Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Bioconjug Chem ; 31(8): 1893-1898, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32667786

RESUMEN

Several antibody-drug conjugates (ADCs) have failed to achieve a sufficiently large therapeutic window in patients due to toxicity induced by unspecific payload release in the circulation or ADC uptake into healthy organs. Herein, we describe the successful engineering of ADCs consisting of novel linkers, which are efficiently and selectively cleaved by the tumor-associated protease legumain. ADCs generated via this approach demonstrate high potency and a preferential activation in tumors compared to healthy tissue, thus providing an additional level of safety. A remarkable tolerance of legumain for different linker peptides, including those with just a single asparagine residue, together with a modifier of the physicochemical metabolite profile, proves the broad applicability of this approach for a tailored design of ADCs.


Asunto(s)
Inmunoconjugados/química , Cinesinas/antagonistas & inhibidores , Animales , Cisteína Endopeptidasas/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Profármacos , Relación Estructura-Actividad , Neoplasias Urológicas/tratamiento farmacológico , Urotelio
2.
Chemistry ; 25(35): 8208-8213, 2019 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-30869180

RESUMEN

Many antibody-drug conjugates (ADCs) have failed to achieve a sufficient therapeutic window in clinical studies either due to target-mediated or off-target toxicities. To achieve an additional safety level, a new class of antibody-prodrug conjugates (APDCs) directed against different targets in solid tumors is here described. The tumor-associated lysosomal endopeptidase legumain with a unique cleavage sequence was utilized for APDC metabolism. Legumain-activatable APDCs were as potent as their cathepsin B-activatable analogues. The peptide sequence susceptible to legumain cleavage was optimized for further discrimination of the formation of active metabolites within tumor cells versus healthy tissues, leveraging different tissue-specific legumain activities. Optimized APDCs with slow legumain-mediated conversion reduced preclinically the levels of active metabolite in healthy organs while retaining high activity against different TWEAKR- and B7H3-expressing tumors.


Asunto(s)
Anticuerpos/química , Antineoplásicos/química , Cisteína Endopeptidasas/metabolismo , Inmunoconjugados/química , Cinesinas/antagonistas & inhibidores , Oligopéptidos/química , Profármacos/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Antígenos B7/genética , Antígenos B7/inmunología , Antígenos B7/metabolismo , Línea Celular Tumoral , Xenoinjertos , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/metabolismo , Ratones
3.
Angew Chem Int Ed Engl ; 57(46): 15243-15247, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30180286

RESUMEN

The number of cytotoxic payload classes successfully employed in antibody-drug conjugates (ADCs) is still rather limited. The identification of ADC payloads with a novel mode of action will increase therapeutic options and potentially increase the therapeutic window. Herein, we describe the utilization of kinesin spindle protein inhibitors (KSPi) as a novel payload class providing highly potent ADCs against different targets, for instance HER-2 or TWEAKR/Fn14. Aspects of technical optimization include the development of different linker attachment sites, the stabilization of ADC linkage to avoid payload deconjugation and finally, the tailor-made design of active metabolites with a long lasting intracellular exposure in the tumor matching the mode of action of KSP inhibition. These KSPi-ADCs are highly potent and selective in vitro and demonstrate in vivo efficacy in a broad panel of tumor models including complete regressions in a patient-derived urothelial cancer model.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Inmunoconjugados/química , Inmunoconjugados/farmacología , Cinesinas/antagonistas & inhibidores , Pirroles/química , Pirroles/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Descubrimiento de Drogas , Humanos , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Pirroles/uso terapéutico
4.
J Med Chem ; 67(4): 2907-2940, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38348661

RESUMEN

The matrix metalloprotease ADAMTS7 has been identified by multiple genome-wide association studies as being involved in the development of coronary artery disease. Subsequent research revealed the proteolytic function of the enzyme to be relevant for atherogenesis and restenosis after vessel injury. Based on a publicly known dual ADAMTS4/ADAMTS5 inhibitor, we have in silico designed an ADAMTS7 inhibitor of the catalytic domain, which served as a starting point for an optimization campaign. Initially our inhibitors suffered from low selectivity vs MMP12. An X-ray cocrystal structure inspired us to exploit amino acid differences in the binding site of MMP12 and ADAMTS7 to improve selectivity. Further optimization composed of employing 5-membered heteroaromatic groups as hydantoin substituents to become more potent on ADAMTS7. Finally, fine-tuning of DMPK properties yielded BAY-9835, the first orally bioavailable ADAMTS7 inhibitor. Further optimization to improve selectivity vs ADAMTS12 seems possible, and a respective starting point could be identified.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Humanos , Proteína ADAMTS7/genética , Proteína ADAMTS7/metabolismo , Estudio de Asociación del Genoma Completo , Metaloproteinasa 12 de la Matriz
5.
Cell Rep ; 36(3): 109394, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289372

RESUMEN

Novel treatment options for metastatic colorectal cancer (CRC) are urgently needed to improve patient outcome. Here, we screen a library of non-characterized small molecules against a heterogeneous collection of patient-derived CRC spheroids. By prioritizing compounds with inhibitory activity in a subset of-but not all-spheroid cultures, NCT02 is identified as a candidate with minimal risk of non-specific toxicity. Mechanistically, we show that NCT02 acts as molecular glue that induces ubiquitination of cyclin K (CCNK) and proteasomal degradation of CCNK and its complex partner CDK12. Knockout of CCNK or CDK12 decreases proliferation of CRC cells in vitro and tumor growth in vivo. Interestingly, sensitivity to pharmacological CCNK/CDK12 degradation is associated with TP53 deficiency and consensus molecular subtype 4 in vitro and in patient-derived xenografts. We thus demonstrate the efficacy of targeted CCNK/CDK12 degradation for a CRC subset, highlighting the potential of drug-induced proteolysis for difficult-to-treat types of cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Proteolisis , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Daño del ADN , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteómica , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA