Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Int J Colorectal Dis ; 39(1): 99, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926205

RESUMO

PURPOSE: Achieving a pathologic complete response (pCR) after neoadjuvant chemoradiotherapy (NCRT) remains a challenge for most patients with rectal cancer. Exploring the potential of combining NCRT with immunotherapy or targeted therapy for those achieving a partial response (PR) offers a promising avenue to enhance treatment efficacy. This study investigated the impact of NCRT on the tumor microenvironment in locally advanced rectal cancer (LARC) patients who exhibited a PR. METHODS: This was a retrospective, observational study. Five patients demonstrating a PR after neoadjuvant treatment for LARC were enrolled in the study. Biopsy samples before treatment and resected specimens after treatment were stained with a panel of 26 antibodies targeting various immune and tumor-related markers, each labeled with distinct metal tags. The labeled samples were then analyzed using the Hyperion imaging system. RESULTS: Heterogeneity within the tumor microenvironment was observed both before and after NCRT. Notably, tumor-associated macrophages, CD4 + T cells, CD8 + T cells, CD56 + natural killer cells, tumor-associated neutrophils, cytokeratin, and E-cadherin exhibited slight increase in abundance within the tumor microenvironment following treatment (change ratios = 0.78, 0.2, 0.27, 0.32, 0.17, 0.46, 0.32, respectively). Conversely, the number of CD14 + monocytes, CD19 + B cells, CD45 + CD4 + T cells, collagen I, α-smooth muscle actin, vimentin, and ß-catenin proteins displayed significant decreases post-treatment (change ratios = 1.73, 1.92, 1.52, 1.25, 1.52, 1.12, 2.66, respectively). Meanwhile, Foxp3 + regulatory cells demonstrated no significant change (change ratio = 0.001). CONCLUSIONS: NCRT has diverse effects on various components of the tumor microenvironment in LARC patients who achieve a PR after treatment. Leveraging combination therapies may optimize treatment outcomes in this patient population.


Assuntos
Terapia Neoadjuvante , Neoplasias Retais , Microambiente Tumoral , Humanos , Neoplasias Retais/terapia , Neoplasias Retais/patologia , Neoplasias Retais/tratamento farmacológico , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Quimiorradioterapia , Resultado do Tratamento , Estudos Retrospectivos
2.
World J Surg Oncol ; 21(1): 339, 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37880729

RESUMO

BACKGROUND: To determine the efficacy of adjuvant radiotherapy for stage II-III biliary tract carcinoma. METHODS: We retrospectively analyzed the data of 37 patients who underwent radical resection of biliary tract carcinomas at the Affiliated Hospital of Inner Mongolia Medical University between 2016 and 2020. We analyzed survival differences between patients who did (n = 17) and did not (n = 20) receive postoperative adjuvant radiotherapy by using Kaplan-Meier analysis. The log-rank test and Cox univariate analysis were used. The Cox proportional risk regression model was used for the multifactorial analysis of factors influencing prognosis. RESULTS: The median survival time (28.9 vs. 14.5 months) and the 1-year (82.40% vs. 55.0%) and 2-year survival rates (58.8% vs. 25.0%) were significantly higher among patients who received adjuvant radiotherapy than among those who did not (χ2 = 6.381, p = 0.012). Multifactorial analysis showed that pathological tumor type (p = 0.004), disease stage (p = 0.021), and adjuvant radiotherapy (p = 0.001) were independent prognostic factors in biliary tract carcinoma. Subgroup analyses showed that compared to no radiotherapy, adjuvant radiotherapy significantly improved median survival time in patients with stage III disease (21.6 vs. 12.7 months; p = 0.017), positive margins (28.9 vs. 10.5 months; p = 0.012), and T3 or T4 tumors (26.8 vs. 16.8 months; p = 0.037). CONCLUSION: Adjuvant radiotherapy significantly improved the survival of patients with biliary tract carcinoma, and is recommended especially for patients with stage III disease, positive surgical margins, or ≥ T3.


Assuntos
Sistema Biliar , Carcinoma , Neoplasias Gastrointestinais , Humanos , Radioterapia Adjuvante/efeitos adversos , Estudos Retrospectivos , Prognóstico , Estadiamento de Neoplasias
3.
Clin Lab ; 68(10)2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36250845

RESUMO

BACKGROUND: Leukopenia is the most common adverse event in chemotherapy, which natural products can prevent and treat. The aim of this study was to investigate the clinical efficacy of potato extract for alleviating chemoradiotherapy-induced leukopenia in cancer patients. METHODS: This was a single-blinded randomized placebo-controlled trial that enrolled 184 cancer patients. The participants were scheduled to undergo chemoradiotherapy in two hospitals, where they were randomized to receive potato extract or a placebo in a 1:1 ratio for a period of 49 days. Change in leukocyte value was considered the primary outcome of this clinical trial. Secondary outcomes included tumor response rate, blood test, and quality of life score. RESULTS: The leukopenia was relieved in the potato extract group compared with the placebo group. Of note, a significant difference in leukopenia between the two groups was found after 14 days (p = 0.04). In addition, there was no statistically significant difference in leucocyte levels in the potato extract group (before and after potato extract treatment; p = 0.13), but in the placebo group, the leukocyte value significantly decreased compared to before treatment (p = 0.06). CONCLUSIONS: Potato extract can alleviate chemoradiotherapy-induced leukopenia in cancer patients. These results show the potential function of potato extract as a protective agent in management of cancer chemoradiotherapy.


Assuntos
Produtos Biológicos , Leucopenia , Neoplasias , Solanum tuberosum , Quimiorradioterapia/efeitos adversos , Quimiorradioterapia/métodos , Método Duplo-Cego , Humanos , Leucopenia/induzido quimicamente , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Qualidade de Vida , Método Simples-Cego , Resultado do Tratamento
4.
Carcinogenesis ; 33(7): 1302-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22610073

RESUMO

Nasopharyngeal carcinoma (NPC) has the highest metastatic potential among head and neck cancers. Distant metastasis is the major cause of treatment failure. The role of interleukin-8 (IL-8) in NPC progression remains unknown. Our multivariate survival analyses of 255 patients with NPC revealed that higher IL-8 expression in primary NPC tissue was an independent prognostic factor for overall survival, disease-free survival, and distant metastasis-free survival of the patients. In vitro study revealed that IL-8 was highly expressed in the established high-metastasis NPC clone S18 relative to the low-metastasis cells. Suppression of IL-8 by short-hairpin RNA reduced the expression of IL-8 in S18 cells and subsequently inhibited migration, invasion, and hepatic metastasis of the cells without influencing cellular growth. Overexpression of IL-8 in S26 cells resulted in increased migration, invasion, and metastasis capabilities of the cells without affecting cellular growth. Exogenous IL-8 enhanced the migration and invasion of low-metastasis CNE-2 cells in a dose-dependent manner. An epithelial-mesenchymal transition (EMT) could be induced by IL-8 in various NPC cell lines. The high level of phosphorylated AKT in S18 cells could be suppressed by knocking down IL-8 expression. Further, IL-8-promoted migration and invasion could be abolished by either the application of the phosphoinositide-3-kinase inhibitor LY294002 or the knock down of AKT expression by using small-interfering RNA. In summary, IL-8 serves as an independent prognostic indicator of overall survival, disease-free survival, and metastasis-free survival for patients with NPC. IL-8 promotes NPC metastasis via autocrine and paracrine means, involving activation of AKT signaling and inducing EMT in NPC cells.


Assuntos
Transição Epitelial-Mesenquimal , Interleucina-8/fisiologia , Neoplasias Nasofaríngeas/patologia , Metástase Neoplásica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Sequência de Bases , Primers do DNA , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Humanos , Neoplasias Nasofaríngeas/enzimologia , Prognóstico , Reação em Cadeia da Polimerase em Tempo Real
5.
Medicine (Baltimore) ; 101(52): e32410, 2022 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-36596010

RESUMO

Sacrococcygeal teratomas (SCT) in adults are extremely rare, and most SCTs are located either mainly outside the pelvis, with a small number of intrapelvic components, or mostly in the pelvis (types III and IV). The etiology of teratomas remains unknown. Most teratomas are benign, and approximately 1 to 2% of teratomas undergo malignant transformation, including squamous cell carcinoma, adenocarcinoma, sarcoma, and other malignancies. Most SCTs grow insidiously, and their symptoms are not easily detected in the early stages. Some cases may only be discovered through physical examination or compression symptoms when the tumor reaches a detectable size. Computed tomography and magnetic resonance imaging have high detection rates for presacral space-occupying lesions and can provide imaging details with guiding significance for the selection of surgical methods. Surgical resection is the preferred treatment option for SCT and can determine the pathological type. Common sacrococcygeal malignancies are mainly immature teratomas and mature teratomas. When the presence of malignant components is confirmed, the treatment model should be adjusted according to pathological type.


Assuntos
Neoplasias Pélvicas , Neoplasias da Coluna Vertebral , Teratoma , Humanos , Adulto , Região Sacrococcígea/patologia , Teratoma/diagnóstico por imagem , Teratoma/cirurgia , Tomografia Computadorizada por Raios X , Neoplasias Pélvicas/diagnóstico por imagem , Neoplasias Pélvicas/cirurgia , Neoplasias Pélvicas/patologia , Pelve/patologia , Neoplasias da Coluna Vertebral/diagnóstico por imagem , Neoplasias da Coluna Vertebral/cirurgia , Neoplasias da Coluna Vertebral/patologia
6.
Biomed Res Int ; 2019: 2920169, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31886195

RESUMO

It has been reported that 18F-FDG uptake is higher in hypoxic cancer cells than in well-oxygenated cells. We demonstrated that 18F-FDG uptake in lung cancer would be affected by high concentration oxygen breathing. Methods. Overnight fasted non-small-cell lung cancer A549 subcutaneous (s.c.) xenografts bearing mice (n = 10) underwent 18F-FDG micro-PET scans, animals breathed room air on day 1, and same animals breathed carbogen (95% O2 + 5% CO2) on the subsequent day. In separated studies, autoradiography and immunohistochemical staining visualization of frozen section of A549 s.c. tumors were applied, and to compare between carbogen-breathing mice and those with air breathing, a combination of 18F-FDG and hypoxia marker pimonidazole was injected 1 h before animal sacrifice, and 18F-FDG accumulation was compared with pimonidazole binding and glucose transporter 1 (GLUT-1) expression. Results. PET studies revealed that tumor 18F-FDG uptake was significantly decreased in carbogen-breathing mice than those with air breathing (P < 0.05). Ex vivo studies confirmed that carbogen breathing significantly decreased hypoxic fraction detected by pimonidazole staining, referring to GLUT-1 expression, and significantly decreased 18F-FDG accumulation in tumors. Conclusions. High concentration of O2 breathing during 18F-FDG uptake phase significantly decreases 18F-FDG uptake in non-small-cell lung cancer A549 xenografts growing in mice.


Assuntos
Dióxido de Carbono , Carcinoma Pulmonar de Células não Pequenas , Fluordesoxiglucose F18/farmacocinética , Neoplasias Pulmonares , Pulmão , Oxigênio , Células A549 , Animais , Dióxido de Carbono/farmacocinética , Dióxido de Carbono/farmacologia , Carcinoma Pulmonar de Células não Pequenas/química , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Modelos Animais de Doenças , Feminino , Fluordesoxiglucose F18/análise , Xenoenxertos , Humanos , Hipóxia/metabolismo , Imuno-Histoquímica , Pulmão/química , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Neoplasias Pulmonares/química , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Oxigênio/farmacocinética , Oxigênio/farmacologia , Tomografia por Emissão de Pósitrons , Distribuição Tecidual
7.
Oncotarget ; 6(12): 10239-52, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25823923

RESUMO

Nasopharyngeal carcinoma (NPC) has the highest metastasis rate among head and neck cancers with unclear mechanism. WNT5A belongs to the WNT family of cysteine-rich secreted glycoproteins. Our previous high-throughput gene expression profiling revealed that WNT5A was up-regulated in highly metastatic cells. In the present study, we first confirmed the elevated expression of WNT5A in metastatic NPC tissues at both the mRNA and protein levels. We then found that WNT5A promoted epithelial-mesenchymal transition (EMT) in NPC cells, induced the accumulation of CD24-CD44+ cells and side population, which are believed to be cancer stem cell characteristics. Moreover, WNT5A promoted the migration and invasion of NPC cells in vitro, while in vivo treatment with recombinant WNT5A promoted lung metastasis. Knocking down WNT5A diminished NPC tumorigenesis in vivo. When elevated expression of WNT5A coincided with the elevated expression of vimentin in the primary NPC, the patients had a poorer prognosis. Among major signaling pathways, protein kinase C (PKC) signaling was activated by WNT5A in NPC cells. A positive feedback loop between WNT5A and phospho-PKC to promote EMT was also revealed. Taken together, these data suggest that WNT5A is an important molecule in promoting stem cell characteristics in NPC, leading to tumorigenesis and metastasis.


Assuntos
Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Wnt/biossíntese , Proteínas Wnt/farmacologia , Animais , Carcinogênese/genética , Carcinoma , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Metástase Neoplásica , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes/farmacologia , Transfecção , Proteínas Wnt/genética , Proteína Wnt-5a
8.
Cell Cycle ; 13(12): 1958-69, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24763226

RESUMO

Nasopharyngeal carcinoma (NPC) is one of the most common malignancies in southern China and Southeast Asia, with the highest metastasis rate among head and neck cancers. The mechanisms underlying NPC progression remain poorly understood. Genome-wide expression profiling on 18 NPC vs. 18 noncancerous nasopharyngeal tissues together with GeneGo pathway analysis and expression verification in NPC cells and tissues revealed a potential role of urokinase-type plasminogen activator receptor (uPAR) in NPC progression, which has not been investigated in NPC. We then observed that uPAR expression is increased in poorly differentiated, highly metastatic NPC cells compared with lowly metastatic cells or differentiated NPC cells. In vitro studies demonstrated that uPAR regulates NPC cell growth, colony formation, migration, and invasion and promotes the epithelial-mesenchymal transition (EMT). Additional tumor xenograft and spontaneous metastasis experiments revealed that uPAR promotes NPC cell growth and metastasis in vivo. The JAK-STAT pathway is involved in uPAR-regulated signaling in NPC cells as determined by immunoblotting. Moreover, uPAR-mediated growth and motility is partially abolished upon treatment with the Jak1/Jak2 inhibitor INCB018424. We suppressed uPA expression in uPAR-overexpressing NPC cells and found that uPAR-mediated cellular growth and motility is not exclusively dependent on uPA. In summary, uPAR is a significant regulator of NPC progression and could serve as a promising therapeutic target.


Assuntos
Processos de Crescimento Celular , Neoplasias Nasofaríngeas/patologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adolescente , Adulto , Idoso , Animais , Carcinoma , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Xenoenxertos , Humanos , Janus Quinases/antagonistas & inibidores , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Metástase Neoplásica , Nitrilas , Pirazóis/farmacologia , Pirimidinas , Fatores de Transcrição STAT/metabolismo
9.
Cancer Res ; 71(8): 3162-72, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21289131

RESUMO

Nasopharyngeal carcinoma (NPC) is known for its high-metastatic potential. Here we report the identification of the proteoglycan serglycin as a functionally significant regulator of metastasis in this setting. Comparative genomic expression profiling of NPC cell line clones with high- and low-metastatic potential revealed the serglycin gene (SRGN) as one of the most upregulated genes in highly metastatic cells. RNAi-mediated inhibition of serglycin expression blocked serglycin secretion and the invasive motility of highly metastatic cells, reducing metastatic capacity in vivo. Conversely, serglycin overexpression in poorly metastatic cells increased their motile behavior and metastatic capacity in vivo. Growth rate was not influenced by serglycin in either highly or poorly metastatic cells. Secreted but not bacterial recombinant serglycin promoted motile behavior, suggesting a critical role for glycosylation in serglycin activity. Serglycin inhibition was associated with reduced expression of vimentin but not other epithelial-mesenchymal transition proteins. In clinical specimens, serglycin expression was elevated significantly in liver metastases from NPC relative to primary NPC tumors. We evaluated the prognostic value of serglycin by immunohistochemical staining of tissue microarrays from 263 NPC patients followed by multivariate analyses. High serglycin expression in primary NPC was found to be an unfavorable independent indicator of distant metastasis-free and disease-free survival. Our findings establish that glycosylated serglycin regulates NPC metastasis via autocrine and paracrine routes, and that it serves as an independent prognostic indicator of metastasis-free survival and disease-free survival in NPC patients.


Assuntos
Proteoglicanas/biossíntese , Proteínas de Transporte Vesicular/biossíntese , Adolescente , Adulto , Idoso , Carcinoma , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal , Feminino , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patologia , Invasividade Neoplásica , Metástase Neoplásica , Proteoglicanas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Transporte Vesicular/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA