Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 23(2): 199-211, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37828728

RESUMO

Topoisomerase I (TOP1) Inhibitors constitute an emerging payload class to engineer antibody-drug conjugates (ADC) as next-generation biopharmaceutical for cancer treatment. Existing ADCs are using camptothecin payloads with lower potency and suffer from limited stability in circulation. With this study, we introduce a novel camptothecin-based linker-payload platform based on the highly potent camptothecin derivative exatecan. First, we describe general challenges that arise from the hydrophobic combination of exatecan and established dipeptidyl p-aminobenzyl-carbamate (PAB) cleavage sites such as reduced antibody conjugation yields and ADC aggregation. After evaluating several linker-payload structures, we identified ethynyl-phosphonamidates in combination with a discrete PEG24 chain to compensate for the hydrophobic PAB-exatecan moiety. Furthermore, we demonstrate that the identified linker-payload structure enables the construction of highly loaded DAR8 ADCs with excellent solubility properties. Head-to-head comparison with Enhertu, an approved camptothecin-based ADC, revealed improved target-mediated killing of tumor cells, excellent bystander killing, drastically improved linker stability in vitro and in vivo and superior in vivo efficacy over four tested dose levels in a xenograft model. Moreover, we show that ADCs based on the novel exatecan linker-payload platform exhibit antibody-like pharmacokinetic properties, even when the ADCs are highly loaded with eight drug molecules per antibody. This ADC platform constitutes a new and general solution to deliver TOP1 inhibitors with highest efficiency to the site of the tumor, independent of the antibody and its target, and is thereby broadly applicable to various cancer indications.


Assuntos
Antineoplásicos , Imunoconjugados , Neoplasias , Humanos , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Imunoconjugados/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Anticorpos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química
2.
Blood ; 141(9): 1023-1035, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35981498

RESUMO

Fms-like tyrosine kinase 3 (FLT3) is often overexpressed or constitutively activated by internal tandem duplication (ITD) and tyrosine kinase domain (TKD) mutations in acute myeloid leukemia (AML). Despite the use of receptor tyrosine kinase inhibitors (TKI) in FLT3-ITD-positive AML, the prognosis of patients is still poor, and further improvement of therapy is required. Targeting FLT3 independent of mutations by antibody-drug conjugates (ADCs) is a promising strategy for AML therapy. Here, we report the development and preclinical characterization of a novel FLT3-targeting ADC, 20D9-ADC, which was generated by applying the innovative P5 conjugation technology. In vitro, 20D9-ADC mediated potent cytotoxicity to Ba/F3 cells expressing transgenic FLT3 or FLT3-ITD, to AML cell lines, and to FLT3-ITD-positive patient-derived xenograft AML cells. In vivo, 20D9-ADC treatment led to a significant tumor reduction and even durable complete remission in AML xenograft models. Furthermore, 20D9-ADC demonstrated no severe hematotoxicity in in vitro colony formation assays using concentrations that were cytotoxic in AML cell line treatment. The combination of 20D9-ADC with the TKI midostaurin showed strong synergy in vitro and in vivo, leading to reduction of aggressive AML cells below the detection limit. Our data indicate that targeting FLT3 with an advanced new-generation ADC is a promising and potent antileukemic strategy, especially when combined with FLT3-TKI in FLT3-ITD-positive AML.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Tirosina Quinase 3 Semelhante a fms/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Mutação
3.
Bioconjug Chem ; 32(6): 1167-1174, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34060308

RESUMO

Multiple, site-specific protein conjugation is increasingly attractive for the generation of antibody-drug conjugates (ADCs). As it is important to control the number and position of cargoes in an ADC, position-selective generation of reactive sites in the protein of interest is required. Formylglycine (FGly) residues are generated by enzymatic conversion of cysteine residues embedded in a certain amino acid sequence motif with a formylglycine-generating enzyme (FGE). The addition of copper ions increases FGE activity leading to the conversion of cysteines within less readily accepted sequences. With this tuned enzyme activity, it is possible to address two different recognition sequences using two aerobic formylglycine-generating enzymes. We demonstrate an improved and facile strategy for the functionalization of a DARPin (designed ankyrin repeat protein) and the single-chain antibody scFv425-Fc, both directed against the epidermal growth factor receptor (EGFR). The single-chain antibody was conjugated with monomethyl auristatin E (MMAE) and carboxyfluorescein (CF) and successfully tested for receptor binding, internalization, and cytotoxicity in cell culture, respectively.


Assuntos
Enzimas/metabolismo , Glicina/análogos & derivados , Imunoconjugados/química , Imunoconjugados/metabolismo , Aerobiose , Repetição de Anquirina , Cobre/química , Fluoresceínas/química , Glicina/metabolismo , Oligopeptídeos/química
4.
Chembiochem ; 22(7): 1205-1209, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33207032

RESUMO

Antibody conjugates have taken a great leap forward as tools in basic and applied molecular life sciences that was enabled by the development of chemoselective reactions for the site-specific modification of proteins. Antibody-oligonucleotide conjugates combine the antibody's target specificity with the reversible, sequence-encoded binding properties of oligonucleotides like DNAs or peptide nucleic acids (PNAs), allowing sequential imaging of large numbers of targets in a single specimen. In this report, we use the Tub-tag® technology in combination with Cu-catalyzed azide-alkyne cycloaddition for the site-specific conjugation of single DNA and PNA strands to an eGFP-binding nanobody. We show binding of the conjugate to recombinant eGFP and subsequent sequence-specific annealing of fluorescently labelled imager strands. Furthermore, we reversibly stain eGFP-tagged proteins in human cells, thus demonstrating the suitability of our conjugation strategy to generate antibody-oligonucleotides for reversible immunofluorescence imaging.


Assuntos
DNA/química , Fragmentos de Imunoglobulinas/química , Microscopia de Fluorescência , Ácidos Nucleicos Peptídicos/química , Alcinos/química , Azidas/química , Catálise , Linhagem Celular , Cobre/química , Reação de Cicloadição , Proteínas de Fluorescência Verde/química , Humanos , Imunoconjugados/química , Imunoconjugados/metabolismo , Fragmentos de Imunoglobulinas/metabolismo , Anticorpos de Domínio Único/química
5.
Chembiochem ; 21(1-2): 113-119, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31661184

RESUMO

Herein, the application of N-hydroxysuccinimide-modified phosphonamidate building blocks for the incorporation of cysteine-selective ethynylphosphonamidates into lysine residues of proteins, followed by thiol addition with small molecules and proteins, is reported. It is demonstrated that the building blocks significantly lower undesired homo-crosslinking side products that can occur with commonly applied succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) under physiological pH. The previously demonstrated stability of the phosphonamidate moiety additionally solves the problem of premature maleimide hydrolysis, which can hamper the efficiency of subsequent thiol addition. Furthermore, a method to separate the phosphonamidate enantiomers to be able to synthesize protein conjugates in a defined configuration has been developed. Finally, the building blocks are applied to the construction of functional antibody-drug conjugates, analogously to FDA-approved, SMCC-linked Kadcyla, and to the synthesis of a functional antibody-protein conjugate.


Assuntos
Amidas/química , Etilenoglicol/química , Proteínas de Fluorescência Verde/química , Ácidos Fosfóricos/química , Succinimidas/química , Linhagem Celular Tumoral , Humanos , Estrutura Molecular
6.
Angew Chem Int Ed Engl ; 58(34): 11631-11636, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31250955

RESUMO

Requirements for novel bioconjugation reactions for the synthesis of antibody-drug conjugates (ADCs) are exceptionally high, since conjugation selectivity as well as the stability and hydrophobicity of linkers and payloads drastically influence the performance and safety profile of the final product. We report Cys-selective ethynylphosphonamidates as new reagents for the rapid generation of efficacious ADCs from native non-engineered monoclonal antibodies through a simple one-pot reduction and alkylation. Ethynylphosphonamidates can be easily substituted with hydrophilic residues, giving rise to electrophilic labeling reagents with tunable solubility properties. We demonstrate that ethynylphosphonamidate-linked ADCs have excellent properties for next-generation antibody therapeutics in terms of serum stability and in vivo antitumor activity.


Assuntos
Antineoplásicos Imunológicos/química , Cisteína/química , Etilenoglicol/química , Imunoconjugados/metabolismo , Organofosfonatos/química , Receptor ErbB-2/imunologia , Trastuzumab/química , Antineoplásicos Imunológicos/imunologia , Humanos , Imunoconjugados/química , Trastuzumab/imunologia , Células Tumorais Cultivadas
7.
Methods Mol Biol ; 2012: 327-355, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31161516

RESUMO

Tubulin tyrosine ligase (TTL) catalyzes the addition of tyrosine derivatives to the C-terminal carboxylic acid of proteins. The enzyme binds to a 14-amino acid recognition sequence, termed Tub-tag, and allows for the introduction of tyrosine derivatives that carry a unique chemical handle. These handles enable subsequent bioorthogonal reactions with a great variety of probes or effector molecules. Clearly, this two-step chemoenzymatic approach, facilitates the site-specific functionalization of proteins. Furthermore, due to its broad substrate tolerance, tubulin tyrosine ligase also enables an enzymatic one-step modification. For example, a coumarin amino acid was utilized to generate fluorescently labeled proteins for advanced applications in imaging and diagnostics. Here we describe the modification of proteins using TTL in detail via a one-step as well as two-step procedure and highlight its practicability for applications in imaging, diagnostics, and cell biology.


Assuntos
Peptídeo Sintases/metabolismo , Proteínas/metabolismo , Aminoácidos/química , Catálise , Linhagem Celular , Humanos , Peptídeo Sintases/química , Processamento de Proteína Pós-Traducional , Proteínas/química , Proteínas Recombinantes , Análise Espectral , Relação Estrutura-Atividade
8.
Org Biomol Chem ; 17(20): 4964-4969, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30932115

RESUMO

We introduce a chemoenzymatic strategy for straightforward in vitro generation of C-terminally linked fusion proteins. Tubulin tyrosine ligase is used for the incorporation of complementary click chemistry handles facilitating subsequent formation of functional bispecific antibody-fragments. This simple strategy may serve as central conjugation hub for a modular protein ligation platform.


Assuntos
Anticorpos/química , Peptídeo Sintases/química , Proteínas Recombinantes de Fusão/química , Anticorpos/metabolismo , Química Click , Estrutura Molecular , Peptídeo Sintases/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo
9.
Angew Chem Int Ed Engl ; 57(24): 7245-7249, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29579347

RESUMO

Formylglycine-generating enzymes are of increasing interest in the field of bioconjugation chemistry. They catalyze the site-specific oxidation of a cysteine residue to the aldehyde-containing amino acid Cα -formylglycine (FGly). This non-canonical residue can be generated within any desired target protein and can subsequently be used for bioorthogonal conjugation reactions. The prototypic formylglycine-generating enzyme (FGE) and the iron-sulfur protein AtsB display slight variations in their recognition sequences. We designed specific tags in peptides and proteins that were selectively converted by the different enzymes. Combination of the different tag motifs within a single peptide or recombinant protein enabled the independent and consecutive introduction of two formylglycine residues and the generation of heterobifunctionalized protein conjugates.


Assuntos
Bactérias/enzimologia , Glicina/análogos & derivados , Linhagem Celular , Cisteína/metabolismo , Glicina/metabolismo , Humanos , Proteínas Ferro-Enxofre/metabolismo , Methanosarcina/enzimologia , Oxirredução , Sulfatases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...