Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(5): 1723-1732, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30559189

RESUMEN

Lipoprotein lipase (LPL) is responsible for the intravascular processing of triglyceride-rich lipoproteins. The LPL within capillaries is bound to GPIHBP1, an endothelial cell protein with a three-fingered LU domain and an N-terminal intrinsically disordered acidic domain. Loss-of-function mutations in LPL or GPIHBP1 cause severe hypertriglyceridemia (chylomicronemia), but structures for LPL and GPIHBP1 have remained elusive. Inspired by our recent discovery that GPIHBP1's acidic domain preserves LPL structure and activity, we crystallized an LPL-GPIHBP1 complex and solved its structure. GPIHBP1's LU domain binds to LPL's C-terminal domain, largely by hydrophobic interactions. Analysis of electrostatic surfaces revealed that LPL contains a large basic patch spanning its N- and C-terminal domains. GPIHBP1's acidic domain was not defined in the electron density map but was positioned to interact with LPL's large basic patch, providing a likely explanation for how GPIHBP1 stabilizes LPL. The LPL-GPIHBP1 structure provides insights into mutations causing chylomicronemia.


Asunto(s)
Lipoproteína Lipasa/metabolismo , Plasma/metabolismo , Receptores de Lipoproteína/metabolismo , Triglicéridos/sangre , Triglicéridos/metabolismo , Animales , Células CHO , Capilares/metabolismo , Línea Celular , Cricetulus , Cristalografía por Rayos X/métodos , Células Endoteliales/metabolismo , Humanos , Hidrólisis , Hipertrigliceridemia/metabolismo
2.
Protein Expr Purif ; 177: 105752, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32949707

RESUMEN

Alpha galactosidase (a-Gal) is an acidic hydrolase that plays a critical role in hydrolyzing the terminal alpha-galactoyl moiety from glycolipids and glycoproteins. There are over a hundred mutations reported for the GLA gene that encodes a-Gal that result in reduced protein synthesis, protein instability, and reduction in function. The deficiencies of a-Gal can cause Fabry disease, a rare lysosomal storage disorder (LSD) caused by the failure to catabolize alpha-d-galactoyl glycolipid moieties. The current standard of care for Fabry disease is enzyme replacement therapy (ERT) where the purified recombinant form of human a-Gal is given to patients. The manufacture of a-Gal is currently performed utilizing traditional large-scale chromatography processes. Developing an affinity resin for the purification of a-Gal would reduce the complexity of the manufacturing process, reduce costs, and potentially produce a higher quality a-Gal. After the evaluation of many small molecules, a commercially available small molecule biomimetic, N-5-Carboxypentyl-1-deoxygalactonojirimycin (N5C-DGJ), was utilized for the development of a novel small molecule biomimetic affinity resin for a-Gal. Affinity purified a-Gal demonstrated a purity greater than 90%, exhibited expected thermal stability and specific activity. Complementing this affinity purification is the development of an elution buffer system that confers an increased thermal stability to a-Gal. The N5C-DGJ affinity resin tolerated sodium hydroxide sanitization with no loss of binding capacity, making it amenable to large scale purification processes and potential use in manufacturing. This novel method for purifying the challenging a-Gal enzyme can be extended to other enzyme replacement therapies.


Asunto(s)
Cromatografía de Afinidad/métodos , Clonación Molecular/métodos , Galactosa/química , Iminopiranosas/química , Animales , Células CHO , Cricetulus , Estabilidad de Enzimas , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo
3.
J Pharmacol Exp Ther ; 366(2): 291-302, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29752426

RESUMEN

Follistatin (FS) is an important regulatory protein, a natural antagonist for transforming growth factor-ß family members activin and myostatin. The diverse biologic roles of the activin and myostatin signaling pathways make FS a promising therapeutic target for treating human diseases exhibiting inflammation, fibrosis, and muscle disorders, such as Duchenne muscular dystrophy. However, rapid heparin-mediated hepatic clearance of FS limits its therapeutic potential. We targeted the heparin-binding loop of FS for site-directed mutagenesis to improve clearance parameters. By generating a series of FS variants with one, two, or three negative amino acid substitutions, we demonstrated a direct and proportional relationship between the degree of heparin-binding affinity in vitro and the exposure in vivo. The triple mutation K(76,81,82)E abolished heparin-binding affinity, resulting in ∼20-fold improved in vivo exposure. This triple mutant retains full functional activity and an antibody-like pharmacokinetic profile, and shows a superior developability profile in physical stability and cell productivity compared with FS variants, which substitute the entire heparin-binding loop with alternative sequences. Our surgical approach to mutagenesis should also reduce the immunogenicity risk. To further lower this risk, we introduced a novel glycosylation site into the heparin-binding loop. This hyperglycosylated variant showed a 10-fold improved exposure and decreased clearance in mice compared with an IgG1 Fc fusion protein containing the native FS sequence. Collectively, our data highlight the importance of improving pharmacokinetic properties by manipulating heparin-binding affinity and glycosylation content and provide a valuable guideline to design desirable therapeutic FS molecules.


Asunto(s)
Folistatina/genética , Folistatina/farmacocinética , Ingeniería de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Femenino , Folistatina/metabolismo , Folistatina/uso terapéutico , Glicosilación , Heparina/metabolismo , Humanos , Ratones , Mutación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Distribución Tisular
4.
Bioconjug Chem ; 25(3): 510-20, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24533768

RESUMEN

Antibody-drug conjugates (ADCs) have been proven clinically to be more effective anti-cancer agents than native antibodies. However, the classical conjugation chemistries to prepare ADCs by targeting primary amines or hinge disulfides have a number of shortcomings including heterogeneous product profiles and linkage instability. We have developed a novel site-specific conjugation method by targeting the native glycosylation site on antibodies as an approach to address these limitations. The native glycans on Asn-297 of antibodies were enzymatically remodeled in vitro using galactosyl and sialyltransferases to introduce terminal sialic acids. Periodate oxidation of these sialic acids yielded aldehyde groups which were subsequently used to conjugate aminooxy functionalized cytotoxic agents via oxime ligation. The process has been successfully demonstrated with three antibodies including trastuzumab and two cytotoxic agents. Hydrophobic interaction chromatography and LC-MS analyses revealed the incorporation of ~1.6 cytotoxic agents per antibody molecule, approximating the number of sialic acid residues. These glyco-conjugated ADCs exhibited target-dependent antiproliferative activity toward antigen-positive tumor cells and significantly greater antitumor efficacy than naked antibody in a Her2-positive tumor xenograft model. These findings suggest that enzymatic remodeling combined with oxime ligation of the native glycans of antibodies offers an attractive approach to generate ADCs with well-defined product profiles. The site-specific conjugation approach presented here provides a viable alternative to other methods, which involve a need to either re-engineer the antibody sequence or develop a highly controlled chemical process to ensure reproducible drug loading.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos/química , Antineoplásicos/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glicosilación , Humanos , Ratones , Ratones SCID , Estructura Molecular , Neoplasias Experimentales/patología , Polisacáridos/química , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Sialiltransferasas/química , Sialiltransferasas/metabolismo , Relación Estructura-Actividad , Trastuzumab
5.
Bioconjug Chem ; 24(3): 408-18, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23350694

RESUMEN

Recombinant human thyroid stimulating hormone (rhTSH or Thyrogen) has been approved for thyroid cancer diagnostics and treatment under a multidose regimen due to its short circulating half-life. To reduce dosing frequency, PEGylation strategies were explored to increase the duration of action of rhTSH. Lysine and N-terminal PEGylation resulted in heterogeneous product profiles with 40% or lower reaction yields of monoPEGylated products. Eleven cysteine mutants were designed based on a structure model of the TSH-TSH receptor (TSHR) complex to create unique conjugation sites on both α and ß subunits for site-specific conjugation. Sequential screening of mutant expression level, oligomerization tendency, and conjugation efficiency resulted in the identification of the αG22C rhTSH mutant for stable expression and scale-up PEGylation. The introduced cysteine in the αG22C rhTSH mutant was partially blocked when isolated from conditioned media and could only be effectively PEGylated after mild reduction with cysteine. This produced a higher reaction yield, ~85%, for the monoPEGylated product. Although the mutation had no effect on receptor binding, PEGylation of αG22C rhTSH led to a PEG size-dependent decrease in receptor binding. Nevertheless, the 40 kDa PEG αG22C rhTSH showed a prolonged duration of action compared to rhTSH in a rat pharmacodynamics model. Reverse-phase HPLC and N-terminal sequencing experiments confirmed site-specific modification at the engineered Cys 22 position on the α-subunit. This work is another demonstration of successful PEGylation of a cysteine-knot protein by an engineered cysteine mutation.


Asunto(s)
Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Tirotropina/administración & dosificación , Tirotropina/química , Secuencia de Aminoácidos , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Tirotropina/genética , Factores de Tiempo
6.
Bioconjug Chem ; 24(12): 2025-35, 2013 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-24161263

RESUMEN

The cation-independent mannose 6-phosphate receptor (CI-MPR) plays a critical role in intracellular transport of lysosomal enzymes as well as the uptake of recombinant proteins. To define the minimal glycan structure determinants necessary for receptor binding and cellular uptake, we synthesized a series of glycans containing mono-, di-, tri-, tetra-, and hexamannoses terminated with either one or two phosphates for conjugating to a model protein, recombinant human acid α-glucosidase. A high affinity interaction with the CI-MPR can be achieved for the enzyme conjugated to a dimannose glycan with a single phosphate. However, tightest binding to a CI-MPR affinity column was observed with a hexamannose structure containing two phosphates. Moreover, maximal cellular uptake and a 5-fold improvement in in vivo potency were achieved when the bisphosphorylated hexamannose glycan is conjugated to the protein by a ß linker. Nevertheless, even a monophosphorylated dimannose glycan conjugate showed stronger binding to the receptor affinity column, higher cellular uptake, and significantly greater in vivo efficacy compared to the unconjugated protein which contains a low level of high affinity glycan structure. These results demonstrate that the phosphorylated dimannose moiety appears to be the minimal structure determinant for enhanced CI-MPR binding and that the orientation of the glycan is critical for maximum receptor interaction. In summary, we have improved the understanding of the mechanism of CI-MPR binding and developed a simple alternative for CI-MPR targeting.


Asunto(s)
Polisacáridos/química , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/metabolismo , alfa-Glucosidasas/metabolismo , Animales , Humanos , Espacio Intracelular/metabolismo , Manosa/química , Mioblastos/citología , Polisacáridos/metabolismo , Unión Proteica , Transporte de Proteínas , Ratas
7.
J Biol Chem ; 286(1): 299-308, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20980263

RESUMEN

Gaucher disease is caused by mutations in the enzyme acid ß-glucosidase (GCase), the most common of which is the substitution of serine for asparagine at residue 370 (N370S). To characterize the nature of this mutation, we expressed human N370S GCase in insect cells and compared the x-ray structure and biochemical properties of the purified protein with that of the recombinant human GCase (imiglucerase, Cerezyme®). The x-ray structure of N370S mutant acid ß-glucosidase at acidic and neutral pH values indicates that the overall folding of the N370S mutant is identical to that of recombinant GCase. Subtle differences were observed in the conformation of a flexible loop at the active site and in the hydrogen bonding ability of aromatic residues on this loop with residue 370 and the catalytic residues Glu-235 and Glu-340. Circular dichroism spectroscopy showed a pH-dependent change in the environment of tryptophan residues in imiglucerase that is absent in N370S GCase. The mutant protein was catalytically deficient with reduced V(max) and increased K(m) values for the substrate p-nitrophenyl-ß-D-glucopyranoside and reduced sensitivity to competitive inhibitors. N370S GCase was more stable to thermal denaturation and had an increased lysosomal half-life compared with imiglucerase following uptake into macrophages. The competitive inhibitor N-(n-nonyl)deoxynojirimycin increased lysosomal levels of both N370S and imiglucerase 2-3-fold by reducing lysosomal degradation. Overall, these data indicate that the N370S mutation results in a normally folded but less flexible protein with reduced catalytic activity compared with imiglucerase.


Asunto(s)
Glucosilceramidasa/química , Glucosilceramidasa/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Animales , Fenómenos Biofísicos , Rastreo Diferencial de Calorimetría , Dominio Catalítico , Línea Celular , Dicroismo Circular , Cristalografía por Rayos X , Estabilidad de Enzimas , Glucosilceramidasa/genética , Semivida , Humanos , Concentración de Iones de Hidrógeno , Espacio Intracelular/enzimología , Modelos Moleculares , Proteínas Mutantes/genética , Ratas
8.
Bioconjug Chem ; 23(12): 2354-64, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23176598

RESUMEN

Vascular endothelial growth factor (VEGF) neutralizing antagonists including antibodies or receptor extracellular domain Fc fusions have been applied clinically to control angiogenesis in cancer, wet age-related macular degeneration, and edema. We report here the generation of high-affinity VEGF-binding domains by chemical linkage of the second domain of the VEGF receptor Flt-1 (D2) in several configurations. Recombinant D2 was expressed with a 13 a.a. C-terminal tag, including a C-terminal cysteine to enable its dimerization by disulfide bond formation or by attachment to divalent PEGs and oligomerization by coupling to multivalent PEGs. Disulfide-linked dimers produced by Cu(2+) oxidation of the free-thiol form of the protein demonstrated picomolar affinity for VEGF in solution, comparable to that of a D2-Fc fusion (sFLT01) and ~50-fold higher than monomeric D2, suggesting the 26 a.a. tag length between the two D2 domains permits simultaneous interaction of both faces of the VEGF homodimer. Extending the separation between the D2 domains by short PEG spacers from 0.35 kD to 5 kD produced a modest ~2-fold increase in affinity over the disulfide, thus defining the optimal distance between the two D2 domains for maximum affinity. By surface plasmon resonance (SPR), a larger (~5-fold) increase in affinity was observed by conjugation of the D2 monomer to the termini of 4-arm PEG, and yielding a product with a larger hydrodynamic radius than sFLT01. The higher affinity displayed by these D2 PEG tetramers than either D2 dimer or sFLT01 was largely a consequence of a slower rate of dissociation, suggesting the simultaneous binding by these tetramers to neighboring surface-bound VEGF. Finally, disulfide-linked D2 dimers showed a greater resistance to autocatalytic fragmentation than sFLT01 under elevated temperature stress, indicating such minimum-sequence constructs may be better suited for sustained-release formulations. Therefore, these constructs represent novel Fc-independent VEGF antagonists with ultrahigh affinity, high stability, and a range of hydrodynamic radii for application to multiple therapeutic targets.


Asunto(s)
Polietilenglicoles/química , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química , Cobre/química , Cisteína/química , Dimerización , Disulfuros/química , Células HEK293 , Humanos , Cinética , Terapia Molecular Dirigida , Peso Molecular , Oxidación-Reducción , Conformación Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
9.
Blood ; 116(2): 270-9, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20194895

RESUMEN

A long-acting factor VIII (FVIII) as a replacement therapy for hemophilia A would significantly improve treatment options for patients with hemophilia A. To develop a FVIII with an extended circulating half-life, but without a reduction in activity, we have engineered 23 FVIII variants with introduced surface-exposed cysteines to which a polyethylene glycol (PEG) polymer was specifically conjugated. Screening of variant expression level, PEGylation yield, and functional assay identified several conjugates retaining full in vitro coagulation activity and von Willebrand factor (VWF) binding.PEGylated FVIII variants exhibited improved pharmacokinetics in hemophilic mice and rabbits. In addition, pharmacokinetic studies in VWF knockout mice indicated that larger molecular weight PEG may substitute for VWF in protecting PEGylated FVIII from clearance in vivo. In bleeding models of hemophilic mice, PEGylated FVIII not only exhibited prolonged efficacy that is consistent with the improved pharmacokinetics but also showed efficacy in stopping acute bleeds comparable with that of unmodified rFVIII. In summary site-specifically PEGylated FVIII has the potential to be a long-acting prophylactic treatment while being fully efficacious for on-demand treatment for patients with hemophilia A.


Asunto(s)
Coagulantes/farmacocinética , Factor VIII/farmacocinética , Hemofilia A/tratamiento farmacológico , Polietilenglicoles/farmacocinética , Animales , Western Blotting , Coagulantes/administración & dosificación , Coagulantes/química , Preparaciones de Acción Retardada , Electroforesis en Gel de Poliacrilamida , Factor VIII/administración & dosificación , Factor VIII/química , Semivida , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Conejos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
Bioconjug Chem ; 22(4): 741-51, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21417264

RESUMEN

Engineering proteins for selective tissue targeting can improve therapeutic efficacy and reduce undesired side effects. The relatively high dose of recombinant human acid α-glucosidase (rhGAA) required for enzyme replacement therapy of Pompe disease may be attributed to less than optimal muscle uptake via the cation-independent mannose 6-phosphate receptor (CI-MPR). To improve muscle targeting, Zhu et al. (1) conjugated periodate oxidized rhGAA with bis mannose 6-phosphate bearing synthetic glycans and achieved 5-fold greater potency in a murine Pompe efficacy model. In the current study, we systematically evaluated multiple strategies for conjugation based on a structural homology model of GAA. Glycan derivatives containing succinimide, hydrazide, and aminooxy linkers targeting free cysteine, lysines, and N-linked glycosylation sites on rhGAA were prepared and evaluated in vitro and in vivo. A novel conjugation method using enzymatic oxidation was developed to eliminate side oxidation of methionine. Conjugates derived from periodate oxidized rhGAA still displayed the greatest potency in the murine Pompe model. The efficiency of conjugation and its effect on catalytic activity were consistent with predictions based on the structural model and supported its use in guiding selection of appropriate chemistries.


Asunto(s)
Polisacáridos/química , Proteínas Recombinantes/metabolismo , alfa-Glucosidasas/metabolismo , Animales , Biocatálisis , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Estructura Molecular , Ácido N-Acetilneuramínico/química , Oxidación-Reducción , Polisacáridos/administración & dosificación , Polisacáridos/metabolismo , Ingeniería de Proteínas , Proteínas Recombinantes/química , alfa-Glucosidasas/administración & dosificación , alfa-Glucosidasas/química
11.
J Pept Sci ; 17(1): 47-55, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21171144

RESUMEN

α-melanocyte stimulating hormone (α-MSH) is a tridecapeptide fragment of pro-opiomelanocortin (POMC) with broad effects on appetite, skin pigmentation, hormonal regulation, and potential roles in both inflammation and autoimmunity. The use of this peptide as an anti-inflammatory agent is limited by its low selectivity between the melanocortin receptors, susceptibility to proteolytic degradation, and rapid clearance from circulation. A retro-inverso (RI) sequence of α-MSH was characterized for receptor activity and resistance to protease. This peptide demonstrated surprisingly high selectivity for binding the melanocortin receptor 1 (MC1R). However, RI-α-MSH exhibited a diminished binding affinity for MC1R compared to α-MSH. Mapping of the residues critical for agonist activity, receptor binding, and selectivity by alanine scanning, identified the same critical core tetrapeptide required for the native peptide. Modest improvements in affinity were obtained by conservative changes employing non-natural amino acids and substitution of the C-terminal sequence with a portion of a MC1R ligand peptide previously identified by phage display. Recombination of these elements yielded a peptide with an identical K(i) as α-MSH at MC1R and a lower EC(50) in Mel-624 melanoma cells. A number of other structural modifications of the RI peptide were found to differ in effect from those reported for the L-form α-MSH, suggesting a significantly altered interaction with the MC1R.


Asunto(s)
Receptor de Melanocortina Tipo 1/metabolismo , alfa-MSH/análogos & derivados , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Melanoma/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Datos de Secuencia Molecular , Receptor de Melanocortina Tipo 1/química , alfa-MSH/química , alfa-MSH/metabolismo
12.
Biotechnol Prog ; 37(1): e3061, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32748555

RESUMEN

Antibody-dependent cellular cytotoxicity (ADCC) is the primary mechanism of actions for several marketed therapeutic antibodies (mAbs) and for many more in clinical trials. The ADCC efficacy is highly dependent on the ability of therapeutic mAbs to recruit effector cells such as natural killer cells, which induce the apoptosis of targeted cells. The recruitment of effector cells by mAbs is negatively affected by fucose modification of N-Glycans on the Fc; thus, utilization of afucosylated mAbs has been a trend for enhanced ADCC therapeutics. Most of afucosylated mAbs in clinical or commercial manufacturing were produced from Fut8-/- Chinese hamster ovary cells (CHO) host cells, generally generating low yields compared to wildtype CHO host. This study details the generation and characterization of two engineered CHOZN® cell lines, in which the enzyme involved in guanosine diphosphate (GDP)-fucose synthesis, GDP mannose-4,6-dehydratase (Gmds) and GDP-L-fucose synthase (FX), was knocked out. The top host cell lines for each of the knockouts, FX-/- and Gmds-/-, were selected based on growth robustness, bulk MSX selection tolerance, production titer, fucosylation level, and cell stability. We tested the production of two proprietary IgG1 mAbs in the engineered host cells, and found that the titers were comparable to CHOZN® cells. The mAbs generated from either KO cell line exhibited loss of fucose modification, leading to significantly boosted FcγRIIIa binding and ADCC effects. Our data demonstrated that both FX-/- and Gmds-/- host cells could replace Fut8-/- CHO cells for clinical manufacturing of antibody therapeutics.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Carbohidrato Epimerasas/antagonistas & inhibidores , Fucosa/metabolismo , Guanosina Difosfato/metabolismo , Hidroliasas/antagonistas & inhibidores , Cetona Oxidorreductasas/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Secuencia de Bases , Células CHO , Sistemas CRISPR-Cas , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/metabolismo , Cricetinae , Cricetulus , Glicosilación , Humanos , Hidroliasas/genética , Hidroliasas/metabolismo , Inmunoglobulina G/inmunología , Cetona Oxidorreductasas/genética , Cetona Oxidorreductasas/metabolismo , Receptores de IgG/metabolismo
13.
MAbs ; 12(1): 1814583, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32892677

RESUMEN

Antibodies mediate effector functions through Fcγ receptor (FcγR) interactions and complement activation, causing cytokine release, degranulation, phagocytosis, and cell death. They are often undesired for development of therapeutic antibodies where only antigen binding or neutralization would be ideal. Effector elimination has been successful with extensive mutagenesis, but these approaches can potentially lead to manufacturability and immunogenicity issues. By switching the native glycosylation site from position 297 to 298, we created alternative antibody glycosylation variants in the receptor interaction interface as a novel strategy to eliminate the effector functions. The engineered glycosylation site at Asn298 was confirmed by SDS-PAGE, mass spectrometry, and X-ray crystallography (PDB code 6X3I). The lead NNAS mutant (S298N/T299A/Y300S) shows no detectable binding to mouse or human FcγRs by surface plasmon resonance analyses. The effector functions of the mutant are completely eliminated when measured in antibody-dependent cell-meditated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays. In vivo, the NNAS mutant made on an antibody against a human lymphocyte antigen does not deplete T cells or B cells in transgenic mice, in contrast to wild-type antibody. Structural study confirms the successful glycosylation switch to the engineered Asn298 site. The engineered glycosylation would clash with approaching FcγRs based on reported Fc-FcγR co-crystal structures. In addition, the NNAS mutants of multiple antibodies retain binding to antigens and neonatal Fc receptor, exhibit comparable purification yields and thermal stability, and display normal circulation half-life in mice and non-human primate. Our work provides a novel approach for generating therapeutic antibodies devoid of any ADCC and CDC activities with potentially lower immunogenicity.


Asunto(s)
Sustitución de Aminoácidos , Activación de Complemento , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad Clase I/inmunología , Fragmentos Fc de Inmunoglobulinas , Mutación Missense , Receptores Fc/inmunología , Animales , Células CHO , Cricetulus , Glicosilación , Células HEK293 , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Receptores Fc/genética
14.
MAbs ; 11(7): 1266-1275, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31199181

RESUMEN

Deamidation evaluation and mitigation is an important aspect of therapeutic antibody developability assessment. We investigated the structure and function of the Asn-Gly deamidation in a human anti-CD52 IgG1 antibody light chain complementarity-determining region 1, and risk mitigation through protein engineering. Antigen binding affinity was found to decrease about 400-fold when Asn33 was replaced with an Asp residue to mimic the deamidation product, suggesting significant impacts on antibody function. Other variants made at Asn33 (N33H, N33Q, N33H, N33R) were also found to result in significant loss of antigen binding affinity. The co-crystal structure of the antigen-binding fragment bound to a CD52 peptide mimetic was solved at 2.2Å (PDB code 6OBD), which revealed that Asn33 directly interacts with the CD52 phosphate group via a hydrogen bond. Gly34, but sits away from the binding interface, rendering it more amendable to mutagenesis without affecting affinity. Saturation mutants at Gly34 were prepared and subjected to forced deamidation by incubation at elevated pH and temperature. Three mutants (G34R, G34K and G34Q) showed increased resistance to deamidation by LC-MS peptide mapping, while maintaining high binding affinity to CD52 antigen measured by Biacore. A complement -dependent cytotoxicity assay indicated that these mutants function by triggering antibody effector function. This study illustrates the importance of structure-based design and extensive mutagenesis to mitigate antibody developability issues.


Asunto(s)
Anticuerpos Monoclonales/química , Antígeno CD52/química , Regiones Determinantes de Complementariedad/química , Inmunoglobulina G/química , Cadenas Ligeras de Inmunoglobulina/química , Amidas/química , Anticuerpos Monoclonales/genética , Citotoxicidad Celular Dependiente de Anticuerpos , Asparagina/genética , Bioingeniería , Antígeno CD52/genética , Antígeno CD52/inmunología , Regiones Determinantes de Complementariedad/genética , Cristalografía por Rayos X , Humanos , Inmunoglobulina G/genética , Cadenas Ligeras de Inmunoglobulina/genética , Mutagénesis Sitio-Dirigida , Mapeo Peptídico , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
15.
Respir Res ; 9: 65, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18811961

RESUMEN

BACKGROUND: Surfactant protein D (SP-D) plays an important role in innate defense against influenza A viruses (IAVs) and other pathogens. METHODS: We tested antiviral activities of recombinant human SP-D against a panel of IAV strains that vary in glycosylation sites on their hemagglutinin (HA). For these experiments a recombinant version of human SP-D of the Met11, Ala160 genotype was used after it was characterized biochemically and structurally. RESULTS: Oligosaccharides at amino acid 165 on the HA in the H3N2 subtype and 104 in the H1N1 subtype are absent in collectin-resistant strains developed in vitro and are important for mediating antiviral activity of SP-D; however, other glycans on the HA of these viral subtypes also are involved in inhibition by SP-D. H3N2 strains obtained shortly after introduction into the human population were largely resistant to SP-D, despite having the glycan at 165. H3N2 strains have become steadily more sensitive to SP-D over time in the human population, in association with addition of other glycans to the head region of the HA. In contrast, H1N1 strains were most sensitive in the 1970s-1980s and more recent strains have become less sensitive, despite retaining the glycan at 104. Two H5N1 strains were also resistant to inhibition by SP-D. By comparing sites of glycan attachment on sensitive vs. resistant strains, specific glycan sites on the head domain of the HA are implicated as important for inhibition by SP-D. Molecular modeling of the glycan attachment sites on HA and the carbohydrate recognition domain of SPD are consistent with these observations. CONCLUSION: Inhibition by SP-D correlates with presence of several glycan attachment sites on the HA. Pandemic and avian strains appear to lack susceptibility to SP-D and this could be a contributory factor to their virulence.


Asunto(s)
Hemaglutininas Virales/metabolismo , Virus de la Influenza A/fisiología , Proteína D Asociada a Surfactante Pulmonar/administración & dosificación , Proteína D Asociada a Surfactante Pulmonar/química , Inactivación de Virus/efectos de los fármacos , Antivirales/administración & dosificación , Antivirales/química , Glicosilación/efectos de los fármacos , Humanos , Virus de la Influenza A/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química
16.
Peptides ; 29(3): 479-86, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17942192

RESUMEN

Vasoactive intestinal peptide (VIP) binds to two receptors, VPAC1 and VPAC2. Non-selective VIP antagonists have been shown to inhibit human cancer cell proliferation and reduce tumor growth in mice. Many human cancers over-express VPAC1 but not VPAC2. We show that VPAC1-selective antagonists can inhibit human cancer cell proliferation and identify five positions in the VPAC1-selective antagonist PG 97-269 that may be responsible for VPAC1 selectivity. Position 16 appears to be particularly critical for selectivity, as demonstrated in the replacement of Arg16 of PG 97-269 with the native VIP amino acid; this single change results in greatly reduced VPAC1 binding and selectivity. Finally, we show that site-specific conjugation with a 22kDa polyethylene glycol (PEG) at the C-terminus of VPAC1-selective antagonists further improves VPAC1-selective binding and has minimal effect on antagonistic activity. Our studies have further solidified VPAC1 as a cancer target and offer the possibility of generating highly potent VPAC1-selective antagonists with minimal number of mutations to reduce the risk of immunogenicity and potentially prolonged duration of action to allow more efficient treatment regimen.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Polietilenglicoles/química , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Electroforesis en Gel de Poliacrilamida , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Unión Proteica/efectos de los fármacos , Radioinmunoensayo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Péptido Intestinal Vasoactivo/química , Péptido Intestinal Vasoactivo/farmacología
17.
Biotechnol Bioeng ; 99(3): 652-65, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17680659

RESUMEN

Glycosylation in the Fc region of antibodies has been shown to play an important role in antibody function. In the current study, glycosylation of human monoclonal antibodies was metabolically modulated using a potent alpha-mannosidase I inhibitor, kifunensine, resulting in the production of antibodies with oligomannose-type N-glycans. Growing Chinese hamster ovary cells for 11 days in batch culture with a single treatment of kifunensine was sufficient to elicit this effect without any significant impact on cell viability or antibody production. Antibodies expressed in the presence of kifunensine at a concentration as low as 60 ng/mL contained mainly oligomannose-type glycans and demonstrated increased ADCC activity and affinity for FcgammaRIIIA, but reduced C1q binding. Although the kifunensine-mediated shift to oligomannose-type glycans could, in theory, result in rapid clearance of the antibody through increased mannose receptor binding, the serum levels of antibody in mice were not significantly altered up to 168 h following injection. The use of kifunensine provides a simple and rapid method for the production of antibodies with increased ADCC without the time-consuming need to re-engineer either the antibody molecule or the host cell line.


Asunto(s)
Alcaloides/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Manosa/química , Manosa/inmunología , Ingeniería de Proteínas/métodos , Animales , Células CHO , Cricetinae , Cricetulus , Fucosa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones
18.
Int J Pharm ; 351(1-2): 1-7, 2008 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-17964744

RESUMEN

A pituitary adenylate cyclase-activating peptide (PACAP) analogue (HSDAVFTDNYTRLRKQVAAKKYLQSIKNKRY, P66) was formulated in several non-aqueous solvents in anticipation of improved shelf-life stability. However, the stability of this peptide in these solvents was found to be as poor as in an aqueous solution. The major degradation reaction in non-aqueous solvents was dimer formation. The proposed mechanism for dimerization was a nucleophilic attack of a basic amino acid on cyclic imide formed by dehydration or deamidation of Asp or Asn. Two approaches were found to be effective in stabilizing the peptide in non-aqueous solvents: (1) acidification of the peptide and (2) use of zinc chloride in the formulation. Stabilization could be attributed to reduction of the nucleophilicity of the reactive groups through protonation and metal-peptide interaction through chelation. The stabilization approaches are applicable only in a non-aqueous environment for this peptide, and possibly for other peptides with similar reactive moieties.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Solventes/química , Cloruros/química , Dimerización , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Concentración de Iones de Hidrógeno , Compuestos de Zinc/química
19.
J Endocrinol ; 192(2): 371-80, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17283237

RESUMEN

Type 2 diabetes is characterized by reduced insulin secretion from the pancreas and overproduction of glucose by the liver. Glucagon-like peptide-1 (GLP-1) promotes glucose-dependent insulin secretion from the pancreas, while glucagon promotes glucose output from the liver. Taking advantage of the homology between GLP-1 and glucagon, a GLP-1/glucagon hybrid peptide, dual-acting peptide for diabetes (DAPD), was identified with combined GLP-1 receptor agonist and glucagon receptor antagonist activity. To overcome its short plasma half-life DAPD was PEGylated, resulting in dramatically prolonged activity in vivo. PEGylated DAPD (PEG-DAPD) increases insulin and decreases glucose in a glucose tolerance test, evidence of GLP-1 receptor agonism. It also reduces blood glucose following a glucagon challenge and elevates fasting glucagon levels in mice, evidence of glucagon receptor antagonism. The PEG-DAPD effects on glucose tolerance are also observed in the presence of the GLP-1 antagonist peptide, exendin(9-39). An antidiabetic effect of PEG-DAPD is observed in db/db mice. Furthermore, PEGylation of DAPD eliminates the inhibition of gastrointestinal motility observed with GLP-1 and its analogues. Thus, PEG-DAPD has the potential to be developed as a novel dual-acting peptide to treat type 2 diabetes, with prolonged in vivo activity, and without the GI side-effects.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptidos/farmacología , Polietilenglicoles/farmacología , Animales , Glucemia/análisis , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Obesidad/sangre , Obesidad/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/metabolismo
20.
Mol Biotechnol ; 34(2): 165-78, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17172662

RESUMEN

Loss of coagulation factor VIII (FVIII) function results in a bleeding disorder, hemophilia A, which requires FVIII replacement therapy. Owing to its large size and complexity, the expression level of recombinant FVIII is two to three orders of magnitude lower than other recombinant proteins produced in mammalian cell lines. To understand cellular factors limiting FVIII expression, we studied the expression of FVIII in a human cell line, HKB11 (a hybrid cell line of HEK293 and a human B cell line). In comparison with other cell lines, such as HEK293 and BHK-21, HKB11 showed increased FVIII expression levels. With unamplified, pooled stable cells, FVIII expression in HKB11 cells was 8- to 30-fold higher than the other cell lines tested. In this study, HKB11 clones expressing varying levels of FVIII were analyzed and FVIII secreted from these clones had similar specific activity. Characterization of these clones by immunofluorescence staining, Western blotting analysis, and flow cytometry showed that high-producing cells not only secreted more active FVIII but also accumulated more FVIII protein intracellularly. FVIII expression appears to be controlled by the rates of transcription, translation, and secretion, but transcription and translation may play more important roles than secretion in determining expression level in HKB11 cells. FACS analysis of live cells showed that the high-producing clones also had more FVIII on the HKB11 cell surface than low-producing cells, thus opening the possibility of using FACS to select high-producing cell lines. Expression levels of the chaperone protein Hsp70 and antiapoptotic proteins such as Bcl-2 and Bcl-xL were similar among HKB11 clones with different FVIII productivity. In conclusion, HKB11 is an efficient host cell line for expression of FVIII and possibly other recombinant proteins. Systematic approaches, such as gene expression profiling by DNA microarray, will be necessary to understand the global changes in the cells producing recombinant proteins.


Asunto(s)
Biotecnología/métodos , Factor VIII/biosíntesis , Proteínas Recombinantes/biosíntesis , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Factor VIII/análisis , Factor VIII/genética , Citometría de Flujo , Proteínas HSP70 de Choque Térmico/análisis , Proteínas HSP70 de Choque Térmico/biosíntesis , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteína bcl-X/análisis , Proteína bcl-X/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA