Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int Rev Cell Mol Biol ; 369: 89-106, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35777866

RESUMEN

Anticalin proteins are a novel class of clinical-stage biopharmaceuticals with high potential in various disease areas. Anticalin proteins, derived from extracellular human lipocalins are single-chain proteins, with a highly stable structure that can be engineered to bind with high specificity and potency to targets of therapeutic relevance. The small size and stable structure support their development as inhalable biologics in the field of respiratory diseases as already demonstrated for PRS-060/AZD1402, an Anticalin protein currently undergoing clinical development for the treatment of asthma. Anticalin proteins provide formatting flexibility which allows fusion with the same or other Anticalin proteins, or with other biologics to generate multivalent, multiparatopic or multispecific fusion proteins. The fusion of Anticalin proteins to antibodies allows the generation of potent therapeutic proteins with new modes of action, such as antibody-Anticalin bispecific proteins with tumor-localized activity. Cinrebafusp alfa and PRS-344/S095012 antibody-Anticalin bispecific proteins were designed to reduce potential systemic toxicity by localizing the activity to the tumor, and are currently in clinical development in immuno-oncology. Furthermore, the ease in generating bi- and multispecifics as well as the small and stable structure prompted the investigation of Anticalin proteins for the CAR T space, opening additional potential treatment options based on Anticalin protein therapies.


Asunto(s)
Productos Biológicos , Neoplasias , Productos Biológicos/uso terapéutico , Desarrollo de Medicamentos , Humanos , Lipocalinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas
2.
Clin Cancer Res ; 28(15): 3387-3399, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35121624

RESUMEN

PURPOSE: While patients responding to checkpoint blockade often achieve remarkable clinical responses, there is still significant unmet need due to resistant or refractory tumors. A combination of checkpoint blockade with further T-cell stimulation mediated by 4-1BB agonism may increase response rates and durability of response. A bispecific molecule that blocks the programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) axis and localizes 4-1BB costimulation to a PD-L1-positive (PD-L1+) tumor microenvironment (TME) or tumor draining lymph nodes could maximize antitumor immunity and increase the therapeutic window beyond what has been reported for anti-4-1BB mAbs. EXPERIMENTAL DESIGN: We generated and characterized the PD-L1/4-1BB bispecific molecule PRS-344/S095012 for target binding and functional activity in multiple relevant in vitro assays. Transgenic mice expressing human 4-1BB were transplanted with human PD-L1-expressing murine MC38 cells to assess in vivo antitumoral activity. RESULTS: PRS-344/S095012 bound to its targets with high affinity and efficiently blocked the PD-1/PD-L1 pathway, and PRS-344/S095012-mediated 4-1BB costimulation was strictly PD-L1 dependent. We demonstrated a synergistic effect of both pathways on T-cell stimulation with the bispecific PRS-344/S095012 being more potent than the combination of mAbs. PRS-344/S095012 augmented CD4-positive (CD4+) and CD8-positive (CD8+) T-cell effector functions and enhanced antigen-specific T-cell stimulation. Finally, PRS-344/S095012 demonstrated strong antitumoral efficacy in an anti-PD-L1-resistant mouse model in which soluble 4-1BB was detected as an early marker for 4-1BB agonist activity. CONCLUSIONS: The PD-L1/4-1BB bispecific PRS-344/S095012 efficiently combines checkpoint blockade with a tumor-localized 4-1BB-mediated stimulation burst to antigen-specific T cells, more potent than the combination of mAbs, supporting the advancement of PRS-344/S095012 toward clinical development. See related commentary by Shu et al., p. 3182.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Humanos , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral
3.
Front Pharmacol ; 12: 759337, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34759826

RESUMEN

Anticalin® proteins have been proven as versatile clinical stage biotherapeutics. Due to their small size (∼20 kDa), they harbor a short intrinsic plasma half-life which can be extended, e.g., by fusion with IgG or Fc. However, for antagonism of co-immunostimulatory Tumor Necrosis Factor Receptor Superfamily (TNFRSF) members in therapy of autoimmune and inflammatory diseases, a monovalent, pharmacokinetically optimized Anticalin protein format that avoids receptor clustering and therefore potential activation is favored. We investigated the suitability of an affinity-improved streptococcal Albumin-Binding Domain (ABD) and the engineered Fab-selective Immunoglobulin-Binding Domain (IgBD) SpGC3Fab for plasma Half-Life Extension (HLE) of an OX40-specific Anticalin and bispecific Duocalin proteins, neutralizing OX40 and a second co-immunostimulatory TNFRSF member. The higher affinity of ABD fusion proteins to human serum albumin (HSA) and Mouse Serum Albumin (MSA), with a 4 to 5-order of magnitude lower KD compared with the binding affinity of IgBD fusions to human/mouse IgG, translated into longer terminal plasma half-lives (t 1/2). Hence, the anti-OX40 Anticalin-ABD protein reached t 1/2 values of ∼40 h in wild-type mice and 110 h in hSA/hFcRn double humanized mice, in contrast to ∼7 h observed for anti-OX40 Anticalin-IgBD in wild-type mice. The pharmacokinetics of an anti-OX40 Anticalin-Fc fusion protein was the longest in both models (t 1/2 of 130 h and 146 h, respectively). Protein formats composed of two ABDs or IgBDs instead of one single HLE domain clearly showed longer presence in the circulation. Importantly, Anticalin-ABD and -IgBD fusions showed OX40 receptor binding and functional competition with OX40L-induced cellular reactivity in the presence of albumin or IgG, respectively. Our results suggest that fusion to ABD or IgBD can be a versatile platform to tune the plasma half-life of Anticalin proteins in response to therapeutic needs.

4.
Clin Cancer Res ; 25(19): 5878-5889, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31138587

RESUMEN

PURPOSE: 4-1BB (CD137) is a key costimulatory immunoreceptor and promising therapeutic target in cancer. To overcome limitations of current 4-1BB-targeting antibodies, we have developed PRS-343, a 4-1BB/HER2 bispecific molecule. PRS-343 is designed to facilitate T-cell costimulation by tumor-localized, HER2-dependent 4-1BB clustering and activation. EXPERIMENTAL DESIGN: PRS-343 was generated by the genetic fusion of 4-1BB-specific Anticalin proteins to a variant of trastuzumab with an engineered IgG4 isotype. Its activity was characterized using a panel of in vitro assays and humanized mouse models. The safety was assessed using ex vivo human cell assays and a toxicity study in cynomolgus monkeys. RESULTS: PRS-343 targets 4-1BB and HER2 with high affinity and binds both targets simultaneously. 4-1BB-expressing T cells are efficiently costimulated when incubated with PRS-343 in the presence of cancer cells expressing HER2, as evidenced by increased production of proinflammatory cytokines (IL2, GM-CSF, TNFα, and IFNγ). In a humanized mouse model engrafted with HER2-positive SK-OV-3 tumor cells and human peripheral blood mononuclear cells, PRS-343 leads to tumor growth inhibition and a dose-dependent increase of tumor-infiltrating lymphocytes. In IND-enabling studies, PRS-343 was found to be well tolerated, with no overt toxicity and no relevant drug-related toxicologic findings. CONCLUSIONS: PRS-343 facilitates tumor-localized targeting of T cells by bispecific engagement of HER2 and 4-1BB. This approach has the potential to provide a more localized activation of the immune system with higher efficacy and reduced peripheral toxicity compared with current monospecific approaches. The reported data led to initiation of a phase I clinical trial with this first-in-class molecule.See related commentary by Su et al., p. 5732.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor , Ratones , Linfocitos T , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral
5.
BioDrugs ; 32(3): 233-243, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29748739

RESUMEN

Anticalin proteins are an emerging class of clinical-stage biopharmaceuticals with high potential as an alternative to antibodies. Anticalin molecules are generated by combinatorial design from natural lipocalins, which are abundant plasma proteins in humans, and reveal a simple, compact fold dominated by a central ß-barrel, supporting four structurally variable loops that form a binding site. Reshaping of this loop region results in Anticalin proteins that can recognize and tightly bind a wide range of medically relevant targets, from small molecules to peptides and proteins, as validated by X-ray structural analysis. Their robust format allows for modification in several ways, both as fusion proteins and by chemical conjugation, for example, to tune plasma half-life. Antagonistic Anticalin therapeutics have been developed for systemic administration (e.g., PRS-080: anti-hepcidin) or pulmonary delivery (e.g. PRS-060/AZD1402: anti-interleukin [IL]-4-Rα). Moreover, Anticalin proteins allow molecular formatting as bi- and even multispecific fusion proteins, especially in combination with antibodies that provide a second specificity. For example, PRS-343, which has recently entered clinical-stage development, combines an agonistic Anticalin targeting the costimulatory receptor 4-1BB with an antibody directed against the cancer antigen human epidermal growth factor receptor 2 (HER2), thus offering a novel treatment option in immuno-oncology.


Asunto(s)
Enfermedad , Quimioterapia/métodos , Lipocalinas/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Biofarmacia , Humanos , Lipocalinas/química , Lipocalinas/genética , Terapia Molecular Dirigida , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Tecnología Farmacéutica
6.
Res Vet Sci ; 93(1): 457-62, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21940026

RESUMEN

Adult stem cells are of particular interest for therapeutic use in the field of regenerative medicine. Adipose-derived mesenchymal stem cells (ASCs) are an attractive stem cell source for all fields of regenerative medicine because adipose tissue - and therewith cells - can easily be harvested from each donor. However, common expansion using fetal bovine serum (FBS) can not be used for clinical applications as xenogenic proteins must be avoided. Adipose tissue from equine, canine and porcine donors was digested with collagenase to isolate ASCs. ASCs were either expanded in a cell culture medium supplemented with FBS or in a serum-free medium (UltraCulture; UC) supplemented with a serum substitute (UltroserG). From all three animal species, the adipogenic and osteogenic differentiation potential of ASCs cultured with different media was analyzed in vitro. Cell proliferation analysis showed a population doubling time of 48-68 h for canine cells, 54-65 h for porcine cells and 54-70 h for equine cells, expanded in different media. Except for porcine ASCs, cells cultured in media supplemented with FBS grew faster than cells expanded in UC medium with UltroserG. Yet, all cells maintained their potential to differentiate into adipocytes and osteoblasts. UltraCulture medium containing UltroserG can for all examined species be recommended if FBS needs to be avoided in the expansion of donor-derived (stem) cells.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular/fisiología , Proliferación Celular , Medios de Cultivo/metabolismo , Células Madre Mesenquimatosas/fisiología , Adipocitos/fisiología , Animales , Recuento de Células/veterinaria , Perros , Femenino , Caballos , Masculino , Osteoblastos/fisiología , Porcinos
7.
J Alzheimers Dis ; 28(1): 49-69, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21955818

RESUMEN

The amyloid-ß lowering capacity of anti-Aß antibodies has been demonstrated in transgenic models of Alzheimer's disease (AD) and in AD patients. While the mechanism of immunotherapeutic amyloid-ß removal is controversial, antibody-mediated sequestration of peripheral Aß versus microglial phagocytic activity and disassembly of cerebral amyloid (or a combination thereof) has been proposed. For successful Aß immunotherapy, we hypothesized that high affinity antibody binding to amyloid-ß plaques and recruitment of brain effector cells is required for most efficient amyloid clearance. Here we report the generation of a novel fully human anti-Aß antibody, gantenerumab, optimized in vitro for binding with sub-nanomolar affinity to a conformational epitope expressed on amyloid-ß fibrils using HuCAL(®) phage display technologies. In peptide maps, both N-terminal and central portions of Aß were recognized by gantenerumab. Remarkably, a novel orientation of N-terminal Aß bound to the complementarity determining regions was identified by x-ray analysis of a gantenerumab Fab-Aß(1-11) complex. In functional assays gantenerumab induced cellular phagocytosis of human amyloid-ß deposits in AD brain slices when co-cultured with primary human macrophages and neutralized oligomeric Aß42-mediated inhibitory effects on long-term potentiation in rat brain. In APP751(swedish)xPS2(N141I) transgenic mice, gantenerumab showed sustained binding to cerebral amyloid-ß and, upon chronic treatment, significantly reduced small amyloid-ß plaques by recruiting microglia and prevented new plaque formation. Unlike other Aß antibodies, gantenerumab did not alter plasma Aß suggesting undisturbed systemic clearance of soluble Aß. These studies demonstrated that gantenerumab preferentially interacts with aggregated Aß in the brain and lowers amyloid-ß by eliciting effector cell-mediated clearance.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Monoclonales/metabolismo , Secuencia de Aminoácidos , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Células CHO , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cricetinae , Cricetulus , Cristalografía por Rayos X , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Fagocitosis/efectos de los fármacos , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley
8.
Biomed Tech (Berl) ; 55(5): 285-90, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20731526

RESUMEN

Cardiac resynchronization therapy provides a treatment option for patients with congestive heart failure. Electrodes are usually advanced via the cardiac venous system (CVS). Placement is often hampered by small vessel diameter or tortuosity. The aim of this study was to develop and test a magnetized stimulation wire (MSW) capable of being navigated into small and tortuous CVS branches. Therefore, a conventional guide wire with a permanent magnet and a single stimulation electrode at its tip was coated with iridium oxide at the distal end and insulated except for the very tip (coating thickness: 500 nm, active uncoated area: 10 mm(2)). The MSW was designed to allow for a remote magnetic steering using a magnetic navigation system (MN) (Stereotaxis™). After in vitro testing of the electrical properties, the MSW was tested in an animal model (n=6 sheep): the MSW was placed in a CVS side branch and navigation characteristics of the MSW were determined. The effective (unipolar) pacing threshold was 4.9±2.4 V (at 2 ms pulse width). The rheobase was 1.89±0.44 V and the chronaxie time was 0.75±0.7 ms. The MSW could be navigated into small CVS branches with reasonable MN properties. This could allow stimulation at CVS sites currently not accessible for conventional stimulation catheters or electrodes.


Asunto(s)
Dispositivos de Terapia de Resincronización Cardíaca , Electrodos Implantados , Sistema de Conducción Cardíaco/fisiología , Magnetismo/instrumentación , Función Ventricular Izquierda/fisiología , Animales , Diseño de Equipo , Análisis de Falla de Equipo , Miniaturización , Proyectos Piloto , Sensibilidad y Especificidad , Ovinos
9.
J Mol Biol ; 376(4): 1182-200, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18191144

RESUMEN

This article describes the generation of the Human Combinatorial Antibody Library HuCAL GOLD. HuCAL GOLD is a synthetic human Fab library based on the HuCAL concept with all six complementarity-determining regions (CDRs) diversified according to the sequence and length variability of naturally rearranged human antibodies. The human antibody repertoire was analyzed in-depth, and individual CDR libraries were designed and generated for each CDR and each antibody family. Trinucleotide mixtures were used to synthesize the CDR libraries in order to ensure a high quality within HuCAL GOLD, and a beta-lactamase selection system was employed to eliminate frame-shifted clones after successive cloning of the CDR libraries. With these methods, a large, high-quality library with more than 10 billion functional Fab fragments was achieved. By using CysDisplay, the antibody fragments are displayed on the tip of the phage via a disulfide bridge between the phage coat protein pIII and the heavy chain of the antibody fragment. Efficient elution of specific phages is possible by adding reducing agents. HuCAL GOLD was challenged with a variety of different antigens and proved to be a reliable source of high-affinity human antibodies with best affinities in the picomolar range, thus functioning as an excellent source of antibodies for research, diagnostic, and therapeutic applications. Furthermore, the data presented in this article demonstrate that CysDisplay is a robust and broadly applicable display technology even for high-throughput applications.


Asunto(s)
Anticuerpos/inmunología , Afinidad de Anticuerpos/inmunología , Técnicas Químicas Combinatorias/métodos , Regiones Determinantes de Complementariedad/inmunología , Sistema Inmunológico/inmunología , Biblioteca de Péptidos , Secuencia de Aminoácidos , Anticuerpos/química , Bacteriófagos , Western Blotting , Clonación Molecular , Regiones Determinantes de Complementariedad/química , Genes , Vectores Genéticos , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/inmunología , Datos de Secuencia Molecular , Conformación Proteica , beta-Lactamasas/genética
10.
J Biol Chem ; 278(40): 38194-205, 2003 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-12842902

RESUMEN

The human combinatorial antibody library Fab 1 (HuCAL-Fab 1) was generated by transferring the heavy and light chain variable regions from the previously constructed single-chain Fv library (Knappik, A., Ge, L., Honegger, A., Pack, P., Fischer, M., Wellnhofer, G., Hoess, A., Wölle, J., Plückthun, A., and Virnekäs, B. (2000) J. Mol. Biol. 296, 57-86), diversified in both complementarity-determining regions 3 into a novel Fab display vector, yielding 2.1 x 10(10) different antibody fragments. The modularity has been retained in the Fab display and screening plasmids, ensuring rapid conversion into various antibody formats as well as antibody optimization using prebuilt maturation cassettes. HuCAL-Fab 1 was challenged against the human fibroblast growth factor receptor 3, a potential therapeutic antibody target, against which, to the best of our knowledge, no functional antibodies could be generated so far. A unique screening mode was designed utilizing recombinant functional proteins and cell lines differentially expressing fibroblast growth factor receptor isoforms diversified in expression and receptor dependence. Specific Fab fragments with subnanomolar affinities were isolated by selection without any maturation steps as determined by fluorescence flow cytometry. Some of the selected Fab fragments completely inhibit target-mediated cell proliferation, rendering them the first monoclonal antibodies against fibroblast growth factor receptors having significant function blocking activity. This study validates HuCAL-Fab 1 as a valuable source for the generation of target-specific antibodies for therapeutic applications.


Asunto(s)
Fragmentos Fab de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/química , Biblioteca de Péptidos , Proteínas Tirosina Quinasas , Receptores de Factores de Crecimiento de Fibroblastos/química , Animales , Anticuerpos , Anticuerpos Monoclonales/química , Unión Competitiva , División Celular , Línea Celular , Separación Celular , Clonación Molecular , Disulfuros , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Epítopos , Escherichia coli/metabolismo , Citometría de Flujo , Biblioteca de Genes , Vectores Genéticos , Humanos , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/química , Región Variable de Inmunoglobulina/química , Concentración 50 Inhibidora , Cinética , Ligandos , Ratones , Plásmidos/metabolismo , Unión Proteica , Isoformas de Proteínas , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Temperatura , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA