Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gynecol Oncol ; 164(1): 154-169, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34799137

RESUMEN

OBJECTIVES: Tumor vasculature is structurally abnormal, with anatomical deformities, reduced pericyte coverage and low tissue perfusion. As a result of this vascular dysfunction, tumors are often hypoxic, which is associated with an aggressive tumor phenotype, and reduced delivery of therapeutic compounds to the tumor. We have previously shown that a peptide containing the thrombospondin-1 type I repeats (3TSR) specifically targets tumor vessels and induces vascular normalization in a mouse model of epithelial ovarian cancer (EOC). However, due to its small size, 3TSR is rapidly cleared from circulation. We now introduce a novel construct with the 3TSR peptide fused to the C-terminus of each of the two heavy chains of the Fc region of human IgG1 (Fc3TSR). We hypothesize that Fc3TSR will have greater anti-tumor activity in vitro and in vivo compared to the native compound. METHODS: Fc3TSR was evaluated in vitro using proliferation and apoptosis assays to investigate differences in efficacy compared to native 3TSR. In light of the multivalency of Fc3TSR, we also investigate whether it induces greater clustering of its functional receptor, CD36. We also compare the compounds in vivo using an orthotopic, syngeneic mouse model of advanced stage EOC. The impact of the two compounds on changes to tumor vasculature morphology was also investigated. RESULTS: Fc3TSR significantly decreased the viability and proliferative potential of EOC cells and endothelial cells in vitro compared to native 3TSR. High-resolution imaging followed by image correlation spectroscopy demonstrated enhanced clustering of the CD36 receptor in cells treated with Fc3TSR. This was associated with enhanced downstream signaling and greater in vitro and in vivo cellular responses. Fc3TSR induced greater vascular normalization and disease regression compared to native 3TSR in an orthotopic, syngeneic mouse model of advanced stage ovarian cancer. CONCLUSION: The development of Fc3TSR which is greater in size, stable in circulation and enhances receptor activation compared to 3TSR, facilitates its translational potential as a therapy in the treatment of metastatic advanced stage ovarian cancer.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Trombospondina 1/uso terapéutico , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/farmacología , Animales , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Neoplasias Ováricas/patología , Trombospondina 1/farmacocinética , Trombospondina 1/farmacología
2.
J Immunol ; 194(3): 878-82, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25548231

RESUMEN

CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.


Asunto(s)
Epigénesis Genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Antígenos de Superficie/metabolismo , Línea Celular Tumoral , Islas de CpG , Metilación de ADN , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Humanos , Inmunofenotipificación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Transgénicos , Neoplasias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
Sensors (Basel) ; 15(12): 31392-427, 2015 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-26703598

RESUMEN

The air quality in urban areas is a major concern in modern cities due to significant impacts of air pollution on public health, global environment, and worldwide economy. Recent studies reveal the importance of micro-level pollution information, including human personal exposure and acute exposure to air pollutants. A real-time system with high spatio-temporal resolution is essential because of the limited data availability and non-scalability of conventional air pollution monitoring systems. Currently, researchers focus on the concept of The Next Generation Air Pollution Monitoring System (TNGAPMS) and have achieved significant breakthroughs by utilizing the advance sensing technologies, MicroElectroMechanical Systems (MEMS) and Wireless Sensor Network (WSN). However, there exist potential problems of these newly proposed systems, namely the lack of 3D data acquisition ability and the flexibility of the sensor network. In this paper, we classify the existing works into three categories as Static Sensor Network (SSN), Community Sensor Network (CSN) and Vehicle Sensor Network (VSN) based on the carriers of the sensors. Comprehensive reviews and comparisons among these three types of sensor networks were also performed. Last but not least, we discuss the limitations of the existing works and conclude the objectives that we want to achieve in future systems.


Asunto(s)
Contaminantes Atmosféricos/análisis , Contaminación del Aire/análisis , Monitoreo del Ambiente/instrumentación , Monitoreo del Ambiente/métodos , Tecnología Inalámbrica/instrumentación , Diseño de Equipo , Gases , Hong Kong , Material Particulado
4.
Transl Oncol ; 16: 101322, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34954456

RESUMEN

Combinatorial immunotherapy approaches are emerging as viable cancer therapeutic strategies for improving patient responses and outcomes. This study investigated whether two such immunotherapies, with complementary mechanisms of action, could enhance antitumor activity in murine tumor models. The immunocytokine NHS-IL12, and surrogate NHS-muIL12, are designed to deliver IL-12 and muIL-12, respectively, to the tumor microenvironment (TME) to activate NK cells and CD8+ T cells and increase their cytotoxic functions. Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domains of the TGF-ß receptor II to function as a TGF-ß "trap" fused to a human IgG1 antibody blocking PD-L1. With this dual-targeting strategy, BA enhances efficacy over that of monotherapies in preclinical studies. In this study, NHS-muIL12 and BA combination therapy enhanced antitumor activity, prolonged survival, and induced tumor-specific antitumor immunity. This combination therapy increased tumor-specific CD8+ T cells and induced immune profiles, consistent with the activation of both adaptive and innate immune systems. In addition, BA reduced lung metastasis in the 4T1 model. Collectively, these findings could support clinical trials designed to investigate NHS-IL12 and BA combination therapy for patients with advanced solid tumors.

5.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35858707

RESUMEN

BACKGROUND: Bintrafusp alfa (BA) is a bifunctional fusion protein designed for colocalized, simultaneous inhibition of two immunosuppressive pathways, transforming growth factor-ß (TGF-ß) and programmed death-ligand 1 (PD-L1), within the tumor microenvironment (TME). We hypothesized that targeting PD-L1 to the tumor by BA colocalizes the TGF-ß trap (TGF-ßRII) to the TME, enabling it to sequester TGF-ß in the tumor more effectively than systemic TGF-ß blockade, thereby enhancing antitumor activity. METHODS: Multiple technologies were used to characterize the TGF-ß trap binding avidity. BA versus combinations of anti-PD-L1 and TGF-ß trap or the pan-TGF-ß antibody fresolimumab were compared in proliferation and two-way mixed lymphocyte reaction assays. Immunophenotyping of tumor-infiltrating lymphocytes (TILs) and RNA sequencing (RNAseq) analysis assessing stromal and immune landscape following BA or the combination therapy were performed in MC38 tumors. TGF-ß and PD-L1 co-expression and their associated gene signatures in MC38 tumors and human lung carcinoma tissue were studied with single-cell RNAseq (scRNAseq) and immunostaining. BA-induced internalization, degradation, and depletion of TGF-ß were investigated in vitro. RESULTS: BA and fresolimumab had comparable intrinsic binding to TGF-ß1, but there was an ~80× avidity-based increase in binding affinity with BA. BA inhibited cell proliferation in TGF-ß-dependent and PD-L1-expressing cells more potently than TGF-ß trap or fresolimumab. Compared with the combination of anti-PD-L1 and TGF-ß trap or fresolimumab, BA enhanced T cell activation in vitro and increased TILs in MC38 tumors, which correlated with efficacy. BA induced distinct gene expression in the TME compared with the combination therapy, including upregulation of immune-related gene signatures and reduced activities in TGF-ß-regulated pathways, such as epithelial-mesenchymal transition, extracellular matrix deposition, and fibrosis. Regulatory T cells, macrophages, immune cells of myeloid lineage, and fibroblasts were key PD-L1/TGF-ß1 co-expressing cells in the TME. scRNAseq analysis suggested BA modulation of the macrophage phenotype, which was confirmed by histological assessment. PD-L1/TGF-ß1 co-expression was also seen in human tumors. Finally, BA induced TGF-ß1 internalization and degradation in the lysosomes. CONCLUSION: BA more effectively blocks TGF-ß by targeting TGF-ß trap to the tumor via PD-L1 binding. Such colocalized targeting elicits distinct and superior antitumor responses relative to single agent combination therapy.


Asunto(s)
Neoplasias Pulmonares , Factor de Crecimiento Transformador beta , Antígeno B7-H1 , Humanos , Factores Inmunológicos , Receptor de Muerte Celular Programada 1 , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Microambiente Tumoral
6.
Cancer Cell ; 39(10): 1388-1403.e10, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34506739

RESUMEN

Localized radiotherapy (RT) induces an immunogenic antitumor response that is in part counterbalanced by activation of immune evasive and tissue remodeling processes, e.g., via upregulation of programmed cell death-ligand 1 (PD-L1) and transforming growth factor ß (TGF-ß). We report that a bifunctional fusion protein that simultaneously inhibits TGF-ß and PD-L1, bintrafusp alfa (BA), effectively synergizes with radiotherapy, leading to superior survival in multiple therapy-resistant murine tumor models with poor immune infiltration. The BA + RT (BART) combination increases tumor-infiltrating leukocytes, reprograms the tumor microenvironment, and attenuates RT-induced fibrosis, leading to reconstitution of tumor immunity and regression of spontaneous lung metastases. Consistently, the beneficial effects of BART are in part reversed by depletion of cytotoxic CD8+ T cells. Intriguingly, targeting of the TGF-ß trap to PD-L1+ endothelium and the M2/lipofibroblast-like cell compartment by BA attenuated late-stage RT-induced lung fibrosis. Together, the results suggest that the BART combination has the potential to eradicate therapy-resistant tumors while sparing normal tissue, further supporting its clinical translation.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Evasión Inmune/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Microambiente Tumoral
7.
Clin Cancer Res ; 14(5): 1487-93, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18316573

RESUMEN

PURPOSE: The half-life of the antiangiogenic molecule endostatin that has been used in clinical trial is short ( approximately 2 h). In addition, approximately 50% of the clinical grade endostatin molecules lack four amino acids at their NH(2) termini. Lack of these amino acids gives rise to a molecule that is devoid of zinc, resulting in no antitumor activity. Our goal was to develop a new version of endostatin that does not show such deficiency. EXPERIMENTAL DESIGN: A recombinant human endostatin conjugated to the Fc domain of IgG was constructed and expressed in mammalian cell culture. The presence of Fc has been shown by previous investigators to play a major role in increasing the half-life of the molecule. Fc-endostatin was tested in tumor-bearing mice, and its half-life was compared with the clinical grade endostatin. RESULTS: The antitumor dose of Fc-endostatin was found to be approximately 100 times less than the clinical grade endostatin. The half-life of Fc-endostatin in the circulation was found to be weeks rather than hours, as observed for endostatin alone. In addition, a U-shaped curve was observed for antitumor activity of endostatin as a function of endostatin concentration delivered to the animals. CONCLUSION: Fc-endostatin is a superior molecule to the original clinical endostatin. Due to its long half-life, the amount of protein required is substantially reduced compared with the clinically tested endostatin. Furthermore, in view of the U-shaped curve of efficacy observed for endostatin, we estimate that the requirement for Fc-endostatin is approximately 700-fold less than endostatin alone. The half-life of endostatin is similar to that of vascular endothelial growth factor-Trap and Avastin, two other antiangiogenic reagents. We conclude that a new clinical trial of endostatin, incorporating Fc, may benefit cancer patients.


Asunto(s)
Endostatinas/inmunología , Endostatinas/farmacocinética , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Melanoma Experimental/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Recombinantes de Fusión/farmacocinética , Animales , Apoptosis , Ensayo de Inmunoadsorción Enzimática , Semivida , Humanos , Técnicas para Inmunoenzimas , Etiquetado Corte-Fin in Situ , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Mutación/genética , Neoplasias Pancreáticas/patología , Proteínas Recombinantes/uso terapéutico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Sci Transl Med ; 10(424)2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343622

RESUMEN

Antibodies targeting immune checkpoints are emerging as potent and viable cancer therapies, but not all patients respond to these as single agents. Concurrently targeting additional immunosuppressive pathways is a promising approach to enhance immune checkpoint blockade, and bifunctional molecules designed to target two pathways simultaneously may provide a strategic advantage over the combination of two single agents. M7824 (MSB0011359C) is a bifunctional fusion protein composed of a monoclonal antibody against programmed death ligand 1 (PD-L1) fused to the extracellular domain of human transforming growth factor-ß (TGF-ß) receptor II, which functions as a "trap" for all three TGF-ß isoforms. We demonstrate that M7824 efficiently, specifically, and simultaneously binds PD-L1 and TGF-ß. In syngeneic mouse models, M7824 suppressed tumor growth and metastasis more effectively than treatment with either an anti-PD-L1 antibody or TGF-ß trap alone; furthermore, M7824 extended survival and conferred long-term protective antitumor immunity. Mechanistically, the dual anti-immunosuppressive function of M7824 resulted in activation of both the innate and adaptive immune systems, which contributed to M7824's antitumor activity. Finally, M7824 was an effective combination partner for radiotherapy or chemotherapy in mouse models. Collectively, our preclinical data demonstrate that simultaneous blockade of the PD-L1 and TGF-ß pathways by M7824 elicits potent and superior antitumor activity relative to monotherapies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos Monoclonales/química , Inmunoterapia/métodos , Ratones , Receptor de Muerte Celular Programada 1/química , Factor de Crecimiento Transformador beta/química
10.
Clin Cancer Res ; 23(19): 5869-5880, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28679778

RESUMEN

Purpose: To determine whether combination therapy with NHS-muIL12 and the anti-programmed death ligand 1 (PD-L1) antibody avelumab can enhance antitumor efficacy in preclinical models relative to monotherapies.Experimental Design: BALB/c mice bearing orthotopic EMT-6 mammary tumors and µMt- mice bearing subcutaneous MC38 tumors were treated with NHS-muIL12, avelumab, or combination therapy; tumor growth and survival were assessed. Tumor recurrence following remission and rechallenge was evaluated in EMT-6 tumor-bearing mice. Immune cell populations within spleen and tumors were evaluated by FACS and IHC. Immune gene expression in tumor tissue was profiled by NanoString® assay and plasma cytokine levels were determined by multiplex cytokine assay. The frequency of tumor antigen-reactive IFNγ-producing CD8+ T cells was evaluated by ELISpot assay.Results: NHS-muIL12 and avelumab combination therapy enhanced antitumor efficacy relative to either monotherapy in both tumor models. Most EMT-6 tumor-bearing mice treated with combination therapy had complete tumor regression. Combination therapy also induced the generation of tumor-specific immune memory, as demonstrated by protection against tumor rechallenge and induction of effector and memory T cells. Combination therapy enhanced cytotoxic NK and CD8+ T-cell proliferation and T-bet expression, whereas NHS-muIL12 monotherapy induced CD8+ T-cell infiltration into the tumor. Combination therapy also enhanced plasma cytokine levels and stimulated expression of a greater number of innate and adaptive immune genes compared with either monotherapy.Conclusions: These data indicate that combination therapy with NHS-muIL12 and avelumab increased antitumor efficacy in preclinical models, and suggest that combining NHS-IL12 and avelumab may be a promising approach to treating patients with solid tumors. Clin Cancer Res; 23(19); 5869-80. ©2017 AACR.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Inmunoglobulina G/administración & dosificación , Inmunoterapia , Interleucina-12/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Inmunoglobulina G/inmunología , Interleucina-12/administración & dosificación , Interleucina-12/genética , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Proteínas Recombinantes de Fusión/inmunología
11.
Protein Eng Des Sel ; 18(1): 1-10, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15790575

RESUMEN

Leptin plays a central role in the homeostasis of body weight through its regulatory effects on appetite and energy expenditure, yet in trials as a therapeutic agent for the treatment of obesity in humans it has been disappointing. The poor clinical efficacy of leptin results from its short circulating half-life, low potency and poor solubility, necessitating large and frequent doses to obtain even modest clinical benefit. Engineered Fc-leptin immunofusins, consisting of the Fc fragment of an immunoglobulin gamma chain followed by leptin, exhibit improved pharmacological properties with very consistent and potent biological activities. Furthermore, in extending the circulating half-life of the protein in vivo from a few minutes for leptin to many hours for Fc-leptin, these proteins have the potential to reduce drastically the dosage and frequency of administration required to obtain clinical benefit. The results of this study show that the engineered leptin immunofusins described here have significantly enhanced pharmacological properties in comparison with the recombinant leptin that was used in clinical trials. As such, they could represent an important step towards a therapeutically superior form of leptin if the disappointing performance of leptin in early clinical trials was due to its poor pharmacological properties rather than any conceptual weakness in the strategy of using leptin for the treatment of obesity and its related disorders.


Asunto(s)
Leptina/genética , Leptina/uso terapéutico , Obesidad/tratamiento farmacológico , Ingeniería de Proteínas , Animales , Secuencia de Bases , Western Blotting , Clonación Molecular , Cartilla de ADN , Esquema de Medicación , Humanos , Leptina/administración & dosificación , Leptina/farmacocinética , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/uso terapéutico
12.
Protein Eng Des Sel ; 18(3): 111-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15820978

RESUMEN

Erythropoietin (Epo) is a cytokine that controls the production of red blood cells (RBCs). Epo acts continuously on RBC precursors to prevent apoptosis, so it is important to maintain high levels of Epo activity when treating anemic patients. We describe here modified human Epo [Epo(NDS)] with mutations His32Gly, Cys33Pro, Trp88Cys and Pro90Ala that result in the rearrangement of the disulfide bonding pattern from Cys29-Cys33 to Cys29-Cys88 and that, in the context of an Fc-Epo(NDS) fusion protein, lead to significantly improved properties. Fc-Epo was secreted from NS/0 myeloma cells as about 35% high molecular weight aggregates, was unstable upon removal of N-linked oligosaccharides and showed poor pharmacokinetics and little efficacy in mice. In contrast, a corresponding Fc-Epo(NDS) was secreted almost exclusively as a unit dimer, was relatively stable to removal of N-linked oligosaccharides, had much improved pharmacokinetic properties and had a significantly improved effect on RBC production. These results indicate that rearrangement of the disulfide bonding pattern in a therapeutic protein can have a significant effect on pharmacokinetics and, potentially, the dosing schedule of a protein drug.


Asunto(s)
Eritropoyetina/química , Eritropoyetina/farmacocinética , Ingeniería de Proteínas/métodos , Receptores Fc/química , Secuencia de Aminoácidos , Animales , Apoptosis , Bovinos , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Codón , Simulación por Computador , Cisteína/química , Dimerización , Disulfuros/química , Eritropoyetina/genética , Glicosilación , Humanos , Cinética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Oligosacáridos/química , Péptidos/química , Conformación Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Ratas , Receptores Fc/genética , Proteínas Recombinantes de Fusión/química , Homología de Secuencia de Aminoácido , Factores de Tiempo , Transfección , Tripsina/química
13.
Clin Cancer Res ; 8(1): 210-6, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11801561

RESUMEN

PURPOSE: Previous studies showed that reducing the interaction of antibody-interleukin 2 immunocytokines with Fc receptors improved their circulating half-life in mice and increased their antitumor activity. We sought to modify sequences that would increase half-life but retain the ability to activate Fc receptor-mediated effector functions. EXPERIMENTAL DESIGN: Modified immunocytokines were assessed in vitro for effector function and protease sensitivity and in vivo for pharmacokinetic and antitumor activities in an syngeneic tumor regression model. RESULTS: Single amino acid changes in the junction sequence between the antibody and interleukin-2 components had dramatic effects on circulating half-life and antitumor activity. This effect was independent of Fc receptor binding to either Fcgamma receptors or the Fc protection receptor, but was associated with changes in susceptibility to intracellular proteases. CONCLUSIONS: We have identified modifications that dramatically improve the circulating half-life of immunocytokines based on increased resistance to intracellular degradation and thus have demonstrated how these molecules can be recycled in and out of an intracellular compartment. Use of these improved immunocytokines with highly favorable pharmacokinetic properties and retained effector functions should lead to more effective treatment of epithelial cancers.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Interleucina-2/inmunología , Interleucina-2/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Animales , Catepsina L , Catepsinas/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/prevención & control , Semivida , Humanos , Inmunoconjugados/farmacocinética , Fragmentos Fc de Inmunoglobulinas/inmunología , Interleucina-2/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Mutación , Proteínas Recombinantes de Fusión/uso terapéutico , Distribución Tisular , Transfección , Trasplante Heterólogo , Células Tumorales Cultivadas
14.
J Interferon Cytokine Res ; 24(9): 560-72, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15450132

RESUMEN

Interferon-alpha (IFN-alpha), in conjunction with ribavirin, is the current standard for the treatment of chronic hepatitis C virus (HCV) infection. This treatment requires frequent dosing, with a significant risk of the development of anti-IFN-alpha neutralizing antibodies that correlates with lack of efficacy or relapse. We have developed an IFN-alpha linked to the Fc region of human IgG1 for improved half-life and less frequent dosing. We have also identified, using a human T cell proliferation assay, three regions of IFN-alpha2b that are potentially immunogenic, and a variant containing a total of six mutations within these regions was made. This variant was made as a fusion to Fc either with or without a flexible linker between the fusion partners. Both configurations of the variant were less active than native IFN-alpha alone, although the variant containing the flexible linker had in vitro antiviral activity within the range of other modified IFN-alphas currently in clinical use. Peptides spanning the modified regions were tested in T cell proliferation assays and found to be less immunogenic than native controls when using peripheral blood mononuclear cells (PBMCs) from both healthy individuals and HCV-infected patients who had been treated previously with IFN-alpha2b.


Asunto(s)
Antivirales/química , Hepatitis C Crónica/tratamiento farmacológico , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Interferón-alfa/genética , Secuencia de Aminoácidos , Antivirales/uso terapéutico , Línea Celular , Epítopos de Linfocito T/análisis , Hepacivirus/efectos de los fármacos , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Fragmentos de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Interferón alfa-2 , Interferón-alfa/química , Interferón-alfa/uso terapéutico , Datos de Secuencia Molecular , Péptidos/genética , Mutación Puntual , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes
15.
Cancer Biol Ther ; 3(6): 551-8, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15044850

RESUMEN

Prostate-Specific Membrane Antigen (PSMA) is a glutamate carboxypeptidase II that is highly expressed by both normal and malignant prostate epithelial cells and by the neovasculature of many tumor types but is not expressed by endothelial cells in normal tissue. PSMA possesses the hydrolytic properties of an N-acetylated alpha-linked acidic dipeptidase (NAALADase) and also functions as a pteroyl poly-gamma-glutamyl carboxypeptidase (i.e., folate hydrolase). Therefore, PSMA can be targeted for activation of peptide-based prodrugs within the extracellular fluid of prostate cancers. In this study, methotrexate-based peptide analogs were evaluated to identify PSMA selective substrates that are also stable to nonspecific hydrolysis in human and mouse plasma. These methotrexate analogs were also characterized for in vitro toxicity against PSMA and nonPSMA producing human cancer cell lines. Analogs containing gamma-linked glutamate residues were most efficiently hydrolyzed by PSMA, but were unstable in plasma. Analogs containing both alpha- and gamma-linked acidic amino acids were less efficiently hydrolyzed by PSMA but were most stable in plasma. Analogs were 5-10 fold more selectively toxic in vitro in the presence of active PSMA. These studies have identified PSMA selective, plasma stable peptide substrates that can be incorporated into prodrugs targeted for activation by PSMA within prostate cancer sites.


Asunto(s)
Antígenos de Superficie/metabolismo , Glutamato Carboxipeptidasa II/metabolismo , Metotrexato/metabolismo , Fragmentos de Péptidos/metabolismo , Profármacos , Neoplasias de la Próstata/metabolismo , Animales , Antígenos de Superficie/genética , Ácido Aspártico/metabolismo , Western Blotting , Líquido Extracelular , Glutamato Carboxipeptidasa II/genética , Ácido Glutámico/metabolismo , Humanos , Cadenas gamma de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/genética , Masculino , Ratones , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Células Tumorales Cultivadas/metabolismo
16.
Microbiol Spectr ; 2(1): AID-0007-2012, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26082127

RESUMEN

Advanced molecular biology techniques developed during the past few decades have allowed the industry to exploit and commercialize the natural defense mechanisms that antibodies provide. This review discusses the latest advances in antibody-engineering technologies to enhance clinical efficacy and outcomes. For the constant regions, the choice of the antibody class and isotype has to be made carefully to suit the therapeutic applications. Engineering of the Fc region, either by direct targeted mutagenesis or by modifying the nature of its N-glycan, has played an important role in recent years in increasing half-life or controlling effector functions. The variable regions of the antibody are responsible for binding affinity and exquisite specificity to the target molecule, which together with the Fc determine the drug's efficacy and influence the drug dose required to obtain the desired effectiveness. A key requirement during antibody development is therefore to affinity mature the variable regions when necessary, so that they bind the therapeutic target with sufficiently high affinity to guarantee effective occupancy over prolonged periods. If the antibody was obtained from a non-human source, such as rodents, a humanization process has to be applied to minimize immunogenicity while maintaining the desired binding affinity and selectivity. Finally, we discuss the next next-generation antibodies, such as antibody-drug conjugates, bispecific antibodies, and immunocytokines, which are being developed to meet future challenges.


Asunto(s)
Anticuerpos/genética , Anticuerpos/inmunología , Ingeniería de Proteínas/métodos , Animales , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
17.
Clin Cancer Res ; 17(11): 3673-85, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531812

RESUMEN

PURPOSE: The goal of the study was to engineer a form of interleukin 2 (IL-2) that, when delivered as a tumor-specific antibody fusion protein, retains the ability to stimulate an antitumor immune response via interaction with the high-affinity IL-2 receptor but has lower toxicity because of the reduced activation of the intermediate-affinity IL-2 receptor. EXPERIMENTAL DESIGN: We investigated changes in the proposed toxin motif of IL-2 by introducing a D20T mutation that has little effect on the activity of free IL-2. We expressed this IL-2 variant as a fusion protein with an antibody (NHS76) that targets the necrotic core of tumors and characterized this molecule (NHS-IL2LT) in vitro and in vivo. RESULTS: NHS-IL2LT was shown to have near normal biological activity in vitro by using T-cell lines expressing the high-affinity IL-2 receptor, but little or no activity by using cell lines expressing only the intermediate IL-2 receptor. Relative to the control antibody fusion protein containing wild-type IL-2, NHS-IL2LT retained antitumor activity against established neuroblastoma and non-small cell lung cancer metastases in syngeneic mouse tumor models but was much better tolerated in immune-competent mice and in cynomolgus monkeys. CONCLUSIONS: The qualities of low toxicity and single-agent efficacy shown suggest that NHS-IL2LT is a good candidate for therapeutic approaches combining standard cytotoxic and immune therapies. In fact, this molecule (also known as Selectikine or EMD 521873) is currently in phase I clinical trial.


Asunto(s)
Interleucina-2/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Región Variable de Inmunoglobulina/metabolismo , Interleucina-2/química , Interleucina-2/genética , Interleucina-2/inmunología , Macaca fascicularis , Ratones , Mieloma Múltiple/inmunología , Mutación , Neuroblastoma/inmunología , Ingeniería de Proteínas , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
18.
Protein Eng Des Sel ; 23(4): 195-202, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20299542

RESUMEN

Bispecific antibodies and asymmetric Fc fusion proteins offer opportunities for important advances in therapeutics. Bivalent IgG depends upon in vivo dimerization of its heavy chains, mediated by homodimeric association of its C(H)3 domains. We have developed a heterodimeric Fc platform that supports the design of bispecific and asymmetric fusion proteins by devising strand-exchange engineered domain (SEED) C(H)3 heterodimers. These derivatives of human IgG and IgA C(H)3 domains create complementary human SEED C(H)3 heterodimers that are composed of alternating segments of human IgA and IgG C(H)3 sequences. The resulting pair of SEED C(H)3 domains preferentially associates to form heterodimers when expressed in mammalian cells. SEEDbody (Sb) fusion proteins consist of [IgG1 hinge]-C(H)2-[SEED C(H)3], that may be genetically linked to one or more fusion partners. This investigation reports on the generation of mono-Fab-Sb and Sb-IL2 monocytokine as models. They were expressed at high levels in NS/0 cells, purified on recombinant protein A resin and were well-behaved in solution. When administered intravenously to mice, Sb pharmacokinetics exhibited the long serum half-life extensions typical of comparable Fc-containing immunofusion and IgG1 controls.


Asunto(s)
Anticuerpos Biespecíficos/química , Fragmentos Fc de Inmunoglobulinas/química , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/química , Secuencia de Aminoácidos , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos , Dimerización , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/química , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia
19.
Clin Cancer Res ; 15(18): 5923-30, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19737959

RESUMEN

PURPOSE: Immunocytokine (IC) hu14.18-IL2 is a fusion protein of humanized antidisialoganglioside (GD2) antibody (hu14.18) and interleukin (IL)-2. Sixty-one melanoma and neuroblastoma patients received IC in phase I/Ib studies. Patient sera were examined in ELISA to determine if an anti-IC antibody response occurred during treatment. EXPERIMENTAL DESIGN: Serum was assayed for anti-idiotypic antibody (anti-id Ab) based on ability to bridge biotinylated hu14.18 to plate-bound hu14.18 and ability to inhibit binding of hu14.18 to GD2 antigen and/or murine anti-idiotypic antibody. ELISA was also used to detect antibodies to the Fc-IL2 end of hu14.18-IL2. RESULTS: Thirty-two patients (52%) developed an anti-idiotypic antibody response (absorbance, >0.7) in the bridge ELISA. Twelve patients (20%) had an intermediate response, whereas 17 patients (28%) were negative (adsorbance, <0.3). The development of antibody to hu14.18-IL2 detected in the bridge ELISA was not related to the dose of hu14.18-IL2. Twenty of 33 adult patients (61%) demonstrated an anti-idiotypic antibody response based on binding inhibition ELISA. The anti-idiotypic response was inversely correlated (P < 0.002) with IC measured during the second course of treatment, indicating that development of anti-idiotypic antibodies interfered with detection of circulating hu14.18-IL2. All patients developed some inhibitory activity in the binding inhibition assay designed to detect antibodies to the Fc-IL2 region of the IC. There was a positive correlation between the peak serum level of IC in course 1 and the anti-Fc-IL2 response. CONCLUSIONS: Patients treated with hu14.18-IL2 developed anti-idiotypic antibodies and anti Fc-IL2 antibodies. No association was seen between development of anti-IC antibodies and clinical toxicity.


Asunto(s)
Interleucina-2/genética , Interleucina-2/inmunología , Melanoma/genética , Melanoma/inmunología , Neuroblastoma/genética , Neuroblastoma/inmunología , Adulto , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Reacciones Antígeno-Anticuerpo , Línea Celular Tumoral , Niño , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-2/administración & dosificación , Interleucina-2/sangre , Interleucina-2/uso terapéutico , Melanoma/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico
20.
Cancer Immunol Immunother ; 56(4): 447-57, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16874486

RESUMEN

IL-12 is a cytokine which showed anti-tumor effects in clinical trials, but also produced serious toxicity. We describe a fusion protein, huBC1-IL12, designed to achieve an improved therapeutic index by specifically targeting IL-12 to tumor and tumor vasculature. huBC-1 is a humanized antibody that targets a cryptic sequence of the human ED-B-containing fibronectin isoform, B-FN, present in the subendothelial extracellular matrix of most aggressive tumors. B-FN is oncofetal and angiogenesis-associated, and is undetectable in most normal adult tissues. The original murine BC-1 antibody has been used successfully for immunoscintigraphy to image brain tumor mass in glioblastoma patients. In huBC1-IL12, each of the IgG heavy chains is genetically fused to the N-terminus of the IL-12 p35 subunit, which in turn is disulfide-bonded to the p40 subunit, resulting in a hexameric molecule of MW of approximately 300 kDa. Since human IL-12 has no biological activity in mice, we produced huBC1-muIL12 as a surrogate molecule for animal tumor models. Despite the relatively poor PK profile of this molecule in mice and the apparent drawbacks of xenogeneic models in SCID mice, which lack T and B cells, one cycle of treatment with huBC1-muIL12 was efficacious in the PC3mm2, A431, and HT29 subcutaneous tumor models and PC3mm2 lung metastasis model. This molecule also was found to have surprisingly low toxicity in immunocompetent mice. A fusion protein that contains human IL-12 (huBC1-huIL12), which is a suitable molecule for investigation as a therapeutic, has also been produced. This protein has been shown to have a longer serum half-life than huBC1-muIL12 in mice, and retains both antigen binding and IL-12 activity in in vitro assays.


Asunto(s)
Fibronectinas/inmunología , Interleucina-12/uso terapéutico , Neoplasias Experimentales/terapia , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Inmunoglobulina G , Cadenas Pesadas de Inmunoglobulina , Inmunohistoquímica , Interleucina-12/inmunología , Interleucina-12/farmacocinética , Masculino , Ratones , Neoplasias Experimentales/inmunología , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/farmacocinética , Isoformas de Proteínas/uso terapéutico , Proteínas Recombinantes de Fusión/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA