1
|
Garcia A, Mathew SO. Racial/Ethnic Disparities and Immunotherapeutic Advances in the Treatment of Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:2446. [PMID: 39001508 PMCID: PMC11240753 DOI: 10.3390/cancers16132446] [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: 05/24/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the leading causes of death among many associated liver diseases. Various conventional strategies have been utilized for treatment, ranging from invasive surgeries and liver transplants to radiation therapy, but fail due to advanced disease progression, late screening/staging, and the various etiologies of HCC. This is especially evident within racially distinct populations, where incidence rates are higher and treatment outcomes are worse for racial/ethnic minorities than their Caucasian counterparts. However, with the rapid development of genetic engineering and molecular and synthetic biology, many novel strategies have presented promising results and have provided potential treatment options. In this review, we summarize past treatments, how they have shaped current treatments, and potential treatment strategies for HCC that may prove more effective in the future.
Collapse
Affiliation(s)
- Alexsis Garcia
- Department of Microbiology, Immunology & Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Stephen O Mathew
- Department of Microbiology, Immunology & Genetics, UNT Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
2
|
Scatozza F, Giardina MM, Valente C, Vigiano Benedetti V, Facchiano A. Anti-Melanoma Effects of Miconazole: Investigating the Mitochondria Involvement. Int J Mol Sci 2024; 25:3589. [PMID: 38612401 PMCID: PMC11011910 DOI: 10.3390/ijms25073589] [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: 01/31/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
Miconazole is an antimycotic drug showing anti-cancer effects in several cancers. However, little is known on its effects in melanoma. A375 and SK-MEL-28 human melanoma cell lines were exposed to miconazole and clotrimazole (up to 100 mM). Proliferation, viability with MTT assay and vascular mimicry were assayed at 24 h treatment. Molecular effects were measured at 6 h, namely, ATP-, ROS-release and mitochondria-related cytofluorescence. A metabolomic profile was also investigated at 6 h treatment. Carnitine was one of the most affected metabolites; therefore, the expression of 29 genes involved in carnitine metabolism was investigated in the public platform GEPIA2 on 461 melanoma patients and 558 controls. After 24 h treatments, miconazole and clotrimazole strongly and significantly inhibited proliferation in the presence of 10% serum on either melanoma cell lines; they also strongly reduced viability and vascular mimicry. After 6 h treatment, ATP reduction and ROS increase were observed, as well as a significant reduction in mitochondria-related fluorescence. Further, in A375, miconazole strongly and significantly altered expression of several metabolites including carnitines, phosphatidyl-cholines, all amino acids and several other small molecules, mostly metabolized in mitochondria. The expression of 12 genes involved in carnitine metabolism was found significantly modified in melanoma patients, 6 showing a significant impact on patients' survival. Finally, miconazole antiproliferation activity on A375 was found completely abrogated in the presence of carnitine, supporting a specific role of carnitine in melanoma protection toward miconazole effect, and was significantly reversed in the presence of caspases inhibitors such as ZVAD-FMK and Ac-DEVD-CHO, and a clear pro-apoptotic effect was observed in miconazole-treated cells, by FACS analysis of Annexin V-FITC stained cells. Miconazole strongly affects proliferation and other biological features in two human melanoma cell lines, as well as mitochondria-related functions such as ATP- and ROS-release, and the expression of several metabolites is largely dependent on mitochondria function. Miconazole, likely acting via carnitine and mitochondria-dependent apoptosis, is therefore suggested as a candidate for further investigations in melanoma treatments.
Collapse
|
3
|
Radwan EM, Abo-Elabass E, Abd El-Baky AE, Alshwyeh HA, Almaimani RA, Almaimani G, Ibrahim IAA, Albogami A, Jaremko M, Alshawwa SZ, Saied EM. Unveiling the antitumor potential of novel N-(substituted-phenyl)-8-methoxycoumarin-3-carboxamides as dual inhibitors of VEGFR2 kinase and cytochrome P450 for targeted treatment of hepatocellular carcinoma. Front Chem 2023; 11:1231030. [PMID: 37601910 PMCID: PMC10436493 DOI: 10.3389/fchem.2023.1231030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Being the sixth most diagnosed cancer and the fourth leading cause of cancer-related deaths worldwide, liver cancer is considered as a serious disease with a high prevalence and poor prognosis. Current anticancer drugs for liver cancer have drawbacks, such as limited efficacy in later stages of the disease, toxicity to healthy cells, and the potential for drug resistance. There is ample evidence that coumarin-based compounds are potent anticancer agents, with numerous analogues currently being investigated in preclinical and clinical studies. The current study aimed to explore the antitumor potency of a new class of 8-methoxycoumarin-3-carboxamides against liver cancer. Toward this aim, we have designed, synthesized, and characterized a new set of N-(substituted-phenyl)-8-methoxycoumarin-3-carboxamide analogues. The assessment of antitumor activity revealed that the synthesized class of compounds possesses substantial cytotoxicity toward Hep-G2 cells when compared to staurosporine, without significant impact on normal cells. Out of the synthesized compounds, compound 7 demonstrated the most potent cytotoxic effect against Hep-G2 cells with an IC50 of 0.75 µM, which was more potent than the drug staurosporine (IC50 = 8.37 µM). The investigation into the mechanism behind the antiproliferative activity of compound 7 revealed that it interferes with DNA replication and induces DNA damage, leading to cell cycle arrest as demonstrated by a significant decrease in the percentage of cells in the G1 and G2/M phases, along with an increase in the percentage of cells in the S phase. Flow cytometric analysis further revealed that compound 7 has the ability to trigger programmed cell death by inducing necrosis and apoptosis in HepG-2 cells. Further explorations into the mechanism of action demonstrated that compound 7 displays a potent dual-inhibitory activity toward cytochrome P450 and vascular endothelial growth factor receptor-2 (VEGFR-2) proteins, as compared to sorafenib drug. Further, detailed computational studies revealed that compound 7 displays a considerable binding affinity toward the binding cavity of VEGFR2 and CYP450 proteins. Taken together, our findings indicate that the newly synthesized class of compounds, particularly compound 7, could serve as a promising scaffold for the development of highly effective anticancer agents against liver cancer.
Collapse
Affiliation(s)
- Eman M. Radwan
- The Division of Organic Chemistry, Chemistry Department, Faculty of Science, Port-Said University, Port-Said, Egypt
| | - Eman Abo-Elabass
- The Division of Biochemistry, Chemistry Department, Faculty of Science, Port-Said University, Port-Said, Egypt
| | - Atef E. Abd El-Baky
- Biochemistry Department, Faculty of Pharmacy, Port-Said University, Port-Said, Egypt
| | - Hussah Abdullah Alshwyeh
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Riyad A. Almaimani
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ghassan Almaimani
- Department of Surgery, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulaziz Albogami
- Biology Department, Faculty of science, Al-Baha University, Al Aqiq, Saudi Arabia
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences (BESE) and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Samar Z. Alshawwa
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| |
Collapse
|
4
|
Wang B, Ma X, Zhang W, Li L, Zan Y, Zhan J, Guo X, Lei M, Ma H. Impact of NOTCH1 polymorphisms on liver cancer in a Chinese Han population. Cell Cycle 2023; 22:1127-1134. [PMID: 36951273 PMCID: PMC10081089 DOI: 10.1080/15384101.2023.2189766] [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: 07/04/2022] [Revised: 02/14/2023] [Accepted: 03/06/2023] [Indexed: 03/24/2023] Open
Abstract
NOTCH1, a member of the Notch family, is up-expression in advanced liver cancer (LC) patients and is associated with tumor sizes, tumor stage, metastasis, and invasion. A few studies have discovered the contribution of NOTCH1 variants to LC risk. Our purpose was to assess the relationship of NOTCH1 rs10521, rs2229971, and rs4489420 to LC risk. We enrolled 709 LC patients and 708 healthy controls. Genotyping was determined through the Agena MassARRAY system. Multiple genetic models by logistic regression were useful for odds ratios (ORs) with 95% confidence intervals (CIs). Rs10521-G (p = 0.009, OR = 0.75, 95% CI: 0.61-0.93), rs2229971-A (p = 0.023, OR = 0.81, 95% CI: 0.67-0.97), and rs4489420-A (p = 0.014, OR = 0.38, 95% CI: 0.16-0.85) might be protective factors for LC occurrence in the Chinese Han population, especially rs10521 and rs2229971 (false-positive report probability (FPRP) <0.2 and statistical power >90%). Interestingly, stratified analysis displayed that the contribution of NOTCH1 polymorphisms to LC risk might be associated with gender, age, smoking, and drinking. Our data first determined that NOTCH1 rs10521-G, rs2229971-A, and rs4489420-A might be protective factors for LC susceptibility.
Collapse
Affiliation(s)
- Baofeng Wang
- Department of Radiation Therapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiaobin Ma
- Department of Oncology, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wenjie Zhang
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, China
| | - Liang Li
- Department of Radiotherapy, Shaanxi Provincial Cancer Hospital Affiliated to Medical College, Xi’an Jiaotong University, Xi’an, China
| | - Ying Zan
- Department of Oncology, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jianshui Zhan
- Department of Human Anatomy and Tissue embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xufeng Guo
- Department of Orthopedic, Xi’an Ninth Hospital, Xi’an, China
| | - Ming Lei
- Department of Radiology, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Hongbing Ma
- Department of Radiation Therapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
5
|
Siswanto FM, Handayani MDN, Firmasyah RD, Oguro A, Imaoka S. Nrf2 Regulates the Expression of CYP2D6 by Inhibiting the Activity of Krüppel-Like Factor 9 (KLF9). Curr Drug Metab 2023; 24:667-681. [PMID: 37916628 DOI: 10.2174/0113892002271342231013095255] [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: 07/31/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023]
Abstract
AIMS The aim of the present study is to gain insight into the biology of Parkinson's disease (PD) and cancer to drive translational advances enabling more effective prevention and/or potential treatments. BACKGROUND The expression of Cytochrome P450 2D6 (CYP2D6) is correlated with various diseases such as PD and cancer; therefore, exploring its regulatory mechanism at transcriptional levels is of interest. NF-E2-related factor 2 (Nrf2) has been known to be responsible for regulating phase II and phase III drug-metabolizing genes. OBJECTIVES The objectives of this study are to investigate the transcriptional regulation of CYP2D6 by Nrf2 and to analyze its role in PD and cancer. METHODS Nrf2 was transiently expressed in human hepatoma Hep3B cells, and the expression of CYP2D6 was examined by RT-qPCR. The promoter activity of CYP2D6 and the DNA binding of Nrf2 were examined by luciferase and ChIP assay, respectively. We then investigated the expression and correlation of Nrf2 and CYP2D6 in the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets. RESULTS In the present study, we demonstrated that Nrf2 down-regulated CYP2D6 mRNA expression in hepatoma Hep3B cells. Mechanistically, Nrf2 binds to the antioxidant responsive element (ARE) in the proximity of krüppel- like factor 9 (KLF9)-binding site within the -550/+51 of CYP2D6 promoter. The inhibition and activation of Nrf2 enhanced and suppressed KLF9 effects on CYP2D6 expression, respectively. The expression levels of Nrf2 and CYP2D6 were upregulated and downregulated in the PD patient GEO datasets compared to the healthy control tissues, and Nrf2 was negatively correlated with CYP2D6. In liver cancer patients, decreased CYP2D6 levels were apparent and associated with a lower probability of survival. CONCLUSION Our work revealed the inhibitory role of Nrf2 in regulating CYP2D6 expression. Moreover, Nrf2- dependent regulation of CYP2D6 can be used as a prognostic factor and therapeutic strategy in PD and liver cancer.
Collapse
Affiliation(s)
- Ferbian Milas Siswanto
- Department of Chemistry and Biochemistry, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
- Department of Biomedical Chemistry, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| | - Maria Dara Novi Handayani
- Department of Chemistry and Biochemistry, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Rita Dewi Firmasyah
- Department of Chemistry and Biochemistry, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Ami Oguro
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Susumu Imaoka
- Department of Biomedical Chemistry, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| |
Collapse
|
6
|
Zhou Y, Gu H, Shao B, Zhang S, Pall H, Peixoto RD, Mok SRS, Zhu G. Glycolysis-related gene dihydrolipoamide acetyltransferase promotes poor prognosis in hepatocellular carcinoma through the Wnt/β-catenin and PI3K/Akt signaling pathways. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1240. [PMID: 36544660 PMCID: PMC9761179 DOI: 10.21037/atm-22-5272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
Background Recent research suggests that dihydrolipoamide acetyltransferase (DLAT), which is a copper-induced cell death-related gene, is involved in multiple biological events in tumors. This study sought to investigate the relationship between DLAT and hepatocellular carcinoma (HCC). Methods In the Cancer Genome Atlas (TCGA) database, we first identified the differentially expressed gene (i.e., DLAT), then confirmed DLAT expression, and found a link between it and the prognosis of HCC patients. An internal validation nomogram was built based on a multivariate Cox regression analysis. Data from the Tumor Immune Estimation Resource (TIMER) database was used to examine the association between DLT and immunological cells. A gene set enrichment analysis (GSEA) was conducted to investigate the probable mechanism of action. Finally, in vitro cytological research was conducted to further examin the involvement of DLAT in HCC-related unfavorable biological events. Results The database screenings showed that DLAT was a differentially expressed molecule; that is, DLAT was more highly expressed in the cancer tissues than normal tissues. TCGA results and Kaplan-Meier-plotter data sets showed that HCC patients with reduced DLAT expression had greater disease-specific survival (DSS), overall survival (OS), and progression-free interval (PFI). The prediction model had a concordance index of 0.659 (0.614-0.704), which indicates high accuracy. According to the TIMER database, tumor cells in the HCC microenvironment may be able to bypass the immune system due to the expression of DLAT. The in vitro cytological tests showed that DLAT knockdown significantly decreased the proliferation and invasion of the HCC cells. It also inhibited the activity of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (Akt) and Wnt/β-catenin signaling pathways. Conclusions Decreased DLAT expression significantly prolongs the OS, PFI, and DSS of HCC patients. DLAT may be employed as a new predictive biomarker for HCC, and may be linked to the immune system in HCC patients. The tumor microenvironment (TME) may have a significant effect on the ability of tumor cells to evade the immune system. The PI3K/Akt and Wnt/β-catenin signaling pathways may affect the prognosis of HCC by interfering with DLAT. Given these findings, HCC may be an ideal target for the development of anti-cancer therapies.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Hepatobiliary Surgery, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China;,Southeast University School of Medicine, Nanjing, China
| | - Haijuan Gu
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Bingfeng Shao
- Department of Hepatobiliary Surgery, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Suqing Zhang
- Department of Hepatobiliary Surgery, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Harpreet Pall
- Department of Pediatrics, Hackensack Meridian School of Medicine, Hackensack Meridian K. Hovnanian Children’s Hospital, Jersey Shore University Medical Center, Neptune, NJ, USA
| | | | - Shaffer R. S. Mok
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Guodong Zhu
- Department of Hepatobiliary Surgery, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| |
Collapse
|
7
|
Jiang P, Xue W, Xi C, Zhuang L, Yuan Z, Liu Z, Sun T, Xu X, Tan Y, Ding W. A new acidic microenvironment related lncRNA signature predicts the prognosis of liver cancer patients. Front Oncol 2022; 12:1016721. [PMID: 36387100 PMCID: PMC9660327 DOI: 10.3389/fonc.2022.1016721] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/18/2022] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The acidic microenvironment (AME), like hypoxia, inflammation, or immunoreaction, is a hallmark of the tumor microenvironment (TME). This work aimed to develop a prediction signature dependent on AME-associated lncRNAs in order to predict the prognosis of LC individuals. METHODS We downloaded RNA-seq information and the corresponding clinical and predictive data from The Cancer Genome Atlas (TCGA) dataset and conducted univariate and multivariate Cox regression analyses to identify AME-associated lncRNAs for the construction of a prediction signature The Kaplan-Meier technique was utilized to determine the overall survival (OS) rate of the high (H)-risk and low (L)-risk groups. Using gene set enrichment analysis (GSEA) the functional variations between the H- and L-risk groups were investigated. The association between the prediction signature and immunological state was investigated using single-sample GSEA (ssGSEA). Additionally, the association between the predicted signature and the therapeutic response of LC individuals was evaluated. Lastly, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to verify the risk model. RESULTS We generated a signature comprised of seven AME-associated lncRNAs (LINC01116, AC002511.2, LINC00426, ARHGAP31-AS1, LINC01060, TMCC1-AS1, AC012065.1). The H-risk group had a worse prognosis than the L- risk group. The AME-associated lncRNA signature might determine the prognosis of individuals with LC independently. The AME-related lncRNA signature shows a greater predictive effectiveness than clinic-pathological factors, with an area under the receiver operating characteristic (ROC) curve of 0.806%. When participants were categorized based on several clinico-pathological characteristics, the OS of high-risk individuals was shorter compared to low-risk patients. GSEA demonstrated that the metabolism of different acids and the PPAR signaling pathway are closely associated with low-risk individuals. The prognostic signature was substantially associated with the immunological status of LC individuals, as determined by ssGSEA. High risk individuals were more sensitive to some immunotherapies (including anti-TNFSF4 anti-SIRPA, anti-CD276 and anti-TNFSF15) and some conventional chemotherapy drugs (including lapatinib and paclitaxel). Finally, the expression levels of the seven lncRNAs comprising the signature were tested by qRT-PCR. CONCLUSIONS A basis for the mechanism of AME-associated lncRNAs in LC is provided by the prediction signature, which also offers clinical therapeutic recommendations for LC individuals.
Collapse
Affiliation(s)
- Peng Jiang
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - Wenbo Xue
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - Cheng Xi
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - Lin Zhuang
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - Zhiping Yuan
- Department of Gastroenterology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Zhilin Liu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Tao Sun
- Department of Hepatopancreatobiliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xuezhong Xu
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - Yulin Tan
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - Wei Ding
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| |
Collapse
|