Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
iScience ; 26(3): 106246, 2023 Mar 17.
Article En | MEDLINE | ID: mdl-36994190

Peritoneal recurrence is the most frequent and lethal recurrence pattern in gastric cancer (GC) with serosal invasion after radical surgery. However, current evaluation methods are not adequate for predicting peritoneal recurrence in GC with serosal invasion. Emerging evidence shows that pathomics analyses could be advantageous for risk stratification and outcome prediction. Herein, we propose a pathomics signature composed of multiple pathomics features extracted from digital hematoxylin and eosin-stained images. We found that the pathomics signature was significantly associated with peritoneal recurrence. A competing-risk pathomics nomogram including carbohydrate antigen 19-9 level, depth of invasion, lymph node metastasis, and pathomics signature was developed for predicting peritoneal recurrence. The pathomics nomogram had favorable discrimination and calibration. Thus, the pathomics signature is a predictive indicator of peritoneal recurrence, and the pathomics nomogram may provide a helpful reference for predicting an individual's risk in peritoneal recurrence of GC with serosal invasion.

2.
Nat Commun ; 13(1): 6903, 2022 11 12.
Article En | MEDLINE | ID: mdl-36371443

The current tumour-node-metastasis (TNM) staging system alone cannot provide adequate information for prognosis and adjuvant chemotherapy benefits in patients with gastric cancer (GC). Pathomics, which is based on the development of digital pathology, is an emerging field that might improve clinical management. Herein, we propose a pathomics signature (PSGC) that is derived from multiple pathomics features of haematoxylin and eosin-stained slides. We find that the PSGC is an independent predictor of prognosis. A nomogram incorporating the PSGC and TNM staging system shows significantly improved accuracy in predicting the prognosis compared to the TNM staging system alone. Moreover, in stage II and III GC patients with a low PSGC (but not in those with a high PSGC), satisfactory chemotherapy benefits are observed. Therefore, the PSGC could serve as a prognostic predictor in patients with GC and might be a potential predictive indicator for decision-making regarding adjuvant chemotherapy.


Stomach Neoplasms , Humans , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/drug therapy , Prognosis , Neoplasm Staging , Chemotherapy, Adjuvant , Nomograms , Retrospective Studies
3.
JAMA Netw Open ; 4(11): e2136388, 2021 11 01.
Article En | MEDLINE | ID: mdl-34846524

Importance: The current TNM staging system provides limited information for prognosis prediction and adjuvant chemotherapy benefits for patients with gastric cancer (GC). Objective: To develop a tumor-associated collagen signature of GC (TACSGC) in the tumor microenvironment to predict prognosis and adjuvant chemotherapy benefits in patients with GC. Design, Setting, and Participants: This retrospective cohort study included a training cohort of 294 consecutive patients treated between January 1, 2012, and December 31, 2013, from Nanfang Hospital, Southern Medical University, People's Republic of China, and a validation cohort of 225 consecutive patients treated between October 1, 2010, and December 31, 2012, from Fujian Provincial Cancer Hospital, Fujian Medical University, People's Republic of China. In total, 146 collagen features in the tumor microenvironment were extracted with multiphoton imaging. A TACSGC was then constructed using the least absolute shrinkage and selection operator Cox proportional hazards regression model in the training cohort. Data analysis was conducted from October 1, 2020, to April 30, 2021. Main Outcomes and Measures: The association of TACSGC with disease-free survival (DFS) and overall survival (OS) was assessed. An independent external cohort was included to validate the results. Interactions between TACSGC and adjuvant chemotherapy were calculated. Results: This study included 519 patients (median age, 57 years [IQR, 49-65 years]; 360 [69.4%] male). A 9 feature-based TACSGC was built. A higher TACSGC level was significantly associated with worse DFS and OS in both the training (DFS: hazard ratio [HR], 3.57 [95% CI, 2.45-5.20]; OS: HR, 3.54 [95% CI, 2.41-5.20]) and validation (DFS: HR, 3.10 [95% CI, 2.26-4.27]; OS: HR, 3.24 [95% CI, 2.33-4.50]) cohorts (continuous variable, P < .001 for all comparisons). Multivariable analyses found that carbohydrate antigen 19-9, depth of invasion, lymph node metastasis, distant metastasis, and TACSGC were independent prognostic predictors of GC, and 2 integrated nomograms that included the 5 predictors were established for predicting DFS and OS. Compared with clinicopathological models that included only the 4 clinicopathological predictors, the integrated nomograms yielded an improved discrimination for prognosis prediction in a C index comparison (training cohort: DFS, 0.80 [95% CI, 0.73-0.88] vs 0.78 [95% CI, 0.71-0.85], P = .03; OS, 0.81 [95% CI, 0.75-0.88] vs 0.80 [95% CI, 0.73-0.86], P = .03; validation cohort: DFS, 0.78 [95% CI, 0.70-0.87] vs 0.76 [95% CI, 0.67-0.84], P = .006; OS, 0.78 [95% CI, 0.69-0.86] vs 0.75 [95% CI, 0.67-0.84], P = .002). Patients with stage II and III GC and low TACSGC levels rather than high TACSGC levels had a favorable response to adjuvant chemotherapy (DFS: HR, 0.65 [95% CI, 0.43-0.96]; P = .03; OS: HR, 0.55 [95% CI, 0.36-0.82]; P = .004; dichotomized variable, P < .001 for interaction for both comparisons). Conclusions and Relevance: The findings suggest that TACSGC provides additional prognostic information for patients with GC and may distinguish patients with stage II and III disease who are more likely to derive benefits from adjuvant chemotherapy.


Adenocarcinoma/drug therapy , Biomarkers, Tumor/blood , Chemotherapy, Adjuvant , Collagen/blood , Collagen/therapeutic use , Disease-Free Survival , Stomach Neoplasms/drug therapy , Adenocarcinoma/physiopathology , Adenocarcinoma/surgery , Aged , China , Cohort Studies , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Stomach Neoplasms/physiopathology , Stomach Neoplasms/surgery , Treatment Outcome
4.
Ann Transl Med ; 9(8): 651, 2021 Apr.
Article En | MEDLINE | ID: mdl-33987349

BACKGROUND: Current preoperative evaluation approaches cannot provide adequate information for the prediction of lymph node (LN) metastasis in colorectal cancer (CRC). Collagen alterations in the tumor microenvironment affect the progression of tumor cells. To more accurately assess the LN status of CRC preoperatively, we developed and validated a collagen signature-based nomogram for predicting LN metastasis in CRC. METHODS: In total, 342 consecutive CRC patients were assigned to the training and validation cohorts. A total of 148 fully quantitative collagen features were extracted based on preoperative biopsies using multiphoton imaging, and the least absolute shrinkage and selection operator method was utilized to construct the collagen signature. A collagen signature-based nomogram was developed by multivariable logistic regression in the training cohort. Nomogram performance was evaluated for its discrimination, calibration, and clinical usefulness and then validated in the validation cohort. The prognostic values of the nomogram were also evaluated. RESULTS: A seven-feature-based collagen signature was built. We found that the collagen signature showed a significant association with LN metastasis in CRC. Additionally, a nomogram incorporating preoperative tumor differentiation, computed tomography-reported T stage and LN status, carcinoembryonic antigen level, carbohydrate antigen 19-9 level and collagen signature was developed. This nomogram had good discrimination and calibration, with AUROCs of 0.826 and 0.846 in the training and validation cohorts, respectively, and had a sensitivity of 86.5%, a specificity of 68.2%, an accuracy of 76.9%, a negative predictive value of 84.9%, and a positive predictive value of 71.2% for all patients. Compared to the clinicopathological model, which consisted of the clinicopathological risk factors for LN metastasis, the collagen signature-based nomogram demonstrated a significantly improved ability to discriminate LN status. Moreover, a nomogram-predicted high-risk subgroup had remarkably reduced survival compared with that of the low-risk subgroup. CONCLUSIONS: The collagen signature in the tumor microenvironment of preoperative biopsies is an independent predictor for LN metastasis in CRC, and the collagen signature-based nomogram is helpful for tailored treatment and prognostic predictions in CRC preoperatively.

5.
Nat Commun ; 12(1): 179, 2021 01 08.
Article En | MEDLINE | ID: mdl-33420057

Accurate prediction of peritoneal metastasis for gastric cancer (GC) with serosal invasion is crucial in clinic. The presence of collagen in the tumour microenvironment affects the metastasis of cancer cells. Herein, we propose a collagen signature, which is composed of multiple collagen features in the tumour microenvironment of the serosa derived from multiphoton imaging, to describe the extent of collagen alterations. We find that a high collagen signature is significantly associated with a high risk of peritoneal metastasis (P < 0.001). A competing-risk nomogram including the collagen signature, tumour size, tumour differentiation status and lymph node metastasis is constructed. The nomogram demonstrates satisfactory discrimination and calibration. Thus, the collagen signature in the tumour microenvironment of the gastric serosa is associated with peritoneal metastasis in GC with serosal invasion, and the nomogram can be conveniently used to individually predict the risk of peritoneal metastasis in GC with serosal invasion after radical surgery.


Collagen/therapeutic use , Lymphatic Metastasis/pathology , Nomograms , Peritoneal Neoplasms/pathology , Serous Membrane/pathology , Stomach Neoplasms/pathology , Cohort Studies , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Peritoneal Neoplasms/surgery , Peritoneum/pathology , Postoperative Period , Serous Membrane/surgery , Stomach Neoplasms/surgery , Tumor Microenvironment
6.
Br J Cancer ; 122(3): 388-396, 2020 02.
Article En | MEDLINE | ID: mdl-31787749

BACKGROUND: Preoperative prediction of lymph node (LN) status is integral to determining the most appropriate treatment strategy for colorectal cancer (CRC). This study aimed to develop and validate a nomogram to predict LN metastasis in CRC preoperatively. METHODS: A total of 530 patients were enrolled and divided into training and validation cohorts. The tumour stroma percentage (TSP) of the preoperative biopsies was assessed. The risk factors for LN metastasis were selected, and a nomogram was constructed subsequently. The performance of the nomogram was assessed by using the AUROC and the calibration curve, and then validated in the validation cohort. RESULTS: High TSP was significantly associated with LN metastasis in both the training and validation cohorts. Computed tomography (CT)-reported T stage, CT-reported LN status, preoperative tumour differentiation, carcinoembryonic antigen, carbohydrate antigen 19-9 and TSP were independent predictors of LN metastasis in CRC. A nomogram incorporating the six predictors was constructed. The nomogram yielded good discrimination and calibration, with an AUROC of 0.846 (95% CI: 0.807-0.886) and 0.809 (95% CI: 0.745-0.872) in the training and validation cohorts, respectively. CONCLUSIONS: Assessment of TSP in the preoperative biopsies provided additional information about the LN status. The nomogram was useful for tailored therapy in CRC preoperatively.


Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Lymph Nodes/pathology , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Adenocarcinoma/surgery , Adenocarcinoma, Mucinous/diagnostic imaging , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/surgery , Aged , Area Under Curve , Biopsy , CA-19-9 Antigen/metabolism , Carcinoembryonic Antigen/metabolism , Carcinoma, Signet Ring Cell/diagnostic imaging , Carcinoma, Signet Ring Cell/metabolism , Carcinoma, Signet Ring Cell/pathology , Carcinoma, Signet Ring Cell/surgery , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/surgery , Female , Humans , Lymph Node Excision , Lymph Nodes/diagnostic imaging , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Nomograms , Preoperative Period , ROC Curve , Reproducibility of Results , Survival Rate , Tomography, X-Ray Computed
7.
Surg Endosc ; 34(11): 5098-5106, 2020 11.
Article En | MEDLINE | ID: mdl-31792690

BACKGROUND: The feasibility of endoscopic dissection for gastric gastrointestinal stromal tumor (gGIST) between 2 and 5 cm in size has been demonstrated. However, its impact on short-term and long-term outcomes, compared with laparoscopic resection, is unknown. The purpose of this study was to compare short-term and long-term outcomes between laparoscopic resection and endoscopic dissection for 2-5-cm gGIST. METHODS: A case-matched study was performed using the propensity score. To overcome selection bias, we performed a 1:1 match using six covariates, including age, sex, BMI, ASA score, tumor size, and tumor location. Short-term and long-term outcomes between laparoscopic resection and endoscopic dissection were compared. RESULTS: A total of 210 patients with 2-5-cm gGIST were enrolled between 2006 and 2017 in our gastrointestinal center. According to the intention-to-treat approach, 165 patients underwent laparoscopic resection, and 45 patients underwent endoscopic dissection. After the propensity score, 45 pairs were balanced and analyzed. There was no significant difference in the baseline characteristics between the laparoscopic and endoscopic groups after matching. The rate of complications was significantly higher in the endoscopic group compared with the laparoscopic group (P < 0.001). Perforations occurred in 16 patients in the endoscopic group (16/45, 35.6%). The postoperative hospital stay was significantly longer in the endoscopic group compared with the laparoscopic group (P < 0.001). There was no significant difference between the two groups in disease-free survival or overall survival. CONCLUSION: Laparoscopic resection is better than endoscopic dissection for 2-5-cm gGIST because of the lower complication rate and shorter hospital stay.


Gastrointestinal Stromal Tumors/surgery , Gastroscopy/methods , Laparoscopy/methods , Stomach Neoplasms/surgery , Adult , Aged , Disease-Free Survival , Dissection , Female , Gastrectomy/methods , Humans , Length of Stay , Male , Middle Aged , Operative Time , Postoperative Period , Propensity Score , Retrospective Studies , Treatment Outcome
8.
JAMA Surg ; 154(3): e185249, 2019 03 01.
Article En | MEDLINE | ID: mdl-30698615

Importance: Lymph node status is the primary determinant in treatment decision making in early gastric cancer (EGC). Current evaluation methods are not adequate for estimating lymph node metastasis (LNM) in EGC. Objective: To develop and validate a prediction model based on a fully quantitative collagen signature in the tumor microenvironment to estimate the individual risk of LNM in EGC. Design, Setting, and Participants: This retrospective study was conducted from August 1, 2016, to May 10, 2018, at 2 medical centers in China (Nanfang Hospital and Fujian Provincial Hospital). Participants included a primary cohort (n = 232) of consecutive patients with histologically confirmed gastric cancer who underwent radical gastrectomy and received a T1 gastric cancer diagnosis from January 1, 2008, to December 31, 2012. Patients with neoadjuvant radiotherapy, chemotherapy, or chemoradiotherapy were excluded. An additional consecutive cohort (n = 143) who received the same diagnosis from January 1, 2011, to December 31, 2013, was enrolled to provide validation. Baseline clinicopathologic data of each patient were collected. Collagen features were extracted in specimens using multiphoton imaging, and the collagen signature was constructed. An LNM prediction model based on the collagen signature was developed and was internally and externally validated. Main Outcomes and Measures: The area under the receiver operating characteristic curve (AUROC) of the prediction model and decision curve were analyzed for estimating LNM. Results: In total, 375 patients were included. The primary cohort comprised 232 consecutive patients, in whom the LNM rate was 16.4% (n = 38; 25 men [65.8%] with a mean [SD] age of 57.82 [10.17] years). The validation cohort consisted of 143 consecutive patients, in whom the LNM rate was 20.9% (n = 30; 20 men [66.7%] with a mean [SD] age of 54.10 [13.19] years). The collagen signature was statistically significantly associated with LNM (odds ratio, 5.470; 95% CI, 3.315-9.026; P < .001). Multivariate analysis revealed that the depth of tumor invasion, tumor differentiation, and the collagen signature were independent predictors of LNM. These 3 predictors were incorporated into the new prediction model, and a nomogram was established. The model showed good discrimination in the primary cohort (AUROC, 0.955; 95% CI, 0.919-0.991) and validation cohort (AUROC, 0.938; 95% CI, 0.897-0.981). An optimal cutoff value was selected in the primary cohort, which had a sensitivity of 86.8%, a specificity of 93.3%, an accuracy of 92.2%, a positive predictive value of 71.7%, and a negative predictive value of 97.3%. The validation cohort had a sensitivity of 90.0%, a specificity of 90.3%, an accuracy of 90.2%, a positive predictive value of 71.1%, and a negative predictive value of 97.1%. Among the 375 patients, a sensitivity of 87.3%, a specificity of 92.1%, an accuracy of 91.2%, a positive predictive value of 72.1%, and a negative predictive value of 96.9% were found. Conclusions and Relevance: This study's findings suggest that the collagen signature in the tumor microenvironment is an independent indicator of LNM in EGC, and the prediction model based on this collagen signature may be useful in treatment decision making for patients with EGC.


Biomarkers, Tumor/metabolism , Collagen/metabolism , Lymphatic Metastasis , Stomach Neoplasms/metabolism , Tumor Microenvironment , China , Female , Gastrectomy , Humans , Male , Middle Aged , Neoplasm Invasiveness , Retrospective Studies , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
9.
Oncol Rep ; 41(1): 154-164, 2019 Jan.
Article En | MEDLINE | ID: mdl-30320346

Transmembrane protein 40 (TMEM40) is a 23­kDa protein and its association with tongue squamous cell carcinoma (TSCC) remains unclear. This study aimed to investigate the expression and clinical significance of TMEM40 in TSCC and its roles in TSCC cells. Immunohistochemical analysis was performed to detect the expression levels of TMEM40 in 60 tongue tissue samples. Furthermore, TMEM40 was overexpressed and inhibited in two TSCC cell lines by transfection with pEZ­M98­TMEM40 plasmid or TMEM40 small interfering RNA, respectively. Cell Counting Kit­8 and colony formation assays were used to investigate the effects of TMEM40 on cell proliferation and colony formation ability, respectively. Flow cytometry was performed to determine cell apoptosis and cycle conditions of transfected cells. Wound­healing and Transwell assays were processed to explore the effects of TMEM40 on cell migration and invasion, respectively. The results indicated that TMEM40 expression levels were significantly increased in TSCC tissues compared with adjacent normal tongue tissues (P<0.01). Clinicopathological analysis revealed that TMEM40 expression was positively correlated with pathological TNM (pTNM) status (P<0.05), histological grade (P<0.001) and clinical stage (P<0.01), but not with sex or age. Results of cell proliferation, apoptosis, migration and invasion assays indicated that when TMEM40 had been successfully overexpressed or knocked down in CAL27 and SCC9 TSCC cell lines, cell growth and invasion increased in the TMEM40 overexpressing cells, while they decreased in TMEM40­knockdown cells. Furthermore, experiments revealed that TMEM40 knockdown resulted in increased levels of p53 and Bax, and decreased levels of MMP­9, which indicated that TMEM40 regulated cell apoptosis and migration via involvement of p53, Bax and MMP­9 in TSCC cells. Our findings indicated that increased expression of TMEM40 contributed to progressive features of TSCC via regulation of p53, Bax and MMP­9.


Carcinoma, Squamous Cell/pathology , Membrane Proteins/metabolism , Tongue Neoplasms/pathology , Up-Regulation , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Matrix Metalloproteinase 9/metabolism , Neoplasm Staging , Signal Transduction , Tongue Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
10.
Oncol Rep ; 38(1): 109-119, 2017 Jul.
Article En | MEDLINE | ID: mdl-28586040

Human Pinx1 protein, associated with shelterin proteins, is widely revealed as a haploinsufficient tumor suppressor. Growing evidence has manifested the deregulation of PinX1 in distinct cancers. Nonetheless, the loss status of PinX1 and its diagnostic, prognostic and clinicopathological significance in Basal-like breast cancer are still unclear. In the present study, the PinX1 expression levels of breast cancer tissues were investigated by qRT-PCR and immunoblotting assays. Then immunohistochemistry (IHC) was performed to detect PinX1 expression on a tissue microarray. The optimal threshold for PinX1 positivity was determined by receiver operating characteristic (ROC) curve analysis. To clarify the probable role of PinX1 in BLBC, the PinX1 knockout and stably over-expressed MDA-MB-231 cell lines were constructed by the CRISPR-Cas9 system and gene transfection. The association of PinX1 expression with cell proliferation, migration and apoptosis of MDA-MB-231 cells were observed by CCK-8 assay, wound healing assay, transwell assay, flow cytometric analysis and immunoblotting of the cleaved caspase-3 protein level. Our results showed that both PinX1 mRNA and protein expression were downregulated in breast cancer tissues (P<0.05). In IHC analysis, the optimal cut-off parameter for PinX1 positive expression was 62.5% (the AUC was 0.749, P<0.01). PinX1 positivity was 76.9% (10/14) in luminal subtypes, 50% (5/10) in Her2-enriched breast cancer and 27.3% (9/33) in basal-like subtypes. Besides, in 59 invasive ductal breast carcinomas, PinX1 expression was inversely related to histology grade (P<0.05) while it was positively associated with PR status (P<0.05) and ER status (P<0.05). These results indicated that low expression of PinX1 correlated with aggressive clinicopathological significance of breast cancer, especially in the basal-like subtype. Besides, we identified that overexpression of PinX1 inhibited the proliferation rates and migration ability and increased the apoptosis rates of BLBC. Our findings demonstrated that low expression of PinX1 was associated with malignant behaviors in basal-like subtype of breast cancer. PinX1 is likely a feasible biomarker and molecular target of BLBC.


Breast Neoplasms/pathology , Carcinoma, Basal Cell/pathology , Carcinoma, Ductal, Breast/pathology , Cell Movement , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/metabolism , Apoptosis , Breast Neoplasms/metabolism , Carcinoma, Basal Cell/metabolism , Carcinoma, Ductal, Breast/metabolism , Case-Control Studies , Cell Cycle Proteins , Cell Proliferation , Female , Humans , Middle Aged , Prognosis , Tumor Cells, Cultured
11.
Int J Clin Exp Pathol ; 10(7): 8050-8057, 2017.
Article En | MEDLINE | ID: mdl-31966657

Transmembrane protein 40 (TMEM40) is a 23-kDa protein in cell membrane. There is no report that TMEM40 is associated with cancer. However, our study found that TMEM40 was high expressed in bladder cancer tissues. Immunohistochemical analyses of TMEM40 expression were performed on a tissue microarray including 72 transitional cell carcinomas and 43 normal bladder tissues to investigate the expression and clinical significance of TMEM40 in bladder cancer. We adopted receiver operating characteristic (ROC) analysis to select the optimal cut-off score. TMEM40 expression was defined positive if above 62.5% of cells were stained, and below it was negative. Then, the expression of TMEM40 in bladder cancer cells was evaluated by quantitative real-time PCR and western blot analysis. A significantly high level of TMEM40 in bladder cancer cells was proved. On the basis of ROC curve analysis, TMEM40 expression was positive in 68.1% (n=49) and negative in 31.9% (n=23) of bladder cancer cases. TMEM40 staining was positive in 2.3% (n=1) and negative in 97.7% (n=42) of normal bladder tissues. It showed that TMEM40 was up-regulated in bladder cancer tissues compared to normal bladder tissues. Moreover, TMEM40 expression was significantly associated with histological grade (P<0.05), clinical stage (P<0.05), pT status (P<0.05), but not age. Our study demonstrates that high TMEM40 expression is associated with bladder cancer, and it could be a diagnostic biomarker for bladder cancer.

...