Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 14(1): 2078466, 2022.
Article in English | MEDLINE | ID: mdl-35634725

ABSTRACT

Antibody-drug conjugates (ADCs) are increasingly powerful medicines for targeted cancer therapy. Inspired by the trend to further improve their therapeutic index by generation of homogenous ADCs, we report here how the clinical-stage GlycoConnect™ technology uses the globally conserved N-glycosylation site to generate stable and site-specific ADCs based on enzymatic remodeling and metal-free click chemistry. We demonstrate how an engineered endoglycosidase and a native glycosyl transferase enable highly efficient, one-pot glycan remodeling, incorporating a novel sugar substrate 6-azidoGalNAc. Metal-free click attachment of an array of cytotoxic payloads was highly optimized, in particular by inclusion of anionic surfactants. The therapeutic potential of GlycoConnect™, in combination with HydraSpace™ polar spacer technology, was compared to that of Kadcyla® (ado-trastuzumab emtansine), showing significantly improved efficacy and tolerability.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Ado-Trastuzumab Emtansine , Immunoconjugates/therapeutic use , Polysaccharides , Therapeutic Index
2.
Antibodies (Basel) ; 7(1)2018 Feb 20.
Article in English | MEDLINE | ID: mdl-31544864

ABSTRACT

Despite tremendous efforts in the field of targeted cancer therapy with antibody-drug conjugates (ADCs), attrition rates have been high. Historically, the priority in ADC development has been the selection of target, antibody, and toxin, with little focus on the nature of the linker. We show here that a short and polar sulfamide spacer (HydraSpace™, Oss, The Netherlands) positively impacts ADC properties in various ways: (a) efficiency of conjugation; (b) stability; and (c) therapeutic index. Different ADC formats are explored in terms of drug-to-antibody ratios (DAR2, DAR4) and we describe the generation of a DAR4 ADC by site-specific attachment of a bivalent linker-payload construct to a single conjugation site in the antibody. A head-to-head comparison of HydraSpace™-containing DAR4 ADCs to marketed drugs, derived from the same antibody and toxic payload components, indicated a significant improvement in both the efficacy and safety of several vivo models, corroborated by in-depth pharmacokinetic analysis. Taken together, HydraSpace™ technology based on a polar sulfamide spacer provides significant improvement in manufacturability, stability, and ADC design, and is a powerful platform to enable next-generation ADCs with enhanced therapeutic index.

4.
Bioconjug Chem ; 26(11): 2233-42, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26061183

ABSTRACT

A robust, generally applicable, nongenetic technology is presented to convert monoclonal antibodies into stable and homogeneous ADCs. Starting from a native (nonengineered) mAb, a chemoenzymatic protocol allows for the highly controlled attachment of any given payload to the N-glycan residing at asparagine-297, based on a two-stage process: first, enzymatic remodeling (trimming and tagging with azide), followed by ligation of the payload based on copper-free click chemistry. The technology, termed GlycoConnect, is applicable to any IgG isotype irrespective of glycosylation profile. Application to trastuzumab and maytansine, both components of the marketed ADC Kadcyla, demonstrate a favorable in vitro and in vivo efficacy for GlycoConnect ADC. Moreover, the superiority of the native glycan as attachment site was demonstrated by in vivo comparison to a range of trastuzumab-based glycosylation mutants. A side-by-side comparison of the copper-free click probes bicyclononyne (BCN) and a dibenzoannulated cyclooctyne (DBCO) showed a surprising difference in conjugation efficiency in favor of BCN, which could be even further enhanced by introduction of electron-withdrawing fluoride substitutions onto the azide. The resulting mAb-conjugates were in all cases found to be highly stable, which in combination with the demonstrated efficacy warrants ADCs with a superior therapeutic index.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoconjugates/chemistry , Polysaccharides/chemistry , Ado-Trastuzumab Emtansine , Antibodies, Monoclonal, Humanized/chemistry , Azides/chemistry , Carbohydrate Sequence , Click Chemistry , Glycosylation , Humans , Maytansine/analogs & derivatives , Maytansine/chemistry , Models, Molecular , Molecular Sequence Data , Protein Stability , Trastuzumab/chemistry
5.
Bioconjug Chem ; 23(3): 392-8, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22372991

ABSTRACT

The 1,3-dipolar cycloaddition of azides with ring-strained alkynes is one of the few bioorthogonal reactions suitable for specific biomolecule labeling in complex biological systems. Nevertheless, azide-independent labeling of proteins by strained alkynes can occur to a varying extent, thereby limiting the sensitivity of assays based on strain-promoted azide-alkyne cycloaddition (SPAAC). In this study, a subset of three cyclooctynes, dibenzocyclooctyne (DIBO), azadibenzocyclooctyne (DIBAC), and bicyclo[6.1.0]nonyne (BCN), was used to evaluate the azide-independent labeling of proteins in vitro. For all three cyclooctynes, we show that thiol-yne addition with reduced peptidylcysteines is responsible for most of the azide-independent polypeptide labeling. The identity of the reaction product was confirmed by LC-MS and NMR analysis. Moreover, we show that undesired thiol-yne reactions can be prevented by alkylating peptidylcysteine thiols with iodoacetamide (IAM). Since IAM is compatible with SPAAC, a more specific azide-dependent labeling is achieved by preincubating proteins containing reduced cysteines with IAM.


Subject(s)
Alkynes/chemistry , Azides/chemistry , Proteins/chemistry , Sulfhydryl Compounds/chemistry , Blotting, Western , Chromatography, Liquid , Cyclization , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry
6.
Amino Acids ; 43(3): 1251-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22180026

ABSTRACT

Transglutaminase 2 (TG2) is a Ca(2+)-dependent enzyme able to catalyze the formation of ε(γ-glutamyl)-lysine crosslinks between polypeptides, resulting in high molecular mass multimers. We have developed a bioorthogonal chemical method for the labeling of TG2 glutamine-donor proteins. As amine-donor substrates we used a set of azide- and alkyne-containing primary alkylamines that allow, after being crosslinked to glutamine-donor proteins, specific labeling of these proteins via the azide-alkyne cycloaddition. We demonstrate that these azide- and alkyne-functionalized TG2 substrates are cell permeable and suitable for specific labeling of TG2 glutamine-donor substrates in HeLa and Movas cells. Both the Cu(I)-catalyzed and strain promoted azide-alkyne cycloaddition proved applicable for subsequent derivatization of the TG2 substrate proteins with the desired probe. This new method for labeling TG2 substrate proteins introduces flexibility in the detection and/or purification of crosslinked proteins, allowing differential labeling of cellular proteins.


Subject(s)
Click Chemistry , GTP-Binding Proteins/chemistry , Transglutaminases/chemistry , Acylation , Alkynes/chemistry , Amines/chemistry , Amino Acid Sequence , Animals , Azides/chemistry , Biocatalysis , Biotin/analogs & derivatives , Biotin/chemistry , Cadaverine/metabolism , Cell Membrane/metabolism , Cross-Linking Reagents/chemistry , Cycloaddition Reaction , Enzyme Inhibitors/chemistry , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , GTP-Binding Proteins/antagonists & inhibitors , HSP20 Heat-Shock Proteins/chemistry , HeLa Cells , Humans , Mice , Peptide Fragments/chemistry , Permeability , Protein Glutamine gamma Glutamyltransferase 2 , Staining and Labeling , Transglutaminases/antagonists & inhibitors
7.
PLoS One ; 6(8): e23067, 2011.
Article in English | MEDLINE | ID: mdl-21901120

ABSTRACT

While inward remodeling of small arteries in response to low blood flow, hypertension, and chronic vasoconstriction depends on type 2 transglutaminase (TG2), the mechanisms of action have remained unresolved. We studied the regulation of TG2 activity, its (sub) cellular localization, substrates, and its specific mode of action during small artery inward remodeling. We found that inward remodeling of isolated mouse mesenteric arteries by exogenous TG2 required the presence of a reducing agent. The effect of TG2 depended on its cross-linking activity, as indicated by the lack of effect of mutant TG2. The cell-permeable reducing agent DTT, but not the cell-impermeable reducing agent TCEP, induced translocation of endogenous TG2 and high membrane-bound transglutaminase activity. This coincided with inward remodeling, characterized by a stiffening of the artery. The remodeling could be inhibited by a TG2 inhibitor and by the nitric oxide donor, SNAP. Using a pull-down assay and mass spectrometry, 21 proteins were identified as TG2 cross-linking substrates, including fibronectin, collagen and nidogen. Inward remodeling induced by low blood flow was associated with the upregulation of several anti-oxidant proteins, notably glutathione-S-transferase, and selenoprotein P. In conclusion, these results show that a reduced state induces smooth muscle membrane-bound TG2 activity. Inward remodeling results from the cross-linking of vicinal matrix proteins, causing a stiffening of the arterial wall.


Subject(s)
Arteries/drug effects , Arteries/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/pharmacology , Transglutaminases/metabolism , Transglutaminases/pharmacology , Animals , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Cell Line , Enzyme Activation/drug effects , GTP-Binding Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins , Reducing Agents/metabolism , Transglutaminases/genetics
8.
PLoS One ; 5(4): e10158, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405018

ABSTRACT

BACKGROUND: The aging related decline of heat shock factor-1 (HSF1) signaling may be causally related to protein aggregation diseases. To model such disease, we tried to cripple HSF1 signaling in the Xenopus tadpole. RESULTS: Over-expression of heat shock factor binding protein-1 did not inhibit the heat shock response in Xenopus. RNAi against HSF1 mRNA inhibited the heat shock response by 70% in Xenopus A6 cells, but failed in transgenic tadpoles. Expression of XHSF380, a dominant-negative HSF1 mutant, was embryonic lethal, which could be circumvented by delaying expression via a tetracycline inducible promoter. HSF1 signaling is thus essential for embryonic Xenopus development. Surprisingly, transgenic expression of the XHSF380 or of full length HSF1, whether driven by a ubiquitous or a neural specific promoter, was not detectable in the larval brain. CONCLUSIONS: Our finding that the majority of neurons, which have little endogenous HSF1, refused to accept transgene-driven expression of HSF1 or its mutant suggests that HSF1 levels are strictly controlled in neuronal tissue.


Subject(s)
Gene Expression Regulation , Heat-Shock Proteins/genetics , Neurons/metabolism , Transcription Factors/genetics , Animals , Brain Chemistry , Embryonic Development/genetics , Larva , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...