Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 166(4): 1041-1054, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27499020

RESUMEN

We used clinical tissue from lethal metastatic castration-resistant prostate cancer (CRPC) patients obtained at rapid autopsy to evaluate diverse genomic, transcriptomic, and phosphoproteomic datasets for pathway analysis. Using Tied Diffusion through Interacting Events (TieDIE), we integrated differentially expressed master transcriptional regulators, functionally mutated genes, and differentially activated kinases in CRPC tissues to synthesize a robust signaling network consisting of druggable kinase pathways. Using MSigDB hallmark gene sets, six major signaling pathways with phosphorylation of several key residues were significantly enriched in CRPC tumors after incorporation of phosphoproteomic data. Individual autopsy profiles developed using these hallmarks revealed clinically relevant pathway information potentially suitable for patient stratification and targeted therapies in late stage prostate cancer. Here, we describe phosphorylation-based cancer hallmarks using integrated personalized signatures (pCHIPS) that shed light on the diversity of activated signaling pathways in metastatic CRPC while providing an integrative, pathway-based reference for drug prioritization in individual patients.


Asunto(s)
Fosfoproteínas/análisis , Neoplasias de la Próstata Resistentes a la Castración/química , Proteoma/análisis , Algoritmos , Humanos , Masculino , Medicina de Precisión , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal , Transcriptoma
2.
Proc Natl Acad Sci U S A ; 121(14): e2320442121, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38536748

RESUMEN

The ability to selectively bind to antigenic peptides and secrete effector molecules can define rare and low-affinity populations of cells with therapeutic potential in emerging T cell receptor (TCR) immunotherapies. We leverage cavity-containing hydrogel microparticles, called nanovials, each coated with peptide-major histocompatibility complex (pMHC) monomers to isolate antigen-reactive T cells. T cells are captured and activated by pMHCs inducing the secretion of effector molecules including IFN-γ and granzyme B that are accumulated on nanovials, allowing sorting based on both binding and function. The TCRs of sorted cells on nanovials are sequenced, recovering paired αß-chains using microfluidic emulsion-based single-cell sequencing. By labeling nanovials having different pMHCs with unique oligonucleotide-barcodes and secretions with oligo-barcoded detection antibodies, we could accurately link TCR sequences to specific targets and rank each TCR based on the corresponding cell's secretion level. Using the technique, we identified an expanded repertoire of functional TCRs targeting viral antigens with high specificity and found rare TCRs with activity against cancer-specific splicing-enhanced epitopes.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Linfocitos T , Péptidos/química , Antígenos de Histocompatibilidad/química , Antígenos
3.
Proc Natl Acad Sci U S A ; 120(28): e2220190120, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37399401

RESUMEN

The MYC proto-oncogene contributes to the pathogenesis of more than half of human cancers. Malignant transformation by MYC transcriptionally up-regulates the core pre-mRNA splicing machinery and causes misregulation of alternative splicing. However, our understanding of how splicing changes are directed by MYC is limited. We performed a signaling pathway-guided splicing analysis to identify MYC-dependent splicing events. These included an HRAS cassette exon repressed by MYC across multiple tumor types. To molecularly dissect the regulation of this HRAS exon, we used antisense oligonucleotide tiling to identify splicing enhancers and silencers in its flanking introns. RNA-binding motif prediction indicated multiple binding sites for hnRNP H and hnRNP F within these cis-regulatory elements. Using siRNA knockdown and cDNA expression, we found that both hnRNP H and F activate the HRAS cassette exon. Mutagenesis and targeted RNA immunoprecipitation implicate two downstream G-rich elements in this splicing activation. Analyses of ENCODE RNA-seq datasets confirmed hnRNP H regulation of HRAS splicing. Analyses of RNA-seq datasets across multiple cancers showed a negative correlation of HNRNPH gene expression with MYC hallmark enrichment, consistent with the effect of hnRNP H on HRAS splicing. Interestingly, HNRNPF expression showed a positive correlation with MYC hallmarks and thus was not consistent with the observed effects of hnRNP F. Loss of hnRNP H/F altered cell cycle progression and induced apoptosis in the PC3 prostate cancer cell line. Collectively, our results reveal mechanisms for MYC-dependent regulation of splicing and point to possible therapeutic targets in prostate cancers.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo F-H , Neoplasias de la Próstata , Masculino , Humanos , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN/genética , Proteínas de Unión al ARN/metabolismo , Exones/genética , Empalme Alternativo/genética , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(47): e2312374120, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37963244

RESUMEN

CAR (chimeric antigen receptor) T cell therapy has shown clinical success in treating hematological malignancies, but its treatment of solid tumors has been limited. One major challenge is on-target, off-tumor toxicity, where CAR T cells also damage normal tissues that express the targeted antigen. To reduce this detrimental side-effect, Boolean-logic gates like AND-NOT gates have utilized an inhibitory CAR (iCAR) to specifically curb CAR T cell activity at selected nonmalignant tissue sites. However, the strategy seems inefficient, requiring high levels of iCAR and its target antigen for inhibition. Using a TROP2-targeting iCAR with a single PD1 inhibitory domain to inhibit a CEACAM5-targeting CAR (CEACAR), we observed that the inefficiency was due to a kinetic delay in iCAR inhibition of cytotoxicity. To improve iCAR efficiency, we modified three features of the iCAR-the avidity, the affinity, and the intracellular signaling domains. Increasing the avidity but not the affinity of the iCAR led to significant reductions in the delay. iCARs containing twelve different inhibitory signaling domains were screened for improved inhibition, and three domains (BTLA, LAIR-1, and SIGLEC-9) each suppressed CAR T function but did not enhance inhibitory kinetics. When inhibitory domains of LAIR-1 or SIGLEC-9 were combined with PD-1 into a single dual-inhibitory domain iCAR (DiCARs) and tested with the CEACAR, inhibition efficiency improved as evidenced by a significant reduction in the inhibitory delay. These data indicate that a delicate balance between CAR and iCAR signaling strength and kinetics must be achieved to regulate AND-NOT gate CAR T cell selectivity.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Linfocitos T , Complejo Hierro-Dextran , Inmunoterapia Adoptiva , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico
5.
Proc Natl Acad Sci U S A ; 120(21): e2221116120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37192158

RESUMEN

Alternative splicing (AS) is prevalent in cancer, generating an extensive but largely unexplored repertoire of novel immunotherapy targets. We describe Isoform peptides from RNA splicing for Immunotherapy target Screening (IRIS), a computational platform capable of discovering AS-derived tumor antigens (TAs) for T cell receptor (TCR) and chimeric antigen receptor T cell (CAR-T) therapies. IRIS leverages large-scale tumor and normal transcriptome data and incorporates multiple screening approaches to discover AS-derived TAs with tumor-associated or tumor-specific expression. In a proof-of-concept analysis integrating transcriptomics and immunopeptidomics data, we showed that hundreds of IRIS-predicted TCR targets are presented by human leukocyte antigen (HLA) molecules. We applied IRIS to RNA-seq data of neuroendocrine prostate cancer (NEPC). From 2,939 NEPC-associated AS events, IRIS predicted 1,651 epitopes from 808 events as potential TCR targets for two common HLA types (A*02:01 and A*03:01). A more stringent screening test prioritized 48 epitopes from 20 events with "neoantigen-like" NEPC-specific expression. Predicted epitopes are often encoded by microexons of ≤30 nucleotides. To validate the immunogenicity and T cell recognition of IRIS-predicted TCR epitopes, we performed in vitro T cell priming in combination with single-cell TCR sequencing. Seven TCRs transduced into human peripheral blood mononuclear cells (PBMCs) showed high activity against individual IRIS-predicted epitopes, providing strong evidence of isolated TCRs reactive to AS-derived peptides. One selected TCR showed efficient cytotoxicity against target cells expressing the target peptide. Our study illustrates the contribution of AS to the TA repertoire of cancer cells and demonstrates the utility of IRIS for discovering AS-derived TAs and expanding cancer immunotherapies.


Asunto(s)
Neoplasias , Precursores del ARN , Masculino , Humanos , Precursores del ARN/metabolismo , Empalme Alternativo , Leucocitos Mononucleares/metabolismo , Receptores de Antígenos de Linfocitos T , Epítopos de Linfocito T , Inmunoterapia , Antígenos de Neoplasias , Péptidos/metabolismo , Neoplasias/genética , Neoplasias/terapia
6.
Nat Immunol ; 14(10): 1073-83, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24013668

RESUMEN

C2H2 zinc fingers are found in several key transcriptional regulators in the immune system. However, these proteins usually contain more fingers than are needed for sequence-specific DNA binding, which suggests that different fingers regulate different genes and functions. Here we found that mice lacking finger 1 or finger 4 of Ikaros exhibited distinct subsets of the hematological defects of Ikaros-null mice. Most notably, the two fingers controlled different stages of lymphopoiesis, and finger 4 was selectively required for tumor suppression. The distinct defects support the hypothesis that only a small number of genes that are targets of Ikaros are critical for each of its biological functions. The subcategorization of functions and target genes by mutagenesis of individual zinc fingers will facilitate efforts to understand how zinc-finger transcription factors regulate development, immunity and disease.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/genética , Leucemia/genética , Linfopoyesis/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Secuencia de Bases , Sitios de Unión , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Inmunoprecipitación de Cromatina , Análisis por Conglomerados , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Factor de Transcripción Ikaros/metabolismo , Inmunofenotipificación , Leucemia/metabolismo , Leucemia/mortalidad , Linfoma/genética , Linfoma/metabolismo , Linfoma/mortalidad , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Motivos de Nucleótidos , Fenotipo , Posición Específica de Matrices de Puntuación , Unión Proteica , Timocitos/metabolismo
7.
Proc Natl Acad Sci U S A ; 119(31): e2203410119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35878026

RESUMEN

Tissue-specific antigens can serve as targets for adoptive T cell transfer-based cancer immunotherapy. Recognition of tumor by T cells is mediated by interaction between peptide-major histocompatibility complexes (pMHCs) and T cell receptors (TCRs). Revealing the identity of peptides bound to MHC is critical in discovering cognate TCRs and predicting potential toxicity. We performed multimodal immunopeptidomic analyses for human prostatic acid phosphatase (PAP), a well-recognized tissue antigen. Three physical methods, including mild acid elution, coimmunoprecipitation, and secreted MHC precipitation, were used to capture a thorough signature of PAP on HLA-A*02:01. Eleven PAP peptides that are potentially A*02:01-restricted were identified, including five predicted strong binders by NetMHCpan 4.0. Peripheral blood mononuclear cells (PBMCs) from more than 20 healthy donors were screened with the PAP peptides. Seven cognate TCRs were isolated which can recognize three distinct epitopes when expressed in PBMCs. One TCR shows reactivity toward cell lines expressing both full-length PAP and HLA-A*02:01. Our results show that a combined multimodal immunopeptidomic approach is productive in revealing target peptides and defining the cloned TCR sequences reactive with prostatic acid phosphatase epitopes.


Asunto(s)
Fosfatasa Ácida , Antígenos de Neoplasias , Receptores de Antígenos de Linfocitos T , Fosfatasa Ácida/metabolismo , Antígenos de Neoplasias/metabolismo , Epítopos , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Leucocitos Mononucleares , Neoplasias/inmunología , Péptidos , Receptores de Antígenos de Linfocitos T/metabolismo
8.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33431692

RESUMEN

T cell receptors (TCRs) are generated by somatic recombination of V/D/J segments to produce up to 1015 unique sequences. Highly sensitive and specific techniques are required to isolate and identify the rare TCR sequences that respond to antigens of interest. Here, we describe the use of mRNA sequencing via cross-linker regulated intracellular phenotype (CLInt-Seq) for efficient recovery of antigen-specific TCRs in cells stained for combinations of intracellular proteins such as cytokines or transcription factors. This method enables high-throughput identification and isolation of low-frequency TCRs specific for any antigen. As a proof of principle, intracellular staining for TNFα and IFNγ identified cytomegalovirus (CMV)- and Epstein-Barr virus (EBV)-reactive TCRs with efficiencies similar to state-of-the-art peptide-MHC multimer methodology. In a separate experiment, regulatory T cells were profiled based on intracellular FOXP3 staining, demonstrating the ability to examine phenotypes based on transcription factors. We further optimized the intracellular staining conditions to use a chemically cleavable primary amine cross-linker compatible with current single-cell sequencing technology. CLInt-Seq for TNFα and IFNγ performed similarly to isolation with multimer staining for EBV-reactive TCRs. We anticipate CLInt-Seq will enable droplet-based single-cell mRNA analysis from any tissue where minor populations need to be isolated by intracellular markers.


Asunto(s)
Factores de Transcripción Forkhead/genética , Interferón gamma/genética , Factor de Necrosis Tumoral alfa/genética , Recombinación V(D)J/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Clonación Molecular , Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Epítopos/inmunología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/patogenicidad , Humanos , ARN Mensajero/genética , RNA-Seq , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Análisis de la Célula Individual , Linfocitos T Reguladores/inmunología , Recombinación V(D)J/inmunología
9.
Immunology ; 168(1): 152-169, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35986643

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease driven by lymphocyte activation against myelin autoantigens in the central nervous system leading to demyelination and neurodegeneration. The deoxyribonucleoside salvage pathway with the rate-limiting enzyme deoxycytidine kinase (dCK) captures extracellular deoxyribonucleosides for use in intracellular deoxyribonucleotide metabolism. Previous studies have shown that deoxyribonucleoside salvage activity is enriched in lymphocytes and required for early lymphocyte development. However, specific roles for the deoxyribonucleoside salvage pathway and dCK in autoimmune diseases such as MS are unknown. Here we demonstrate that dCK activity is necessary for the development of clinical symptoms in the MOG35-55 and MOG1-125 experimental autoimmune encephalomyelitis (EAE) mouse models of MS. During EAE disease, deoxyribonucleoside salvage activity is elevated in the spleen and lymph nodes. Targeting dCK with the small molecule dCK inhibitor TRE-515 limits disease severity when treatments are started at disease induction or when symptoms first appear. EAE mice treated with TRE-515 have significantly fewer infiltrating leukocytes in the spinal cord, and TRE-515 blocks activation-induced B and T cell proliferation and MOG35-55 -specific T cell expansion without affecting innate immune cells or naïve T and B cell populations. Our results demonstrate that targeting dCK limits symptoms in EAE mice and suggest that dCK activity is required for MOG35-55 -specific lymphocyte activation-induced proliferation.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Ratones , Desoxicitidina Quinasa/genética , Linfocitos/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
10.
Proc Natl Acad Sci U S A ; 117(1): 563-572, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871155

RESUMEN

Small cell carcinoma of the bladder (SCCB) is a rare and lethal phenotype of bladder cancer. The pathogenesis and molecular features are unknown. Here, we established a genetically engineered SCCB model and a cohort of patient SCCB and urothelial carcinoma samples to characterize molecular similarities and differences between bladder cancer phenotypes. We demonstrate that SCCB shares a urothelial origin with other bladder cancer phenotypes by showing that urothelial cells driven by a set of defined oncogenic factors give rise to a mixture of tumor phenotypes, including small cell carcinoma, urothelial carcinoma, and squamous cell carcinoma. Tumor-derived single-cell clones also give rise to both SCCB and urothelial carcinoma in xenografts. Despite this shared urothelial origin, clinical SCCB samples have a distinct transcriptional profile and a unique transcriptional regulatory network. Using the transcriptional profile from our cohort, we identified cell surface proteins (CSPs) associated with the SCCB phenotype. We found that the majority of SCCB samples have PD-L1 expression in both tumor cells and tumor-infiltrating lymphocytes, suggesting that immune checkpoint inhibitors could be a treatment option for SCCB. We further demonstrate that our genetically engineered tumor model is a representative tool for investigating CSPs in SCCB by showing that it shares a similar a CSP profile with clinical samples and expresses SCCB-up-regulated CSPs at both the mRNA and protein levels. Our findings reveal distinct molecular features of SCCB and provide a transcriptional dataset and a preclinical model for further investigating SCCB biology.


Asunto(s)
Carcinoma de Células Pequeñas/patología , Carcinoma de Células Transicionales/patología , Transformación Celular Neoplásica/genética , Neoplasias de la Vejiga Urinaria/patología , Vejiga Urinaria/patología , Urotelio/patología , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/terapia , Carcinoma de Células Transicionales/genética , Carcinoma de Células Transicionales/terapia , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Cistectomía , Conjuntos de Datos como Asunto , Células Epiteliales , Regulación Neoplásica de la Expresión Génica , Ingeniería Genética , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Cultivo Primario de Células , RNA-Seq , Vejiga Urinaria/citología , Vejiga Urinaria/cirugía , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/terapia , Urotelio/citología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Proc Natl Acad Sci U S A ; 117(10): 5269-5279, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32086391

RESUMEN

We sought to define the landscape of alternative pre-mRNA splicing in prostate cancers and the relationship of exon choice to known cancer driver alterations. To do so, we compiled a metadataset composed of 876 RNA-sequencing (RNA-Seq) samples from five publicly available sources representing a range of prostate phenotypes from normal tissue to drug-resistant metastases. We subjected these samples to exon-level analysis with rMATS-turbo, purpose-built software designed for large-scale analyses of splicing, and identified 13,149 high-confidence cassette exon events with variable incorporation across samples. We then developed a computational framework, pathway enrichment-guided activity study of alternative splicing (PEGASAS), to correlate transcriptional signatures of 50 different cancer driver pathways with these alternative splicing events. We discovered that Myc signaling was correlated with incorporation of a set of 1,039 cassette exons enriched in genes encoding RNA binding proteins. Using a human prostate epithelial transformation assay, we confirmed the Myc regulation of 147 of these exons, many of which introduced frameshifts or encoded premature stop codons. Our results connect changes in alternative pre-mRNA splicing to oncogenic alterations common in prostate and many other cancers. We also establish a role for Myc in regulating RNA splicing by controlling the incorporation of nonsense-mediated decay-determinant exons in genes encoding RNA binding proteins.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Codón de Terminación/genética , Simulación por Computador , Conjuntos de Datos como Asunto , Resistencia a Antineoplásicos/genética , Exones , Femenino , Mutación del Sistema de Lectura , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-myc/genética , RNA-Seq , Transducción de Señal , Programas Informáticos
12.
Proc Natl Acad Sci U S A ; 117(22): 12315-12323, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32424106

RESUMEN

The androgen receptor (AR) antagonist enzalutamide is one of the principal treatments for men with castration-resistant prostate cancer (CRPC). However, not all patients respond, and resistance mechanisms are largely unknown. We hypothesized that genomic and transcriptional features from metastatic CRPC biopsies prior to treatment would be predictive of de novo treatment resistance. To this end, we conducted a phase II trial of enzalutamide treatment (160 mg/d) in 36 men with metastatic CRPC. Thirty-four patients were evaluable for the primary end point of a prostate-specific antigen (PSA)50 response (PSA decline ≥50% at 12 wk vs. baseline). Nine patients were classified as nonresponders (PSA decline <50%), and 25 patients were classified as responders (PSA decline ≥50%). Failure to achieve a PSA50 was associated with shorter progression-free survival, time on treatment, and overall survival, demonstrating PSA50's utility. Targeted DNA-sequencing was performed on 26 of 36 biopsies, and RNA-sequencing was performed on 25 of 36 biopsies that contained sufficient material. Using computational methods, we measured AR transcriptional function and performed gene set enrichment analysis (GSEA) to identify pathways whose activity state correlated with de novo resistance. TP53 gene alterations were more common in nonresponders, although this did not reach statistical significance (P = 0.055). AR gene alterations and AR expression were similar between groups. Importantly, however, transcriptional measurements demonstrated that specific gene sets-including those linked to low AR transcriptional activity and a stemness program-were activated in nonresponders. Our results suggest that patients whose tumors harbor this program should be considered for clinical trials testing rational agents to overcome de novo enzalutamide resistance.


Asunto(s)
Antineoplásicos/administración & dosificación , Resistencia a Antineoplásicos , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/administración & dosificación , Receptores Androgénicos/genética , Anciano , Anciano de 80 o más Años , Benzamidas , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Nitrilos , Feniltiohidantoína/administración & dosificación , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo
13.
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
14.
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
15.
Proc Natl Acad Sci U S A ; 115(19): E4473-E4482, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686080

RESUMEN

Prostate cancer is a heterogeneous disease composed of divergent molecular and histologic subtypes, including prostate adenocarcinoma (PrAd) and neuroendocrine prostate cancer (NEPC). While PrAd is the major histology in prostate cancer, NEPC can evolve from PrAd as a mechanism of treatment resistance that involves a transition from an epithelial to a neurosecretory cancer phenotype. Cell surface markers are often associated with specific cell lineages and differentiation states in normal development and cancer. Here, we show that PrAd and NEPC can be broadly discriminated by cell-surface profiles based on the analysis of prostate cancer gene expression datasets. To overcome a dependence on predictions of human cell-surface genes and an assumed correlation between mRNA levels and protein expression, we integrated transcriptomic and cell-surface proteomic data generated from a panel of prostate cancer cell lines to nominate cell-surface markers associated with these cancer subtypes. FXYD3 and CEACAM5 were validated as cell-surface antigens enriched in PrAd and NEPC, respectively. Given the lack of effective treatments for NEPC, CEACAM5 appeared to be a promising target for cell-based immunotherapy. As a proof of concept, engineered chimeric antigen receptor T cells targeting CEACAM5 induced antigen-specific cytotoxicity in NEPC cell lines. Our findings demonstrate that the surfaceomes of PrAd and NEPC reflect unique cancer differentiation states and broadly represent vulnerabilities amenable to therapeutic targeting.


Asunto(s)
Antígenos de Superficie/análisis , Antígenos de Superficie/inmunología , Carcinoma Neuroendocrino/terapia , Neoplasias de la Próstata/terapia , Proteoma/análisis , Linfocitos T/trasplante , Transcriptoma , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Antígeno Carcinoembrionario/metabolismo , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/inmunología , Carcinoma Neuroendocrino/metabolismo , Diferenciación Celular , Células Cultivadas , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Próstata/inmunología , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Proteoma/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
16.
Genes Dev ; 27(14): 1539-44, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23873937

RESUMEN

Despite several recent studies addressing the cells of origin for prostate cancer, there is still considerable discussion in the field regarding the most relevant target populations for transformation. Tissue regeneration studies have pointed to a basal cell origin for mouse and human prostate cancer. In contrast, genetically engineered mouse models demonstrate that cells within both the basal and luminal layers can initiate murine prostate cancer. Based on differences between these two approaches, we propose that further work should address the requirement for microenvironmental components such as immune or mesenchymal cells on epithelial cell types of origin for prostate cancer.


Asunto(s)
Transformación Celular Neoplásica/patología , Microambiente Celular/fisiología , Neoplasias de la Próstata/patología , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/patología , Células Epiteliales/patología , Humanos , Inflamación/patología , Masculino , Ratones , Neoplasias de la Próstata/genética
17.
Genes Dev ; 26(20): 2271-85, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23070813

RESUMEN

The cell surface protein Trop2 is expressed on immature stem/progenitor-like cells and is overexpressed in many epithelial cancers. However the biological function of Trop2 in tissue maintenance and tumorigenesis remains unclear. In this study, we demonstrate that Trop2 is a regulator of self-renewal, proliferation, and transformation. Trop2 controls these processes through a mechanism of regulated intramembrane proteolysis that leads to cleavage of Trop2, creating two products: the extracellular domain and the intracellular domain. The intracellular domain of Trop2 is released from the membrane and accumulates in the nucleus. Heightened expression of the Trop2 intracellular domain promotes stem/progenitor self-renewal through signaling via ß-catenin and is sufficient to initiate precursor lesions to prostate cancer in vivo. Importantly, we demonstrate that loss of ß-catenin or Trop2 loss-of-function cleavage mutants abrogates Trop2-driven self-renewal and hyperplasia in the prostate. These findings suggest that heightened expression of Trop2 is selected for in epithelial cancers to enhance the stem-like properties of self-renewal and proliferation. Defining the mechanism of Trop2 function in self-renewal and transformation is essential to identify new therapeutic strategies to block Trop2 activation in cancer.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transducción de Señal , Células Madre/citología , beta Catenina/metabolismo , Animales , Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/patología , Regulación de la Expresión Génica , Humanos , Hiperplasia/patología , Selectina L , Masculino , Ratones , Neoplasias de la Próstata/patología , Proteolisis
18.
Proc Natl Acad Sci U S A ; 113(16): 4482-7, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044116

RESUMEN

The cell of origin for prostate cancer remains a subject of debate. Genetically engineered mouse models have demonstrated that both basal and luminal cells can serve as cells of origin for prostate cancer. Using a human prostate regeneration and transformation assay, our group previously demonstrated that basal cells can serve as efficient targets for transformation. Recently, a subpopulation of multipotent human luminal cells defined by CD26 expression that retains progenitor activity in a defined organoid culture was identified. We transduced primary human prostate basal and luminal cells with lentiviruses expressing c-Myc and activated AKT1 (myristoylated AKT1 or myrAKT1) to mimic theMYCamplification andPTENloss commonly detected in human prostate cancer. These cells were propagated in organoid culture before being transplanted into immunodeficient mice. We found that c-Myc/myrAKT1-transduced luminal xenografts exhibited histological features of well-differentiated acinar adenocarcinoma, with strong androgen receptor (AR) and prostate-specific antigen (PSA) expression. In contrast, c-Myc/myrAKT1-transduced basal xenografts were histologically more aggressive, with a loss of acinar structures and low/absent AR and PSA expression. Our findings imply that distinct subtypes of prostate cancer may arise from luminal and basal epithelial cell types subjected to the same oncogenic insults. This study provides a platform for the functional evaluation of oncogenes in basal and luminal epithelial populations of the human prostate. Tumors derived in this fashion with defined genetics can be used in the preclinical development of targeted therapeutics.


Asunto(s)
Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Epiteliales/patología , Xenoinjertos , Humanos , Calicreínas/biosíntesis , Calicreínas/genética , Lentivirus , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Organoides/metabolismo , Organoides/patología , Fosfohidrolasa PTEN/biosíntesis , Fosfohidrolasa PTEN/genética , Próstata/patología , Antígeno Prostático Específico/biosíntesis , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Transducción Genética
19.
Proc Natl Acad Sci U S A ; 113(2): E172-81, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26621741

RESUMEN

Mutationally activated kinases play an important role in the progression and metastasis of many cancers. Despite numerous oncogenic alterations implicated in metastatic prostate cancer, mutations of kinases are rare. Several lines of evidence suggest that nonmutated kinases and their pathways are involved in prostate cancer progression, but few kinases have been mechanistically linked to metastasis. Using a mass spectrometry-based phosphoproteomics dataset in concert with gene expression analysis, we selected over 100 kinases potentially implicated in human metastatic prostate cancer for functional evaluation. A primary in vivo screen based on overexpression of candidate kinases in murine prostate cells identified 20 wild-type kinases that promote metastasis. We queried these 20 kinases in a secondary in vivo screen using human prostate cells. Strikingly, all three RAF family members, MERTK, and NTRK2 drove the formation of bone and visceral metastasis confirmed by positron-emission tomography combined with computed tomography imaging and histology. Immunohistochemistry of tissue microarrays indicated that these kinases are highly expressed in human metastatic castration-resistant prostate cancer tissues. Our functional studies reveal the strong capability of select wild-type protein kinases to drive critical steps of the metastatic cascade, and implicate these kinases in possible therapeutic intervention.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Proteínas Quinasas/metabolismo , Vísceras/patología , Animales , Neoplasias Óseas/patología , Huesos/patología , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Lentivirus , Pulmón/metabolismo , Masculino , Ratones , Ratones SCID , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Proteómica , Familia-src Quinasas/metabolismo
20.
Proc Natl Acad Sci U S A ; 113(42): E6457-E6466, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27694579

RESUMEN

Metastatic castration-resistant prostate cancer (CRPC) is the primary cause of prostate cancer-specific mortality. Defining new mechanisms that can predict recurrence and drive lethal CRPC is critical. Here, we demonstrate that localized high-risk prostate cancer and metastatic CRPC, but not benign prostate tissues or low/intermediate-risk prostate cancer, express high levels of nuclear Notch homolog 1, translocation-associated (Notch1) receptor intracellular domain. Chronic activation of Notch1 synergizes with multiple oncogenic pathways altered in early disease to promote the development of prostate adenocarcinoma. These tumors display features of epithelial-to-mesenchymal transition, a cellular state associated with increased tumor aggressiveness. Consistent with its activation in clinical CRPC, tumors driven by Notch1 intracellular domain in combination with multiple pathways altered in prostate cancer are metastatic and resistant to androgen deprivation. Our study provides functional evidence that the Notch1 signaling axis synergizes with alternative pathways in promoting metastatic CRPC and may represent a new therapeutic target for advanced prostate cancer.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Biomarcadores , Línea Celular Tumoral , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Expresión Génica , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Inmunohistoquímica , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos , Clasificación del Tumor , Metástasis de la Neoplasia , Fenotipo , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Carga Tumoral , Quinasas raf/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA