101
|
Li W, Lv S, Liu G, Lu N, Jiang Y, Liang H, Xia W, Xiang Y, Xie C, He J. Epstein-Barr virus DNA seropositivity links distinct tumoral heterogeneity and immune landscape in nasopharyngeal carcinoma. Front Immunol 2023; 14:1124066. [PMID: 36860875 PMCID: PMC9968721 DOI: 10.3389/fimmu.2023.1124066] [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: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Background Epstein-Barr virus (EBV) DNA seronegative (Sero-) and seropositive (Sero+) nasopharyngeal carcinoma (NPC) are distinctly different disease subtypes. Patients with higher baseline EBV DNA titers seem to benefit less from anti-PD1 immunotherapy, but underlying mechanisms remain unclear. Tumor microenvironment (TME) characteristics could be the important factor affecting the efficacy of immunotherapy. Here, we illuminated the distinct multicellular ecosystems of EBV DNA Sero- and Sero+ NPCs from cellular compositional and functional perspectives at single-cell resolution. Method We performed single-cell RNA sequencing analyses of 28,423 cells from ten NPC samples and one non-tumor nasopharyngeal tissue. The markers, function, and dynamics of related cells were analyzed. Results We found that tumor cells from EBV DNA Sero+ samples exhibit low-differentiation potential, stronger stemness signature, and upregulated signaling pathways associated with cancer hallmarks than that of EBV DNA Sero- samples. Transcriptional heterogeneity and dynamics in T cells were associated with EBV DNA seropositivity status, indicating different immunoinhibitory mechanisms employed by malignant cells depending on EBV DNA seropositivity status. The low expression of classical immune checkpoints, early-triggered cytotoxic T-lymphocyte response, global activation of IFN-mediated signatures, and enhanced cell-cell interplays cooperatively tend to form a specific immune context in EBV DNA Sero+ NPC. Conclusions Collectively, we illuminated the distinct multicellular ecosystems of EBV DNA Sero- and Sero+ NPCs from single-cell perspective. Our study provides insights into the altered tumor microenvironment of NPC associated with EBV DNA seropositivity, which will help direct the development of rational immunotherapy strategies.
Collapse
Affiliation(s)
- Wangzhong Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Shuhui Lv
- Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Guoying Liu
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Nian Lu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yaofei Jiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Hu Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Weixiong Xia
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yanqun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,*Correspondence: Jianxing He, ; Changqing Xie, ; Yanqun Xiang,
| | - Changqing Xie
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States,*Correspondence: Jianxing He, ; Changqing Xie, ; Yanqun Xiang,
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China,*Correspondence: Jianxing He, ; Changqing Xie, ; Yanqun Xiang,
| |
Collapse
|
102
|
Sun P, Zhang H, Shi J, Xu M, Cheng T, Lu B, Yang L, Zhang X, Huang J. KRTCAP2 as an immunological and prognostic biomarker of hepatocellular carcinoma. Colloids Surf B Biointerfaces 2023; 222:113124. [PMID: 36634487 DOI: 10.1016/j.colsurfb.2023.113124] [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: 11/09/2022] [Revised: 12/27/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
Alterations in protein glycosylation affect tumor progression and immune responses in the tumor microenvironment. Keratinocyte-associated protein 2 (KRTCAP2) encodes the corresponding proteins involved in N-glycosylation. The clinical predictive significance and immune role of KRTCAP2 in hepatocellular carcinoma (HCC) largely remain elusive. Combining bioinformatics tools and multiplex immunohistochemistry analysis, we evaluated the KRTCAP2 expression in the HCC tumor microenvironment. The results showed that KRTCAP2 mRNA and protein expression were markedly increased in HCC tissues. Furthermore, high KRTCAP2 expression was an independent predictive factor of unfavorable prognosis in HCC. Moreover, high KRTCAP2 protein expression was associated with a lower proportion of CD8+ T cells and CD68+ macrophages in the stroma region. There was also a lower proportion of CD8+ T cells in the tumor region with high KRTCAP2 protein expression. Specifically, KRTCAP2 expression showed an inverse relationship with programmed cell death ligand-1 in HCC. Analysis of immunophenoscore showed that the low KRTCAP2 expression group had a stronger ability to predict response to immune checkpoint inhibitors. In conclusion, KRTCAP2 had a significant prognostic value for HCC and was correlated with the immune microenvironment. Our findings suggest that KRTCAP2 is a prognostic marker for HCC patients with potential clinical implications for predicting immunotherapeutic responsiveness.
Collapse
Affiliation(s)
- Pingping Sun
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Hui Zhang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jiawen Shi
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Manyu Xu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Tong Cheng
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Bing Lu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Lei Yang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaojing Zhang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jianfei Huang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
103
|
Guo B, Chen Q, Liu Z, Chen X, Zhu P. Adjuvant therapy following curative treatments for hepatocellular carcinoma: current dilemmas and prospects. Front Oncol 2023; 13:1098958. [PMID: 37139151 PMCID: PMC10149944 DOI: 10.3389/fonc.2023.1098958] [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: 11/15/2022] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
Curative surgical treatments, mainly liver resection, are still one of the optimal options for patients with early-, mid-, and even progression-stage hepatocellular carcinoma (HCC). However, the recurrence rate within 5 years after surgery is as high as 70%, especially in patients with high risk factors for recurrence, most of whom experience early recurrence within 2 years. Effective adjuvant therapy may improve prognosis, previous studies found that adjuvant transarterial chemoembolization, antiviral, and traditional Chinese medicine et al. were helpful in preventing HCC recurrence. Nevertheless, due to controversial results or lack of high-level evidence, there is no standardized postoperative management protocol worldwide at present. Continued exploration of effective postoperative adjuvant treatments to improve surgical prognosis is necessary.
Collapse
Affiliation(s)
- Bin Guo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Zhicheng Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Peng Zhu,
| |
Collapse
|
104
|
Ai Y, Wang H, Zheng Q, Li S, Liu J, Huang J, Tang J, Meng X. Add fuel to the fire: Inflammation and immune response in lung cancer combined with COVID-19. Front Immunol 2023; 14:1174184. [PMID: 37033918 PMCID: PMC10076709 DOI: 10.3389/fimmu.2023.1174184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
The corona virus disease 2019 (COVID-19) global pandemic has had an unprecedented and persistent impact on oncological practice, especially for patients with lung cancer, who are more vulnerable to the virus than the normal population. Indeed, the onset, progression, and prognosis of the two diseases may in some cases influence each other, and inflammation is an important link between them. The original chronic inflammatory environment of lung cancer patients may increase the risk of infection with COVID-19 and exacerbate secondary damage. Meanwhile, the acute inflammation caused by COVID-19 may induce tumour progression or cause immune activation. In this article, from the perspective of the immune microenvironment, the pathophysiological changes in the lungs and whole body of these special patients will be summarised and analysed to explore the possible immunological storm, immunosuppression, and immune escape phenomenon caused by chronic inflammation complicated by acute inflammation. The effects of COVID-19 on immune cells, inflammatory factors, chemokines, and related target proteins in the immune microenvironment of tumours are also discussed, as well as the potential role of the COVID-19 vaccine and immune checkpoint inhibitors in this setting. Finally, we provide recommendations for the treatment of lung cancer combined with COVID-19 in this special group.
Collapse
Affiliation(s)
- Yanling Ai
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hengyi Wang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiao Zheng
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Songtao Li
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingwen Liu
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ju Huang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jianyuan Tang, ; Xiangrui Meng,
| | - Xiangrui Meng
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jianyuan Tang, ; Xiangrui Meng,
| |
Collapse
|
105
|
Fan B, Zheng X, Wang Y, Hu X. Predicting prognosis and clinical efficacy of immune checkpoint blockade therapy via interferon-alpha response in muscle-invasive bladder cancer. Pathol Oncol Res 2023; 29:1611117. [PMID: 37082269 PMCID: PMC10110843 DOI: 10.3389/pore.2023.1611117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
Background: Immune checkpoint blockade (ICB) can prompt durable and robust responses in multiple cancers, involving muscle-invasive bladder cancer (MIBC). However, only a limited fraction of patients received clinical benefit. Clarifying the determinants of response and exploring corresponding predictive biomarkers is key to improving outcomes. Methods: Four independent formerly published cohorts consisting of 641 MIBC patients were enrolled in this study. We first analyzed the associations between various cancer hallmarks and ICB therapy response in two immunotherapeutic cohorts to identify the leading prognostic hallmark in MIBC. Furthermore, advanced machine learning methods were performed to select robust and promising predictors from genes functioning in the above leading pathway. The predictive ability of selected genes was also validated in multiple MIBC cohorts. Results: We identified and verified IFNα response as the leading cancer hallmark indicating better treatment responses, favorable overall survival, and an inflamed tumor microenvironment with higher infiltration of immune effector cells in MIBC patients treated with ICB therapy. Subsequently, two commonly selected genes, CXCL10 and LAMP3, implied better therapy response and the CXCL10highLAMP3high patients would benefit more from ICB therapy, which was comprehensively validated from the perspective of gene expression, clinical response, patient survival and immune features. Conclusion: Higher IFNα response primarily predicted better ICB therapeutic responses and reflected an inflamed microenvironment in MIBC. A composite of CXCL10 and LAMP3 expression could serve as promising predictive biomarkers for ICB therapeutic responses and be beneficial for clinical decision-making in MIBC.
Collapse
Affiliation(s)
- Bohan Fan
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Xin Zheng
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Urology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yicun Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
- *Correspondence: Xiaopeng Hu,
| |
Collapse
|
106
|
Lu W, Xie B, Tan G, Dai W, Ren J, Pervaz S, Li K, Li F, Wang Y, Wang M. Elafin is related to immune infiltration and could predict the poor prognosis in ovarian cancer. Front Endocrinol (Lausanne) 2023; 14:1088944. [PMID: 36742380 PMCID: PMC9893492 DOI: 10.3389/fendo.2023.1088944] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most lethal gynecologic malignancy, yet the clinical results for OC patients are still variable. Therefore, we examined how elafin expression affects the patients' prognoses and immunotherapy responses in OC, which may facilitate treatment selection and improve prognosis. METHODS The elafin mRNA expression profile was downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus. Elafin's prognostic potential and its relationship with clinical variables were investigated using Kaplan-Meier survival curves, time-dependent receiver operating characteristic curves as well as univariate and multivariate Cox regression models. As validation, protein expression in the tumor and adjacent tissues of OC patients was investigated by using immunohistochemistry (IHC). Comprehensive analyses were then conducted to explore the correlation between immune infiltration and elafin expression. RESULTS A higher mRNA expression of elafin was associated with an unfavorable prognosis in TCGA cohort and was validated in GSE31245 and IHC. Moreover, elafin was indicated as an independent risk factor for OC. A significantly higher protein expression of elafin was detected in the adjacent tissues of OC patients with shorter overall survival (OS). The immune-related pathways were mainly enriched in the high-elafin-mRNA-expression group. However, the mRNA expression of elafin was favorably correlated with indicators of the immune filtration and immunotherapy response, which also proved better immunotherapy outcomes. CONCLUSION The high elafin expression was associated with an unfavorable OS, while it also indicated better immunotherapy responses. Thus, the detection of elafin is beneficial to diagnosis and treatment selection.
Collapse
Affiliation(s)
- Weiyu Lu
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Biao Xie
- Department of Biostatistics, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Guangqing Tan
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Wanying Dai
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Jingyi Ren
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Sadaf Pervaz
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Kun Li
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Meijiao Wang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
- *Correspondence: Meijiao Wang,
| |
Collapse
|
107
|
Li Q, Han J, Yang Y, Chen Y. PD-1/PD-L1 checkpoint inhibitors in advanced hepatocellular carcinoma immunotherapy. Front Immunol 2022; 13:1070961. [PMID: 36601120 PMCID: PMC9806143 DOI: 10.3389/fimmu.2022.1070961] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has a high prevalence and mortality rate worldwide. Sorafenib monotherapy has been the standard of first-line treatment for advanced HCC for a long time, but there are still many shortcomings. In recent years, with the deepening of research on tumor immune microenvironment, researchers have begun to explore new approaches in immunotherapy, and the introduction of immune checkpoint inhibitors has brought fundamental changes to the treatment of HCC. Programmed cell death protein 1 (PD-1) is an immune checkpoint molecule that plays an important role in down-regulating immune system function and promoting tolerance. Programmed cell death ligand 1 (PDL-1) is involved in tumor immune evasion by binding to PD-1, resulting in failure of treatment. Currently, immunotherapy targeting the PD-1/PD-L1 axis has achieved unprecedented success in HCC, but it also faces great challenges, with its low remission rate still to be solved. For most patients with HCC, the PD-1/PD-L1 pathway is not the only rate limiting factor of antitumor immunity, and blocking only the PD-1/PD-L1 axis is not enough to stimulate an effective antitumor immune response; thus, combination therapy may be a better option. In this study, changes in the immune microenvironment of HCC patients were reviewed to clarify the feasibility of anti-PD-1/PD-L1 therapy, and a series of monotherapy and combination therapy clinical trials were summarized to verify the safety and efficacy of this newly developed treatment in patients with advanced HCC. Furthermore, we focused on hyperprogressive disease and drug resistance to gain a better understanding of PD-1/PD-L1 blockade as a promising treatment.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Han
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yonglin Yang
- Department of Infectious Diseases, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| | - Yu Chen
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
108
|
Xu RC, Wang F, Sun JL, Abuduwaili W, Zhang GC, Liu ZY, Liu TT, Dong L, Shen XZ, Zhu JM. A novel murine model of combined hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J Transl Med 2022; 20:579. [PMID: 36494846 PMCID: PMC9733131 DOI: 10.1186/s12967-022-03791-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Primary liver cancer (PLC) is a common gastrointestinal malignancy worldwide. While hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are two major pathologic types of PLC, combined HCC and ICC (cHCC-ICC) is a relatively rare subtype that shares both hepatocyte and cholangiocyte differentiation. However, the molecular feature of this unique tumor remains elusive because of its low incidence and lack of a suitable animal model. Herein, we generated a novel spontaneous cHCC-ICC model using a Sleeping Beauty-dependent transposon plasmid co-expressing oncogenic Myc and AKT1 and a CRISPR-Cas9 plasmid expressing single-guide RNA targeting p53 into mouse hepatocytes via in situ electroporation. The histological and transcriptional analysis confirmed that this model exhibits cHCC-ICC features and activates pathways committing cHCC-ICC formation, such as TGF-β, WNT, and NF-κB. Using this model, we further screened and identified LAMB1, a protein involved in cell adhesion and migration, as a potential therapeutic target for cHCC-ICC. In conclusion, our work presents a novel genetic cHCC-ICC model and provides new insights into cHCC-ICC.
Collapse
Affiliation(s)
- Ru-Chen Xu
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Fu Wang
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Jia-Lei Sun
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Weinire Abuduwaili
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Guang-Cong Zhang
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Zhi-Yong Liu
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Tao-Tao Liu
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Ling Dong
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| | - Xi-Zhong Shen
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China ,grid.11841.3d0000 0004 0619 8943Key Laboratory of Medical Molecular Virology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ji-Min Zhu
- grid.413087.90000 0004 1755 3939Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, 180 Fenglin Rd., Shanghai, 200032 China ,grid.413087.90000 0004 1755 3939Shanghai Institute of Liver Diseases, Shanghai, China
| |
Collapse
|
109
|
Li W, Xu N, Meng X, Yuan H, Yu T, Miao Q, Yang H, Hai B, Xiao W, Zhang X. SLC17A9-PTHLH-EMT axis promotes proliferation and invasion of clear renal cell carcinoma. iScience 2022; 26:105764. [PMID: 36590170 PMCID: PMC9800294 DOI: 10.1016/j.isci.2022.105764] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/16/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
SLC17A9 is a vesicular ATP transport protein that plays an important role in determining cell functions and the onset and progression of different diseases. In this study, SLC17A9 was initially identified as a potential diagnostic and prognostic risk biomarker for clear cell renal cell carcinoma (ccRCC). Then, the aberrant expression levels of SLC17A9 were confirmed in both the cell lines and clinical tissues. Mechanistically, SLC17A9 could upregulate the expression of PTHLH, thus promoting epithelial-mesenchymal transition (EMT) in ccRCC. Functionally, SLC17A9 knockdown inhibited the proliferation, migration, and invasion activity of renal cancer cells, whereas its overexpression led to stronger cell viability and more malignant phenotype in vitro. The overexpression of SLC17A9 in vivo could significantly contribute to the growth of tumors. Finally, we found that SLC17A9 might be related to the drug resistance of vorinostat. Cumulatively, this study demonstrated that the SLC17A9-PTHLH-EMT axis could promote the progression of ccRCC.
Collapse
Affiliation(s)
- Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Ning Xu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Hongwei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Tiexi Yu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Qi Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bo Hai
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China,Corresponding author
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China,Corresponding author
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China,Corresponding author
| |
Collapse
|
110
|
Identification of Glucose Metabolism-Related Genes in the Progression from Nonalcoholic Fatty Liver Disease to Hepatocellular Carcinoma. Genet Res (Camb) 2022; 2022:8566342. [PMID: 36407083 PMCID: PMC9649330 DOI: 10.1155/2022/8566342] [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: 09/07/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a manifestation of hepatic metabolic syndrome that varies in severity. Hepatocellular carcinoma progresses from NAFLD when there is heterogeneity in the infiltration of immune cells and molecules. A precise molecular classification of NAFLD remains lacking, allowing further exploration of the link between NAFLD and hepatocellular carcinoma. In this work, a weighted gene coexpression network analysis was used to identify two coexpression modules based on multiple omics data used to differentiate NAFLD subtypes. Additionally, key genes in the process of glucose metabolism and NAFLD were used to construct a prognostic model in a cohort of patients with hepatocellular carcinoma. Furthermore, the specific expression of signature genes in hepatocellular carcinoma cells was analyzed using a single-cell RNA sequencing approach. A total of 19 liver tissues of NAFLD patients were obtained from the GEO database, and 81 glucose metabolism-related genes were downloaded from the CTD database. In addition, based on nine signature genes, we constructed a prognostic model to divide the HCC cohort into high and low-risk groups. We also demonstrated a significant correlation between prognostic models and clinical phenotypes. Furthermore, we integrated single-cell RNA-sequencing data and immunology data to assess potential relationships between different molecular subtypes and hepatocellular carcinoma. Finally, our study discovered that the glucose metabolism pathway may play an important role in the process of NAFLD-hepatocellular carcinoma. In addition, three glucose metabolism-related genes (SERPINE1, VCAN, and TFPI2) may be the potential targets for the immunotherapy of patients with NAFLD-hepatocellular carcinoma.
Collapse
|
111
|
Wang T, Lin M, Mao J, Tian L, Gan H, Hu X, Yan L, Long H, Cai J, Zheng X, Xiao Y, Li D, Shuai X, Pang P. Inflammation-Regulated Nanodrug Sensitizes Hepatocellular Carcinoma to Checkpoint Blockade Therapy by Reprogramming the Tumor Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:49542-49554. [PMID: 36314479 DOI: 10.1021/acsami.2c14448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Immune checkpoint blockade (ICB) utilizing programmed death ligand-1 (PD-L1) antibody is a promising treatment strategy in solid tumors. However, in fact, more than half of hepatocellular carcinoma (HCC) patients are unresponsive to PD-L1-based ICB treatment due to multiple immune evasion mechanisms such as the hyperactivation of inflammation pathway, excessive tumor-associated macrophages (TAMs) infiltration, and insufficient infiltration of T cells. Herein, an inflammation-regulated nanodrug was designed to codeliver NF-κB inhibitor curcumin and PD-L1 antibody to reprogram the tumor microenvironment (TME) and activate antitumor immunity. The nanodrug accumulated in TME by an enhanced permeability and retention effect, where it left antibody to block PD-L1 on the membrane of tumor cells and TAMs due to pH-responsiveness. Simultaneously, a new curcumin-encapsulated nanodrug was generated, which was easily absorbed by either tumor cells or TAMs to inhibit the nuclear factor kappa-B (NF-κB) signal and related immunosuppressive genes. The inflammation-regulated nanodrug possessed good biocompatibility. Simultaneously, it reprogrammed TME effectively and exhibited an effective anticancer effect in immunocompetent mice. Overall, this study provided a potent strategy to improve the efficiency of ICB-based treatment for HCC.
Collapse
Affiliation(s)
- TianCheng Wang
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - MinZhao Lin
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - JunJie Mao
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - LiRong Tian
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - HaiRun Gan
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - XinYan Hu
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - LeYe Yan
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - HaoYu Long
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - JianXun Cai
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - XiaoDi Zheng
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - YuDong Xiao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha 410012, China
| | - Dan Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - XinTao Shuai
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - PengFei Pang
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| |
Collapse
|
112
|
Choi SI, Yin J. Prospective approaches to enhancing CAR T cell therapy for glioblastoma. Front Immunol 2022; 13:1008751. [PMID: 36275671 PMCID: PMC9582117 DOI: 10.3389/fimmu.2022.1008751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor. The poor clinical outcome and overall ineffectiveness of current standard treatments, including surgery, chemotherapy, and radiation, highlight the urgent need for alternative tumor-specific therapies for GBM. Chimeric antigen receptor (CAR) T cell therapy is a revolutionary therapeutic strategy for hematological malignancies, but the optimal potency of CAR T cell therapy for solid tumors, especially GBM, has not been achieved. Although CAR T cell therapeutic strategies for GBM have been assessed in clinical trials, the current antitumor activity of CAR T cells remains insufficient. In this review, we present our perspective on genetically modifying CAR constructs, overcoming T cell dysfunctions, and developing additional treatments that can improve CAR T cell effectiveness, such as functionality, persistence, and infiltration into tumor sites. Effectively improved CAR T cells may offer patients with GBM new treatment opportunities, and this review is intended to provide a comprehensive overview for researchers to develop potent CAR T cells using genetic engineering or combinatorial preparations.
Collapse
Affiliation(s)
- Sun Il Choi
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
| | - Jinlong Yin
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- *Correspondence: Jinlong Yin,
| |
Collapse
|
113
|
Xiong J, Wang QQ. Mechanisms and strategies to overcome immunotherapy resistance in hepatobiliary malignancies. Hepatobiliary Pancreat Dis Int 2022; 21:430-439. [PMID: 35907687 DOI: 10.1016/j.hbpd.2022.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023]
Abstract
Unprecedented advances have been achieved in hepatobiliary cancer treatment with immune checkpoint blockade (ICB). However, the efficacy of ICB in patients with hepatobiliary malignancies is still limited. Resistance to immunotherapies is often orchestrated by complicated tumor-host-microenvironment interactions but could also occur after initial efficacy, mostly when only partial responses are obtained. Clarification of cancer-resistance mechanisms will be beneficial to provide the rationale for the administration of personalized drugs. Here, we review the factors related to resistance to immune-targeted therapies in hepatobiliary malignancies and discuss the potential strategies for overcoming resistance and future directions of immunotherapy development.
Collapse
Affiliation(s)
- Jia Xiong
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Qing-Qing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China; The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
114
|
Targeting JWA for Cancer Therapy: Functions, Mechanisms and Drug Discovery. Cancers (Basel) 2022; 14:cancers14194655. [PMID: 36230577 PMCID: PMC9564207 DOI: 10.3390/cancers14194655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary JWA has been identified as a potential therapeutic target for several cancers. In this review, we summarize the tumor suppressive functions of the JWA gene and its role in anti-cancer drug development. The focus is on elucidating the key regulatory proteins up and downstream of JWA and their signaling networks. We also discuss current strategies for targeting JWA (JWA peptides, small molecule agonists, and JWA-targeted Pt (IV) prodrugs). Abstract Tumor heterogeneity limits the precision treatment of targeted drugs. It is important to find new tumor targets. JWA, also known as ADP ribosylation factor-like GTPase 6 interacting protein 5 (ARL6IP5, GenBank: AF070523, 1998), is a microtubule-associated protein and an environmental response gene. Substantial evidence shows that JWA is low expressed in a variety of malignancies and is correlated with overall survival. As a tumor suppressor, JWA inhibits tumor progression by suppressing multiple oncogenes or activating tumor suppressor genes. Low levels of JWA expression in tumors have been reported to be associated with multiple aspects of cancer progression, including angiogenesis, proliferation, apoptosis, metastasis, and chemotherapy resistance. In this review, we will discuss the structure and biological functions of JWA in tumors, examine the potential therapeutic strategies for targeting JWA and explore the directions for future investigation.
Collapse
|
115
|
Zhang P, Ono A, Fujii Y, Hayes CN, Tamura Y, Miura R, Shirane Y, Nakahara H, Yamauchi M, Uchikawa S, Uchida T, Teraoka Y, Fujino H, Nakahara T, Murakami E, Miki D, Kawaoka T, Okamoto W, Makokha GN, Imamura M, Arihiro K, Kobayashi T, Ohdan H, Fujita M, Nakagawa H, Chayama K, Aikata H. The presence of Vessels Encapsulating Tumor Clusters is associated with an immunosuppressive tumor microenvironment in hepatocellular carcinoma. Int J Cancer 2022; 151:2278-2290. [DOI: 10.1002/ijc.34247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Peiyi Zhang
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Atsushi Ono
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Yasutoshi Fujii
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - C. Nelson Hayes
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Yosuke Tamura
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Ryoichi Miura
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Yuki Shirane
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Hikaru Nakahara
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Masami Yamauchi
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Shinsuke Uchikawa
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Takuro Uchida
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Yuji Teraoka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Hatsue Fujino
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Takashi Nakahara
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Eisuke Murakami
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Daiki Miki
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Wataru Okamoto
- Cancer Treatment Center Hiroshima University Hospital Hiroshima Japan
| | - Grace Naswa Makokha
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Koji Arihiro
- Department of Anatomical Pathology Hiroshima University Hospital Hiroshima Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences Hiroshima University
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences Hiroshima University
| | - Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences Yokohama Japan
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences Yokohama Japan
| | - Kazuaki Chayama
- Collaborative Research Laboratory of Medical Innovation, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
- Research Center for Hepatology and Gastroenterology Hiroshima University Hiroshima Japan
- RIKEN Center for Integrative Medical Sciences Yokohama Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| |
Collapse
|