1
|
Gao B, Gou X, Feng C, Zhang Y, Gu H, Chai F, Wang Y, Ye Y, Hong N, Hu G, Sun B, Cheng J, Yang H. Identification of cancer-associated fibrolast subtypes and distinctive role of MFAP5 in CT-detected extramural venous invasion in gastric cancer. Transl Oncol 2025; 51:102188. [PMID: 39531783 PMCID: PMC11600027 DOI: 10.1016/j.tranon.2024.102188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Extramural venous invasion (EMVI) detected by computed tomography has been identified as an independent risk factor for distant metastasis in patients with advanced gastric cancer (GC). Cancer-associated fibroblasts (CAFs) are critical for remodeling the tumor microenvironment in GCs. Here, we report that MFAP5+ CAFs promote the formation of EMVI imaging in GC. We detected gene expression in pathological samples from 13 advanced GC patients with EMVI. Radiogenomics results showed the degree of CAFs infiltration was directly proportional to the EMVI score and EMT pathway in GC patients. Single-cell sequencing data analysis results showed that MFAP5+CAFs subtypes in GC were negatively correlated with patient prognosis and were enriched in tumor lactylation modification and EMT pathways. Immunohistochemistry results showed that the expression of MFAP5, L-lactyl and EMT markers in GC tissues was proportional to the EMVI score. CAF from gastric cancer tissue was extracted using collagenase method and co-cultured with GC cell line in vitro. After lentivirus knockdown of MFAP5 in CAFs, the levels of L-lactoyl and histone lactylation modifications were significantly reduced, and the sphere-forming and vascularization abilities of CAFs were significantly inhibited. Cell function experiments showed that MFAP5+ CAFs can affect the EMT, metastasis and invasion capabilities of GC cells. In vivo experimental results of the nude mouse in situ EMVI model suggest that MFAP5+ CAF may promote the formation of EMVI imaging features in GC by regulating lactylation modification. This innovative work may provide important new references for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Bo Gao
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Xinyi Gou
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Huining Gu
- Department of Immunology, School of Basic Medical Sciences, Peking University and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Guohua Hu
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Boshi Sun
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China.
| | - Hao Yang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
2
|
Yang H, Gou X, Feng C, Zhang Y, Sun B, Peng P, Wang Y, Hong N, Ye Y, Cheng J, Gao B. Overall survival prediction of gastric cancer using the gene signature of CT-detected extramural venous invasion combined with M2 macrophages infiltration. J Transl Med 2024; 22:829. [PMID: 39252063 PMCID: PMC11382430 DOI: 10.1186/s12967-024-05628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/18/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND CT-detected Extramural venous invasion (EMVI) is known as an independent risk factor for distant metastasis in patients with advanced gastric cancer (GC). However, the molecular basis is not clear. In colorectal cancer, M2 macrophages plays a vital role in determining EMVI. This study aimed to investigate the relationship between CT-detected EMVI and the M2 macrophages as well as prognosis predictionusing a radiogenomic approach. METHOD We utilized EMVI-related genes (from mRNA sequencing of 13 GC samples correlated with EMVI score by spearman analysis, P < 0.01) to overlap the co-expression genes of WGCNA module and M2 macrophages related genes (from mRNA data of 371 GC patients in TCGA database), generating a total of 136 genes. An EMVI-M2-prognosis-related hub gene signature was constructed by COX and least absolute shrinkage and selection operator (LASSO) analysis from a training cohort TCGA database (n = 371) and validated it in a validation cohort from GEO database (n = 357). High- and low-risk groups were divided by hub gene (EGFLAM and GNG11) signature-derived risk scores. We assessed its predictive ability through Kaplan-Meier (K-M) curve and COX analysis. Furthermore, we utilized ESTIMATE to detect tumor mutation burden (TMB) and evaluate sensitivity to immune checkpoint inhibitors (ICIs). Expression of hub genes was tested using western blotting and immunohistochemistry (IHC) analysis. RESULTS The overall survival (OS) was significantly reduced in the high-risk group (Training/Validation: AUC = 0.701/0.620; P < 0.001/0.003). Furthermore, the risk score was identified as an independent predictor of OS in multivariate COX regression analyses (Training/Validation: HR = 1.909/1.928; 95% CI: 1.225-2.974/1.308-2.844). The low-risk group exhibited significantly higher TMB levels (P = 1.6e- 07) and greater sensitivity to ICIs. Significant higher expression of hub-genes was identified on multiple GC cell lines and original samples. Hub-genes knockdown in gastric cancer cell lines inhibited their proliferation, metastatic and invasive capacity to varying degrees. In vivo experiments indicate that EGFLAM, as one of the hub genes, its high expression can serve as a biomarker for low response to immunotherapy. CONCLUSION Our study demonstrated EMVI-M2 gene signature could effectively predict the prognosis of GC tissue, reflecting the relationship between EMVI and M2 macrophages.
Collapse
Affiliation(s)
- Hao Yang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xinyi Gou
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Yuanyuan Zhang
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Boshi Sun
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Peng
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China.
| | - Bo Gao
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
3
|
Chen Z, Zhang G, Liu Y, Zhu K. Radiomics analysis in predicting vascular invasion in gastric cancer based on enhanced CT: a preliminary study. BMC Cancer 2024; 24:1020. [PMID: 39152398 PMCID: PMC11330039 DOI: 10.1186/s12885-024-12793-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Vascular invasion (VI) is closely related to the metastasis, recurrence, prognosis, and treatment of gastric cancer. Currently, predicting VI preoperatively using traditional clinical examinations alone remains challenging. This study aims to explore the value of radiomics analysis based on preoperative enhanced CT images in predicting VI in gastric cancer. METHODS We retrospectively analyzed 194 patients with gastric adenocarcinoma who underwent enhanced CT examination. Based on pathology analysis, patients were divided into the VI group (n = 43) and the non-VI group (n = 151). Radiomics features were extracted from arterial phase (AP) and portal venous phase (PP) CT images. The radiomics score (Rad-score) was then calculated. Prediction models based on image features, clinical factors, and a combination of both were constructed. The diagnostic efficiency and clinical usefulness of the models were evaluated using receiver operating characteristic (ROC) curves and decision curve analysis (DCA). RESULTS The combined prediction model included the Rad-score of AP, the Rad-score of PP, Ki-67, and Lauren classification. In the training group, the area under the curve (AUC) of the combined prediction model was 0.83 (95% CI 0.76-0.89), with a sensitivity of 64.52% and a specificity of 92.45%. In the validation group, the AUC was 0.80 (95% CI 0.67-0.89), with a sensitivity of 66.67% and a specificity of 88.89%. DCA indicated that the combined prediction model might have a greater net clinical benefit than the clinical model alone. CONCLUSION The integrated models, incorporating enhanced CT radiomics features, Ki-67, and clinical factors, demonstrate significant predictive capability for VI. Moreover, the radiomics model has the potential to optimize personalized clinical treatment selection and patient prognosis assessment.
Collapse
Affiliation(s)
- Zhicheng Chen
- Department of Radiology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 100004, China
- Department of Radiology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Guangfeng Zhang
- Department of Radiology, Children's Hospital Affiliated to Shandong University, 23976 Jingshi road, Huaiyin District, Jinan, 250000, China
- Department of Radiology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Yi Liu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Kexin Zhu
- Department of Radiology, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| |
Collapse
|
4
|
Yang H, Gou X, Feng C, Zhang Y, Chai F, Hong N, Ye Y, Wang Y, Gao B, Cheng J. Computed tomography-detected extramural venous invasion-related gene signature: a potential negative biomarker of immune checkpoint inhibitor treatment in patients with gastric cancer. J Transl Med 2023; 21:4. [PMID: 36604653 PMCID: PMC9814439 DOI: 10.1186/s12967-022-03845-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND To investigate the association between computed tomography (CT)-detected extramural venous invasion (EMVI)-related genes and immunotherapy resistance and immune escape in patients with gastric cancer (GC). METHODS Thirteen patients with pathologically proven locally advanced GC who had undergone preoperative abdominal contrast-enhanced CT and radical resection surgery were included in this study. Transcriptome sequencing was multidetector performed on the cancerous tissue obtained during surgery, and EMVI-related genes (P value for association < 0.001) were selected. A single-sample gene set enrichment analysis algorithm was also used to divide all GC samples (n = 377) in The Cancer Genome Atlas (TCGA) database into high and low EMVI-immune related groups based on immune-related differential genes. Cluster analysis was used to classify EMVI-immune-related genotypes, and survival among patients was validated in TCGA and Gene Expression Omnibus (GEO) cohorts. The EMVI scores were calculated using principal component analysis (PCA), and GC samples were divided into high and low EMVI score groups. Microsatellite instability (MSI) status, tumor mutation burden (TMB), response rate to immune checkpoint inhibitors (ICIs), immune escape were compared between the high and low EMVI score groups. Hub gene of the model in pan-cancer analysis was also performed. RESULTS There were 17 EMVI-immune-related genes used for cluster analysis. PCA identified 8 genes (PCH17, SEMA6B, GJA4, CD34, ACVRL1, SOX17, CXCL12, DYSF) that were used to calculate EMVI scores. High EMVI score groups had lower MSI, TMB and response rate of ICIs, status but higher immune escape status. Among the 8 genes used for EMVI scores, CXCL12 and SOX17 were at the core of the protein-protein interaction (PPI) network and had a higher priority in pan-cancer analysis. Immunohistochemical analysis showed that the expression of CXCL12 and SOX17 was significantly higher in CT-detected EMVI-positive samples than in EMVI-negative samples (P < 0.0001). CONCLUSION A CT-detected EMVI gene signature could be a potential negative biomarker for ICIs treatment, as the signature is negatively correlated with TMB, and MSI, resulting in poorer prognosis.
Collapse
Affiliation(s)
- Hao Yang
- grid.412463.60000 0004 1762 6325Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyi Gou
- grid.411634.50000 0004 0632 4559Department of Radiology, Peking University People’s Hospital, 11 Xizhimen South St., Beijing, 100044 China
| | - Caizhen Feng
- grid.411634.50000 0004 0632 4559Department of Radiology, Peking University People’s Hospital, 11 Xizhimen South St., Beijing, 100044 China
| | - Yinli Zhang
- grid.411634.50000 0004 0632 4559Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Fan Chai
- grid.411634.50000 0004 0632 4559Department of Radiology, Peking University People’s Hospital, 11 Xizhimen South St., Beijing, 100044 China
| | - Nan Hong
- grid.411634.50000 0004 0632 4559Department of Radiology, Peking University People’s Hospital, 11 Xizhimen South St., Beijing, 100044 China
| | - Yingjiang Ye
- grid.411634.50000 0004 0632 4559Department of Gastrointestinal Surgery, Peking University People’s Hospital, Beijing, China
| | - Yi Wang
- grid.411634.50000 0004 0632 4559Department of Radiology, Peking University People’s Hospital, 11 Xizhimen South St., Beijing, 100044 China
| | - Bo Gao
- grid.411634.50000 0004 0632 4559Department of General Surgery, Peking University People’s Hospital, Beijing, 100044 China
| | - Jin Cheng
- grid.411634.50000 0004 0632 4559Department of Radiology, Peking University People’s Hospital, 11 Xizhimen South St., Beijing, 100044 China
| |
Collapse
|
5
|
Gastric adenocarcinoma: A review of the TNM classification system and ways of spreading. RADIOLOGIA 2023; 65:66-80. [PMID: 36842787 DOI: 10.1016/j.rxeng.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 02/28/2023]
Abstract
Gastric cancer is the fifth most common cancer in the world. The most common histologic subtype is adenocarcinoma. Gastric adenocarcinomas are staged using the American Joint Committee on Cancer's 8th TNM classification. The perigastric ligaments, mesentery, omentum, and potential spaces between the parietal and visceral peritoneal linings play are important structures for staging. The spread of disease is influenced by the location of the tumor within the stomach, as well as by the anatomy related to the ligaments and lymph vessels. CT is the imaging modality of choice for the preoperative clinical staging of gastric cancer, and it is essential for planning treatment. To be able to do an adequate imaging workup, radiologists need to know the different pathways through which gastric cancer can spread: lymphatic, subperitoneal, direct invasion, transperitoneal, hematogenous, and extramural venous invasion.
Collapse
|
6
|
Adenocarcinoma gástrico: revisión del TNM y de las vías de diseminación. RADIOLOGIA 2022. [DOI: 10.1016/j.rx.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
7
|
Guan Z, Zhang XY, Li XT, Sun RJ, Lu QY, Wu AW, Sun YS. Correlation and prognostic value of CT-detected extramural venous invasion and pathological lymph-vascular invasion in colon cancer. Abdom Radiol (NY) 2022; 47:1232-1243. [PMID: 35133470 DOI: 10.1007/s00261-022-03414-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE To explore the association between CT-detected extramural vascular invasion (ctEMVI) and lymph-vascular invasion (LVI) in colon cancer, and analyze the prognostic value of ctEMVI in different conditions of LVI. METHODS This single-center, retrospective study included 448 colon cancer patients from January 2015 to December 2017. Preoperative CT features and clinical and pathological data were collected. Associations between ctEMVI and LVI were tested. Univariate and multivariate logistic regression was performed. Multivariate Cox regression was performed adjusted with propensity score(PS). Kaplan-Meier method was used to compare survival differences between the ctEMVI and LVI groups. A 1:1 patient pairing was conducted using PS matching to assess the prognostic effect of ctEMVI in LVI subgroups. RESULTS Among the 448 patients, there were 261 men and 187 women, with an average age of 63 ± 12 years. The coincidence rate of ctEMVI and LVI was 73.9%. The k coefficient for identifying ctEMVI was 0.84. ctEMVI and LVI were both independent risk factors for overall survival (ctEMVI: HR 2.8, 95% CI 1.5-5.5; LVI: HR 2.2, 95% CI 1.2-4.1) and metastasis-free survival (ctEMVI: HR 3.3, 95% CI 1.7-6.4; LVI: HR 2.4, 95% CI 1.3-4.5) adjusted with PS. In the LVI(+) subgroup, the prognosis of ctEMVI(+) was significantly worse than that of ctEMVI(-); in the LVI(-) subgroup, the prognosis of different ctEMVI states was similar. CONCLUSION ctEMVI is an independent prognostic risk factor and has different prognostic value in different LVI states. It is recommended to perform the evaluation in routine work, especially for patients with positive LVI.
Collapse
Affiliation(s)
- Zhen Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Xiao-Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Rui-Jia Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Qiao-Yuan Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Ai-Wen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China.
| |
Collapse
|
8
|
Yang S, Zou X, Li J, Zhang A, Zhu L, Hu X, Li C. The Application Value of ceMDCT in the Diagnosis of Gastric Cancer Extramural Vascular Invasion and Its Influencing Factors. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:4239600. [PMID: 35035833 PMCID: PMC8759867 DOI: 10.1155/2022/4239600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 12/04/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the value of enhanced multislice spiral CT (ceMDCT) in the diagnosis of extramural vascular invasion of gastric cancer and the influencing factors of extramural vascular invasion. There are different methods used in this paper. METHOD 131 patients with primary gastric cancer treated in our hospital from January 2017 to May 2019 were selected. All patients underwent surgical resection and ceMDCT examination before operation. RESULT There were 40 cases with extramural vascular invasion of gastric cancer by surgical pathological diagnosis. The kappa value of ceMDCT in diagnosing extramural vascular invasion of gastric cancer was 0.947, and the consistency was excellent. The diagnostic sensitivity, specificity, positive predictive value, and negative predictive value were 100.00%, 96.70%, 93.02%, and 100.00%, respectively. The proportions of T3-T4, tumour diameter ≥5.0 cm, and growth pattern of proximal nodular + diffuse type in patients with gastric cancer extramural vascular invasion were 92.50%, 85.00%, and 65.00%, respectively, which were significantly higher than those in patients without extramural vascular invasion (P < 0.05). The logistic regression analysis results showed that T3-T4, tumour diameter ≥5.0 cm, proximal nodular + diffuse growth pattern were the risk factors for extrahepatic vascular invasion in gastric cancer (OR = 3.751, 2.901, and 3.367, P < 0.05). CONCLUSION ceMDCT has good application value in diagnosing gastric cancer extramural vascular invasion. The occurrence of gastric cancer extramural vascular invasion is affected by T staging, tumour diameter, and tumour growth pattern.
Collapse
Affiliation(s)
- Shifeng Yang
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| | - Xiaoming Zou
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| | - Jiacheng Li
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| | - Ange Zhang
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| | - Lei Zhu
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| | - Xiaolong Hu
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| | - Changjian Li
- Department of Gastrointestinal Surgery, The Second Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Gao B, Feng C, Chai F, Wei S, Hong N, Ye Y, Wang Y, Cheng J. CT-detected extramural venous invasion-related gene signature for the overall survival prediction in patients with gastric cancer. Cancer Med 2021; 10:7816-7830. [PMID: 34510798 PMCID: PMC8559479 DOI: 10.1002/cam4.4266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/25/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background Computed tomography (CT)‐detected extramural venous invasion (EMVI) has been identified as an independent factor that can be used for risk stratification and prediction of prognosis in patients with gastric cancer (GC). Overall survival (OS) is identified as the most important prognostic indicator for GC patients. However, the molecular mechanism of EMVI development and its potential relationship with OS in GC are not fully understood. In this radiogenomics‐based study, we sought to investigate the molecular mechanism underlying CT‐detected EMVI in patients with GC, and aimed to construct a genomic signature based on EMVI‐related genes with the goal of using this signature to predict the OS. Materials and Methods Whole mRNA genome sequencing of frozen tumor samples from 13 locally advanced GC patients was performed to identify EMVI‐related genes. EMVI‐prognostic hub genes were selected based on overlapping EMVI‐related differentially expressed genes and OS‐related genes, using a training cohort of 176 GC patients who were included in The Cancer Genome Atlas database. Another 174 GC patients from this database comprised the external validation cohort. A risk stratification model using a seven‐gene signature was constructed through the use of a least absolute shrinkage and selection operator Cox regression model. Results Patients with high risk score showed significantly reduced OS (training cohort, p = 1.143e‐04; validation cohort, p = 2.429e‐02). Risk score was an independent predictor of OS in multivariate Cox regression analyses (training cohort, HR = 2.758; 95% CI: 1.825–4.169; validation cohort, HR = 2.173; 95% CI: 1.347–3.505; p < 0.001 for both). Gene functions/pathways of the seven‐gene signature mainly included cell proliferation, cell adhesion, regulation of metal ion transport, and epithelial to mesenchymal transition. Conclusions A CT‐detected EMVI‐related gene model could be used to predict the prognosis in GC patients, potentially providing clinicians with additional information regarding appropriate therapeutic strategy and medical decision‐making.
Collapse
Affiliation(s)
- Bo Gao
- Department of General Surgery, Peking University People's Hospital, Beijing, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Shengcai Wei
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
10
|
Development and evaluation of a ceMDCT-based preoperative risk stratification model to predict disease-free survival after radical surgery in patients with gastric cancer. Abdom Radiol (NY) 2021; 46:4079-4089. [PMID: 33811513 DOI: 10.1007/s00261-021-03049-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE To develop and evaluate a preoperative risk stratification model for predicting disease-free survival (DFS) based on contrast-enhanced multidetector computed tomography (ceMDCT) images in patients with gastric cancer (GC) undergoing radical surgery. METHODS We retrospectively enrolled patients with GC who underwent ceMDCT followed by radical surgery. A preoperative risk stratification model was constructed (including risk factor selection, risk status scoring, and risk level assignment) using Cox proportional hazard regression and log-rank analyses in the training cohort; the model was tested in the validation cohort. A nomogram was used to compare the preoperative risk stratification model with a postoperative DFS prediction model. RESULTS A total of 462 patients (training/validation: 271/191) were included. The ceMDCT features of T category (score of 0 or 2), N category (0, 1, 2, or 3), extramural vessel invasion (0 or 2), and tumor location (0 or 1) were selected to construct the preoperative risk stratification model, with 4 risk levels defined based on risk score. There were significant differences in DFS among the risk levels in both cohorts (p < 0.001). The predictive value of the preoperative model was similar to that of the postoperative model, with concordance indices of 0.791 (95% CI, 0.743-0.837) and 0.739 (95% CI, 0.666-0.812), respectively, in the training cohort and 0.762 (95% CI, 0.696-0.828) and 0.738 (95% CI, 0.684-0.792), respectively, in the validation cohort. CONCLUSION A preoperative risk stratification model based on ceMDCT images could be used to predict DFS and thus classify GC cases into various risk levels.
Collapse
|
11
|
Zhu Y, Zhou Y, Zhang W, Xue L, Li Y, Jiang J, Zhong Y, Wang S, Jiang L. Value of quantitative dynamic contrast-enhanced and diffusion-weighted magnetic resonance imaging in predicting extramural venous invasion in locally advanced gastric cancer and prognostic significance. Quant Imaging Med Surg 2021; 11:328-340. [PMID: 33392032 DOI: 10.21037/qims-20-246] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Extramural venous invasion (EMVI) has been found to be related to poor prognosis in gastric cancer. Preoperative diagnosis of EMVI is challenging, as it can only be detected by surgical pathology. The present study aimed to investigate the value of quantitative dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and diffusion-weighted imaging (DWI) in predicting EMVI preoperatively, and to determine the relationship between prediction results and prognosis in patients with locally advanced gastric cancer (LAGC). Methods Between January, 2015, and June, 2017, 79 LAGC patients underwent MRI preoperatively were enrolled in this study. Pathological EMVI (pEMVI) was used as the gold standard for diagnosis. The differences in quantitative DCE-MRI and DWI parameters between groups with different pEMVI status were analyzed. Multivariate logistic regression was used to build the combined prediction model for pEMVI with statistically significant quantitative parameters. The performance of the model for predicting pEMVI was evaluated using receiver operating characteristic (ROC) analysis. Patients were grouped based on MRI-predicted EMVI (mrEMVI). Kaplan-Meier analysis was used to investigate the relationship between mrEMVI and 2-year recurrence-free survival (RFS). Results Of the 79 LAGC patients who underwent MRI, 29 were pEMVI positive and 50 were pEMVI negative. Among the patients' clinical and pathological characteristics, only postoperative staging showed a significant difference between the 2 groups (P=0.015). The pEMVI-positive group had higher volume transfer constant (Ktrans) and rate constant (kep), and lower apparent diffusion coefficient (ADC) values than the negative group (0.189 vs. 0.082 min-1, 0.687 vs. 0.475 min-1, and 1.230×10-3 vs. 1.463×10-3 mm2/s, respectively; P<0.05). Quantitative parameters, Ktrans and kep, and ADC values, were independently associated with pEMVI which odds ratio values were 3.66, 2.65, and 0.30 (P<0.05), respectively, using multivariate logistic regression. ROC analysis showed that the area under the curve, sensitivity, specificity, positive predictive value, and negative predictive value in predicting pEMVI using combined Ktrans, kep, and ADC values were 0.879, 72.4%, 96%, 91.3%, and 85.7%, respectively. A total of 23 cases were considered to be mrEMVI positive, and 56 cases were considered to be mrEMVI negative, according to the predictive results. The median RFS of the mrEMVI-positive group was significant lower than the negative group (21.7 vs. 31.2 months), and the 2-year RFS rate in the mrEMVI-positive group was significantly lower than that of the negative group (43.6% vs. 72.5%, P=0.010). Conclusions The quantitative DCE-MRI parameters, Ktrans and kep, and DWI parameter, ADC, are independent predictors of pEMVI in LAGC; mrEMVI was confirmed to be a poor prognostic predictor for RFS.
Collapse
Affiliation(s)
- Yongjian Zhu
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yutao Zhou
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Li
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Jiang
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sicong Wang
- GE Healthcare, Life Sciences, Beijing, China
| | - Liming Jiang
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Yang YT, Dong SY, Zhao J, Wang WT, Zeng MS, Rao SX. CT-detected extramural venous invasion is corelated with presence of lymph node metastasis and progression-free survival in gastric cancer. Br J Radiol 2020; 93:20200673. [PMID: 33002375 DOI: 10.1259/bjr.20200673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE This study aimed to investigate if CT-detected extramural venous invasion (ctEMVI) was associated with the presence of lymph node metastasis (LNM) and survival outcomes in patients with gastric cancer. METHODS We retrospectively reviewed 105 patients with pathologically proved gastric cancer who underwent pre-operative CT examinations and received radical gastrectomy with extended lymphadenectomy. Differences in CT characteristics between the LNM-positive and -negative groups were assessed by two observers. Binary logistic regression analysis was performed to determine the risk factors of lymph node metastasis in gastric cancer. Progression-free survival analysis was performed by Kaplan-Meier method. RESULTS Two observers reached good inter-reader agreements in ctEMVI and ctN status with κ values of 0.711 and 0.751, respectively. The frequency of ctEMVI-positive status was 58.1% (61/105) in patients with gastric cancer. The LNM-positive group showed higher possibility of ctEMVI-positive status (81.7% vs 26.7%, p<0.001), larger tumor volume (mean volume, 40.77 vs 22.09 mL, p<0.001), poor tumor margin (45.0% vs 26.7% , p = 0.054) and high enhancement on arterial phase (43.3% vs 26.7%, p = 0.023) and venous phase (60.0% vs 44.4%, p = 0.048), than LNM-negative group. In multivariate analysis, ctEMVI status and tumor volume were identified as independent risk factors for lymph node metastasis with odds ratio (OR) of 9.804 (95% CI, 3.076-31.246; p<0.001) and 1.030 (95% CI, 1.001-1.060; p = 0.044). CT-detected EMVI presented better diagnostic efficiency for lymph node metastasis than CT-defined N status, with sensitivity (81.7% vs 70.0%), specificity (73.3% vs 71.1%), accuracy (78.1% vs 70.5), PPV (80.3% vs 76.4%), and NPV (75.0% vs 64.0%), respectively. Kaplan-Meier curves showed that patients with positive ctEMVI findings has lower PFS rate than patients with negative ctEMVI findings (Log-rank test, p = 0.007). CONCLUSION CT-detected EMVI was significantly associated with lymph node metastasis and progression free survival in patients with gastric cancer. Compared to CT-defined N status, ctEMVI provided superior diagnostic performance to predict pathologic Nstatus. ADVANCES IN KNOWLEDGE Our study proved that CT-detected EMVI is a promising imaging marker to predict lymph node metastasis and poor prognosis, which may contribute to the precise evaluation of gastric cancer before surgery.
Collapse
Affiliation(s)
- Yu-Tao Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - San-Yuan Dong
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Jue Zhao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Wen-Tao Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Meng-Su Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Sheng-Xiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| |
Collapse
|
13
|
Wang S, Feng C, Dong D, Li H, Zhou J, Ye Y, Liu Z, Tian J, Wang Y. Preoperative computed tomography-guided disease-free survival prediction in gastric cancer: a multicenter radiomics study. Med Phys 2020; 47:4862-4871. [PMID: 32592224 DOI: 10.1002/mp.14350] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/24/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Preoperative and noninvasive prognosis evaluation remains challenging for gastric cancer. Novel preoperative prognostic biomarkers should be investigated. This study aimed to develop multidetector-row computed tomography (MDCT)-guided prognostic models to direct follow-up strategy and improve prognosis. METHODS A retrospective dataset of 353 gastric cancer patients were enrolled from two centers and allocated to three cohorts: training cohort (n = 166), internal validation cohort (n = 83), and external validation cohort (n = 104). Quantitative radiomic features were extracted from MDCT images. The least absolute shrinkage and selection operator penalized Cox regression was adopted to construct a radiomic signature. A radiomic nomogram was established by integrating the radiomic signature and significant clinical risk factors. We also built a preoperative tumor-node-metastasis staging model for comparison. All models were evaluated considering the abilities of risk stratification, discrimination, calibration, and clinical use. RESULTS In the two validation cohorts, the established four-feature radiomic signature showed robust risk stratification power (P = 0.0260 and 0.0003, log-rank test). The radiomic nomogram incorporated radiomic signature, extramural vessel invasion, clinical T stage, and clinical N stage, outperforming all the other models (concordance index = 0.720 and 0.727) with good calibration and decision benefits. Also, the 2-yr disease-free survival (DFS) prediction was most effective (time-dependent area under curve = 0.771 and 0.765). Moreover, subgroup analysis indicated that the radiomic signature was more sensitive in risk stratifying patients with advanced clinical T/N stage. CONCLUSIONS The proposed MDCT-guided radiomic signature was verified as a prognostic factor for gastric cancer. The radiomic nomogram was a noninvasive auxiliary model for preoperative individualized DFS prediction, holding potential in promoting treatment strategy and clinical prognosis.
Collapse
Affiliation(s)
- Siwen Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Beijing, 100044, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hailin Li
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Zhou
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
14
|
Chen X, Yang Z, Yang J, Liao Y, Pang P, Fan W, Chen X. Radiomics analysis of contrast-enhanced CT predicts lymphovascular invasion and disease outcome in gastric cancer: a preliminary study. Cancer Imaging 2020; 20:24. [PMID: 32248822 PMCID: PMC7132895 DOI: 10.1186/s40644-020-00302-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND To determine whether radiomics features based on contrast-enhanced CT (CECT) can preoperatively predict lymphovascular invasion (LVI) and clinical outcome in gastric cancer (GC) patients. METHODS In total, 160 surgically resected patients were retrospectively analyzed, and seven predictive models were constructed. Three radiomics predictive models were built from radiomics features based on arterial (A), venous (V) and combination of two phase (A + V) images. Then, three Radscores (A-Radscore, V-Radscore and A + V-Radscore) were obtained. Another four predictive models were constructed by the three Radscores and clinical risk factors through multivariate logistic regression. A nomogram was developed to predict LVI by incorporating A + V-Radscore and clinical risk factors. Kaplan-Meier curve and log-rank test were utilized to analyze the outcome of LVI. RESULTS Radiomics related to tumor size and intratumoral inhomogeneity were the top-ranked LVI predicting features. The related Radscores showed significant differences according to LVI status (P < 0.01). Univariate logistic analysis identified three clinical features (T stage, N stage and AJCC stage) and three Radscores as LVI predictive factors. The Clinical-Radscore (namely, A + V + C) model that used all these factors showed a higher performance (AUC = 0.856) than the clinical (namely, C, including T stage, N stage and AJCC stage) model (AUC = 0.810) and the A + V-Radscore model (AUC = 0.795) in the train cohort. For patients without LVI and with LVI, the median progression-free survival (PFS) was 11.5 and 8.0 months (P < 0.001),and the median OS was 20.2 and 17.0 months (P = 0.3), respectively. In the Clinical-Radscore-predicted LVI absent and LVI present groups, the median PFS was 11.0 and 8.0 months (P = 0.03), and the median OS was 20.0 and 18.0 months (P = 0.05), respectively. N stage, LVI status and Clinical-Radscore-predicted LVI status were associated with disease-specific recurrence or mortality. CONCLUSIONS Radiomics features based on CECT may serve as potential markers to successfully predict LVI and PFS, but no evidence was found that these features were related to OS. Considering that it is a single central study, multi-center validation studies will be required in the future to verify its clinical feasibility.
Collapse
Affiliation(s)
- Xiaofeng Chen
- Department of Radiology, Meizhou People's Hospital, Meizhou, Guangdong, 514031, People's Republic of China
| | - Zhiqi Yang
- Department of Radiology, Meizhou People's Hospital, Meizhou, Guangdong, 514031, People's Republic of China
| | - Jiada Yang
- Department of Radiology, Meizhou People's Hospital, Meizhou, Guangdong, 514031, People's Republic of China
| | - Yuting Liao
- GE Healthcare, Guangzhou, Guangdong, People's Republic of China, 510623
| | - Peipei Pang
- GE Healthcare, Hangzhou, Zhejiang, People's Republic of China, 311100
| | - Weixiong Fan
- Department of Radiology, Meizhou People's Hospital, Meizhou, Guangdong, 514031, People's Republic of China
| | - Xiangguang Chen
- Department of Radiology, Meizhou People's Hospital, Meizhou, Guangdong, 514031, People's Republic of China.
| |
Collapse
|
15
|
CT-Detected Extramural Vessel Invasion and Regional Lymph Node Involvement in Stage T4a Gastric Cancer for Predicting Progression-Free Survival. AJR Am J Roentgenol 2019; 212:1030-1036. [PMID: 30779670 DOI: 10.2214/ajr.18.20342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE. This study aimed to investigate the 3-year progression-free survival (PFS) of patients with stage T4a gastric cancer with extramural vessel invasion (EMVI) detected with contrast-enhanced (CE) MDCT. In addition, we investigated the possibility that CT of EMVI could improve clinical nodal (N) staging. MATERIALS AND METHODS. This retrospective study included 143 patients with T4a gastric cancer. Clinical staging was performed with CE-MDCT. All patients underwent radical gastrectomy with extended lymphadenectomy, adjuvant chemotherapy, and conventional follow-up visits. Potential prognostic factors, including CE-MDCT-detected N status, pathologic N status, EMVI detected at CT, tumor location or growth pattern, histologic type or tumor differentiation, and tumor size, were recorded. Survival estimates for PFS were obtained using the Kaplan-Meier product limit for the following patient subgroups: EMVI positive-N positive, EMVI positive-N negative, EMVI negative-N positive, and EMVI negative-N negative. Hazard ratios for 3-year PFS were generated using a Cox proportional hazard regression analysis. RESULTS. The frequency of EMVI detected at CT was 55.9% (80/143). The 3-year PFS rates were 25.0% for the EMVI positive-N positive group, 53.1% for the EMVI positive-N negative group, 75.6% for the EMVI negative-N positive group, and 64.7% for the EMVI negative-N negative group. The EMVI positive-N positive subgroup 3-year PFS rate was significantly lower than that of the other three groups (p < 0.05, log-rank test). Using Cox proportional hazards regression analysis, EMVI positive-N positive status was found to be an independent factor for reduced 3-year PFS, with a hazard ratio of 2.169 (95% CI, 1.300-3.618; p = 0.003). CONCLUSION. EMVI detected at CT, combined with N status detected with CE-MDCT, could be used as a valuable preoperative prognostic factor in patients with T4a gastric cancer.
Collapse
|
16
|
Yedururi S, Kang H, Cox VL, Chawla S, Le O, Loyer EM, Marcal L. Tumor thrombus in the venous drainage pathways of primary, recurrent and metastatic disease on routine oncologic imaging studies: beyond hepatocellular and renal cell carcinomas. Br J Radiol 2019; 92:20180478. [PMID: 30844299 DOI: 10.1259/bjr.20180478] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Radiologists routinely evaluate for tumor thrombus in the portal and hepatic veins in patients with hepatocellular carcinoma and in the renal vein and inferior vena cava in patients with renal cell carcinoma. However, tumor thrombus occurs in association with numerous other tumor types, e.g. colorectal carcinoma and pancreatic neuroendocrine tumor. Furthermore tumor thrombi are not limited to the primary tumor but also seen with local recurrence and metastatic disease. While less recognized, these thrombi nevertheless affect patterns of recurrence and prognosis. Their detection is critical for accurate local staging and early detection of local recurrence and metastatic disease. The purpose of this pictorial review is to draw the attention of radiologists to the less familiar manifestations of tumor thrombus, review the imaging findings and illustrate the clinical significance of these thrombi.
Collapse
Affiliation(s)
- Sireesha Yedururi
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| | - HyunSeon Kang
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| | - Veronica L Cox
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| | - Sumedha Chawla
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| | - Ott Le
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| | - Evelyne M Loyer
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| | - Leonardo Marcal
- 1 Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center Houston , Texas , USA
| |
Collapse
|
17
|
Yedururi S, Chawla S, Amini B, Wei W, Salem UI, Morani AC, Wang WL, Gorlick R, Lewis VO, Daw NC. Tumor thrombus in the large veins draining primary pelvic osteosarcoma on cross sectional imaging. Eur J Radiol 2018; 105:49-55. [PMID: 30017298 PMCID: PMC6056011 DOI: 10.1016/j.ejrad.2018.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/07/2018] [Accepted: 05/20/2018] [Indexed: 11/23/2022]
Abstract
PURPOSE To evaluate the frequency of tumor thrombus in the large veins draining primary pelvic osteosarcoma on early cross-sectional imaging studies and its effect on patient survival. MATERIALS AND METHODS Our retrospective study included all patients with primary pelvic osteosarcoma treated at our facility between January 2000 and May 2014, who were ≤ 45 years of age, and had adequate imaging studies and clinical follow up. Four radiologists evaluated for tumor in the large draining veins on initial CT, MRI and PET/CTs. A consensus evaluation by the four radiologists together with findings on operative reports, pathology reports or follow-up imaging was used as the reference standard. RESULTS Thirty-nine patients with primary pelvic osteosarcoma met final inclusion criteria. Tumor thrombus was identified in the large draining veins in 10 of the 22 (45%) patients who underwent tumor resection and 10 of the 17 (59%) who did not. In the 22 patients who underwent tumor resection, tumor thrombus was significantly associated with worse overall survival (p = 0.03). CONCLUSIONS Tumor thrombus in the large draining veins is identified in a significant proportion of initial imaging studies in patients with pelvic osteosarcoma, and is associated with worse overall survival in patients who undergo tumor resection.
Collapse
Affiliation(s)
- Sireesha Yedururi
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1473, Houston, TX, 77030, United States.
| | - Sumedha Chawla
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1473, Houston, TX, 77030, United States.
| | - Behrang Amini
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1473, Houston, TX, 77030, United States.
| | - Wei Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1411, Houston, TX, 77030, United States.
| | - Usama I Salem
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1473, Houston, TX, 77030, United States.
| | - Ajaykumar C Morani
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1473, Houston, TX, 77030, United States.
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 0085, Houston, TX, 77030, United States.
| | - Richard Gorlick
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 0087, Houston, TX, 77030, United States.
| | - Valerae O Lewis
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1448, Houston, TX, 77030, United States; Pelvic Sarcoma Center of Excellence, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 1448, Houston, TX, 77030, United States.
| | - Najat C Daw
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Street, Unit 0087, Houston, TX, 77030, United States.
| |
Collapse
|