Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Transl Med ; 20(1): 386, 2022 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-36058929

RESUMO

BACKGROUND: Although the anti-programmed death-1 (PD-1) inhibitor plus chemotherapy combination has been approved as the standard first-line treatment for advanced gastric cancer, a proportion of patients do not significantly benefit from this therapy. Who would respond poorly to this treatment and the underlying mechanisms of treatment failure are far from clear. METHODS: We retrospectively analyzed the associations between the peripheral basophils at baseline and clinical outcomes in 63 advanced gastric cancer patients treated with anti-PD-1 plus chemotherapy and 54 patients treated with chemotherapy alone. Immunohistochemistry and immunofluorescence staining in gastric cancer samples were utilized to investigate the basophil-related immunophenotype. RESULTS: The optimal cutoff of basophil count to distinguish responders to anti-PD-1 plus chemotherapy from non-responders was 20.0/µL. Compared with the low basophil group (≤ 20.0/µL, n = 40), the high basophil group (> 20.0/µL, n = 23) had a significantly lower objective response rate (ORR 17.4% vs. 67.5%, p = 0.0001), worse progression-free survival (median PFS 4.0 vs. 15.0 months, p = 0.0003), and worse overall survival (median OS not reached, p = 0.027). Multivariate analyses identified a basophil count of > 20.0/µL as an independent risk factor for a worse ORR (OR 0.040, 95% CI 0.007-0.241, p = 0.0004), worse PFS (HR 3.720, 95% CI 1.823-7.594, p = 0.0003) and worse OS (HR 3.427, 95% CI 1.698-6.917, p = 0.001). In contrast, there was no significant association between peripheral basophil counts and tumor response or survival in the chemotherapy-alone group (p > 0.05). In primary gastric cancer samples, we observed a correlation between higher peripheral basophil counts and the accumulation of tumor-infiltrating basophils (r = 0.6833, p = 0.005). Tumor-infiltrating basophils were found to be spatially proximate to M2 macrophages within TME and positively correlated with tumor M2 macrophage infiltration (r = 0.7234, p = 0.0023). The peripheral basophil counts also had a significant positive correlation with tumor-infiltrating M2 macrophage counts (r = 0.6584, p = 0.003). Further validation in tumor samples treated with the neoadjuvant anti-PD-1 inhibitor plus chemotherapy combination suggests that the peripheral basophils, tumor infiltration of basophils, and M2 macrophages were significantly more abundant in non-responders than in responders (p = 0.0333, p = 0.0007, and p = 0.0066, respectively). CONCLUSIONS: The peripheral basophil count was observed to be a potential biomarker of anti-PD-1 efficacy for advanced gastric cancer. Moreover, basophils may induce an immune-evasive tumor microenvironment by increasing M2 macrophage infiltration, which could be a potential immunotherapeutic target for advanced gastric cancer.


Assuntos
Neoplasias Pulmonares , Neoplasias Gástricas , Basófilos , Humanos , Contagem de Leucócitos , Macrófagos , Estudos Retrospectivos , Neoplasias Gástricas/tratamento farmacológico , Microambiente Tumoral
2.
Nutr Metab (Lond) ; 20(1): 7, 2023 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-36750880

RESUMO

BACKGROUND: Lipid metabolism dysregulation is a prominent metabolic alteration in various cancers. The study aimed to explore the association of plasma lipid metabolism profiles with overall survival (OS) for gastric cancer (GC) patients who received gastrectomy. METHODS: GC patients who were treated with gastrectomy and measured with plasma lipid metabolism profiles using proton nuclear magnetic resonance (1H-NMR) spectroscopy in Nanfang Hospital between January 1, 2017, and October 31, 2018, were recruited. The Least Absolute Shrinkage and Selection Operator (LASSO) regression model was used to analyze variables selected by univariate analysis for OS. An index of plasma lipid metabolism profiles, named plasma lipid metabolism index (PLMI), was constructed by variables' coefficients in LASSO regression to explore its association with OS and its role in the prediction model. RESULTS: A total of 158 GC patients were included in this study. Four of the 110 lipid profiles, including LDL-5 Apo-B, LDL-4 Cholesterol, HDL-4 Apo-A2, and HDL-4 Free Cholesterol, were selected to construct the PLMI. The optimal cut-off value of PLMI for OS was used to classify the population into two subgroups, the high PLMI group (≥ - 0.163) and the low PLMI group (< - 0.163). The high PLMI group had a shorter OS (p = 0.0034) and was the independent risk factor for OS (Hazard Ratio = 2.13, 95% Confidence Interval (CI): 1.07-4.22, p = 0.031) after adjusting for perineural invasion and tumor stage. In subsets of the I-III stage and treating postoperative chemotherapy, high PLMI also had an unfavorable correlation with OS (p = 0.016 and p = 0.0086, respectively). The nomogram prediction models of both the training cohort and validation cohort showed good calibration and discrimination with the concordance indexes of 0.806 (95% CI, 0.732-0.880) in the training cohort and 0.794 (95% CI, 0.725-0.862) in the validation cohort. CONCLUSIONS: This study found that the index derived from the LDL-5 Apo-B, LDL-4 Cholesterol, HDL-4 Apo-A2, and HDL-4 Free Cholesterol, was significantly associated with overall survival, suggesting that regulating lipid metabolisms might improve the prognosis for GC patients.

3.
Int J Surg ; 109(12): 4162-4172, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37720943

RESUMO

BACKGROUND: The clinical benefit of conversion surgery following immunochemotherapy in patients with stage IV gastric cancer (GC) remains uncertain. This study aims to clarify the clinical outcomes of conversion surgery for such patients. METHODS: This retrospective cohort study enroled consecutive patients with stage IV GC treated with a combination of immune checkpoint inhibitors and chemotherapy and/or anti-human epidermal growth factor receptor-2 targeted therapy as first-line therapy. Cumulative survival curves were estimated using Kaplan-Meier method. Logistic regression and Cox regression analyses were conducted to identify factors associated with conversion surgery and survival, respectively. RESULTS: Among the 136 patients included in the study. The disease control rate was 72.1% (98/136), with objective response rate in 58.8% (80/136) and complete response rate in 5.9% (8/136). Among 98 patients with disease control, 56 patients underwent palliative immunochemotherapy with median progression-free survival (PFS) and overall survival at 9.2 and 16.2 months, respectively; the remaining 42 patients underwent conversion surgery, yielding an unreached median PFS over a 19.0-month median follow-up, accompanied by 1-year overall survival and PFS rates of 96.6% and 89.1%, respectively. The R0 resection rate reached 90.5% (38/42). 7 out of 42 patients achieved pathological complete response, of whom three patients demonstrated human epidermal growth factor receptor-2 positivity. No serious complications leading to death were observed during the perioperative period. Multivariate analysis indicated that programmed death ligand 1 combined positive score greater than or equal to 5 (odds ratio, 0.22; 95% CI, 0.08-0.57; P =0.002) favored successful conversion surgery, while signet ring cell carcinoma (hazard ratio, 6.29; 95% CI, 1.56-25.36; P =0.010) was the poor prognostic factor associated with survival in patients who underwent conversion surgery. CONCLUSIONS: Conversion surgery holds the potential for significant survival benefits in stage IV GC patients who have achieved a favourable clinical response to immunochemotherapy. Individuals with signet ring cell carcinoma may experience increased post-conversion surgery recurrence.


Assuntos
Carcinoma de Células em Anel de Sinete , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/cirurgia , Inibidores de Checkpoint Imunológico/uso terapêutico , Estudos Retrospectivos , Gastrectomia/métodos , Receptores ErbB/uso terapêutico
4.
J Immunol Res ; 2022: 6011632, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35340583

RESUMO

Background: Microbes have been shown to contribute to gastric cancer (GC), gastric bacteria and viruses are associated with gastric carcinogenesis. However, the relationship between gastric fungi and GC is still unclear. Our aim was to evaluate the gastric fungal microbiota in the GC microenvironment. Methods: Gastric fungal microbiome profiling was performed with internal transcribed spacer (ITS) rDNA sequencing in primary tumor and corresponding paired normal mucosal tissues from 61 GC patients. Differences in microbial composition, taxa diversity, and predicted function were further analyzed. Results: Dysbiosis of gastric mucosal fungal microbiome was observed between the tumor and normal groups in GC. The tumor group had a higher abundance of certain taxa than the normal group. In the taxa classification, the abundances of Pezizomycetes, Sordariales, Chaetomiaceae, and Rozellomycota were lower in the tumor group than in the normal group. At the genus level, Solicoccozyma (P = 0.033) was found in higher abundance and was differentially enriched in the tumor group with Lefse analysis. Additionally, Solicoccozyma accounted for 0.3% of gastric fungi in the GC microenvironment. Twenty-seven of the 61 GC patients showed positive Solicoccozyma expression in tumors. Solicoccozyma-positive expression in tumors was associated with the Bormann classification and nerve invasion. Solicoccozyma was considered a gastric fungal marker to classify stage I and stage II-IV GC patients with an area under the receiver-operating curve (AUC) of 0.7061, as well as to classify the nerve invasive and nonnerve invasive tumors from GC patients with an AUC of 0.6978. Functional prediction indicated that the positive expression of Solicoccozyma in tumors was associated with the amino acid- and carbohydrate-related metabolic pathways in GC. Conclusions: This study revealed a novel perspective on the role of Solicoccozyma in tumors and a theoretical basis for therapeutic targets against GC.


Assuntos
Micobioma , Neoplasias , Disbiose/microbiologia , Mucosa Gástrica/patologia , Humanos , Neoplasias/patologia , Estômago/microbiologia , Microambiente Tumoral
5.
Eur J Surg Oncol ; 48(5): 1039-1045, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34836729

RESUMO

INTRODUCTION: The prognostic implication of circulating tumor cell (CTC) -associated white blood cell clusters (CTC-WBC clusters) in patients with gastric cancer (GC) after radical gastrectomy is not well defined. METHODS: The prognostic value of the CTC-WBC clusters was evaluated retrospectively in an independent cohort of GC patients with radical gastrectomy from Nanfang Hospital, Southern Medical University, China, between March 1, 2018, and September 31, 2019. The cohort was grouped into two groups: CTC-WBC group and CTC group. The CTC-WBC clusters and CTCs in blood were detected by technology of Canapatrol™ CTC filtration system. The Kaplan-Meier method was used to generate survival curve and compare the disease-free survival and OS. Cox regression model was used for multivariate analyses. RESULTS: Two hundred and seventeen patients were included for analyses, 29 patients presenting CTC-WBC clusters positive (CTC-WBC group) and 188 patients presenting exclusively CTCs (CTC group). Depth of tumor invasion was statistically different between two groups (P = 0.043), and the other clinicopathological features between the two groups were similar. Kaplan-Meier analysis showed that positive CTC-WBC cluster patients had significantly shorter OS than patients with exclusively CTC (P = 0.037). Cox regression analysis revealed that CTC-WBC cluster was an independent factor (Hazard Ratio = 2.553, 95% Confidence Interval: 1.008-6.465, P = 0.048) for OS after adjustment of age, gender, number of CTCs, type of CTCs, and tumor stage. CONCLUSION: The presence of CTC-WBC clusters is associated with poor OS in the GC patients after radical surgery regardless of tumor stage. Our data suggest alternative prognostic model needs to be further investigated.


Assuntos
Células Neoplásicas Circulantes , Neoplasias Gástricas , Biomarcadores Tumorais , Gastrectomia , Humanos , Leucócitos/patologia , Células Neoplásicas Circulantes/patologia , Prognóstico , Estudos Retrospectivos , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia
6.
J Immunol Res ; 2022: 7863480, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35859928

RESUMO

Background: Little is known about the relation between perioperative inflammatory changes and long-term survival in cancer patients. The aim of the study was to assess the association of perioperative serum interleukin-6 (IL6) and tumor necrosis factor-α (TNFα) levels with the 5-year overall survival in locally advanced gastric cancer. Methods: The 135 eligible patients in one center of Nanfang Hospital were retrieved from CLASS-01 trial (NCT01609309), an open-label, multicenter, randomized clinical noninferiority trial conducted at 14 centers in China. Serum IL6 and TNFα levels were tested before surgery, and on postoperative day (POD) 1, POD3, and POD5, respectively, referring to IL6_0, IL6_1, IL6_3, and IL6_5 and TNFα_0, TNFα_1, TNFα_3, and TNFα_5. Kaplan-Meier methods and COX models were used for survival analysis. Results: High levels of IL6_0 (≥3.67 pg/mL) and TNFα_0 (≥14.8 pg/mL) presented worse disease-free survival (DFS) (P = 0.0057 for IL6_0 and P = 0.0014 for TNFα_0) and overall survival (OS) (P = 0.0021 for IL6_0 and P = 0.0019 for TNFα_0). Both high IL6_0 and high IL6_5 levels indicated worse prognosis than other combinations (P = 0.0045 for DFS and P = 0.0022 for OS). In multivariate analysis, both high IL6_0 and high IL6_5 levels were significantly associated with poor DFS (HR = 4.29, 95% CI: 1.42-12.95, P = 0.01) and OS (HR = 4.11, 95% CI: 1.35-12.49, P = 0.013) after adjustment of tumor stage and TNFα_0. Also, high IL6_5 level was identified as the independent-related factor for postoperative infectious complications (OR = 2.69, 95% CI: 1.03-7.01, P = 0.043). Conclusions: Perioperative high serum IL6 and TNFα levels are negatively associated with 5-year survival outcomes in patients with locally advanced gastric cancer, indicating the potential survival benefits from perioperative anti-inflammatory treatment.


Assuntos
Interleucina-6/sangue , Neoplasias Gástricas , Humanos , Prognóstico , Modelos de Riscos Proporcionais , Neoplasias Gástricas/patologia , Fator de Necrose Tumoral alfa
7.
J Immunol Res ; 2022: 2430759, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36405009

RESUMO

Background: Microbiotas affected the prognosis of cancer patients by regulating programmed death ligand-1 (PD-L1) expression. However, the relationship between gastric fungi and PD-L1 expression is still unclear in gastric cancer (GC). We aimed at exploring the association of gastric fungi with PD-L1 expression and overall survival in GC. Methods: A total of 61 GC patients were divided into the two groups based on the PD-L1 combined positive scores (CPS). Fungal profiling was performed by internal transcribed spacer rDNA sequencing, and the survival analyses were performed by Kaplan-Meier curves. Results: We observed a taxonomic difference of fungi between the PD-L1-High (CPS ≥ 10) and PD-L1-Low group (CPS < 10) by principal coordinates analysis (PCoA) (P = 0.014 for Bray-Curtis and P = 0.042 for Jaccard). Malassezia had a higher abundance in the PD-L1-High group compared to the PD-L1-Low group (P = 0.045). Malassezia globosa elevated significantly in the PD-L1-High group. GC patients with PD-L1 low expression and low abundance of Malassezia globosa had a longer overall survival (OS) than others (P = 0.047). Malassezia globosa was associated with PD-L1 expression (Odds Ratio = 3.509, 95% Confidence Interval: 1.056-11.656, P = 0.040). Malassezia globosa was associated with the tumor size (P = 0.031) and PD-L1 status (P = 0.024). GC patients with a high abundance of Malassezia globosa had shorter OS than others (P = 0.028). Malassezia globosa was an independent factor (Hazard Ratio = 3.080, 95% Confidence Interval: 1.140-8.323, P = 0.027) for OS after adjusting for tumor stage. Malassezia globosa was figured out to be associated with- fatty acid and lipid biosynthesis and degradation via LIPASYN pathway. Conclusions. Malassezia globosa was identified as a PD-L1 expression-associated gastric fungus and associated with OS of GC patients, which calls for more studies to further explore its potential in PD-L1/PD-1 targeted immunotherapy.


Assuntos
Malassezia , Neoplasias Gástricas , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias Gástricas/patologia , Malassezia/metabolismo , Biomarcadores Tumorais/metabolismo
8.
Front Immunol ; 13: 1015549, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36211361

RESUMO

Background: The specific efficacy of immunotherapy for patients with liver metastases of gastric cancer is unclear. This study set out to explore the treatment response and related prognostic factors for patients with liver metastases of gastric cancer treated with immunotherapy. Patients and methods: This retrospective cohort study included 135 patients with unresectable advanced gastric cancer. According to the presence of liver metastases and/or first-line treatment with immunotherapy, patients were divided into the following three groups: I-LM(-) group(patients without liver metastases treated with immunotherapy, n=66), I-LM(+) group(patients with liver metastases treated with immunotherapy, n=36), C-LM(+) group(patients with liver metastases treated with chemotherapy and/or target therapy, n=33). Cox regression analyses were used to identify factors associated with survival in all patients and the three groups, respectively. Results: For the patients with liver metastases treated with immunotherapy, multivariate analysis showed that only the presence of peritoneal metastases was significantly associated with shorter PFS [hazard ratios (HR), 3.23; 95% CI, 1.12-9.32; P=0.030] and the patients with peritoneal metastases had shorter median PFS than patients without peritoneal metastases(3.1 vs 18.4 months; P=0.004), while the objective response rate was 100% in patients with HER2-positive (2 complete radiographic responses and 2 partial responses; 3 of 4 patients were still ongoing benefits [median follow-up time, 15.3 months ; interquartile range(IQR), 6.3-17.9 months]). Conclusions: The findings suggest that patients with various types of gastric cancer liver metastases respond differently to immune checkpoint inhibitors, HER2-positive patients may derive clinical benefits from immune checkpoint inhibitors, while the presence of peritoneal metastases is associated with resistance.


Assuntos
Neoplasias Hepáticas , Neoplasias Peritoneais , Neoplasias Gástricas , Apoptose , Humanos , Inibidores de Checkpoint Imunológico , Imunoterapia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Neoplasias Peritoneais/terapia , Prognóstico , Estudos Retrospectivos , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA