Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
MAGMA ; 36(4): 621-629, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36495411

RESUMEN

OBJECTIVE: To analyze the association of apparent diffusion coefficient (ADC) values measured by readout-segmented echo-planar imaging (rs-EPI) using different simultaneous multislice (SMS) acceleration factors and the differentiation of rectal cancer grade. MATERIALS AND METHODS: Patients with non-mucinous rectal adenocarcinoma diagnosed by biopsy (endoscope-guided biopsy or surgical resection) were retrospectively collected, and each patient underwent an MRI examination. ADC values of rs-EPI, 2 × SMS rs-EPI, and 3 × SMS rs-EPI were recorded as ADC1, ADC2, and ADC3, respectively. RESULTS: The scanning time of 2 × SMS rs-EPI was 60 s, 56.2% shorter than 137 s of rs-EPI sequence, while that of 3 × SMS rs-EPI was 51 s, 72.8% less than that of rs-EPI time. The ADC value of the three groups dropped with the decrease in cancer grade (p < 0.05). The AUC values of ADC1, ADC2, and ADC3 in predicting highly differentiated rectal cancer were 0.74, 0.729, and 0.687, respectively. The difference in AUC values between ADC1 and ADC2 was not statistically significant (p = 0.889). DISCUSSION: SMS technology with an acceleration factor of 2 could be applied clinically to evaluate the pathological differentiation of rectal cancer grade.


Asunto(s)
Imagen Eco-Planar , Neoplasias del Recto , Humanos , Imagen Eco-Planar/métodos , Estudios Retrospectivos , Imagen de Difusión por Resonancia Magnética/métodos , Neoplasias del Recto/diagnóstico por imagen , Neoplasias del Recto/patología , Biopsia
2.
World J Surg Oncol ; 20(1): 142, 2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35509066

RESUMEN

BACKGROUND: Microcystic adnexal carcinoma (MAC) is a skin cancer with challenges in diagnosis and management. This study was aimed to detect molecular alterations of MAC and guide its pathologic diagnosis and treatment. METHODS: We performed transcriptome analysis on 5 MAC and 5 normal skin tissues, identified the differentially expressed genes, and verified them by immunohistochemistry. RESULTS: Three hundred four differentially expressed genes (DEGs) in MAC were identified by next-generation transcriptome sequencing, among which 225 genes were upregulated and 79 genes were downregulated. Four genes of the calcium signaling pathway, including calcium voltage-gated channel subunit alpha 1 S (CACNA1S), ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1 (ATP2A1), ryanodine receptor 1 (RYR1), and myosin light chain kinase 3 (MYLK3), were upregulated and then been verified by immunohistochemistry. The expression of CACNA1S, ATP2A1, RYR1, and MYLK3 was upregulated in MAC compared with normal sweat glands and syringoma tumor cells and was generally negative in trichoepithelioma and infundibulocystic type basal cell carcinoma. CONCLUSIONS: The four genes of the calcium signaling pathway were upregulated in MAC at both RNA and protein levels. CACNA1S, ATP2A1, RYR1, and MYLK3 may be new diagnostic molecular markers and therapeutic targets for MAC.


Asunto(s)
Canal Liberador de Calcio Receptor de Rianodina , Neoplasias Cutáneas , Señalización del Calcio , Carcinoma Basocelular , Perfilación de la Expresión Génica , Humanos , Neoplasias de Anexos y Apéndices de Piel , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Neoplasias Cutáneas/patología
3.
Cell Oncol (Dordr) ; 2023 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-37847338

RESUMEN

PURPOSE: Tumors bearing mismatch repair deficiency (MMRd) are characterized by a high load of neoantigens and are believed to trigger immunogenic reactions upon immune checkpoint blockade treatment such as anti-PD-1/PD-L1 therapy. However, the mechanisms are still ill-defined, as multiple cancers with MMRd exhibit variable responses to immune checkpoint inhibitors (ICIs). In endometrial cancer (EC), a distinct tumor microenvironment (TME) exists that may correspond to treatment-related efficacies. We aimed to characterize EC patients with aberrant MMR pathways to identify molecular subtypes predisposed to respond to ICI therapies. METHODS: We applied digital spatial profiling, a high-plex spatial transcriptomic approach covering over 1,800 genes, to obtain a highly resolved TME landscape in 45 MMRd-EC patients. We cross-validated multiple biomarkers identified using immunohistochemistry and multiplexed immunofluorescence using in-study and independent cohorts totaling 123 MMRd-EC patients and validated our findings using external TCGA data from microsatellite instability endometrial cancer (MSI-EC) patients. RESULTS: High-plex spatial profiling identified a 14-gene signature in the MMRd tumor-enriched regions stratifying tumors into "hot", "intermediate" and "cold" groups according to their distinct immune profiles, a finding highly consistent with the corresponding CD8 + T-cell infiltration status. Our validation studies further corroborated an existing coregulatory network involving HLA class I and DNMT3A potentially bridged through dynamic crosstalk incorporating CCL5. CONCLUSION: Our study confirmed the heterogeneous TME status within MMRd-ECs and showed that these ECs can be stratified based on potential biomarkers such as HLA class I, DNMT3A and CD8 in pathological settings for improved ICI therapeutic efficacy in this subset of patients.

4.
Front Oncol ; 11: 640018, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33816285

RESUMEN

OBJECTIVE: Although Polymerase-epsilon (POLE)-mutated and mismatch repair (MMR)-deficient endometrial cancers (ECs) are considered as promising candidates for anti-PD-1/PD-L1 therapy, selecting only these patients may exclude other patients who could potentially respond to this treatment strategy, highlighting the need of additional biomarkers for better patient selection. This study aims to evaluate potential predictive biomarkers for anti-PD-1/PD-L1 therapy in addition to POLE mutation (POLEm) and MMR deficiency (MMRd). METHODS: We performed next generation sequencing for POLE from 202 ECs, and immunohistochemistry for MLH1, MSH2, MSH6, PMS2, CD3, CD8, PD-1 and PD-L1 on full-section slides from these ECs. We assessed the association of POLEm and MMRd with clinicopathologic features, expression of check point proteins, and density of tumor-infiltrating lymphocytes (TILs). Prognostic impact of these immune markers was also evaluated. RESULTS: POLEm, MMRd and high-grade tumors exhibited elevated level of TILs. Increased expression of PD-1 and PD-L1 was observed in MMRd and high-grade ECs. A subgroup of MMR proficient ECs also harbored increased density of TILs, and positive expression of PD-1 and PD-L1. In addition, negative expression of checkpoint proteins and high density of TILs in combination was associated with good prognosis. CONCLUSIONS: Candidates for PD-1 blockade may extend beyond POLEm and MMRd ECs, additional factors such as tumor grade, and combination of TILs levels and expression of checkpoint proteins may need to be considered for better patient selection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA