Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Immunol Rev ; 270(1): 178-92, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26864112

RESUMEN

The activation of the immune system for a selective removal of tumor cells represents an attractive strategy for the treatment of metastatic malignancies, which cannot be cured by existing methodologies. In this review, we examine the design and therapeutic potential of immunocytokines and bispecific antibodies, two classes of bifunctional products which can selectively activate the immune system at the tumor site. Certain protein engineering aspects, such as the choice of the antibody format, are common to both classes of therapeutic agents and can have a profound impact on tumor homing performance in vivo of individual products. However, immunocytokines and bispecific antibodies display different mechanisms of action. Future research activities will reveal whether an additive of even synergistic benefit can be obtained from the judicious combination of these two types of biopharmaceutical agents.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Citocinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/genética , Especificidad de Anticuerpos , Antígenos de Neoplasias/inmunología , Ensayos Clínicos como Asunto , Citocinas/química , Citocinas/genética , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Neoplasias/metabolismo , Unión Proteica , Ingeniería de Proteínas
2.
J Biol Chem ; 291(43): 22496-22508, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27582495

RESUMEN

The Sso7d protein from the hyperthermophilic archaeon Sulfolobus solfataricus is an attractive binding scaffold because of its small size (7 kDa), high thermal stability (Tm of 98 °C), and absence of cysteines and glycosylation sites. However, as a DNA-binding protein, Sso7d is highly positively charged, introducing a strong specificity constraint for binding epitopes and leading to nonspecific interaction with mammalian cell membranes. In the present study, we report charge-neutralized variants of Sso7d that maintain high thermal stability. Yeast-displayed libraries that were based on this reduced charge Sso7d (rcSso7d) scaffold yielded binders with low nanomolar affinities against mouse serum albumin and several epitopes on human epidermal growth factor receptor. Importantly, starting from a charge-neutralized scaffold facilitated evolutionary adaptation of binders to differentially charged epitopes on mouse serum albumin and human epidermal growth factor receptor, respectively. Interestingly, the distribution of amino acids in the small and rigid binding surface of enriched rcSso7d-based binders is very different from that generally found in more flexible antibody complementarity-determining region loops but resembles the composition of antibody-binding energetic hot spots. Particularly striking was a strong enrichment of the aromatic residues Trp, Tyr, and Phe in rcSso7d-based binders. This suggests that the rigidity and small size of this scaffold determines the unusual amino acid composition of its binding sites, mimicking the energetic core of antibody paratopes. Despite the high frequency of aromatic residues, these rcSso7d-based binders are highly expressed, thermostable, and monomeric, suggesting that the hyperstability of the starting scaffold and the rigidness of the binding surface confer a high tolerance to mutation.


Asunto(s)
Proteínas Arqueales/química , Proteínas de Unión al ADN/química , Calor , Sulfolobus solfataricus/química , Aminoácidos Aromáticos/química , Aminoácidos Aromáticos/genética , Animales , Proteínas Arqueales/genética , Sitios de Unión , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Ratones , Estabilidad Proteica , Sulfolobus solfataricus/genética
3.
Exp Hematol ; 95: 31-45, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33484750

RESUMEN

Precise replacement of diseased or dysfunctional organs is the goal of regenerative medicine and has appeared to be a distant goal for a long time. In the field of hematopoietic stem cell transplantation, this goal is now becoming tangible as gene-editing technologies and novel conditioning agents are entering the clinical arena. Targeted immunologic depletion of hematopoietic stem cells (HSCs), which are at the very root of the hematopoietic system, will enable more selective and potentially more effective hematopoietic stem cell transplantation in patients with hematological diseases. In contrast to current conditioning regimes based on ionizing radiation and chemotherapy, immunologic conditioning will spare mature hematopoietic cells and cause substantially less inflammation and unspecific collateral damage to other organs. Biological agents that target the stem cell antigen CD117 are the frontrunners for this purpose and have exhibited preclinical activity in depletion of healthy HSCs. The value of anti-CD117 antibodies as conditioning agents is currently being evaluated in early clinical trials. Whereas mild, antibody-based immunologic conditioning concepts might be appropriate for benign hematological disorders in which incomplete replacement of diseased cells is sufficient, higher efficacy will be required for treatment and elimination of hematologic stem cell malignancies such as acute myeloid leukemia and myelodysplastic syndrome. Antibody-drug conjugates, bispecific T-cell engaging and activating antibodies (TEAs), or chimeric antigen receptor (CAR) T cells might offer increased efficacy compared with naked antibodies and yet higher tolerability and safety compared with current genotoxic conditioning approaches. Here, we summarize the current state regarding immunologic conditioning concepts for the treatment of HSC disorders and outline potential future developments.


Asunto(s)
Antineoplásicos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Inmunoterapia/métodos , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Acondicionamiento Pretrasplante/métodos , Animales , Anticuerpos Biespecíficos/uso terapéutico , Protocolos de Ensayos Clínicos como Asunto , Hematopoyesis Clonal , Terapia Combinada , Enfermedades Genéticas Congénitas/terapia , Neoplasias Hematológicas/terapia , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Inmunoterapia Adoptiva , Inmunotoxinas/uso terapéutico , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/terapia , Preleucemia/terapia , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/inmunología , Medición de Riesgo , Inmunodeficiencia Combinada Grave/terapia
4.
Leukemia ; 34(10): 2688-2703, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32358567

RESUMEN

Acute myeloid leukemia (AML) initiating and sustaining cells maintain high cell-surface similarity with their cells-of-origin, i.e., hematopoietic stem and progenitor cells (HSPCs), and identification of truly distinguishing leukemia-private antigens has remained elusive to date. To nonetheless utilize surface antigen-directed immunotherapy in AML, we here propose targeting both, healthy and malignant human HSPC, by chimeric antigen receptor (CAR) T-cells with specificity against CD117, the cognate receptor for stem cell factor. This approach should spare most mature hematopoietic cells and would require CAR T termination followed by subsequent transplantation of healthy HSPCs to rescue hematopoiesis. We successfully generated anti-CD117 CAR T-cells from healthy donors and AML patients. Anti-CD117 CAR T-cells efficiently targeted healthy and leukemic CD117-positive cells in vitro. In mice xenografted with healthy human hematopoiesis, they eliminated CD117-expressing, but not CD117-negative human cells. Importantly, in mice xenografted with primary human CD117-positive AML, they eradicated disease in a therapeutic setting. Administration of ATG in combination with rituximab, which binds to the co-expressed CAR T-cell transduction/selection marker RQR8, led to CAR T-cell depletion. Thus, we here provide the first proof of concept for the generation and preclinical efficacy of CAR T-cells directed against CD117-expressing human hematopoietic cells.


Asunto(s)
Inmunoterapia Adoptiva , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Biomarcadores , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/terapia , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Depleción Linfocítica , Ratones , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cell Rep ; 30(4): 997-1012.e6, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995768

RESUMEN

Control of established chronic lymphocytic choriomeningitis virus (LCMV) infection requires the production of neutralizing antibodies, but it remains unknown how the ensemble of antibodies evolves during ongoing infection. Here, we analyze the evolution of antibody responses during acute or chronic LCMV infection, combining quantitative functional assays and time-resolved antibody repertoire sequencing. We establish that antibody responses initially converge in both infection types on a functional and repertoire level, but diverge later during chronic infection, showing increased clonal diversity, the appearance of mouse-specific persistent clones, and distinct phylogenetic signatures. Chronic infection is characterized by a longer-lasting germinal center reaction and a continuous differentiation of plasma cells, resulting in the emergence of higher-affinity plasma cells exhibiting increased antibody secretion rates. Taken together, our findings reveal the emergence of a personalized antibody response in chronic infection and support the concept that maintaining B cell diversity throughout chronic LCMV infection correlates with the development of infection-resolving antibodies.


Asunto(s)
Anticuerpos Antivirales/inmunología , Diversidad de Anticuerpos/genética , Evolución Clonal/inmunología , Inmunidad Humoral/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Enfermedad Aguda , Animales , Formación de Anticuerpos/genética , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Enfermedad Crónica , Evolución Clonal/genética , Centro Germinal/metabolismo , Inmunoglobulina G/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Endogámicos C57BL , Filogenia , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Hipermutación Somática de Inmunoglobulina
6.
J Invest Dermatol ; 139(6): 1339-1348, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30543899

RESUMEN

The recombinant murine IgG2a antibody TA99, directed against a melanoma antigen, was used to study combination modalities that potentiate antibody-dependent cell cytotoxicity. As previously reported, IgG2a(TA99) was extremely efficacious in preventing the growth of B16 lung metastases. However, the same antibody mediated only minimal tumor growth retardation when used to treat established neoplastic masses. The therapeutic activity of IgG2a(TA99) could be substantially enhanced by co-administration with an antibody-cytokine fusion (TA99-murine tumor necrosis factor [mTNF]), consisting of the TA99 antibody in single-chain variable fragment format fused to murine TNF. This fusion protein efficiently killed endothelial cells in vitro and displayed only minimal activity against B16 melanoma cells. In vivo, TA99-mTNF boosted the influx of natural killer cells and macrophages into B16 melanoma lesions. Therapy studies with two different administration schedules showed that the combination of TA99-mTNF and IgG2a(TA99) was superior to the individual products used as single agents. The combination treatment converted most of the tumor mass into a necrotic lesion, but a vital tumor rim eventually regrew, even when dacarbazine was included in the therapeutic regimen. The treatment modality described in this article may be applicable to the treatment of melanoma patients, given the specificity of the gp75 antigen and its conservation across species.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoconjugados/administración & dosificación , Melanoma Experimental/tratamiento farmacológico , Glicoproteínas de Membrana/inmunología , Oxidorreductasas/inmunología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Anticuerpos Monoclonales de Origen Murino/genética , Anticuerpos Monoclonales de Origen Murino/aislamiento & purificación , Células CHO , Línea Celular Tumoral/trasplante , Cricetulus , Esquema de Medicación , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Inmunoconjugados/genética , Inmunoconjugados/aislamiento & purificación , Inmunoglobulina G/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/aislamiento & purificación
7.
Protein Sci ; 26(10): 1942-1952, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28691351

RESUMEN

Designed armadillo repeat proteins (dArmRP) are α-helical solenoid repeat proteins with an extended peptide binding groove that were engineered to develop a generic modular technology for peptide recognition. In this context, the term "peptide" not only denotes a short unstructured chain of amino acids, but also an unstructured region of a protein, as they occur in termini, loops, or linkers between folded domains. Here we report two crystal structures of dArmRPs, in complex with peptides fused either to the N-terminus of Green Fluorescent Protein or to the C-terminus of a phage lambda protein D. These structures demonstrate that dArmRPs bind unfolded peptides in the intended conformation also when they constitute unstructured parts of folded proteins, which greatly expands possible applications of the dArmRP technology. Nonetheless, the structures do not fully reflect the binding behavior in solution, that is, some binding sites remain unoccupied in the crystal and even unexpected peptide residues appear to be bound. We show how these differences can be explained by restrictions of the crystal lattice or the composition of the crystallization solution. This illustrates that crystal structures have to be interpreted with caution when protein-peptide interactions are characterized, and should always be correlated with measurements in solution.


Asunto(s)
Proteínas del Dominio Armadillo/química , Proteínas del Dominio Armadillo/metabolismo , Péptidos/química , Péptidos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas del Dominio Armadillo/genética , Cristalización , Modelos Moleculares , Péptidos/genética , Conformación Proteica , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/genética
8.
Sci Rep ; 7(1): 5831, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28724936

RESUMEN

Ras is at the hub of signal transduction pathways controlling cell proliferation and survival. Its mutants, present in about 30% of human cancers, are major drivers of oncogenesis and render tumors unresponsive to standard therapies. Here we report the engineering of a protein scaffold for preferential binding to K-Ras G12D. This is the first reported inhibitor to achieve nanomolar affinity while exhibiting specificity for mutant over wild type (WT) K-Ras. Crystal structures of the protein R11.1.6 in complex with K-Ras WT and K-Ras G12D offer insight into the structural basis for specificity, highlighting differences in the switch I conformation as the major defining element in the higher affinity interaction. R11.1.6 directly blocks interaction with Raf and reduces signaling through the Raf/MEK/ERK pathway. Our results support greater consideration of the state of switch I and provide a novel tool to study Ras biology. Most importantly, this work makes an unprecedented contribution to Ras research in inhibitor development strategy by revealing details of a targetable binding surface. Unlike the polar interfaces found for Ras/effector interactions, the K-Ras/R11.1.6 complex reveals an extensive hydrophobic interface that can serve as a template to advance the development of high affinity, non-covalent inhibitors of K-Ras oncogenic mutants.


Asunto(s)
Ingeniería de Proteínas , Proteínas Recombinantes/farmacología , Proteínas ras/antagonistas & inhibidores , Secuencia de Aminoácidos , Células HEK293 , Humanos , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica , Proteínas Recombinantes/química , Proteínas ras/metabolismo
9.
J Mol Biol ; 428(20): 4228-4241, 2016 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-27448945

RESUMEN

Quantifying protein location and concentration is critical for understanding function in situ. Scaffold conjugated to environment-sensitive fluorophore (SuCESsFul) biosensors, in which a reporting fluorophore is conjugated to a binding scaffold, can, in principle, detect analytes of interest with high temporal and spatial resolution. However, their adoption has been limited due to the extensive empirical screening required for their development. We sought to establish design principles for this class of biosensor by characterizing over 400 biosensors based on various protein analytes, binding proteins, and fluorophores. We found that the brightest readouts are attained when a specific binding pocket for the fluorophore is present on the analyte. Also, interaction of the fluorophore with the binding protein it is conjugated to can raise background fluorescence, considerably limiting sensor dynamic range. Exploiting these two concepts, we designed biosensors that attain a 100-fold increase in fluorescence upon binding to analyte, an order of magnitude improvement over the previously best-reported SuCESsFul biosensor. These design principles should facilitate the development of improved SuCESsFul biosensors.


Asunto(s)
Técnicas Biosensibles/métodos , Proteínas/análisis , Fluorescencia , Análisis Espacio-Temporal
10.
J Mol Biol ; 426(21): 3606-18, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25073100

RESUMEN

Fibrin formation from fibrinogen is a rare process in the healthy organism but is a pathological feature of thrombotic events, cancer and a wide range of inflammatory conditions. We have designed and constructed an antibody phage display library (containing 13 billion clones) for the selective recognition of the N-terminal peptide of fibrin alpha chain. The key structural feature for selective fibrin binding was a K94E mutation in the VH domain. From this library, an antibody was isolated (termed AP2), which recognizes the five N-terminal amino acids of fibrin with high affinity (Kd=44nM), but does not bind to fibrinogen. The AP2 antibody could be expressed in various formats (scFv, small immune protein and IgG) and inhibited fibrin clot formation in a concentration-dependent manner. Moreover, the AP2 antibody stained the fibrin-rich provisional stroma in solid tumors but did not exhibit any detectable staining toward normal tissues. Using a radioiodinated antibody preparation and quantitative biodistribution studies in tumor-bearing mice, AP2 was shown to selectively localize to fibrin-rich F9 murine teratocarcinomas, but not to SKRC-52 human kidney cancer xenografts. Collectively, the experiments indicate that the AP2 antibody recognizes fibrin in vitro and in vivo. The antibody may facilitate the development of fibrin-specific therapeutic agents.


Asunto(s)
Anticuerpos Monoclonales/química , Coagulación Sanguínea , Fibrina/química , Neoplasias/inmunología , Biblioteca de Péptidos , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Fibrinógeno/química , Humanos , Inmunoglobulina G/química , Ratones , Datos de Secuencia Molecular , Mutación , Trasplante de Neoplasias , Neoplasias/química , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie , Trombosis
11.
MAbs ; 6(1): 130-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24256717

RESUMEN

The development of efficient strategies for generating fully human monoclonal antibodies with unique functional properties that are exploitable for tailored therapeutic interventions remains a major challenge in the antibody technology field. Here, we present a methodology for recovering such antibodies from antigen-encountered human B cell repertoires. As the source for variable antibody genes, we cloned immunoglobulin G (IgG)-derived B cell repertoires from lymph nodes of 20 individuals undergoing surgery for head and neck cancer. Sequence analysis of unselected "LYmph Node Derived Antibody Libraries" (LYNDAL) revealed a naturally occurring distribution pattern of rearranged antibody sequences, representing all known variable gene families and most functional germline sequences. To demonstrate the feasibility for selecting antibodies with therapeutic potential from these repertoires, seven LYNDAL from donors with high serum titers against herpes simplex virus (HSV) were panned on recombinant glycoprotein B of HSV-1. Screening for specific binders delivered 34 single-chain variable fragments (scFvs) with unique sequences. Sequence analysis revealed extensive somatic hypermutation of enriched clones as a result of affinity maturation. Binding of scFvs to common glycoprotein B variants from HSV-1 and HSV-2 strains was highly specific, and the majority of analyzed antibody fragments bound to the target antigen with nanomolar affinity. From eight scFvs with HSV-neutralizing capacity in vitro,the most potent antibody neutralized 50% HSV-2 at 4.5 nM as a dimeric (scFv)2. We anticipate our approach to be useful for recovering fully human antibodies with therapeutic potential.


Asunto(s)
Anticuerpos Antivirales , Clonación Molecular , Biblioteca de Genes , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/inmunología , Inmunoglobulina G , Región Variable de Inmunoglobulina , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Femenino , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Masculino , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA