1
|
Li J, Yang C, Zhang Y, Hong X, Jiang M, Zhu Z, Li J. Deciphering aging-associated prognosis and heterogeneity in gastric cancer through a machine learning-driven approach. iScience 2025; 28:112316. [PMID: 40256325 PMCID: PMC12008712 DOI: 10.1016/j.isci.2025.112316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with a high mortality rate and limited treatment options. Aging significantly contributes to tumor progression, and GC was confirmed as an aging-related heterogeneous disease. This study established an aging-associated index (AAI) using a machine learning-derived gene panel to stratify GC patients. High AAI scores associated with poor prognosis and indicated potential benefits from adjuvant chemotherapy, while showing resistance to immunotherapy. Single-cell transcriptome analysis revealed that AAI was enriched in monocyte cells within the tumor microenvironment. Two distinct molecular subtypes of GC were identified through unsupervised clustering, leading to the development of a subtype-specific regulatory network highlighting SOX7 and ELK3 as potential therapeutic targets. Drug sensitivity analyses indicated that patients with high ELK3 expression may respond to FDA-approved drugs (axitinib, dacarbazine, crizotinib, and vincristine). Finally, a user-friendly Shiny application was created to facilitate access to the prognostic model and molecular subtype classifier for GC.
Collapse
Affiliation(s)
- Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chuanlai Yang
- Department of Science and Technology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Yunxiao Zhang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mingye Jiang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhongxu Zhu
- Biomics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Guangdong, Shenzhen, China
| |
Collapse
|
2
|
Simbulan-Rosenthal CM, Islam N, Haribabu Y, Alobaidi R, Shalamzari A, Graham G, Kuo LW, Sykora P, Rosenthal DS. CD133 Stimulates Cell Proliferation via the Upregulation of Amphiregulin in Melanoma. Cells 2024; 13:777. [PMID: 38727313 PMCID: PMC11083289 DOI: 10.3390/cells13090777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
CD133, a cancer stem cell (CSC) marker in tumors, including melanoma, is associated with tumor recurrence, chemoresistance, and metastasis. Patient-derived melanoma cell lines were transduced with a Tet-on vector expressing CD133, generating doxycycline (Dox)-inducible cell lines. Cells were exposed to Dox for 24 h to induce CD133 expression, followed by RNA-seq and bioinformatic analyses, revealing genes and pathways that are significantly up- or downregulated by CD133. The most significantly upregulated gene after CD133 was amphiregulin (AREG), validated by qRT-PCR and immunoblot analyses. Induced CD133 expression significantly increased cell growth, percentage of cells in S-phase, BrdU incorporation into nascent DNA, and PCNA levels, indicating that CD133 stimulates cell proliferation. CD133 induction also activated EGFR and the MAPK pathway. Potential mechanisms highlighting the role(s) of CD133 and AREG in melanoma CSC were further delineated using AREG/EGFR inhibitors or siRNA knockdown of AREG mRNA. Treatment with the EGFR inhibitor gefitinib blocked CD133-induced cell growth increase and MAPK pathway activation. Importantly, siRNA knockdown of AREG reversed the stimulatory effects of CD133 on cell growth, indicating that AREG mediates the effects of CD133 on cell proliferation, thus serving as an attractive target for novel combinatorial therapeutics in melanoma and cancers with overexpression of both CD133 and AREG.
Collapse
Affiliation(s)
- Cynthia M Simbulan-Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Nusrat Islam
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Yogameenakshi Haribabu
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Ryyan Alobaidi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Azadeh Shalamzari
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Garrett Graham
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Li-Wei Kuo
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| | - Peter Sykora
- Amelia Technologies, LLC., Washington, DC 20001, USA;
| | - Dean S Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (N.I.); (Y.H.); (R.A.); (A.S.); (G.G.); (L.-W.K.)
| |
Collapse
|
3
|
Identification of a Biomarker Combination for Survival Stratification in pStage II/III Gastric Cancer after Curative Resection. Cancers (Basel) 2022; 14:cancers14184427. [PMID: 36139587 PMCID: PMC9497152 DOI: 10.3390/cancers14184427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Gastric cancer (GC) is the fifth most common cancer worldwide and the fourth most common cause of cancer-related deaths, with a high frequency of recurrence and metastasis, and a poor prognosis. This study presents a novel combination of four proteins (PDGFRB, INHBA, MMP11, and galectin-10) in GC tissues that have been identified as useful survival stratification markers in patients with pStage II/III GC after curative resection by quantitative polymerase chain reaction (qPCR), proteomic analysis, and immunohistochemistry (IHC). Abstract Background: We sought to identify an optimal combination of survival risk stratification markers in patients with pathological (p) stage II/III gastric cancer (GC) after curative resection. Methods: We measured the expression levels of 127 genes in pStage II/III GC tissues of two patient cohorts by quantitative polymerase chain reaction (qPCR) and the expression of 1756 proteins between two prognosis (good and poor) groups by proteomic analysis to identify candidate survival stratification markers. Further, immunohistochemistry (IHC) using tumor microarrays (TMAs) in another cohort of patients was performed to identify an optimal biomarker combination for survival stratification in GC patients. Results: secreted protein acidic and rich in cysteine (SPARC), erb-b2 receptor tyrosine kinase 2 (ERBB2), inhibin subunit beta A (INHBA), matrix metallopeptidase-11 (MMP11), tumor protein p53 (TP53), and platelet-derived growth factor receptor-beta (PDGFRB) were identified as candidate biomarkers from qPCR analysis, and SPARC and galectin-10 were obtained from the proteomic analysis. The combination of PDGFRB, INHBA, MMP11, and galectin-10 was identified as the optimal combination of survival risk stratification markers. Conclusions: A combination of four proteins in GC tissues may serve as useful survival risk stratification markers in patients with pStage II/III GC following curative resection. Our results may facilitate future multicenter prospective clinical trials.
Collapse
|
4
|
Gastric cancer biomarker analysis in patients treated with different adjuvant chemotherapy regimens within SAMIT, a phase III randomized controlled trial. Sci Rep 2022; 12:8509. [PMID: 35595817 PMCID: PMC9123164 DOI: 10.1038/s41598-022-12439-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 03/23/2022] [Indexed: 12/23/2022] Open
Abstract
Biomarkers for selecting gastric cancer (GC) patients likely to benefit from sequential paclitaxel treatment followed by fluorinated-pyrimidine-based adjuvant chemotherapy (sequential paclitaxel) were investigated using tissue samples of patients recruited into SAMIT, a phase III randomized controlled trial. Total RNA was extracted from 556 GC resection samples. The expression of 105 genes was quantified using real-time PCR. Genes predicting the benefit of sequential paclitaxel on overall survival, disease-free survival, and cumulative incidence of relapse were identified based on the ranking of p-values associated with the interaction between the biomarker and sequential paclitaxel or monotherapy groups. Low VSNL1 and CD44 expression predicted the benefit of sequential paclitaxel treatment for all three endpoints. Patients with combined low expression of both genes benefitted most from sequential paclitaxel therapy (hazard ratio = 0.48 [95% confidence interval, 0.30-0.78]; p < 0.01; interaction p-value < 0.01). This is the first study to identify VSNL1 and CD44 RNA expression levels as biomarkers for selecting GC patients that are likely to benefit from sequential paclitaxel treatment followed by fluorinated-pyrimidine-based adjuvant chemotherapy. Our findings may facilitate clinical trials on biomarker-oriented postoperative adjuvant chemotherapy for patients with locally advanced GC.
Collapse
|
5
|
Sung JY, Cheong JH. The Matrisome Is Associated with Metabolic Reprograming in Stem-like Phenotypes of Gastric Cancer. Cancers (Basel) 2022; 14:cancers14061438. [PMID: 35326589 PMCID: PMC8945874 DOI: 10.3390/cancers14061438] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Our results suggested a correlation between the metabolic reprogramming associated with the high-matrisome group and stem-like phenotype in gastric cancer. Carbohydrate sulfotransferase 7 was found to be associated with the signaling transduction of overexpressed oncogenes and tumor suppressor genes in the high-matrisome group. The high expression of glycosaminoglycan biosynthesis-chondroitin sulfate metabolic pathway genes was associated with poor prognosis. Abstract The extracellular matrix (ECM) is an important regulator of all cellular functions, and the matrisome represents a major component of the tumor microenvironment. The matrisome is an essential component comprising genes encoding ECM glycoproteins, collagens, and proteoglycans; however, its role in cancer progression and the development of stem-like molecular subtypes in gastric cancer is unknown. We analyzed gastric cancer data from five molecular subtypes (n = 497) and found that metabolic reprograming differs based on the state of the matrisome. Approximately 95% of stem-like cancer type samples of gastric cancer were in the high-matrisome category, and energy metabolism was considerably increased in the high-matrisome group. Particularly, high glycosaminoglycan biosynthesis-chondroitin sulfate metabolic reprograming was associated with an unfavorable prognosis. Glycosaminoglycan biosynthesis-chondroitin sulfate metabolic reprograming may occur according to the matrisome status and contribute to the development of stem-like phenotypes. Our analysis suggests the possibility of precision medicine for anticancer therapies.
Collapse
Affiliation(s)
- Ji-Yong Sung
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: (J.-Y.S.); (J.-H.C.)
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: (J.-Y.S.); (J.-H.C.)
| |
Collapse
|
6
|
Tazawa H, Suzuki T, Saito A, Ishikawa A, Komo T, Sada H, Shimada N, Hadano N, Onoe T, Sudo T, Shimizu Y, Kuraoka K, Tashiro H. Utility of TMPRSS4 as a Prognostic Biomarker and Potential Therapeutic Target in Patients with Gastric Cancer. J Gastrointest Surg 2022; 26:305-313. [PMID: 34379296 PMCID: PMC8821072 DOI: 10.1007/s11605-021-05101-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/14/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND Transmembrane serine protease 4 (TMPRSS4) belongs to the family of type II transmembrane serine proteases that are known to be upregulated in many malignant tumors. However, there is a paucity of studies documenting the clinical impact and biological effects of TMPRSS4 on gastric cancer (GC) patients who underwent surgery. METHODS Tissues samples were obtained from 105 patients with GC who underwent gastrectomy followed by adjuvant chemotherapy, excluding those at stage I. The expression of TMPRSS4 was examined through immunohistochemical analysis. The association between TMPRSS4 expression and clinico-pathological features as well as prognosis was assessed. Moreover, the effects of TMPRSS4 expression on cell migration and sensitivity to 5-FU were investigated. RESULTS The expression rate of TMPRSS4 was 56.3% (59/105) in GC cases. The expression of TMPRSS4 was positively correlated with the depth of tumor (T) and venous (V) invasion. The 5-year overall survival (OS) and recurrence-free survival (RFS) rates of the TMPRSS4-positive group was significantly lower than that of the TMPRSS4-negative group (p=0.0001 and p=0.005, respectively). Especially, there was significant differences in OS and RFS of patients with stage III cancer between the two groups (p=0.0064 and 0.012, respectively). Multivariate analysis demonstrated that TMPRSS4 expression and the stage of cancer were crucial prognostic factors for RFS. TMPRSS4-silenced GC cells exhibited increased sensitivity to 5-FU when compared with the non-specific control siRNA-transfected cells. CONCLUSION TMPRSS4 can be considered as a potential prognostic biomarker, especially for stage III, and a promising therapeutic target for GC.
Collapse
Affiliation(s)
- Hirofumi Tazawa
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan.
| | - Takahisa Suzuki
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akihisa Saito
- Department of Diagnostic Pathology, Kure Medical Center・Chugoku Cancer Center, Kure, Hiroshima, Japan
| | - Akira Ishikawa
- Department of Diagnostic Pathology, Kure Medical Center・Chugoku Cancer Center, Kure, Hiroshima, Japan
| | - Toshiaki Komo
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Haruki Sada
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Norimitsu Shimada
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Naoto Hadano
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Takashi Onoe
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Takeshi Sudo
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Yosuke Shimizu
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan
| | - Kazuya Kuraoka
- Department of Diagnostic Pathology, Kure Medical Center・Chugoku Cancer Center, Kure, Hiroshima, Japan
| | - Hirotaka Tashiro
- Department of Surgery, Kure Medical Center・Chugoku Cancer Center, 737-0023, 3-1, Kure, Hiroshima, Japan.
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
7
|
Bolitho C, Moscova M, Baxter RC, Marsh DJ. Amphiregulin increases migration and proliferation of epithelial ovarian cancer cells by inducing its own expression via PI3-kinase signaling. Mol Cell Endocrinol 2021; 533:111338. [PMID: 34062166 DOI: 10.1016/j.mce.2021.111338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/09/2022]
Abstract
The epidermal growth factor receptor (EGFR) is overexpressed in many types of cancer, including epithelial ovarian cancer (EOC), and its expression has been found to correlate with advanced stage and poor prognosis. The EGFR ligand amphiregulin (AREG) has been investigated as a target for human cancer therapy and is known to have an autocrine role in many cancers. A cytokine array identified AREG as one of several cytokines upregulated by EGF in a phosphatidylinositol 3-kinase (PI3-K) dependent manner in EOC cells. To investigate the functional role of AREG in EOC, its effect on cellular migration and proliferation was assessed in two EOC cells lines, OV167 and SKOV3. AREG increased both migration and proliferation of EOC cell line models through activation of PI3-K signaling, but independent of mitogen activated protein kinase (MAPK) signaling. Through an AREG autocrine loop mediated via PI3-K, upregulation of AREG led to increased levels of both AREG transcript and secreted AREG, while downregulation of endogenous AREG decreased the ability of exogenous AREG to induce cell migration and proliferation. Further, inhibition of endogenous AREG activity or metalloproteinase activity decreased EGF-induced EOC migration and proliferation, indicating a role for soluble endogenous AREG in mediating the functional effects of EGFR in inducing migration and proliferation in EOC.
Collapse
Affiliation(s)
- Christine Bolitho
- University of Sydney, Kolling Institute, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Michelle Moscova
- University of Sydney, Kolling Institute, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia; School of Medical Sciences, University of New South Wales, Sydney, Kensington, NSW, 2052, Australia
| | - Robert C Baxter
- University of Sydney, Kolling Institute, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Deborah J Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology, Sydney, Ultimo, NSW, 2007, Australia; Northern Clinical School, Kolling Institute, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| |
Collapse
|
8
|
Smith TAD, AbdelKarem OA, Irlam-Jones JJ, Lane B, Valentine H, Bibby BAS, Denley H, Choudhury A, West CML. Selection of endogenous control genes for normalising gene expression data derived from formalin-fixed paraffin-embedded tumour tissue. Sci Rep 2020; 10:17258. [PMID: 33057113 PMCID: PMC7560892 DOI: 10.1038/s41598-020-74380-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022] Open
Abstract
Quantitative real time polymerase chain reaction (qPCR) data are normalised using endogenous control genes. We aimed to: (1) demonstrate a pathway to identify endogenous control genes for qPCR analysis of formalin-fixed paraffin-embedded (FFPE) tissue using bladder cancer as an exemplar; and (2) examine the influence of probe length and sample age on PCR amplification and co-expression of candidate genes on apparent expression stability. RNA was extracted from prospective and retrospective samples and subject to qPCR using TaqMan human endogenous control arrays or single tube assays. Gene stability ranking was assessed using coefficient of variation (CoV), GeNorm and NormFinder. Co-expressed genes were identified from The Cancer Genome Atlas (TCGA) using the on-line gene regression analysis tool GRACE. Cycle threshold (Ct) values were lower for prospective (19.49 ± 2.53) vs retrospective (23.8 ± 3.32) tissues (p < 0.001) and shorter vs longer probes. Co-expressed genes ranked as the most stable genes in the TCGA cohort by GeNorm when analysed together but ranked lower when analysed individually omitting co-expressed genes indicating bias. Stability values were < 1.5 for the 20 candidate genes in the prospective cohort. As they consistently ranked in the top ten by CoV, GeNorm and Normfinder, UBC, RPLP0, HMBS, GUSB, and TBP are the most suitable endogenous control genes for bladder cancer qPCR.
Collapse
Affiliation(s)
- Tim A D Smith
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK.
| | - Omneya A AbdelKarem
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
- Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, El-Hadra, Alexandria, Egypt
| | - Joely J Irlam-Jones
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
| | - Brian Lane
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
| | - Helen Valentine
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
| | - Becky A S Bibby
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
| | - Helen Denley
- Pathology Centre, Shrewsbury and Telford NHS Trust, Royal Shrewsbury Hospital, Shrewsbury, SY3 8XQ, UK
| | - Ananya Choudhury
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
| | - Catharine M L West
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Academic Health Centre, Christie Hospital NHS Found Trust, Manchester, M20 4BX, UK
| |
Collapse
|
9
|
Yang Z, Sun Y, Liu R, Shi Y, Ding S. Plasma long noncoding RNAs PANDAR, FOXD2-AS1, and SMARCC2 as potential novel diagnostic biomarkers for gastric cancer. Cancer Manag Res 2019; 11:6175-6184. [PMID: 31308753 PMCID: PMC6613614 DOI: 10.2147/cmar.s201935] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/11/2019] [Indexed: 01/02/2023] Open
Abstract
Background Gastric cancer is still a common cancer worldwide. Investigation of potential plasma biomarkers for gastric cancer diagnosis is essential for prevention strategies and early intervention for gastric cancer-control planning. Objectives This study was aimed to explore the lncRNAs' promoter of CDKN1A antisense DNA-damage-activated RNA (PANDAR), FOXD2-AS1, and SMARCC2 as potential novel diagnostic biomarkers for gastric cancer. Method 109 gastric cancer patients and 106 healthy controls were involved in this study. Plasma lncRNAs PANDAR, FOXD2-AS1, and SMARCC2 were detected by real-time PCR. Student's t-test, Mann-Whitney U test, and Chi-square test were used to verify the differences of clinical variables between two groups. Receiver operating characteristic curve (ROC) was used to evaluate the diagnostic value of every biomarker. Multivariable analysis of risk factors for gastric cancer was performed using logistic regression analysis. Results There were significant differences in age, gender, carcinoembryonic antigen (CEA), carbohydrate antigen (CA) 153 between gastric cancer and healthy controls (P<0.05). Compared with healthy subjects, the levels of plasma lncRNAs PANDAR, FOXD2-AS1, and SMARCC2 were all significantly higher in gastric cancer patients (P<0.05). These lncRNAs were significantly associated with clinicopathological parameters of gastric cancer, like pathological differentiation, TNM stage, and/or lymph nodes metastasis, and/or invasion depth (P<0.05). The AUC for lncRNA PANDAR was 0.767, for FOXD2-AS1 was 0.700, for SMARCC2 was 0.748, and the AUC of the combinative diagnostic value of these three lncRNAs was 0.839. Adjusted by other variables, these lncRNAs' expressions were significantly associated with gastric cancer. Conclusions Plasma lncRNAs PANDAR, FOXD2-AS1, and SMARCC2 might be appropriate diagnostic biomarkers for gastric cancer.
Collapse
Affiliation(s)
- Ziwei Yang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yanfei Sun
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Rongfeng Liu
- Department of Clinical Laboratory Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, People's Republic of China
| |
Collapse
|
10
|
van den Ende T, Ter Veer E, Mali RMA, van Berge Henegouwen MI, Hulshof MCCM, van Oijen MGH, van Laarhoven HWM. Prognostic and Predictive Factors for the Curative Treatment of Esophageal and Gastric Cancer in Randomized Controlled Trials: A Systematic Review and Meta-Analysis. Cancers (Basel) 2019; 11:E530. [PMID: 31013858 PMCID: PMC6521055 DOI: 10.3390/cancers11040530] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/05/2019] [Accepted: 04/09/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND An overview of promising prognostic variables and predictive subgroups concerning the curative treatment of esophageal and gastric cancer from randomized controlled trials (RCTs) is lacking. Therefore, we conducted a systematic review and meta-analysis. METHODS PubMed, EMBASE, CENTRAL, and ASCO/ESMO conferences were searched up to March 2019 for RCTs on the curative treatment of esophageal or gastric cancer with data on prognostic and/or predictive factors for overall survival. Prognostic factors were deemed potentially clinically relevant according to the following criteria; (1) statistically significant (p < 0.05) in a multivariate analysis, (2) reported in at least 250 patients, and (3) p < 0.05, in ≥ 33% of the total number of patients in RCTs reporting this factor. Predictive factors were potentially clinically-relevant if (1) the p-value for interaction between subgroups was <0.20 and (2) the hazard ratio in one of the subgroups was significant (p < 0.05). RESULTS For gastric cancer, 39 RCTs were identified (n = 13,530 patients) and, for esophageal cancer, 33 RCTs were identified (n = 8618 patients). In total, we identified 23 potentially clinically relevant prognostic factors for gastric cancer and 16 for esophageal cancer. There were 15 potentially clinically relevant predictive factors for gastric cancer and 10 for esophageal cancer. CONCLUSION The identified prognostic and predictive factors can be included and analyzed in future RCTs and be of guidance for nomograms. Further validation should be performed in large patient cohorts.
Collapse
Affiliation(s)
- Tom van den Ende
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, (UMC) location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Emil Ter Veer
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, (UMC) location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Rosa M A Mali
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, (UMC) location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Mark I van Berge Henegouwen
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, (UMC) location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Maarten C C M Hulshof
- Department of Radiotherapy, Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC), location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, (UMC) location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, (UMC) location AMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Narimatsu T, Kambara T, Abe H, Uematsu T, Tokura Y, Suzuki I, Sakamoto K, Takei K, Nishihara D, Nakamura G, Kokubun H, Yuki H, Betsunoh H, Kamai T. 5-Fluorouracil-based adjuvant chemotherapy improves the clinical outcomes of patients with lymphovascular invasion of upper urinary tract cancer and low expression of dihydropyrimidine dehydrogenase. Oncol Lett 2019; 17:4429-4436. [PMID: 30944635 PMCID: PMC6444440 DOI: 10.3892/ol.2019.10086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/19/2019] [Indexed: 12/04/2022] Open
Abstract
Lymphovascular invasion (LVI) by urothelial carcinoma of the upper urinary tract (UC-UUT) is associated with an unfavorable prognosis. However, a high proportion of patients with UC-UUT are unable to receive the recommended doses of cisplatin-based adjuvant chemotherapy due to advanced age or renal dysfunction resulting from nephroureterectomy. Tegafur-uracil is an oral form of 5-fluorouracil whose efficacy is influenced by the activities of enzymes associated with its metabolism, such as dihydropyrimidine dehydrogenase (DPD), orotatephosphoribosyltransferase (OPRT) and thymidylate synthase (TS). The aim of the present study was to investigate the efficacy of adjuvant 5-fluorouracil chemotherapy for UC-UUT with LVI, and to assess the expression of enzymes associated with 5-fluorouracil metabolism as promising biomarkers of therapy efficacy. The present study retrospectively investigated 52 cases of UC-UUT. Following nephroureterectomy, tegafur-uracil was administered to 15 out of 30 patients with LVI who were not eligible for cisplatin-based adjuvant chemotherapy. Levels of DPD, OPRT and TS expression in tumor specimens were determined by reverse transcription-quantitative polymerase chain reaction, and their associations with the efficacy of adjuvant 5-fluorouracil chemotherapy were analyzed. The levels of DPD, OPRT and TS expression were not associated with pathological factors or outcome, although a higher expression of TS was associated with a poorer outcome. Adjuvant 5-fluorouracil chemotherapy significantly improved the outcome of patients with lower DPD expression. However, the levels of OPRT and TS expression did not influence therapeutic efficacy. Adjuvant 5-fluorouracil chemotherapy appears to be effective for lymphovascular-invasive UC-UUT in patients with lower DPD expression.
Collapse
Affiliation(s)
- Takahiro Narimatsu
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Tsunehito Kambara
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Hideyuki Abe
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Toshitaka Uematsu
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Yuumi Tokura
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Issei Suzuki
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Kazumasa Sakamoto
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Kouhei Takei
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Daisaku Nishihara
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Gaku Nakamura
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Hidetoshi Kokubun
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Hideo Yuki
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Hironori Betsunoh
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Takao Kamai
- Department of Urology, Dokkyo Medical University, Tochigi 321-0293, Japan
| |
Collapse
|
12
|
Harada H, Hosoda K, Moriya H, Mieno H, Ema A, Ushiku H, Washio M, Nishizawa N, Ishii S, Yokota K, Tanaka Y, Kaida T, Soeno T, Kosaka Y, Watanabe M, Yamashita K. Cancer-specific promoter DNA methylation of Cysteine dioxygenase type 1 (CDO1) gene as an important prognostic biomarker of gastric cancer. PLoS One 2019; 14:e0214872. [PMID: 30934021 PMCID: PMC6443169 DOI: 10.1371/journal.pone.0214872] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND There have been few available prognostic biomarkers in gastric cancer. We rigorously assessed the clinical relevance of promoter DNA methylation of Cysteine dioxygenase type 1 (CDO1) gene, a cancer-specific aberration, in human gastric cancer. METHODS Quantitative CDO1 methylation value (TaqMeth V) was initially calculated in 138 gastric cancer patients operated in 2005, and its clinical significance was elucidated. As a subsequent expanded set, 154 gastric cancer patients with pathological stage (pStage) II / III with no postoperative therapy were validated between 2000 and 2010. RESULTS (1) Median TaqMeth V of CDO1 gene methylation of gastric cancer was 25.6, ranging from 0 to 120.9. As pStage progressed, CDO1 TaqMeth V became higher (p < 0.0001). (2) The optimal cut-off value was determined to be 32.6; gastric cancer patients with high CDO1 gene methylation showed a significantly worse prognosis than those with low CDO1 gene methylation (p < 0.0001). (3) A multivariate cox proportional hazards model identified high CDO1 gene methylation (p = 0.033) as an independent prognostic factor. (4) The results were recapitulated in the expanded set in pStage III, where high CDO1 gene methylation group had a significantly worse prognosis than low CDO1 gene methylation group (p = 0.0065). Hematogenous metastasis was unique in pStage III with high CDO1 gene methylation (p = 0.0075). (5) Anchorage independent growth was reduced in several gastric cancer cell lines due to forced expression of the CDO1 gene, suggesting that abnormal CDO1 gene expression may represent distant metastatic ability. CONCLUSIONS Promoter DNA hypermethylation of CDO1 gene was rigorously validated as an important prognostic biomarker in primary gastric cancer with specific stage.
Collapse
Affiliation(s)
- Hiroki Harada
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Kei Hosoda
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Hiromitsu Moriya
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Hiroaki Mieno
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Akira Ema
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Hideki Ushiku
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Marie Washio
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Nobuyuki Nishizawa
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Satoru Ishii
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Kazuko Yokota
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Yoko Tanaka
- Department of Breast and Endocrine Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Takeshi Kaida
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Takafumi Soeno
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Yoshimasa Kosaka
- Department of Breast and Endocrine Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Masahiko Watanabe
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Keishi Yamashita
- Department of Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
- Division of Advanced Surgical Oncology, Department of Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Minami-ku, Sagamihara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
13
|
Zhao W, Ding G, Wen J, Tang Q, Yong H, Zhu H, Zhang S, Qiu Z, Feng Z, Zhu J. Correlation between Trop2 and amphiregulin coexpression and overall survival in gastric cancer. Cancer Med 2017; 6:994-1001. [PMID: 28256068 PMCID: PMC5430091 DOI: 10.1002/cam4.1018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/14/2016] [Accepted: 12/27/2016] [Indexed: 12/28/2022] Open
Abstract
Gastric cancer (GC) is a multistep and multistage disease and the majority of GC cells could overexpressed one or more oncogenes. Trop2 and amphiregulin (AREG) are both overexpressed in various epithelial cell cancers and have the role in the increases tumor cells division and metastasis. However, little is known about the function and correlation of two oncogenes coexpressed in GC. The expression level of these two genes in 791 cases of GC tissues were tested, the correlations between two genes expression and clinical pathological characteristics and overall survival in GC patients through immunohistochemistry (IHC) were analyzed. This study also explored the mRNA expression level of two genes in 26 cases of freshly GC tissues by qRT‐PCR. The results indicated that Trop2+/AREG+ coexpression was higher in GC tissues than in adjacent tissues. Trop2+/AREG+ protein coexpression were associated with Tumor Node Metastasis (TNM) stage (χ2 = 50.345, P < 0.001), tumor size (χ2 = 40.349, P < 0.001), lymph node metastases (χ2 = 26.481, P < 0.001), and distant metastases (χ2 = 8.387, P = 0.039). GC patients with Trop2+ and AREG+ protein coexpression had poor overall survival rates (HR = 3.682, 95% CI = 2.038–6.654, P < 0.001). The expression level of Trop2/AREG were positively correlated (r 0.254 and P < 0.001). The result of the mRNA expression was similar to that of the protein expression. Overall, Trop2 and AREG could be seen as prognostic cobiomarker in GC and combined detection of Trop2 and AREG could be viewed as helpful in predicting the prognosis of the GC patients.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China.,School of Public Health, Nantong University, Nantong, 226019, China
| | - Guipeng Ding
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Jinbo Wen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Qi Tang
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, 210029, China
| | - Hongmei Yong
- Department of Oncology, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second People's Hospital, Huai'an, 223001, China
| | - Huijun Zhu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, 226019, China
| | - Shu Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, 226019, China
| | - Zhenning Qiu
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, 210029, China
| | - Zhenqing Feng
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China.,Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China.,Key Laboratory of Cancer Biomarkers, Prevent and Treatment, Cancer Center, Nanjing Medical University, Nanjing, 210029, China
| | - Jin Zhu
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, 210029, China.,Huadong Medical Institute of Biotechniques, Nanjing, 210029, China
| |
Collapse
|
14
|
Wang B, Yong H, Zhu H, Ni D, Tang S, Zhang S, Wang W, Zhou Y, Zhao W, Ding G, Zhu J, Li X, Feng Z. Abnormal amphiregulin expression correlates with gastric cancer prognosis. Oncotarget 2016; 7:76684-76692. [PMID: 27713123 PMCID: PMC5363540 DOI: 10.18632/oncotarget.12436] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/27/2016] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is a global health issue with a high mortality rate. Early diagnosis and tracking of GC is a challenge due to a lack of reliable tools. Amphiregulin (AREG) is a member of the epidermal growth factor (EGF) family that activates growth signaling upon binding of EGF receptors. Elevated AREG expression is associated with various pathological conditions, including cancer. Here, we investigated whether increased AREG expression is a disease indicator and/or prognostic biomarker for GC. We used tissue microarray and quantitative real-time polymerase chain reaction to assess AREG expression in clinical tissue specimens at various stages of GC and a conducted bioinformatics analysis to evaluate the value of AREG over-expression as a GC biomarker. We found that both mRNA and protein expression of AREG were increased in the tissues of GC patients when compared to tissues from non-cancer patients or normal tissues. High expression of AREG was also associated with GC clinicopathological characteristics and poor survival. Thus, over-expression of AREG could serve as a novel GC biomarker, and active surveillance of its expression could be a novel approach to GC diagnosis and monitoring.
Collapse
Affiliation(s)
- Bing Wang
- Center for Pathology and Laboratory Medicine, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
- Department of Oncology, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
| | - Hongmei Yong
- Department of Oncology, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second People's Hospital, Huai'an, Jiangsu, China
| | - Huijun Zhu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Daguang Ni
- Center for Pathology and Laboratory Medicine, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
| | - Sijie Tang
- Center for Pathology and Laboratory Medicine, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
| | - Shu Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wei Wang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yan Zhou
- Department of Oncology, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
| | - Wei Zhao
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Guipeng Ding
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Xiaohua Li
- Center for Pathology and Laboratory Medicine, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
- School of Medicine, Jiangsu University, Jiangsu, China
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenqing Feng
- Department of Oncology, Zhangjiagang Ao Yang Hospital, Zhangjiagang, Jiangsu, China
- Department of Pathology, Nanjing Medical University, Nanjing, China
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, China
- Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|