Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Prenat Diagn ; 38(13): 1069-1078, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30357877

RESUMEN

OBJECTIVE: To gather additional data on the ability to detect subchromosomal abnormalities of various sizes in single fetal cells isolated from maternal blood, using low-coverage shotgun next-generation sequencing for cell-based noninvasive prenatal testing (NIPT). METHOD: Fetal trophoblasts were recovered from approximately 30 mL of maternal blood using maternal white blood cell depletion, density-based cell separation, immunofluorescence staining, and high-resolution scanning. These trophoblastic cells were picked as single cells and underwent whole genome amplification for subsequent genome-wide copy number analysis and genotyping to confirm the fetal origin of the cells. RESULTS: Applying our fetal cell isolation method to a series of 125 maternal blood samples, we detected on average 4.17 putative fetal cells/sample. The series included 15 cases with clinically diagnosed fetal aneuploidies and five cases with subchromosomal abnormalities. This method was capable of detecting findings that were 1 to 2 Mb in size, and all were concordant with the microarray or karyotype data obtained on a fetal sample. A minority of fetal cells showed evidence of genome degradation likely related to apoptosis. CONCLUSION: We demonstrate that this cell-based NIPT method has the capacity to reliably diagnose fetal chromosomal abnormalities down to 1 to 2 Mb in size.


Asunto(s)
Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN , Eliminación de Gen , Duplicación de Gen , Trofoblastos/metabolismo , Adulto , Separación Celular , Deleción Cromosómica , Femenino , Técnica del Anticuerpo Fluorescente , Genotipo , Humanos , Cariotipo , Masculino , Embarazo , Diagnóstico Prenatal , Análisis de la Célula Individual , Análisis de Matrices Tisulares , Adulto Joven
2.
Prenat Diagn ; 36(11): 1009-1019, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27616633

RESUMEN

OBJECTIVE: The goal was to develop methods for detection of chromosomal and subchromosomal abnormalities in fetal cells in the mother's circulation at 10-16 weeks' gestation using analysis by array comparative genomic hybridization (CGH) and/or next-generation sequencing (NGS). METHOD: Nucleated cells from 30 mL of blood collected at 10-16 weeks' gestation were separated from red cells by density fractionation and then immunostained to identify cytokeratin positive and CD45 negative trophoblasts. Individual cells were picked and subjected to whole genome amplification, genotyping, and analysis by array CGH and NGS. RESULTS: Fetal cells were recovered from most samples as documented by Y chromosome PCR, short tandem repeat analysis, array CGH, and NGS including over 30 normal male cells, one 47,XXY cell from an affected fetus, one trisomy 18 cell from an affected fetus, nine cells from a trisomy 21 case, three normal cells and one trisomy 13 cell from a case with confined placental mosaicism, and two chromosome 15 deletion cells from a case known by CVS to have a 2.7 Mb de novo deletion. CONCLUSION: We believe that this is the first report of using array CGH and NGS whole genome sequencing to detect chromosomal abnormalities in fetal trophoblastic cells from maternal blood. © 2016 The Authors. Prenatal Diagnosis published by John Wiley & Sons, Ltd.


Asunto(s)
Aberraciones Cromosómicas , Hibridación Genómica Comparativa , Pruebas de Detección del Suero Materno/métodos , Análisis de Secuencia de ADN , Trofoblastos/citología , Variaciones en el Número de Copia de ADN , Estudios de Factibilidad , Femenino , Voluntarios Sanos , Humanos , Masculino , Embarazo
3.
Target Oncol ; 17(3): 329-341, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35696014

RESUMEN

BACKGROUND: Reliable biomarkers that can be serially monitored to predict treatment response to immune checkpoint inhibitors (ICIs) are still an unmet need. Here, we present a multiplex immunofluorescence (IF) assay that simultaneously detects circulating tumor cells (CTCs) and assesses CTC expression of programmed death ligand-1 (PD-L1) and interferon regulatory factor 1 (IRF-1) as a candidate biomarker related to ICI use. OBJECTIVE: To assess the potential of CTC PD-L1 and IRF-1 expression as candidate biomarkers for patients with advanced epithelial solid tumors receiving ICIs. PATIENTS AND METHODS: We tested the IF CTC assay in a pilot study of 28 patients with advanced solid tumors who were starting ICI. Blood for CTC evaluation was obtained prior to starting ICI, after a single cycle of therapy, and at the time of radiographic assessment or treatment discontinuation. RESULTS: At baseline, patients with 0-1 CTCs had longer progression-free survival (PFS) compared to patients with ≥ 2 CTCs (4.3 vs 1.3 months, p = 0.01). The presence of any PD-L1+ CTCs after a single dose of ICI portended shorter PFS compared to patients with no CTCs or PD-L1- CTCs (1.2 vs 4.2 months, p = 0.02); the presence of any PD-L1+ or IRF-1+ CTCs at time of imaging assessment or treatment discontinuation also was associated with shorter PFS (1.9 vs 5.5 months, p < 0.01; 1.6 vs 4.7 months, p = 0.05). CTC PD-L1 and IRF-1 expression did not correlate with tumor tissue PD-L1 or IRF-1 expression. Strong IRF-1 expression in tumor tissue was associated with durable (≥ 1 year) radiographic response (p = 0.02). CONCLUSIONS: Based on these results, CTC PD-L1 and IRF-1 expression is of interest in identifying ICI resistance and warrants further study.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Células Neoplásicas Circulantes , Antígeno B7-H1 , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Humanos , Inhibidores de Puntos de Control Inmunológico , Factor 1 Regulador del Interferón/metabolismo , Biopsia Líquida , Neoplasias Pulmonares/tratamiento farmacológico , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Proyectos Piloto
4.
Cancer Immunol Immunother ; 59(4): 587-98, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19826812

RESUMEN

Tumor-associated macrophages (TAM) are very abundant in tumors and are thought to play a major role in promoting tumor growth. The generation of TAM is positively regulated by several cytokines, including colony stimulating factor-1 (CSF-1) and monocyte chemoattractant protein-1 (CCL2). However, endogenous factors that suppress the generation of TAM within tumors have not been previously identified. An earlier study showed that endogenously produced type I interferons (IFN) suppressed tumor growth via their effects on hematopoietic cells rather than through direct effects on tumor cells. Therefore, we used mouse tumor models to investigate the effects of endogenously produced type I IFNs on the generation of TAM. We found using immunohistochemistry and flow cytometry that TAM density was significantly increased in tumors of mice lacking the type I IFN receptor (IFN-alpha/betaR(-/-) mice) compared to wild type mice. Moreover, the increase in TAM density was associated with a significant increase in tumor growth rate and angiogenesis. The phenotype of TAM was similar in IFN-alpha/betaR(-/-) mice and wild type mice and tumors in both mice produced similar amounts of CSF-1 and CCL2. However, in vitro assays indicated that low concentrations of type I IFNs significantly inhibited the generation of bone marrow macrophages in response to CSF-1. These findings indicate that endogenously produced type I IFNs suppress the generation of TAM, which may in turn account for inhibition of tumor growth and angiogenesis.


Asunto(s)
Movimiento Celular , Interferón Tipo I/fisiología , Macrófagos/efectos de los fármacos , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Animales , Western Blotting , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnicas para Inmunoenzimas , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/prevención & control , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Cancer Immunol Immunother ; 59(3): 367-78, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19701748

RESUMEN

Clinical studies over the past several years have reported that metastasis-free survival times in humans and dogs with osteosarcoma are significantly increased in patients that develop chronic bacterial osteomyelitis at their surgical site. However, the immunological mechanism by which osteomyelitis may suppress tumor growth has not been investigated. Therefore, we used a mouse model of osteomyelitis to assess the effects of bone infection on innate immunity and tumor growth. A chronic Staphylococcal osteomyelitis model was established in C3H mice and the effects of infection on tumor growth of syngeneic DLM8 osteosarcoma were assessed. The effects of infection on tumor angiogenesis and innate immunity, including NK cell and monocyte responses, were assessed. We found that osteomyelitis significantly inhibited the growth of tumors in mice, and that the effect was independent of the infecting bacterial type, tumor type, or mouse strain. Depletion of NK cells or monocytes reversed the antitumor activity elicited by infection. Moreover, infected mice had a significant increase in circulating monocytes and numbers of tumor associated macrophages. Infection suppressed tumor angiogenesis but did not affect the numbers of circulating endothelial cells. Therefore, we concluded that chronic localized bacterial infection could elicit significant systemic antitumor activity dependent on NK cells and macrophages.


Asunto(s)
Osteomielitis/complicaciones , Osteosarcoma/complicaciones , Osteosarcoma/inmunología , Infecciones Estafilocócicas/complicaciones , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Inmunidad Innata , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos , Monocitos/inmunología , Neoplasias , Neovascularización Patológica , Osteosarcoma/patología
6.
J Vet Intern Med ; 21(4): 764-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17708397

RESUMEN

BACKGROUND: Hemangiosarcoma (HSA) is a highly metastatic and often rapidly fatal tumor in dogs. At present, conventional adjuvant chemotherapy provides only a modest survival benefit for treated dogs. Continuous oral administration of low-dose chemotherapy (LDC) has been suggested as an alternative to conventional chemotherapy protocols. Therefore, we evaluated the safety and effectiveness of LDC using a combination of cyclophosphamide, etoposide, and piroxicam as adjuvant therapy for dogs with stage II HSA. HYPOTHESIS: We hypothesized that oral adjuvant therapy with LDC could be safely administered to dogs with HSA and that survival times would be comparable to those attained with conventional doxorubicin (DOX) chemotherapy. ANIMALS: Nine dogs with stage II splenic HSA were enrolled in the LDC study. Treatment outcomes were also evaluated retrospectively for 24 dogs with stage II splenic HSA treated with DOX chemotherapy. METHODS: Nine dogs with stage II splenic HSA were treated with LDC over a 6-month period. Adverse effects and treatment outcomes were determined. The pharmacokinetics of orally administered etoposide were determined in 3 dogs. Overall survival times and disease-free intervals were compared between the 9 LDC-treated dogs and 24 DOX-treated dogs. RESULTS: Dogs treated with LDC did not develop severe adverse effects, and long-term treatment over 6 months was well-tolerated. Oral administration of etoposide resulted in detectable plasma concentrations that peaked between 30 and 60 minutes after dosing. Both the median overall survival time and the median disease-free interval in dogs treated with LDC were 178 days. By comparison, the overall survival time and disease-free interval in dogs treated with DOX were 133 and 126 days, respectively. CONCLUSIONS: Continuous orally administered LDC may be an effective alternative to conventional high-dose chemotherapy for adjuvant therapy of dogs with HSA.


Asunto(s)
Enfermedades de los Perros/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Etopósido/administración & dosificación , Etopósido/uso terapéutico , Hemangiosarcoma/veterinaria , Neoplasias del Bazo/veterinaria , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Perros , Doxorrubicina/administración & dosificación , Etopósido/farmacocinética , Femenino , Hemangiosarcoma/tratamiento farmacológico , Masculino , Neoplasias del Bazo/tratamiento farmacológico
7.
J Vet Intern Med ; 21(1): 113-20, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17338158

RESUMEN

BACKGROUND: Hemangiosarcoma (HSA) is a highly metastatic and often rapidly fatal tumor of dogs. At present, adjuvant chemotherapy is the only proven effective treatment for dogs with HSA, though the benefits from chemotherapy are modest. Administration of immunotherapy together with chemotherapy has also been reported to improve survival in dogs with HSA. Therefore, we evaluated safety and immunologic responses to a novel tumor vaccine administered together with doxorubicin chemotherapy in dogs with different stages of HSA. HYPOTHESIS: That tumor vaccination could be safely and effectively combined with doxorubicin chemotherapy for treatment of dogs with HSA. ANIMALS: Twenty-eight dogs with various stages of HSA were enrolled in the study. METHODS: The HSA vaccine was prepared with lysates of allogeneic canine HSA cell lines mixed with an adjuvant composed of liposome-DNA complexes. Dogs received a series of 8 immunizations administered over a 22-week period, and most also received chemotherapy. Clinical adverse effects were noted, immune responses were measured by enzyme-linked immunosorbent assay (ELISA) and flow cytometry, and survival times were calculated. RESULTS: The most common adverse effects observed in vaccinated dogs also treated with doxorubicin chemotherapy were diarrhea and anorexia. Vaccinated dogs were found to mount strong humoral immune responses against a control antigen and, most dogs also mounted antibody responses against canine HSA cells. Thirteen dogs with stage II splenic HSA that received the tumor vaccine plus doxorubicin chemotherapy had an overall median survival time of 182 days. CONCLUSIONS: We conclude that an allogeneic tumor lysate vaccine is safe in dogs with HSA and can elicit humoral immune responses in dogs that are receiving concurrent doxorubicin chemotherapy.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Hemangiosarcoma/veterinaria , Neoplasias Cutáneas/veterinaria , Neoplasias del Bazo/veterinaria , Animales , Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/efectos adversos , Quimioterapia Adyuvante/veterinaria , Perros , Doxorrubicina/uso terapéutico , Quimioterapia Combinada , Neoplasias Cardíacas/tratamiento farmacológico , Neoplasias Cardíacas/veterinaria , Hemangiosarcoma/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/veterinaria , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias del Bazo/tratamiento farmacológico
8.
Methods Mol Biol ; 1634: 163-172, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28819849

RESUMEN

The RareCyte platform addresses important technology limitations of current circulating tumor cell (CTC) collection methods, and expands CTC interrogation to include advanced phenotypic characterization and single-cell molecular analysis. In this respect, it represents the "next generation" of cell-based liquid biopsy technologies. In order to identify and analyze CTCs, RareCyte has developed an integrated sample preparation, imaging and individual cell retrieval process. The first step in the process, AccuCyte®, allows the separation, collection, and transfer to a slide the nucleated cell fraction of the blood that contains CTCs. Separation and collection are based on cell density-rather than size or surface molecular expression-and are performed within a closed system, without wash or lysis steps, enabling high CTC recovery. Here, we describe our technique for nucleated cell collection from a blood sample, and the spreading of these nucleated cells onto glass slides permitting immunofluorescent staining, cell identification, and individual cell picking described in subsequent chapters. In addition to collection of rare cells such as CTCs, AccuCyte also collects cells of the circulating immune system onto archivable slides as well as plasma from the same sample.


Asunto(s)
Separación Celular/métodos , Células Inmovilizadas/patología , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patología , Análisis de la Célula Individual/métodos , Recuento de Células , Línea Celular Tumoral , Separación Celular/instrumentación , Células Inmovilizadas/metabolismo , Centrifugación/instrumentación , Centrifugación/métodos , Diseño de Equipo , Humanos , Neoplasias/sangre , Neoplasias/patología , Células Neoplásicas Circulantes/metabolismo , Juego de Reactivos para Diagnóstico/normas , Análisis de la Célula Individual/instrumentación
9.
Cancer Res ; 73(1): 74-85, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23161490

RESUMEN

Vaccination with antigens expressed by tumors is one strategy for stimulating enhanced T-cell responses against tumors. However, these peptide vaccines rarely result in efficient expansion of tumor-specific T cells or responses that protect against tumor growth. Mimotopes, or peptide mimics of tumor antigens, elicit increased numbers of T cells that crossreact with the native tumor antigen, resulting in potent antitumor responses. Unfortunately, mimotopes may also elicit cells that do not crossreact or have low affinity for tumor antigen. We previously showed that one such mimotope of the dominant MHC class I tumor antigen of a mouse colon carcinoma cell line stimulates a tumor-specific T-cell clone and elicits antigen-specific cells in vivo, yet protects poorly against tumor growth. We hypothesized that boosting the mimotope vaccine with the native tumor antigen would focus the T-cell response elicited by the mimotope toward high affinity, tumor-specific T cells. We show that priming T cells with the mimotope, followed by a native tumor-antigen boost, improves tumor immunity compared with T cells elicited by the same prime with a mimotope boost. Our data suggest that the improved tumor immunity results from the expansion of mimotope-elicited tumor-specific T cells that have increased avidity for the tumor antigen. The enhanced T cells are phenotypically distinct and enriched for T-cell receptors previously correlated with improved antitumor immunity. These results suggest that incorporation of native antigen into clinical mimotope vaccine regimens may improve the efficacy of antitumor T-cell responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos BALB C
10.
PLoS One ; 8(1): e53789, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23342002

RESUMEN

Mucosal-associated invariant T cells are a unique population of T cells that express a semi-invariant αß TCR and are restricted by the MHC class I-related molecule MR1. MAIT cells recognize uncharacterized ligand(s) presented by MR1 through the cognate interaction between their TCR and MR1. To understand how the MAIT TCR recognizes MR1 at the surface of APCs cultured both with and without bacteria, we undertook extensive mutational analysis of both the MAIT TCR and MR1 molecule. We found differential contribution of particular amino acids to the MAIT TCR-MR1 interaction based upon the presence of bacteria, supporting the hypothesis that the structure of the MR1 molecules with the microbial-derived ligand(s) differs from the one with the endogenous ligand(s). Furthermore, we demonstrate that microbial-derived ligand(s) is resistant to proteinase K digestion and does not extract with common lipids, suggesting an unexpected class of antigen(s) might be recognized by this unique lymphocyte population.


Asunto(s)
Escherichia coli/fisiología , Antígenos de Histocompatibilidad Clase I/metabolismo , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/microbiología , Animales , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Línea Celular , Técnicas de Cocultivo , Endopeptidasa K/metabolismo , Escherichia coli/citología , Escherichia coli/inmunología , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/genética , Metabolismo de los Lípidos , Ratones , Antígenos de Histocompatibilidad Menor , Modelos Moleculares , Mutagénesis , Mutación , Unión Proteica , Conformación Proteica , Proteolisis , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA