Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(44): 15741-5, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25331893

RESUMEN

There are many transmembrane receptor-like proteins whose ligands have not been identified. A strategy for finding ligands when little is known about their tissue source is to screen each extracellular protein individually expressed in an array format by using a sensitive functional readout. Taking this approach, we have screened a large collection (3,191 proteins) of extracellular proteins for their ability to activate signaling of an orphan receptor, leukocyte tyrosine kinase (LTK). Only two related secreted factors, FAM150A and FAM150B (family with sequence similarity 150 member A and member B), stimulated LTK phosphorylation. FAM150A binds LTK extracellular domain with high affinity (K(D) = 28 pM). FAM150A stimulates LTK phosphorylation in a ligand-dependent manner. This strategy provides an efficient approach for identifying functional ligands for other orphan receptors.


Asunto(s)
Citocinas/metabolismo , Proteoma/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/fisiología , Citocinas/genética , Femenino , Células HEK293 , Humanos , Masculino , Fosforilación/fisiología , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Proteoma/genética , Proteómica , Proteínas Tirosina Quinasas Receptoras/genética
2.
Cancer Res ; 82(22): 4288-4298, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36112781

RESUMEN

T cell-engaging bispecific antibodies (TCB) are highly potent therapeutics that can recruit and activate cytotoxic T cells to stimulate an antitumor immune response. However, the development of TCBs against solid tumors has been limited by significant on-target toxicity to normal tissues. Probody therapeutics have been developed as a novel class of recombinant, protease-activated antibody prodrugs that are "masked" to reduce antigen binding in healthy tissues but can become conditionally unmasked by proteases that are preferentially active in the tumor microenvironment (TME). Here, we describe the preclinical efficacy and safety of CI107, a Probody TCB targeting EGFR and CD3. In vitro, the protease-activated, unmasked CI107 effectively bound EGFR and CD3 expressed on the surface of cells and induced T-cell activation, cytokine release, and cytotoxicity toward tumor cells. In contrast, dually masked CI107 displayed a >500-fold reduction in antigen binding and >15,000-fold reduction in cytotoxic activity. In vivo, CI107 potently induced dose-dependent tumor regression of established colon cancer xenografts in mice engrafted with human peripheral blood mononuclear cells. Furthermore, the MTD of CI107 in cynomolgus monkeys was more than 60-fold higher than that of the unmasked TCB, and much lower levels of toxicity were observed in animals receiving CI107. Therefore, by localizing activity to the TME and thus limiting toxicity to normal tissues, this Probody TCB demonstrates the potential to expand clinical opportunities for TCBs as effective anticancer therapies for solid tumor indications. SIGNIFICANCE: A conditionally active EGFR-CD3 T cell-engaging Probody therapeutic expands the safety window of bispecific antibodies while maintaining efficacy in preclinical solid tumor settings.


Asunto(s)
Anticuerpos Biespecíficos , Complejo CD3 , Neoplasias del Colon , Receptores ErbB , Animales , Humanos , Ratones , Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3/antagonistas & inhibidores , Neoplasias del Colon/terapia , Receptores ErbB/antagonistas & inhibidores , Leucocitos Mononucleares/metabolismo , Péptido Hidrolasas/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Cancer Ther ; 21(8): 1326-1336, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35666803

RESUMEN

Probody therapeutics (Pb-Txs) are conditionally activated antibody-drug conjugates (ADCs) designed to remain inactive until proteolytically activated in the tumor microenvironment, enabling safer targeting of antigens expressed in both tumor and normal tissue. Previous attempts to target CD71, a highly expressed tumor antigen, have failed to establish an acceptable therapeutic window due to widespread normal tissue expression. This study evaluated whether a probody-drug conjugate targeting CD71 can demonstrate a favorable efficacy and tolerability profile in preclinical studies for the treatment of cancer. CX-2029, a Pb-Tx conjugated to maleimido-caproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E, was developed as a novel cancer therapeutic targeting CD71. Preclinical studies were performed to evaluate the efficacy and safety of this anti-CD71 PDC in patient-derived xenograft (PDX) mouse models and cynomolgus monkeys, respectively. CD71 expression was detected at high levels by IHC across a broad range of tumor and normal tissues. In vitro, the masked Pb-Tx form of the anti-CD71 PDC displayed a >50-fold reduced affinity for binding to CD71 on cells compared with protease-activated, unmasked anti-CD71 PDC. Potent in vivo tumor growth inhibition (stasis or regression) was observed in >80% of PDX models (28/34) at 3 or 6 mg/kg. Anti-CD71 PDC remained mostly masked (>80%) in circulation throughout dosing in cynomolgus monkeys at 2, 6, and 12 mg/kg and displayed a 10-fold improvement in tolerability compared with an anti-CD71 ADC, which was lethal. Preclinically, anti-CD71 PDC exhibits a highly efficacious and acceptable safety profile that demonstrates the utility of the Pb-Tx platform to target CD71, an otherwise undruggable target. These data support further clinical development of the anti-CD71 PDC CX-2029 as a novel cancer therapeutic.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Neoplasias , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Plomo , Macaca fascicularis/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Pharmacol Ther ; 109(2): 383-393, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32681519

RESUMEN

CX-072 is an anti-PD-L1 (programmed death ligand 1) Probody therapeutic (Pb-Tx) designed to be preferentially activated by proteases in the tumor microenvironment and not in healthy tissue. Here, we report the model-informed drug development of CX-072. A quantitative systems pharmacology (QSP) model that captured known mechanisms of Pb-Tx activation, biodistribution, elimination, and target engagement was used to inform clinical translation. The QSP model predicted that a trough level of masked CX-072 (intact CX-072) of 13-99 nM would correspond to a targeted, 95% receptor occupancy in the tumor. The QSP model predictions appeared consistent with preliminary human single-dose pharmacokinetic (PK) data following CX-072 0.03-30.0 mg/kg as monotherapy: CX-072 circulated predominantly as intact CX-072 with minimal evidence of target-mediated drug disposition. A preliminary population PK (POPPK) analysis based upon 130 subjects receiving 0.03-30.0 mg/kg as monotherapy included a provision for a putative time-dependent and dose-dependent antidrug antibody (ADA) effect on clearance (CL) with a mixture model. Preliminary POPPK estimates for intact CX-072 time-invariant CL and volume of distribution were 0.306 L/day and 4.84 L, respectively. Exposure-response analyses did not identify statistically significant relationships with best change from baseline sum of measurements and either adverse events of grade ≥ 3 or of special interest. Simulations suggested that > 95% of patients receiving CX-072 10 mg/kg every two weeks would exceed the targeted trough level regardless of ADA, and that dose adjustment by body weight was not necessary, supporting a fixed 800 mg dose for evaluation in phase II.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/metabolismo , Relación Dosis-Respuesta a Droga , Desarrollo de Medicamentos/métodos , Humanos , Masculino , Modelos Biológicos , Distribución Tisular/fisiología , Microambiente Tumoral/efectos de los fármacos
5.
Cancer Immunol Res ; 9(12): 1451-1464, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34635485

RESUMEN

Immune-checkpoint blockade has revolutionized cancer treatment. However, most patients do not respond to single-agent therapy. Combining checkpoint inhibitors with other immune-stimulating agents increases both efficacy and toxicity due to systemic T-cell activation. Protease-activatable antibody prodrugs, known as Probody therapeutics (Pb-Tx), localize antibody activity by attenuating capacity to bind antigen until protease activation in the tumor microenvironment. Herein, we show that systemic administration of anti-programmed cell death ligand 1 (anti-PD-L1) and anti-programmed cell death protein 1 (anti-PD-1) Pb-Tx to tumor-bearing mice elicited antitumor activity similar to that of traditional PD-1/PD-L1-targeted antibodies. Pb-Tx exhibited reduced systemic activity and an improved nonclinical safety profile, with markedly reduced target occupancy on peripheral T cells and reduced incidence of early-onset autoimmune diabetes in nonobese diabetic mice. Our results confirm that localized PD-1/PD-L1 inhibition by Pb-Tx can elicit robust antitumor immunity and minimize systemic immune-mediated toxicity. These data provide further preclinical rationale to support the ongoing development of the anti-PD-L1 Pb-Tx CX-072, which is currently in clinical trials.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/uso terapéutico , Inmunoterapia/métodos , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Microambiente Tumoral
6.
Expert Opin Biol Ther ; 20(2): 163-171, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31779489

RESUMEN

Introduction: The toxicity of potent new biological therapies for cancer has limited their utility. By improving tumor specificity, antibody prodrugs can widen or even create a therapeutic window for anticancer agents that are difficult or impossible to use otherwise because of poor tolerability.Areas covered: This review will describe the current status of the field of antibody prodrugs, focusing on ProbodyTM therapeutics, including the principles behind their design, application to a variety of different antibody-based therapies, preclinical examples of their activity and safety, and early results of Phase 1 trials.Expert opinion: Proof of concept for the antibody prodrug approach, which is defined as demonstration of potent antitumor activity with improved safety, has been extensively established preclinically as well as preliminarily in early clinical trials in human patients. However, experience with antibody prodrugs is limited, and important challenges remain. Principal among them are how to design the molecules to provide the most effective protection from toxicities while preserving efficacy, how to optimize clinical pharmacology, and how to determine which among the many possible clinical applications is the best use of this promising technology.


Asunto(s)
Anticuerpos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Profármacos/uso terapéutico , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias , Neoplasias/epidemiología , Neoplasias/inmunología
7.
SLAS Discov ; 25(9): 1047-1063, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32713278

RESUMEN

The identification of novel peptide hormones by functional screening is challenging because posttranslational processing is frequently required to generate biologically active hormones from inactive precursors. We developed an approach for functional screening of novel potential hormones by expressing them in endocrine host cells competent for posttranslational processing. Candidate preprohormones were selected by bioinformatics analysis, and stable endocrine host cell lines were engineered to express the preprohormones. The production of mature hormones was demonstrated by including the preprohormones insulin and glucagon, which require the regulated secretory pathway for production of the active forms. As proof of concept, we screened a set of G-protein-coupled receptors (GPCRs) and identified protein FAM237A as a specific activator of GPR83, a GPCR implicated in central nervous system and regulatory T-cell function. We identified the active form of FAM237A as a C-terminally cleaved, amidated 9 kDa secreted protein. The related protein FAM237B, which is 64% homologous to FAM237A, demonstrated similar posttranslational modification and activation of GPR83, albeit with reduced potency. These results demonstrate that our approach is capable of identifying and characterizing novel hormones that require processing for activity.


Asunto(s)
Hormonas Peptídicas/aislamiento & purificación , Biblioteca de Péptidos , Transporte de Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Humanos , Ligandos , Hormonas Peptídicas/genética , Hormonas Peptídicas/inmunología , Unión Proteica/genética , Transporte de Proteínas/inmunología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/inmunología , Transducción de Señal/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Biochimie ; 122: 62-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26546838

RESUMEN

Probody™ therapeutics are recombinant, proteolytically-activated antibody prodrugs, engineered to remain inert until activated locally by tumor-associated proteases. Probody therapeutics exploit the fundamental dysregulation of extracellular protease activity that exists in tumors relative to healthy tissue. Leveraging the ability of a Probody therapeutic to bind its target at the site of disease after proteolytic cleavage, we developed a novel method for profiling protease activity in living animals. Using NIR optical imaging, we demonstrated that a non-labeled anti-EGFR Probody therapeutic can become activated and compete for binding to tumor cells in vivo with a labeled anti-EGFR monoclonal antibody. Furthermore, by inhibiting matriptase activity in vivo with a blocking-matriptase antibody, we show that the ability of the Probody therapeutic to bind EGFR in vivo was dependent on protease activity. These results demonstrate that in vivo imaging of Probody therapeutic activation can be used for screening and characterization of protease activity in living animals, and provide a method that avoids some of the limitations of prior methods. This approach can improve our understanding of the activity of proteases in disease models and help to develop efficient strategies for cancer diagnosis and treatment.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Profármacos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Unión Competitiva , Línea Celular Tumoral , Cetuximab/química , Cetuximab/metabolismo , Cetuximab/farmacología , Diagnóstico por Imagen/métodos , Receptores ErbB/metabolismo , Femenino , Colorantes Fluorescentes/química , Humanos , Neoplasias Pulmonares/metabolismo , Ratones Desnudos , Péptido Hidrolasas/metabolismo , Profármacos/química , Profármacos/metabolismo , Unión Proteica , Reproducibilidad de los Resultados , Succinimidas/química , Factores de Tiempo
9.
Sci Transl Med ; 5(178): 178ra39, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23536011

RESUMEN

The fibroblast growth factor (FGF) pathway promotes tumor growth and angiogenesis in many solid tumors. Although there has long been interest in FGF pathway inhibitors, development has been complicated: An effective FGF inhibitor must block the activity of multiple mitogenic FGF ligands but must spare the metabolic hormone FGFs (FGF-19, FGF-21, and FGF-23) to avoid unacceptable toxicity. To achieve these design requirements, we engineered a soluble FGF receptor 1 Fc fusion protein, FP-1039. FP-1039 binds tightly to all of the mitogenic FGF ligands, inhibits FGF-stimulated cell proliferation in vitro, blocks FGF- and vascular endothelial growth factor (VEGF)-induced angiogenesis in vivo, and inhibits in vivo growth of a broad range of tumor types. FP-1039 antitumor response is positively correlated with RNA levels of FGF2, FGF18, FGFR1c, FGFR3c, and ETV4; models with genetic aberrations in the FGF pathway, including FGFR1-amplified lung cancer and FGFR2-mutated endometrial cancer, are particularly sensitive to FP-1039-mediated tumor inhibition. FP-1039 does not appreciably bind the hormonal FGFs, because these ligands require a cell surface co-receptor, klotho or ß-klotho, for high-affinity binding and signaling. Serum calcium and phosphate levels, which are regulated by FGF-23, are not altered by administration of FP-1039. By selectively blocking nonhormonal FGFs, FP-1039 treatment confers antitumor efficacy without the toxicities associated with other FGF pathway inhibitors.


Asunto(s)
Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Inmunoglobulina G/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/uso terapéutico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/uso terapéutico , Calcio/sangre , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Fosfatos/sangre , Proteínas Recombinantes de Fusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA