Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Res ; 12(7): 921-943, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38683145

RESUMEN

The prognosis of patients with acute myeloid leukemia (AML) is limited, especially for elderly or unfit patients not eligible for hematopoietic stem cell (HSC) transplantation. The disease is driven by leukemic stem cells (LSCs), which are characterized by clonal heterogeneity and resistance to conventional therapy. These cells are therefore believed to be a major cause of progression and relapse. We designed MP0533, a multispecific CD3-engaging designed ankyrin repeat protein (DARPin) that can simultaneously bind to three antigens on AML cells (CD33, CD123, and CD70), aiming to enable avidity-driven T cell-mediated killing of AML cells coexpressing at least two of the antigens. In vitro, MP0533 induced selective T cell-mediated killing of AML cell lines, as well as patient-derived AML blasts and LSCs, expressing two or more target antigens, while sparing healthy HSCs, blood, and endothelial cells. The higher selectivity also resulted in markedly lower levels of cytokine release in normal human blood compared to single antigen-targeting T-cell engagers. In xenograft AML mice models, MP0533 induced tumor-localized T-cell activation and cytokine release, leading to complete eradication of the tumors while having no systemic adverse effects. These studies show that the multispecific-targeting strategy used with MP0533 holds promise for improved selectivity toward LSCs and efficacy against clonal heterogeneity, potentially bringing a new therapeutic option to this group of patients with a high unmet need. MP0533 is currently being evaluated in a dose-escalation phase 1 study in patients with relapsed or refractory AML (NCT05673057).


Asunto(s)
Leucemia Mieloide Aguda , Células Madre Neoplásicas , Linfocitos T , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/patología , Animales , Ratones , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Subunidad alfa del Receptor de Interleucina-3/inmunología , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Complejo CD3/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica
2.
Nat Biotechnol ; 40(12): 1845-1854, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35864170

RESUMEN

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with potential resistance to existing drugs emphasizes the need for new therapeutic modalities with broad variant activity. Here we show that ensovibep, a trispecific DARPin (designed ankyrin repeat protein) clinical candidate, can engage the three units of the spike protein trimer of SARS-CoV-2 and inhibit ACE2 binding with high potency, as revealed by cryo-electron microscopy analysis. The cooperative binding together with the complementarity of the three DARPin modules enable ensovibep to inhibit frequent SARS-CoV-2 variants, including Omicron sublineages BA.1 and BA.2. In Roborovski dwarf hamsters infected with SARS-CoV-2, ensovibep reduced fatality similarly to a standard-of-care monoclonal antibody (mAb) cocktail. When used as a single agent in viral passaging experiments in vitro, ensovibep reduced the emergence of escape mutations in a similar fashion to the same mAb cocktail. These results support further clinical evaluation of ensovibep as a broad variant alternative to existing targeted therapies for Coronavirus Disease 2019 (COVID-19).


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Cricetinae , Humanos , SARS-CoV-2/genética , Proteínas de Repetición de Anquirina Diseñadas , Microscopía por Crioelectrón , Anticuerpos Monoclonales/uso terapéutico , Terapéutica Combinada de Anticuerpos , Anticuerpos Neutralizantes
3.
J Immunol Res ; 2020: 7375947, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32832572

RESUMEN

PD-1/PD-L1 blockade has revolutionized the field of immunooncology. Despite the relative success, the response rate to anti-PD-1 therapy requires further improvements. Our aim was to explore the enhancement of T-cell function by using novel PD-1-blocking proteins and compare with clinically approved monoclonal antibodies (mAbs). We isolated T-cells from the ascites and tumor of 17 patients with advanced epithelial ovarian cancer (EOC) and analyzed the effects using the mAbs nivolumab and pembrolizumab and two novel engineered ankyrin repeat proteins (DARPin® proteins). PD-1 blockade with either mAb or DARPin® molecule significantly increased the release of IFN-γ, granzyme B, IL-2, and TNF-α, demonstrating successful reinvigoration. The monovalent DARPin® protein was less effective compared to its bivalent equivalent, demonstrating that bivalency brings an additional benefit to PD-1 blockade. Overall, we found a higher fold increase of lymphokine secretion in response to the PD-1 blockade by tumor-derived T-cells; however, the absolute amounts were significantly lower compared to the release from ascites-derived T-cells. Our results demonstrate that PD-1 blockade can only partially reinvigorate functionally suppressed T-cells from EOC patients. This warrants further investigation preferably in combination with other therapeutics. The study provides an early pilot proof-of-concept for the potential use of DARPin® proteins as eligible alternative scaffold proteins to block PD-1.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunomodulación/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Biomarcadores de Tumor , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Femenino , Humanos , Clasificación del Tumor , Estadificación de Neoplasias , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo
4.
Protein Eng Des Sel ; 30(9): 583-591, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29088432

RESUMEN

A long systemic half-life is key for therapeutic proteins. To that end we have generated serum albumin-binding designed ankyrin repeat domains. These domains bind serum albumin of different species with nanomolar affinities, and have significantly improved pharmacokinetic properties both in mouse and cynomolgus monkey compared to non-serum albumin-binding DARPin® domains. In addition, they exhibit high thermal stability and long storage stability, which is an essential feature for their use in drug development. Covalently linking a serum albumin-binding DARPin® domain to domains with other target specificities results in improvements of multiple orders of magnitude in exposure and terminal half-life, both in mouse and cynomolgus monkey. Pharmacokinetic assessment of such constructs revealed terminal half-life values ranging from 27 h to 80 h in mouse, and from 2.6 days to 20 days in cynomolgus monkey. Extrapolation by allometric scaling on these findings suggests terminal half-life values of 5-50 days in human, indicating that pharmacokinetic properties in the range of monoclonal antibodies can be achieved with DARPin® drug candidates. Such serum albumin-binding DARPin® domains are thus valuable tools for the generation of multi-functional drugs with an extended in vivo half-life.


Asunto(s)
Repetición de Anquirina , Vectores Genéticos/química , Proteínas Recombinantes de Fusión/farmacocinética , Albúmina Sérica/genética , Animales , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/metabolismo , Semivida , Humanos , Concentración de Iones de Hidrógeno , Macaca fascicularis , Ratones , Unión Proteica , Estabilidad Proteica , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Albúmina Sérica/metabolismo
5.
Angiogenesis ; 16(1): 101-11, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22983424

RESUMEN

The next-generation ophthalmic anti-VEGF therapeutics must aim at being superior to the currently available agents with regard to potency and improved drug delivery, while still being stable and safe to use at elevated concentrations. We show here the generation of a set of highly potent VEGF-A antagonistic DARPins (designed ankyrin repeat proteins) delivering these properties. DARPins with single-digit picomolar affinity to human VEGF-A were generated using ribosome display selections. Specific and potent human VEGF-A binding was confirmed by ELISA and endothelial cell sprouting assays. Cross-reactivity with VEGF-A of several species was confirmed by ELISA. Intravitreally injected DARPin penetrated into the retina and reduced fluorescein extravasation in a rabbit model of vascular leakage. In addition, topical DARPin application was found to diminish corneal neovascularization in a rabbit suture model, and to suppress laser-induced neovascularization in a rat model. Even at elevated doses, DARPins were safe to use. The fact that several DARPins are highly active in various assays illustrates the favorable class behavior of the selected binders. Anti-VEGF-A DARPins thus represent a novel class of highly potent and specific drug candidates for the treatment of neovascular eye diseases in both the posterior and the anterior eye chamber.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Repetición de Anquirina , Proteínas Recombinantes/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Administración Tópica , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ojo/irrigación sanguínea , Ojo/efectos de los fármacos , Ojo/patología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inyecciones Intravítreas , Ratones , Soluciones Oftálmicas/farmacología , Soluciones Oftálmicas/uso terapéutico , Unión Proteica/efectos de los fármacos , Conejos , Ratas , Ratas Endogámicas BN , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Mol Cell Biol ; 32(19): 3802-13, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22801374

RESUMEN

Vascular endothelial growth factors (VEGFs) activate three receptor tyrosine kinases, VEGFR-1, -2, and -3, which regulate angiogenic and lymphangiogenic signaling. VEGFR-2 is the most prominent receptor in angiogenic signaling by VEGF ligands. The extracellular part of VEGF receptors consists of seven immunoglobulin homology domains (Ig domains). Earlier studies showed that domains 2 and 3 (D23) mediate ligand binding, while structural analysis of dimeric ligand/receptor complexes by electron microscopy and small-angle solution scattering revealed additional homotypic contacts in membrane-proximal Ig domains D4 and D7. Here we show that D4 and D7 are indispensable for receptor signaling. To confirm the essential role of these domains in signaling, we isolated VEGFR-2-inhibitory "designed ankyrin repeat proteins" (DARPins) that interact with D23, D4, or D7. DARPins that interact with D23 inhibited ligand binding, receptor dimerization, and receptor kinase activation, while DARPins specific for D4 or D7 did not prevent ligand binding or receptor dimerization but effectively blocked receptor signaling and functional output. These data show that D4 and D7 allosterically regulate VEGFR-2 activity. We propose that these extracellular-domain-specific DARPins represent a novel generation of receptor-inhibitory drugs for in vivo applications such as targeting of VEGFRs in medical diagnostics and for treating vascular pathologies.


Asunto(s)
Sitio Alostérico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Animales , Expresión Génica , Humanos , Estructura Terciaria de Proteína , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
7.
J Immunol Methods ; 313(1-2): 140-8, 2006 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-16730021

RESUMEN

Ribosome display is a powerful in vitro technology for the selection and directed evolution of proteins. The ribosome display process exploits cell-free translation to achieve coupling of phenotype and genotype by the production of stabilised ribosome complexes in which translated proteins and their encoding mRNA remain attached to the ribosome. Current ribosome display systems that are well proven, by the evolution of high affinity antibodies and the optimisation of defined protein characteristics, use an Escherichia coli cell extract for in vitro translation and display of an mRNA library. Recently, a cell-free translation system has been produced by combining recombinant E. coli protein factors with purified 70S ribosomes. We have applied this development in cell-free translation technology to ribosome display by using the reconstituted system to generate stabilised ribosome complexes for selection. We show that higher cDNA yields are recovered from ribosome display selections when using a reconstituted translation system and the degree of improvement seen is selection specific. These effects are likely to reflect higher mRNA and protein stability and potentially other advantages that may include protein specific improvements in expression. Reconstituted translation systems therefore enable a highly efficient, robust and accessible prokaryotic ribosome display technology.


Asunto(s)
Biosíntesis de Proteínas/genética , Ribosomas/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Escherichia coli/química , Hormona de Crecimiento Humana/genética , Humanos , Fragmentos de Inmunoglobulinas/genética , Región Variable de Inmunoglobulina/genética , Insulina/inmunología , Interleucina-13/inmunología , Datos de Secuencia Molecular , Biblioteca de Péptidos , Estabilidad del ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribosomas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA