2348, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31380623

RESUMEN

The normal electron-demand Diels-Alder (DA) cycloaddition is a classic transformation routinely used in synthesis; however, applications in biological systems are limited. Here, we report a spiro[2.4]hepta-4,6-diene-containing noncanonical amino acid (SCpHK) capable of efficient incorporation into antibodies and subsequent coupling with maleimide via a DA reaction. SCpHK was stable throughout protein expression in mammalian cells and enabled covalent attachment of maleimide drug-linkers yielding DA antibody-drug conjugates (DA-ADCs) with nearly quantitative conversion in a one-step process. The uncatalyzed DA reaction between SCpHK and maleimide in aqueous buffer was rapid (1.8-5.4 M-1 s-1), and the antibody-drug adduct was stable in rat serum for at least 1 week at 37 °C. Anti-EphA2 DA-ADCs containing AZ1508 or SG3249 maleimide drug-linkers were potent inhibitors of tumor growth in PC3 tumor models in vivo. The DA bioconjugation strategy described here represents a simple method to produce site-specific and stable ADCs with maleimide drug-linkers.


Asunto(s)
Inmunoconjugados/química , Maleimidas/química , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Cricetulus , Reacción de Cicloadición , Humanos , Inmunoconjugados/farmacología , Modelos Moleculares , Células PC-3 , Conformación Proteica , Compuestos de Espiro/química
5.
Bioconjug Chem ; 30(4): 1232-1243, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30912649

RESUMEN

Despite some clinical success with antibody-drug conjugates (ADCs) in patients with solid tumors and hematological malignancies, improvements in ADC design are still desirable due to the narrow therapeutic window of these compounds. Tumor-targeting antibody fragments have distinct advantages over monoclonal antibodies, including more rapid tumor accumulation and enhanced penetration, but are subject to rapid clearance. Half-life extension technologies such as PEGylation and albumin-binding domains (ABDs) have been widely used to improve the pharmacokinetics of many different types of biologics. PEGylation improves pharmacokinetics by increasing hydrodynamic size to reduce renal clearance, whereas ABDs extend half-life via FcRn-mediated recycling. In this study, we used an anti-oncofetal antigen 5T4 diabody conjugated with a highly potent cytotoxic pyrrolobenzodiazepine (PBD) warhead to assess and compare the effects of PEGylation and albumin binding on the in vivo efficacy of antibody fragment drug conjugates. Conjugation of 2× PEG20K to a diabody improved half-life from 40 min to 33 h, and an ABD-diabody fusion protein exhibited a half-life of 45 h in mice. In a xenograft model of breast cancer MDA-MB-436, the ABD-diabody-PBD showed greater tumor growth suppression and better tolerability than either PEG-diabody-PBD or diabody-PBD. These results suggest that the mechanism of half-life extension is an important consideration for designing cytotoxic antitumor agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Ensayo de Inmunoadsorción Enzimática , Femenino , Semivida , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Ratones , Ratones Desnudos , Polietilenglicoles/química , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Angew Chem Int Ed Engl ; 58(25): 8489-8493, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31018033

RESUMEN

Here, we describe a diene-containing noncanonical amino acid (ncAA) capable of undergoing fast and selective normal electron-demand Diels-Alder (DA) reactions following its incorporation into antibodies. A cyclopentadiene derivative of lysine (CpHK) served as the reactive handle for DA transformations and the substrate for genetic incorporation. CpHK incorporated into antibodies with high efficiency and was available for maleimide conjugation or self-reaction depending on position in the amino acid sequence. CpHK at position K274 reacted with the maleimide drug-linker AZ1508 at a rate of ≈79 m-1 s-1 to produce functional antibody-drug conjugates (ADCs) in a one-step process. Incorporation of CpHK at position S239 resulted in dimerization, which covalently linked antibody heavy chains together. The diene ncAA described here is capable of producing therapeutic protein conjugates with clinically validated and widely available maleimide compounds, while also enabling proximity-based stapling through a DA dimerization reaction.


Asunto(s)
Alcadienos/química , Aminoácidos/química , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Maleimidas/química , Reacción de Cicloadición , Dimerización , Humanos , Modelos Moleculares , Estructura Molecular
7.
J Biol Chem ; 292(10): 4361-4370, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28100773

RESUMEN

Soluble ligands have commonly been targeted by antibody therapeutics for cancers and other diseases. Although monoclonal antibodies targeting such ligands can block their interactions with their cognate receptors, they can also significantly increase the half-life of their ligands by FcRn-mediated antibody recycling, thereby evading ligand renal clearance and requiring increasingly high antibody doses to neutralize the increasing pool of target. To overcome this issue, we generated a bispecific/biparatopic antibody (BiSAb) that targets two different epitopes on IL-6 to block IL-6-mediated signaling. The BiSAb formed large immune complexes with IL-6 that can bind Fcγ receptors on phagocytic cells and are rapidly internalized. In addition, rapid clearance of the BiSAb·IL-6 complex was observed in mice while the parental antibodies prolonged the serum half-life of IL-6. Intravital imaging of the liver in mice confirmed that the rapid clearance of these large immune complexes was associated with Fcγ receptor-dependent binding to Kupffer cells in the liver. The approach described here provides a general strategy for therapeutic antibodies with the ability to not only neutralize but also actively drive clearance of their soluble antigens.


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/metabolismo , Complejo Antígeno-Anticuerpo/inmunología , Interleucina-6/antagonistas & inhibidores , Receptores de IgG/metabolismo , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/inmunología , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Interleucina-6/inmunología , Macrófagos del Hígado/citología , Macrófagos del Hígado/metabolismo , Hígado/citología , Hígado/metabolismo , Ratones , Unión Proteica , Receptores de IgG/inmunología
8.
Bioconjug Chem ; 29(7): 2406-2414, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29932647

RESUMEN

The thiol-maleimide linkage is widely used for antibody-drug conjugate (ADC) production; however, conjugation of maleimide-drugs could be improved by simplified procedures and reliable conjugate stability. Here, we report the evaluation of electron-rich and cyclic dienes that can be appended to antibodies and reacted with maleimide-containing drugs through the Diels-Alder (DA) reaction. Drug conjugation is fast and quantitative due to reaction acceleration in water, and the linkage is more stable in serum than in the corresponding thiol-maleimide adduct with the same drug. ADCs produced using the DA reaction (DAADCs) are effective in vitro and in vivo, demonstrating the utility of this reaction in producing effective biotherapeutics. Given the large number of commercially available maleimide compounds, this conjugation approach could be readily applied to the production of a wide range of antibody (or protein) conjugates.


Asunto(s)
Reacción de Cicloadición/métodos , Inmunoconjugados/química , Maleimidas/química , Alquenos , Anticuerpos/química , Reactivos de Enlaces Cruzados/química , Estabilidad de Medicamentos , Maleimidas/uso terapéutico , Preparaciones Farmacéuticas/química
9.
Bioorg Med Chem Lett ; 28(23-24): 3617-3621, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30389292

RESUMEN

Codelivery of multiple therapeutic agents with different anticancer mechanisms can overcome drug resistance as well as generate additive or synergistic anticancer effects that may enhance the antitumor efficacy. Antibody-drug conjugates (ADCs) can be used for highly specific delivery of multiple therapeutic agents with different anticancer mechanisms, though more research is required towards designing flexible platforms on which dual drug ADCs could be prepared. Herein, we describe the synthesis of a heterotrifunctional linker that could be used to construct flexible platforms for preparing dual-cytotoxic drug conjugates in a site-specific manner. As a proof of concept, we synthesized dual drug ADCs carrying monomethyl auristain E (MMAE, tubulin polymerization inhibitor) and pyrrolobenzodiazepine dimer (PBD, DNA minor groove alkylator). We then evaluated the dual drug ADCs for in vitro efficacy and confirmed the dual mechanism of action.


Asunto(s)
Inmunoconjugados/química , Moduladores de Tubulina/química , Aminobenzoatos/química , Anticuerpos Monoclonales/química , Antineoplásicos Alquilantes/química , Benzodiazepinas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Química Clic , Humanos , Inmunoconjugados/farmacología , Oligopéptidos/química , Pirroles/química
10.
J Proteome Res ; 16(10): 3672-3687, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28876938

RESUMEN

Chinese hamster ovary cells represent the dominant host for therapeutic recombinant protein production. However, few large-scale data sets have been generated to characterize this host organism and derived CHO cell lines at the proteomics level. Consequently, an extensive label-free quantitative proteomics analysis of two cell lines (CHO-S and CHO DG44) and two Chinese hamster tissues (liver and ovary) was used to identify a total of 11 801 unique proteins containing at least two unique peptides. 9359 unique proteins were identified specifically in the cell lines, representing a 56% increase over previous work. Additionally, 6663 unique proteins were identified across liver and ovary tissues, providing the first Chinese hamster tissue proteome. Protein expression was more conserved within cell lines during both growth phases than across cell lines, suggesting large genetic differences across cell lines. Overall, both gene ontology and KEGG pathway analysis revealed enrichment of cell-cycle activity in cells. In contrast, upregulated molecular functions in tissue include glycosylation and lipid transporter activity. Furthermore, cellular components including Golgi apparatus are upregulated in both tissues. In conclusion, this large-scale proteomics analysis enables us to delineate specific changes between tissues and cells derived from these tissues, which can help explain specific tissue function and the adaptations cells incur for applications in biopharmaceutical productions.


Asunto(s)
Células CHO/metabolismo , Proteoma/genética , Proteómica , Proteínas Recombinantes/genética , Animales , Cricetinae , Cricetulus/genética , Cricetulus/metabolismo , Proteínas Recombinantes/metabolismo , Espectrometría de Masas en Tándem
11.
Mol Pharm ; 14(5): 1501-1516, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28245132

RESUMEN

Antibody-drug conjugates (ADCs) are a class of biopharmaceuticals that combine the specificity of antibodies with the high-potency of cytotoxic drugs. Engineering cysteine residues in the antibodies using mutagenesis is a common method to prepare site-specific ADCs. With this approach, solvent accessible amino acids in the antibody have been selected for substitution with cysteine for conjugating maleimide-bearing cytotoxic drugs, resulting in homogeneous and stable site-specific ADCs. Here we describe a cysteine engineering approach based on the insertion of cysteines before and after selected sites in the antibody, which can be used for site-specific preparation of ADCs. Cysteine-inserted antibodies have expression level and monomeric content similar to the native antibodies. Conjugation to a pyrrolobenzodiazepine dimer (SG3249) resulted in comparable efficiency of site-specific conjugation between cysteine-inserted and cysteine-substituted antibodies. Cysteine-inserted ADCs were shown to have biophysical properties, FcRn, and antigen binding affinity similar to the cysteine-substituted ADCs. These ADCs were comparable for serum stability to the ADCs prepared using cysteine-mutagenesis and had selective and potent cytotoxicity against human prostate cancer cells. Two of the cysteine-inserted variants abolish binding of the resulting ADCs to FcγRs in vitro, thereby potentially preventing non-target mediated uptake of the ADCs by cells of the innate immune system that express FcγRs, which may result in mitigating off-target toxicities. A selected cysteine-inserted ADC demonstrated potent dose-dependent anti-tumor activity in a xenograph tumor mouse model of human breast adenocarcinoma expressing the oncofetal antigen 5T4.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Cisteína/química , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Desnudos , Trastuzumab/química , Trastuzumab/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Biol Chem ; 290(24): 14979-85, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25925951

RESUMEN

We report the three-dimensional structure of human interferon α-2A (IFN-α2A) bound to the Fab fragment of a therapeutic monoclonal antibody (sifalimumab; IgG1/κ). The structure of the corresponding complex was solved at a resolution of 3.0 Å using molecular replacement and constitutes the first reported structure of a human type I IFN bound to a therapeutic antibody. This study revealed the major contribution made by the first complementarity-determining region in each of sifalimumab light and heavy chains. These data also provided the molecular basis for sifalimumab mechanism of action. We propose that its interferon-neutralizing properties are the result of direct competition for IFN-α2A binding to the IFN receptor subunit 1 (IFNAR1) and do not involve inhibiting IFN-α2A binding to the IFN receptor subunit 2 (IFNAR2).


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Interferón-alfa/efectos de los fármacos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Cristalografía por Rayos X , Interferón alfa-2 , Interferón-alfa/inmunología , Modelos Moleculares , Conformación Proteica , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/inmunología
13.
Biomacromolecules ; 17(5): 1818-33, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27007881

RESUMEN

Targeted nanomedicines are a promising technology for treatment of disease; however, preparation and characterization of well-defined protein-nanoparticle systems remain challenging. Here, we describe a platform technology to prepare antibody binding fragment (Fab)-bearing nanoparticles and an accompanying real-time cell-based assay to determine their cellular uptake compared to monoclonal antibodies (mAbs) and Fabs. The nanoparticle platform was composed of core-cross-linked polyion complex (PIC) micelles prepared from azide-functionalized PEG-b-poly(amino acids), that is, azido-PEG-b-poly(l-lysine) [N3-PEG-b-PLL] and azido-PEG-b-poly(aspartic acid) [N3-PEG-b-PAsp]. These PIC micelles were 30 nm in size and contained approximately 10 polymers per construct. Fabs were derived from an antibody binding the EphA2 receptor expressed on cancer cells and further engineered to contain a reactive cysteine for site-specific attachment and a cleavable His tag for purification from cell culture expression systems. Azide-functionalized micelles and thiol-containing Fab were linked using a heterobifunctional cross-linker (FPM-PEG4-DBCO) that contained a fluorophenyl-maleimide for stable conjugation to Fabs thiols and a strained alkyne (DBCO) group for coupling to micelle azide groups. Analysis of Fab-PIC micelle conjugates by fluorescence correlation spectroscopy, size exclusion chromatography, and UV-vis absorbance determined that each nanoparticle contained 2-3 Fabs. Evaluation of cellular uptake in receptor positive cancer cells by real-time fluorescence microscopy revealed that targeted Fab-PIC micelles achieved higher cell uptake than mAbs and Fabs, demonstrating the utility of this approach to identify targeted nanoparticle constructs with unique cellular internalization properties.


Asunto(s)
Anticuerpos Monoclonales/química , Reactivos de Enlaces Cruzados/química , Fragmentos Fab de Inmunoglobulinas/química , Nanopartículas/química , Polímeros/química , Neoplasias de la Próstata/metabolismo , Receptor EphA2/metabolismo , Anticuerpos Monoclonales/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Masculino , Micelas , Polímeros/metabolismo , Células Tumorales Cultivadas
14.
J Immunol ; 192(4): 1480-90, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24442430

RESUMEN

B cell activation is regulated by a variety of signals. CD19 positively regulates B cell activation, augmenting signals delivered through the BCR complex. In contrast, CD32b contains an ITIM and negatively regulates BCR signaling. Importantly, there are drugs currently in clinical trials and preclinical development that cross-link CD32b to molecules within the BCR complex. We wanted to address how single engagement versus cotargeting these molecules affects human B cell function. When B cells from healthy individuals were activated by signals that mimic a T cell response (IL-21 costimulation), ligation of CD32b, but not CD19, inhibited B cell expansion and plasma cell (PC) differentiation. In contrast, when B cells were activated through TLR, anti-CD19, but not anti-CD32b, blunted the response. However, when both CD19 and CD32b were coengaged by a bispecific anti-CD19×CD32b Ab, both types of stimuli were potently inhibited. Cross-linking CD19 with CD32b also inhibited Ab-independent functions of B cells, such as HLA upregulation, cytokine production, and the ability of B cells to prime CD4(+) T cells. Finally, although cross-linking CD19 and CD32b inhibited PC differentiation of primary B cells, it did not alter Ig production from pre-established PCs. These data elucidate the mechanism by which a complex set of signals determines the fate of B cell responsiveness. Although signals through CD19 influence TLR-driven activation, CD32b impacts the magnitude of the response following IL-21 costimulation. Therefore, simultaneous targeting of multiple surface molecules may be a necessary approach to comprehensively modulate B cell activation in vivo.


Asunto(s)
Antígenos CD19/metabolismo , Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Células Plasmáticas/metabolismo , Receptores de IgG/metabolismo , Anticuerpos/inmunología , Antígenos CD19/biosíntesis , Antígenos CD19/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Muerte Celular/inmunología , Diferenciación Celular , Células Cultivadas , Reactivos de Enlaces Cruzados , Humanos , Memoria Inmunológica/inmunología , Interleucinas/metabolismo , Unión Proteica/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de IgG/biosíntesis , Receptores de IgG/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo
15.
J Biol Chem ; 289(11): 7812-24, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24469444

RESUMEN

We report the three-dimensional structure of human neonatal Fc receptor (FcRn) bound concurrently to its two known ligands. More particularly, we solved the crystal structure of the complex between human FcRn, wild-type human serum albumin (HSA), and a human Fc engineered for improved pharmacokinetics properties (Fc-YTE). The crystal structure of human FcRn bound to wild-type HSA alone is also presented. HSA domain III exhibits an extensive interface of contact with FcRn, whereas domain I plays a lesser role. A molecular explanation for the HSA recycling mechanism is provided with the identification of FcRn His(161) as the only potential direct contributor to the corresponding pH-dependent process. At last, this study also allows an accurate structural definition of residues considered for decades as important to the human IgG/FcRn interaction and reveals Fc His(310) as a significant contributor to pH-dependent binding. Finally, we explain various structural mechanisms by which several Fc mutations (including YTE) result in increased human IgG binding to FcRn. Our study provides an unprecedented relevant understanding of the molecular basis of human Fc interaction with human FcRn.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/química , Inmunoglobulina G/química , Receptores Fc/química , Albúmina Sérica/química , Cristalización , Cristalografía por Rayos X , Células HEK293 , Humanos , Enlace de Hidrógeno , Concentración de Iones de Hidrógeno , Fragmentos Fc de Inmunoglobulinas/química , Ligandos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química
16.
J Biol Chem ; 289(43): 29874-80, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25210036

RESUMEN

MEDI4893 is a neutralizing human monoclonal antibody that targets α-toxin (AT) and is currently undergoing evaluation in the field of Staphylococcus aureus-mediated diseases. We have solved the crystal structure of MEDI4893 Fab bound to monomeric AT at a resolution of 2.56 Å and further characterized its epitope using various engineered AT variants. We have found that MEDI4893 recognizes a novel epitope in the so-called "rim" domain of AT and exerts its neutralizing effect through a dual mechanism. In particular, MEDI4893 not only sterically blocks binding of AT to its cell receptor but also prevents it from adopting a lytic heptameric trans-membrane conformation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Pruebas de Neutralización , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales Humanizados , Anticuerpos Neutralizantes/química , Anticuerpos ampliamente neutralizantes , Línea Celular , Cristalografía por Rayos X , Mapeo Epitopo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Modelos Moleculares , Unión Proteica , Proteínas Recombinantes/química , Resonancia por Plasmón de Superficie
17.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2354-61, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26527150

RESUMEN

The three-dimensional structure of a human IgG1 Fc fragment bound to wild-type human FcγRI is reported. The structure of the corresponding complex was solved at a resolution of 2.4 Šusing molecular replacement; this is the highest resolution achieved for an unmutated FcγRI molecule. This study highlights the critical structural and functional role played by the second extracellular subdomain of FcγRI. It also explains the long-known major energetic contribution of the Fc `LLGG' motif at positions 234-237, and particularly of Leu235, via a `lock-and-key' mechanism. Finally, a previously held belief is corrected and a differing view is offered on the recently proposed direct role of Fc carbohydrates in the corresponding interaction. Structural evidence is provided that such glycan-related effects are strictly indirect.


Asunto(s)
Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Receptores de IgG/química , Receptores de IgG/metabolismo , Línea Celular , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Polisacáridos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
18.
Bioconjug Chem ; 26(10): 2085-96, 2015 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-26340339

RESUMEN

Antibody-drug conjugates (ADCs) have emerged as an important class of therapeutics for cancer treatment that combine the target specificity of antibodies with the killing activity of anticancer chemotherapeutics. Early conjugation technologies relied upon random conjugation to either lysine or cysteine residues, resulting in heterogeneous ADCs. Recent technology advancements have resulted in the preparation of homogeneous ADCs through the site-specific conjugation at engineered cysteines, glycosylated amino acids, and bioorthogonal unnatural amino acids. Here we describe for the first time the conjugation of an anti-mitotic drug to an antibody following the mild and selective oxidation of a serine residue engineered at the N-terminus of the light chain. Using an alkoxyamine-derivatized monomethyl auristatine E payload, we have prepared a hydrolytically stable ADC that retains binding to its antigen and displays potent in vitro cytotoxicity and in vivo tumor growth inhibition.


Asunto(s)
Anticuerpos/química , Anticuerpos/farmacología , Ingeniería de Proteínas/métodos , Animales , Anticuerpos/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Humanos , Hidrólisis , Ratones Desnudos , Oximas/química , Estabilidad Proteica , Ratas , Receptor EphA2/inmunología , Receptor EphA2/metabolismo , Serina/química , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Pharm ; 12(9): 3490-501, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26176328

RESUMEN

Targeting Eph (erythropoietin producing hepatoma) receptors with monoclonal antibodies is being explored as therapy for several types of cancer. To test whether simultaneous targeting of EphA2, EphA4, and EphB4 would be an effective approach to cancer therapy, we generated a recombinant trispecific antibody using the variable domain genes of anti-EphA2, anti-EphA4, and anti-EphB4 monoclonal antibodies. A multidisciplinary approach combining biochemical, biophysical, and cellular-based assays was used to characterize the trispecific antibody in vitro and in vivo. Here we demonstrate that the trispecific antibody is expressed at high levels by mammalian cells, monodispersed in solution, thermostable, capable of simultaneously binding the three receptors, and able to activate the three targets effectively as evidenced by receptor internalization and degradation both in vitro and in vivo. Furthermore, pharmacokinetic analysis using tumor-bearing nude mice showed that the trispecific antibody remains in the circulation similarly to its respective parental antibodies. These results indicate that simultaneous blockade of EphA2, EphA4, and EphB4 could be an attractive approach to cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos/inmunología , Diseño de Fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Receptor EphA2/antagonistas & inhibidores , Receptor EphA4/antagonistas & inhibidores , Receptor EphB4/antagonistas & inhibidores , Animales , Rastreo Diferencial de Calorimetría , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Receptor EphA2/inmunología , Receptor EphA4/inmunología , Receptor EphB4/inmunología , Resonancia por Plasmón de Superficie , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Artículo en Inglés | MEDLINE | ID: mdl-23519809

RESUMEN

Staphylococcus aureus alpha toxin (AT) has been crystallized in complex with the Fab fragment of a human antibody (MEDI4893). This constitutes the first reported crystals of AT bound to an antibody. The monoclinic crystals belonged to space group P21, with unit-cell parameters a=85.52, b=148.50, c=93.82 Å, ß=99.82°. The diffraction of the crystals extended to 2.56 Šresolution. The asymmetric unit contained two MEDI4893 Fab-AT complexes. This corresponds to a crystal volume per protein weight (VM) of 2.3 Å3 Da(-1) and a solvent content of 47%. The three-dimensional structure of this complex will contribute to an understanding of the molecular basis of the interaction of MEDI4893 with AT. It will also shed light on the mechanism of action of this antibody, the current evaluation of which in the field of S. aureus-mediated diseases makes it a particularly interesting case study. Finally, this study will provide the three-dimensional structure of AT in a monomeric state for the first time.


Asunto(s)
Anticuerpos Antiidiotipos/química , Anticuerpos Monoclonales/química , Toxinas Bacterianas/química , Fragmentos Fab de Inmunoglobulinas/química , Staphylococcus aureus/química , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Monoclonales/inmunología , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/aislamiento & purificación , Cristalización , Cristalografía por Rayos X , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA