Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 615(7953): 697-704, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890230

RESUMEN

Neoantigens are peptides derived from non-synonymous mutations presented by human leukocyte antigens (HLAs), which are recognized by antitumour T cells1-14. The large HLA allele diversity and limiting clinical samples have restricted the study of the landscape of neoantigen-targeted T cell responses in patients over their treatment course. Here we applied recently developed technologies15-17 to capture neoantigen-specific T cells from blood and tumours from patients with metastatic melanoma with or without response to anti-programmed death receptor 1 (PD-1) immunotherapy. We generated personalized libraries of neoantigen-HLA capture reagents to single-cell isolate the T cells and clone their T cell receptors (neoTCRs). Multiple T cells with different neoTCR sequences (T cell clonotypes) recognized a limited number of mutations in samples from seven patients with long-lasting clinical responses. These neoTCR clonotypes were recurrently detected over time in the blood and tumour. Samples from four patients with no response to anti-PD-1 also demonstrated neoantigen-specific T cell responses in the blood and tumour to a restricted number of mutations with lower TCR polyclonality and were not recurrently detected in sequential samples. Reconstitution of the neoTCRs in donor T cells using non-viral CRISPR-Cas9 gene editing demonstrated specific recognition and cytotoxicity to patient-matched melanoma cell lines. Thus, effective anti-PD-1 immunotherapy is associated with the presence of polyclonal CD8+ T cells in the tumour and blood specific for a limited number of immunodominant mutations, which are recurrently recognized over time.


Asunto(s)
Antígenos de Neoplasias , Linfocitos T CD8-positivos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Melanoma , Humanos , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Antígenos HLA/inmunología , Metástasis de la Neoplasia , Medicina de Precisión , Edición Génica , Sistemas CRISPR-Cas , Mutación
2.
Nature ; 615(7953): 687-696, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36356599

RESUMEN

T cell receptors (TCRs) enable T cells to specifically recognize mutations in cancer cells1-3. Here we developed a clinical-grade approach based on CRISPR-Cas9 non-viral precision genome-editing to simultaneously knockout the two endogenous TCR genes TRAC (which encodes TCRα) and TRBC (which encodes TCRß). We also inserted into the TRAC locus two chains of a neoantigen-specific TCR (neoTCR) isolated from circulating T cells of patients. The neoTCRs were isolated using a personalized library of soluble predicted neoantigen-HLA capture reagents. Sixteen patients with different refractory solid cancers received up to three distinct neoTCR transgenic cell products. Each product expressed a patient-specific neoTCR and was administered in a cell-dose-escalation, first-in-human phase I clinical trial ( NCT03970382 ). One patient had grade 1 cytokine release syndrome and one patient had grade 3 encephalitis. All participants had the expected side effects from the lymphodepleting chemotherapy. Five patients had stable disease and the other eleven had disease progression as the best response on the therapy. neoTCR transgenic T cells were detected in tumour biopsy samples after infusion at frequencies higher than the native TCRs before infusion. This study demonstrates the feasibility of isolating and cloning multiple TCRs that recognize mutational neoantigens. Moreover, simultaneous knockout of the endogenous TCR and knock-in of neoTCRs using single-step, non-viral precision genome-editing are achieved. The manufacture of neoTCR engineered T cells at clinical grade, the safety of infusing up to three gene-edited neoTCR T cell products and the ability of the transgenic T cells to traffic to the tumours of patients are also demonstrated.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Edición Génica , Neoplasias , Medicina de Precisión , Receptores de Antígenos de Linfocitos T , Linfocitos T , Transgenes , Humanos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Biopsia , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Síndrome de Liberación de Citoquinas/complicaciones , Progresión de la Enfermedad , Encefalitis/complicaciones , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Mutación , Neoplasias/complicaciones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Seguridad del Paciente , Medicina de Precisión/efectos adversos , Medicina de Precisión/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transgenes/genética , Antígenos HLA/inmunología , Sistemas CRISPR-Cas
3.
Cell Rep ; 28(10): 2728-2738.e7, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31484081

RESUMEN

Neoantigen-specific T cells are increasingly viewed as important immunotherapy effectors, but physically isolating these rare cell populations is challenging. Here, we describe a sensitive method for the enumeration and isolation of neoantigen-specific CD8+ T cells from small samples of patient tumor or blood. The method relies on magnetic nanoparticles that present neoantigen-loaded major histocompatibility complex (MHC) tetramers at high avidity by barcoded DNA linkers. The magnetic particles provide a convenient handle to isolate the desired cell populations, and the barcoded DNA enables multiplexed analysis. The method exhibits superior recovery of antigen-specific T cell populations relative to literature approaches. We applied the method to profile neoantigen-specific T cell populations in the tumor and blood of patients with metastatic melanoma over the course of anti-PD1 checkpoint inhibitor therapy. We show that the method has value for monitoring clinical responses to cancer immunotherapy and might help guide the development of personalized mutational neoantigen-specific T cell therapies and cancer vaccines.


Asunto(s)
Antígenos de Neoplasias/sangre , Melanoma/sangre , Melanoma/inmunología , Linfocitos T/inmunología , Biopsia , Células HEK293 , Humanos , Inmunoterapia , Células Jurkat , Cinética , Linfocitos Infiltrantes de Tumor/inmunología , Nanopartículas de Magnetita/química , Complejo Mayor de Histocompatibilidad , Melanoma/patología , Melanoma/secundario , Ácidos Nucleicos/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Reproducibilidad de los Resultados , Tomografía Computarizada por Rayos X
4.
Lab Chip ; 19(18): 3011-3021, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31502632

RESUMEN

Adaptive immunity is based on peptide antigen recognition. Our ability to harness the immune system for therapeutic gain relies on the discovery of the T cell receptor (TCR) genes that selectively target antigens from infections, mutated proteins, and foreign agents. Here we present a method that selectively labels peptide antigen-specific CD8+ T cells using magnetic nanoparticles functionalized with peptide-MHC tetramers, isolates these specific cells within an integrated microfluidic device, and directly amplifies the TCR genes for sequencing. Critically, the identity of the peptide recognized by the TCR is preserved, providing the link between peptide and gene. The platform requires inputs on the order of just 100 000 CD8+ T cells, can be multiplexed for simultaneous analysis of multiple peptides, and performs sorting and isolation on chip. We demonstrate 1000-fold sensitivity enhancement of detecting antigen-specific TCRs relative to bulk analysis and simultaneous capture of two virus antigen-specific TCRs from a population of T cells.


Asunto(s)
Antígenos/genética , Técnicas Analíticas Microfluídicas , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T CD8-positivos , Células Cultivadas , Humanos , Nanopartículas de Magnetita/química , Técnicas Analíticas Microfluídicas/instrumentación , Tamaño de la Partícula , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Nat Methods ; 16(2): 191-198, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700902

RESUMEN

CD8+ T cells recognize and eliminate tumors in an antigen-specific manner. Despite progress in characterizing the antitumor T cell repertoire and function, the identification of target antigens remains a challenge. Here we describe the use of chimeric receptors called signaling and antigen-presenting bifunctional receptors (SABRs) in a cell-based platform for T cell receptor (TCR) antigen discovery. SABRs present an extracellular complex comprising a peptide and major histocompatibility complex (MHC), and induce intracellular signaling via a TCR-like signal after binding with a cognate TCR. We devised a strategy for antigen discovery using SABR libraries to screen thousands of antigenic epitopes. We validated this platform by identifying the targets recognized by public TCRs of known specificities. Moreover, we extended this approach for personalized neoantigen discovery.


Asunto(s)
Presentación de Antígeno , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Células Presentadoras de Antígenos/citología , Antígenos/química , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD8-positivos/citología , Clonación Molecular , Técnicas de Cocultivo , Epítopos/química , Reacciones Falso Positivas , Biblioteca de Genes , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Inmunoterapia/métodos , Células Jurkat , Células K562 , Lectinas Tipo C/metabolismo , Complejo Mayor de Histocompatibilidad , Oligonucleótidos/genética , Péptidos/química
6.
Nat Methods ; 16(2): 183-190, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700903

RESUMEN

T cell receptor (TCR) ligand discovery is essential for understanding and manipulating immune responses to tumors. We developed a cell-based selection platform for TCR ligand discovery that exploits a membrane transfer phenomenon called trogocytosis. We discovered that T cell membrane proteins are transferred specifically to target cells that present cognate peptide-major histocompatibility complex (MHC) molecules. Co-incubation of T cells expressing an orphan TCR with target cells collectively presenting a library of peptide-MHCs led to specific labeling of cognate target cells, enabling isolation of these target cells and sequencing of the cognate TCR ligand. We validated this method for two clinically employed TCRs and further used the platform to identify the cognate neoepitope for a subject-derived neoantigen-specific TCR. Thus, target cell trogocytosis is a robust tool for TCR ligand discovery that will be useful for studying basic tumor immunology and identifying new targets for immunotherapy.


Asunto(s)
Antígenos/química , Técnicas Genéticas , Receptores de Antígenos de Linfocitos T/química , Linfocitos T/citología , Inmunidad Adaptativa , Animales , Biotinilación , ADN/análisis , Epítopos/química , Biblioteca de Genes , Células HEK293 , Humanos , Inmunoterapia , Células Jurkat , Células K562 , Ligandos , Ratones , Péptidos/química , Fagocitosis , Linfocitos T/inmunología
7.
Proc Natl Acad Sci U S A ; 115(45): E10702-E10711, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30348802

RESUMEN

Tumor-specific T cell receptor (TCR) gene transfer enables specific and potent immune targeting of tumor antigens. Due to the prevalence of the HLA-A2 MHC class I supertype in most human populations, the majority of TCR gene therapy trials targeting public antigens have employed HLA-A2-restricted TCRs, limiting this approach to those patients expressing this allele. For these patients, TCR gene therapy trials have resulted in both tantalizing successes and lethal adverse events, underscoring the need for careful selection of antigenic targets. Broad and safe application of public antigen-targeted TCR gene therapies will require (i) selecting public antigens that are highly tumor-specific and (ii) targeting multiple epitopes derived from these antigens by obtaining an assortment of TCRs restricted by multiple common MHC alleles. The canonical cancer-testis antigen, NY-ESO-1, is not expressed in normal tissues but is aberrantly expressed across a broad array of cancer types. It has also been targeted with A2-restricted TCR gene therapy without adverse events or notable side effects. To enable the targeting of NY-ESO-1 in a broader array of HLA haplotypes, we isolated TCRs specific for NY-ESO-1 epitopes presented by four MHC molecules: HLA-A2, -B07, -B18, and -C03. Using these TCRs, we pilot an approach to extend TCR gene therapies targeting NY-ESO-1 to patient populations beyond those expressing HLA-A2.


Asunto(s)
Proteínas de Homeodominio/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Receptores de Antígenos de Linfocitos T/aislamiento & purificación , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Clonación Molecular , Humanos
8.
Proc Natl Acad Sci U S A ; 115(8): 1877-1882, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29437954

RESUMEN

HIV controllers (HCs) are individuals who can naturally control HIV infection, partially due to potent HIV-specific CD8+ T cell responses. Here, we examined the hypothesis that superior function of CD8+ T cells from HCs is encoded by their T cell receptors (TCRs). We compared the functional properties of immunodominant HIV-specific TCRs obtained from HLA-B*2705 HCs and chronic progressors (CPs) following expression in primary T cells. T cells transduced with TCRs from HCs and CPs showed equivalent induction of epitope-specific cytotoxicity, cytokine secretion, and antigen-binding properties. Transduced T cells comparably, albeit modestly, also suppressed HIV infection in vitro and in humanized mice. We also performed extensive molecular dynamics simulations that provided a structural basis for similarities in cytotoxicity and epitope cross-reactivity. These results demonstrate that the differential abilities of HIV-specific CD8+ T cells from HCs and CPs are not genetically encoded in the TCRs alone and must depend on additional factors.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Epítopos de Linfocito T/genética , Infecciones por VIH/inmunología , VIH-1/inmunología , Receptores de Antígenos de Linfocitos T/genética , Clonación Molecular , Regulación de la Expresión Génica/inmunología , Células HEK293 , Antígeno HLA-B27 , Humanos , Células Jurkat
9.
PLoS One ; 13(1): e0191634, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29360859

RESUMEN

For adoptive cell transfer (ACT) immunotherapy of tumor-reactive T cells, an effective therapeutic outcome depends upon cell dose, cell expansion in vivo through a minimally differentiated phenotype, long term persistence, and strong cytolytic effector function. An incomplete understanding of the biological coupling between T cell expansion, differentiation, and response to stimulation hinders the co-optimization of these factors. We report on a biophysical investigation of how the short-term kinetics of T cell functional activation, through molecular stimulation and cell-cell interactions, competes with phenotype differentiation. T cells receive molecular stimulation for a few minutes to a few hours in bulk culture. Following this priming period, the cells are then analyzed at the transcriptional level, or isolated as single cells, with continuing molecular stimulation, within microchambers for analysis via 11-plex secreted protein assays. We resolve a rapid feedback mechanism, promoted by T cell-T cell contact interactions, which strongly amplifies T cell functional performance while yielding only minimal phenotype differentiation. When tested in mouse models of ACT, optimally primed T cells lead to complete tumor eradication. A similar kinetic process is identified in CD8+ and CD4+ T cells collected from a patient with metastatic melanoma.


Asunto(s)
Traslado Adoptivo , Inmunofenotipificación , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Femenino , Citometría de Flujo , Xenoinjertos , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
10.
Cell ; 172(3): 549-563.e16, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29275860

RESUMEN

The immune system can mount T cell responses against tumors; however, the antigen specificities of tumor-infiltrating lymphocytes (TILs) are not well understood. We used yeast-display libraries of peptide-human leukocyte antigen (pHLA) to screen for antigens of "orphan" T cell receptors (TCRs) expressed on TILs from human colorectal adenocarcinoma. Four TIL-derived TCRs exhibited strong selection for peptides presented in a highly diverse pHLA-A∗02:01 library. Three of the TIL TCRs were specific for non-mutated self-antigens, two of which were present in separate patient tumors, and shared specificity for a non-mutated self-antigen derived from U2AF2. These results show that the exposed recognition surface of MHC-bound peptides accessible to the TCR contains sufficient structural information to enable the reconstruction of sequences of peptide targets for pathogenic TCRs of unknown specificity. This finding underscores the surprising specificity of TCRs for their cognate antigens and enables the facile indentification of tumor antigens through unbiased screening.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos de Neoplasias/inmunología , Neoplasias Colorrectales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Anciano , Animales , Antígenos de Neoplasias/química , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Antígenos HLA-A/química , Antígenos HLA-A/inmunología , Humanos , Masculino , Persona de Mediana Edad , Biblioteca de Péptidos , Células Sf9 , Spodoptera
11.
Curr Opin Biotechnol ; 48: 142-152, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28494274

RESUMEN

Immunotherapies are yielding effective treatments for several previously untreatable cancers. Until recently, vaccines and adoptive cell therapies have been designed to target public tumor antigens common to multiple patients rather than private antigens specific to a single patient. Due to the difficulty of identifying public antigens that are expressed exclusively on tumor cells, these studies have yielded both clinical successes and serious immune-related adverse events. Multiple avenues of research now underscore the centrality of tumor-specific mutated private antigens to endogenous anti-tumor immunity. Immunotherapies that target these neoantigens may enable safer and more durable tumor regression, but personalized targeting presents a number of challenges. Foremost among these is to develop processes that accelerate advancement from neoantigen discovery to use of these neoantigens as vaccines or as targets for adoptive cell therapies. Exome sequencing has facilitated discovery of neoantigens for melanoma and other highly mutated cancers. New technologies - possibly proceeding from T cell receptor repertoire sequencing - are needed to identify antigens for cancers with low mutational burden and few neoantigens. In this review, we discuss progress toward personalizing T cell-mediated immunotherapy for cancer as well as challenges going forward.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Humanos
12.
Nat Methods ; 14(5): 521-530, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28369043

RESUMEN

Studies of human T cell development require robust model systems that recapitulate the full span of thymopoiesis, from hematopoietic stem and progenitor cells (HSPCs) through to mature T cells. Existing in vitro models induce T cell commitment from human HSPCs; however, differentiation into mature CD3+TCR-αß+ single-positive CD8+ or CD4+ cells is limited. We describe here a serum-free, artificial thymic organoid (ATO) system that supports efficient and reproducible in vitro differentiation and positive selection of conventional human T cells from all sources of HSPCs. ATO-derived T cells exhibited mature naive phenotypes, a diverse T cell receptor (TCR) repertoire and TCR-dependent function. ATOs initiated with TCR-engineered HSPCs produced T cells with antigen-specific cytotoxicity and near-complete lack of endogenous TCR Vß expression, consistent with allelic exclusion of Vß-encoding loci. ATOs provide a robust tool for studying human T cell differentiation and for the future development of stem-cell-based engineered T cell therapies.


Asunto(s)
Órganos Artificiales , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Organoides/citología , Linfocitos T/citología , Timo/citología , Biotecnología/métodos , Células Madre Hematopoyéticas/inmunología , Humanos , Organoides/inmunología , Células Madre/citología , Células Madre/inmunología , Linfocitos T/inmunología , Timo/inmunología
13.
Biotechniques ; 62(3): 123-130, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28298179

RESUMEN

Peptide-major histocompatibility complex (pMHC) multimers enable the detection, characterization, and isolation of antigen-specific T-cell subsets at the single-cell level via flow cytometry and fluorescence microscopy. These labeling reagents exploit a multivalent scaffold to increase the avidity of individually weak T-cell receptor (TCR)-pMHC interactions. Dextramers are an improvement over the original streptavidin-based tetramer technology because they are more multivalent, improving sensitivity for rare, low-avidity T cells, including self/tumor-reactive clones. However, commercial pMHC dextramers are expensive, and in-house production is very involved for a typical biology research laboratory. Here, we present a simple, inexpensive protocol for preparing pMHC dextramers by doping in biotinylated dextran during conventional tetramer preparation. We use these pMHC dextramers to identify patient-derived, tumor-reactive T cells. We apply the same dextran doping technique to prepare TCR dextramers and use these novel reagents to yield new insight into MHC I-mediated antigen presentation.


Asunto(s)
Biotecnología/métodos , Dextranos/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Biotina , Dextranos/química , Citometría de Flujo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Células HEK293 , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/genética , Humanos , Células Jurkat , Células K562 , Péptidos/química , Péptidos/genética , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Estreptavidina , Transducción Genética
14.
Elife ; 52016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27823582

RESUMEN

T cells engineered to express a tumor-specific αß T cell receptor (TCR) mediate anti-tumor immunity. However, mispairing of the therapeutic αß chains with endogenous αß chains reduces therapeutic TCR surface expression and generates self-reactive TCRs. We report a general strategy to prevent TCR mispairing: swapping constant domains between the α and ß chains of a therapeutic TCR. When paired, domain-swapped (ds)TCRs assemble with CD3, express on the cell surface, and mediate antigen-specific T cell responses. By contrast, dsTCR chains mispaired with endogenous chains cannot properly assemble with CD3 or signal, preventing autoimmunity. We validate this approach in cell-based assays and in a mouse model of TCR gene transfer-induced graft-versus-host disease. We also validate a related approach whereby replacement of αß TCR domains with corresponding γδ TCR domains yields a functional TCR that does not mispair. This work enables the design of safer TCR gene therapies for cancer immunotherapy.


Asunto(s)
Genes Codificadores de los Receptores de Linfocitos T , Terapia Genética/efectos adversos , Terapia Genética/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Animales , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped , Ratones , Dominios Proteicos , Recombinación Genética
15.
Methods Enzymol ; 502: 241-71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22208988

RESUMEN

Celiac sprue is an inflammatory disease of the small intestine caused by dietary gluten and treated by adherence to a life-long gluten-free diet. The recent identification of immunodominant gluten peptides, the discovery of their cogent properties, and the elucidation of the mechanisms by which they engender immunopathology in genetically susceptible individuals have advanced our understanding of the molecular pathogenesis of this complex disease, enabling the rational design of new therapeutic strategies. The most clinically advanced of these is oral enzyme therapy, in which enzymes capable of proteolyzing gluten (i.e., glutenases) are delivered to the alimentary tract of a celiac sprue patient to detoxify ingested gluten in situ. In this chapter, we discuss the key challenges for discovery and preclinical development of oral enzyme therapies for celiac sprue. Methods for lead identification, assay development, gram-scale production and formulation, and lead optimization for next-generation proteases are described and critically assessed.


Asunto(s)
Enfermedad Celíaca/tratamiento farmacológico , Endopeptidasas/metabolismo , Terapia Enzimática , Glútenes/metabolismo , Mucosa Intestinal/metabolismo , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Administración Oral , Secuencia de Aminoácidos , Animales , Bioensayo , Enfermedad Celíaca/enzimología , Enfermedad Celíaca/patología , Cromatografía Liquida , Clonación Molecular , Dieta Sin Gluten , Sistemas de Liberación de Medicamentos , Endopeptidasas/genética , Escherichia coli , Humanos , Intestino Delgado/metabolismo , Espectrometría de Masas , Modelos Biológicos , Datos de Secuencia Molecular , Ratas , Proteínas Recombinantes/genética
16.
PLoS One ; 5(4): e10228, 2010 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-20419103

RESUMEN

BACKGROUND: Based on clinical, histopathological and serological similarities to human celiac disease (CD), we recently established the rhesus macaque model of gluten sensitivity. In this study, we further characterized this condition based on presence of anti-tissue transglutaminase 2 (TG2) antibodies, increased intestinal permeability and transepithelial transport of a proteolytically resistant, immunotoxic, 33-residue peptide from alpha(2)-gliadin in the distal duodenum of gluten-sensitive macaques. METHODOLOGY/PRINCIPAL FINDINGS: Six rhesus macaques were selected for study from a pool of 500, including two healthy controls and four gluten-sensitive animals with elevated anti-gliadin or anti-TG2 antibodies as well as history of non-infectious chronic diarrhea. Pediatric endoscope-guided pinch biopsies were collected from each animal's distal duodenum following administration of a gluten-containing diet (GD) and again after remission by gluten-free diet (GFD). Control biopsies always showed normal villous architecture, whereas gluten-sensitive animals on GD exhibited histopathology ranging from mild lymphocytic infiltration to villous atrophy, typical of human CD. Immunofluorescent microscopic analysis of biopsies revealed IgG+ and IgA+ plasma-like cells producing antibodies that colocalized with TG2 in gluten-sensitive macaques only. Following instillation in vivo, the Cy-3-labeled 33-residue gluten peptide colocalized with the brush border protein villin in all animals. In a substantially enteropathic macaque with "leaky" duodenum, the peptide penetrated beneath the epithelium into the lamina propria. CONCLUSIONS/SIGNIFICANCE: The rhesus macaque model of gluten sensitivity not only resembles the histopathology of CD but it also may provide a model for studying intestinal permeability in states of epithelial integrity and disrepair.


Asunto(s)
Epitelio/metabolismo , Gliadina/inmunología , Glútenes/inmunología , Fragmentos de Péptidos/inmunología , Animales , Enfermedad Celíaca , Modelos Animales de Enfermedad , Duodeno/inmunología , Duodeno/metabolismo , Duodeno/patología , Gliadina/análisis , Gliadina/metabolismo , Glútenes/administración & dosificación , Glútenes/farmacología , Humanos , Macaca , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/metabolismo , Permeabilidad , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transporte de Proteínas
17.
Chem Biol ; 16(8): 868-81, 2009 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-19716477

RESUMEN

New tools are needed for managing celiac sprue, a lifelong immune disease of the small intestine. Ongoing drug trials are also prompting a search for noninvasive biomarkers of gluten-induced intestinal change. We have synthesized and characterized noninflammatory gluten peptide analogs in which key Gln residues are replaced by Asn or His. Like their proinflammatory counterparts, these biomarkers are resistant to gastrointestinal proteases, susceptible to glutenases, and permeable across enterocyte barriers. Unlike gluten peptides, however, they are not appreciably recognized by transglutaminase, HLA-DQ2, or disease-specific T cells. In vitro and animal studies show that the biomarkers can detect intestinal permeability changes as well as glutenase-catalyzed gastric detoxification of gluten. Accordingly, controlled clinical studies are warranted to evaluate the use of these peptides as probes for abnormal intestinal permeability in celiac patients and for glutenase efficacy in clinical trials and practice.


Asunto(s)
Enfermedad Celíaca/metabolismo , Glútenes/metabolismo , Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Biomarcadores/metabolismo , Enfermedad Celíaca/inmunología , Glútenes/química , Glútenes/farmacología , Humanos , Marcaje Isotópico , Masculino , Datos de Secuencia Molecular , Péptidos/química , Ratas , Ratas Wistar , Linfocitos T/inmunología , Transglutaminasas/inmunología , Transglutaminasas/metabolismo
18.
PLoS One ; 4(7): e6313, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19621078

RESUMEN

BACKGROUND AND AIMS: Celiac sprue is a life-long disease characterized by an intestinal inflammatory response to dietary gluten. A gluten-free diet is an effective treatment for most patients, but accidental ingestion of gluten is common, leading to incomplete recovery or relapse. Food-grade proteases capable of detoxifying moderate quantities of dietary gluten could mitigate this problem. METHODS: We evaluated the gluten detoxification properties of two food-grade enzymes, aspergillopepsin (ASP) from Aspergillus niger and dipeptidyl peptidase IV (DPPIV) from Aspergillus oryzae. The ability of each enzyme to hydrolyze gluten was tested against synthetic gluten peptides, a recombinant gluten protein, and simulated gastric digests of whole gluten and whole-wheat bread. Reaction products were analyzed by mass spectrometry, HPLC, ELISA with a monoclonal antibody that recognizes an immunodominant gluten epitope, and a T cell proliferation assay. RESULTS: ASP markedly enhanced gluten digestion relative to pepsin, and cleaved recombinant alpha2-gliadin at multiple sites in a non-specific manner. When used alone, neither ASP nor DPPIV efficiently cleaved synthetic immunotoxic gluten peptides. This lack of specificity for gluten was especially evident in the presence of casein, a competing dietary protein. However, supplementation of ASP with DPPIV enabled detoxification of moderate amounts of gluten in the presence of excess casein and in whole-wheat bread. ASP was also effective at enhancing the gluten-detoxifying efficacy of cysteine endoprotease EP-B2 under simulated gastric conditions. CONCLUSIONS: Clinical studies are warranted to evaluate whether a fixed dose ratio combination of ASP and DPPIV can provide near-term relief for celiac patients suffering from inadvertent gluten exposure. Due to its markedly greater hydrolytic activity against gluten than endogenous pepsin, food-grade ASP may also augment the activity of therapeutically relevant doses of glutenases such as EP-B2 and certain prolyl endopeptidases.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Alimentos , Glútenes/metabolismo , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Glútenes/química , Hidrólisis , Espectrometría de Masas , Datos de Secuencia Molecular
19.
J Pharmacol Exp Ther ; 329(2): 657-68, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19218531

RESUMEN

Celiac sprue is a T-cell-mediated enteropathy elicited in genetically susceptible individuals by dietary gluten proteins. To initiate and propagate inflammation, proteolytically resistant gluten peptides must be translocated across the small intestinal epithelium and presented to DQ2-restricted T cells, but the effectors enabling this translocation under normal and inflammatory conditions are not well understood. We demonstrate that a fluorescently labeled antigenic 33-mer gluten peptide is translocated intact across a T84 cultured epithelial cell monolayer and that preincubation of the monolayer with media from gluten-stimulated, celiac patient-derived intestinal T cells enhances the apical-to-basolateral flux of this peptide in a dose-dependent, saturable manner. The permeability-enhancing activity of activated T-cell media is inhibited by blocking antibodies against either interferon-gamma or its receptor and is recapitulated using recombinant interferon-gamma. At saturating levels of interferon-gamma, activated T-cell media does not further increase transepithelial peptide flux, indicating the primacy of interferon-gamma as an effector of increased epithelial permeability during inflammation. Reducing the assay temperature to 4 degrees C reverses the effect of interferon-gamma but does not reduce basal peptide flux occurring in the absence of interferon-gamma, suggesting active transcellular transport of intact peptides is increased during inflammation. A panel of disease-relevant gluten peptides exhibited an inverse correlation between size and transepithelial flux but no apparent sequence constraints. Anti-interferon-gamma therapy may mitigate the vicious cycle of gluten-induced interferon-gamma secretion and interferon-gamma-mediated enhancement of gluten peptide flux but is unlikely to prevent translocation of gluten peptides in the absence of inflammatory conditions.


Asunto(s)
Enfermedad Celíaca/inmunología , Glútenes/farmacología , Interferón gamma/metabolismo , Mucosa Intestinal/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Linfocitos T/efectos de los fármacos , Transporte Biológico , Western Blotting , Enfermedad Celíaca/metabolismo , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Glútenes/farmacocinética , Humanos , Interferón gamma/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Fragmentos de Péptidos/farmacocinética , Permeabilidad , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
PLoS One ; 3(5): e2294, 2008 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-18509534

RESUMEN

BACKGROUND AND AIMS: Celiac disease is a permanent intolerance to gluten prolamins from wheat, barley, rye and, in some patients, oats. Partially digested gluten peptides produced in the digestive tract cause inflammation of the small intestine. High throughput, immune-based assays using monoclonal antibodies specific for these immunotoxic peptides would facilitate their detection in food and enable monitoring of their enzymatic detoxification. Two monoclonal antibodies, G12 and A1, were developed against a highly immunotoxic 33-mer peptide. The potential of each antibody for quantifying food toxicity for celiac patients was studied. METHODS: Epitope preferences of G12 and A1 antibodies were determined by ELISA with gluten-derived peptide variants of recombinant, synthetic or enzymatic origin. RESULTS: The recognition sequences of G12 and A1 antibodies were hexameric and heptameric epitopes, respectively. Although G12 affinity for the 33-mer was superior to A1, the sensitivity for gluten detection was higher for A1. This observation correlated to the higher number of A1 epitopes found in prolamins than G12 epitopes. Activation of T cell from gluten digested by glutenases decreased equivalently to the detection of intact peptides by A1 antibody. Peptide recognition of A1 included gliadin peptides involved in the both the adaptive and innate immunological response in celiac disease. CONCLUSIONS: The sensitivity and epitope preferences of the A1 antibody resulted to be useful to detect gluten relevant peptides to infer the potential toxicity of food for celiac patients as well as to monitor peptide modifications by transglutaminase 2 or glutenases.


Asunto(s)
Enfermedad Celíaca/fisiopatología , Alimentos/toxicidad , Glútenes/toxicidad , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Secuencia de Bases , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Gliadina/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...