1
|
Liu X, Qin J, Nie J, Sun H, Pan Y, Wang S. Reclassifying TNM stage I/II colorectal cancer into two subgroups with different overall survival, tumor microenvironment, and response to immune checkpoint blockade treatment. Front Genet 2022; 13:948920. [PMID: 36212126 PMCID: PMC9532767 DOI: 10.3389/fgene.2022.948920] [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: 05/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The traditional TNM staging system is often insufficient to differentiate the survival discrepancies of colorectal cancer (CRC) patients at TNM stage I/II. Our study aimed to reclassify stage I/II CRC patients into several subgroups with different prognoses and explore their suitable therapeutic methods. Methods: Single-cell RNA (scRNA) sequencing data, bulk RNA sequencing data, and clinicopathological information of CRC patients were enrolled from the TCGA and GEO databases. The tumor microenvironment of CRC tissues was accessed by the ESTIMATE algorithm. The prognostic genes were identified by Cox regression analysis. GO and KEGG analyses were conducted in the DAVID database. GSEA analysis was performed for annotation of the correlated gene sets. Results: We successfully reclassified stage I/II CRC patients into two subgroups and discovered that patients in cluster-2 underwent worse overall survival than those in cluster-1. GSEA analysis showed that immune-associated gene sets were positively enriched in cluster-2. Besides, the differentially expressed genes (DEGs) between cluster-1 and cluster-2 patients also participated in immune-related biological processes and signaling pathways. Moreover, we found that more immune cells infiltrated the microenvironment of cluster-2 patients compared to that of cluster-1 patients, such as Tregs and tumor-associated macrophages. ScRNA sequencing analysis uncovered that most of the enriched immune-associated signaling in cluster-2 patients was mainly attributed to these upregulated immune cells whose infiltration levels were also high in CRC tissues rather than in normal tissues. In addition, we demonstrated that the expression of immune checkpoint genes was significantly higher in cluster-2 patients compared to cluster-1 patients. ScRNA sequencing analysis revealed that the infiltrated CD8+T cells in CRC were naïve T cells and can be activated into effector T cells after immune checkpoint blockade (ICB) treatment. Conclusion: TNM stage I/II CRC patients can be divided into two subgroups, which have different overall survival rates, tumor microenvironment, and response to ICB therapy.
Collapse
Affiliation(s)
- Xiangxiang Liu
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Qin
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junjie Nie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- *Correspondence: Yuqin Pan, ; Shukui Wang,
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- *Correspondence: Yuqin Pan, ; Shukui Wang,
| |
Collapse
|
2
|
Sharpe BP, Hayden A, Manousopoulou A, Cowie A, Walker RC, Harrington J, Izadi F, Breininger SP, Gibson J, Pickering O, Jaynes E, Kyle E, Saunders JH, Parsons SL, Ritchie AA, Clarke PA, Collier P, Mongan NP, Bates DO, Yacqub-Usman K, Garbis SD, Walters Z, Rose-Zerilli M, Grabowska AM, Underwood TJ. Phosphodiesterase type 5 inhibitors enhance chemotherapy in preclinical models of esophageal adenocarcinoma by targeting cancer-associated fibroblasts. Cell Rep Med 2022; 3:100541. [PMID: 35732148 PMCID: PMC9244979 DOI: 10.1016/j.xcrm.2022.100541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/14/2021] [Accepted: 01/28/2022] [Indexed: 12/03/2022]
Abstract
The chemotherapy resistance of esophageal adenocarcinomas (EACs) is underpinned by cancer cell extrinsic mechanisms of the tumor microenvironment (TME). We demonstrate that, by targeting the tumor-promoting functions of the predominant TME cell type, cancer-associated fibroblasts (CAFs) with phosphodiesterase type 5 inhibitors (PDE5i), we can enhance the efficacy of standard-of-care chemotherapy. In ex vivo conditions, PDE5i prevent the transdifferentiation of normal fibroblasts to CAF and abolish the tumor-promoting function of established EAC CAFs. Using shotgun proteomics and single-cell RNA-seq, we reveal PDE5i-specific regulation of pathways related to fibroblast activation and tumor promotion. Finally, we confirm the efficacy of PDE5i in combination with chemotherapy in close-to-patient and in vivo PDX-based model systems. These findings demonstrate that CAFs drive chemotherapy resistance in EACs and can be targeted by repurposing PDE5i, a safe and well-tolerated class of drug administered to millions of patients world-wide to treat erectile dysfunction.
Collapse
Affiliation(s)
- Benjamin P Sharpe
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Annette Hayden
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | | | - Andrew Cowie
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Robert C Walker
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Jack Harrington
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Fereshteh Izadi
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK; Centre for NanoHealth, Swansea University Medical School, Singleton Campus, Swansea SA2 8PP, UK
| | - Stella P Breininger
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Jane Gibson
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Oliver Pickering
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Eleanor Jaynes
- University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Ewan Kyle
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - John H Saunders
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Salford Royal NHS Foundation Trust, Salford M6 8HD, UK
| | - Simon L Parsons
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Nottingham University Hospitals NHS Trust, Hucknall Road, Nottingham NG5 1PB, UK
| | - Alison A Ritchie
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Philip A Clarke
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Pamela Collier
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nigel P Mongan
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA; Biodiscovery Institute, School of Veterinary Medicine and Science, University of Nottingham, Nottingham NG5 1PB, UK
| | - David O Bates
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kiren Yacqub-Usman
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Zoë Walters
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Matthew Rose-Zerilli
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Anna M Grabowska
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
| |
Collapse
|
3
|
Haince JF, Joubert P, Bach H, Ahmed Bux R, Tappia PS, Ramjiawan B. Metabolomic Fingerprinting for the Detection of Early-Stage Lung Cancer: From the Genome to the Metabolome. Int J Mol Sci 2022; 23:ijms23031215. [PMID: 35163138 PMCID: PMC8835988 DOI: 10.3390/ijms23031215] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
The five-year survival rate of lung cancer patients is very low, mainly because most newly diagnosed patients present with locally advanced or metastatic disease. Therefore, early diagnosis is key to the successful treatment and management of lung cancer. Unfortunately, early detection methods of lung cancer are not ideal. In this brief review, we described early detection methods such as chest X-rays followed by bronchoscopy, sputum analysis followed by cytological analysis, and low-dose computed tomography (LDCT). In addition, we discussed the potential of metabolomic fingerprinting, compared to that of other biomarkers, including molecular targets, as a low-cost, high-throughput blood-based test that is both feasible and affordable for early-stage lung cancer screening of at-risk populations. Accordingly, we proposed a paradigm shift to metabolomics as an alternative to molecular and proteomic-based markers in lung cancer screening, which will enable blood-based routine testing and be accessible to those patients at the highest risk for lung cancer.
Collapse
Affiliation(s)
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Pathology, Laval University, Quebec, QC G1V 4G5, Canada;
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| | - Rashid Ahmed Bux
- BioMark Diagnostics Inc., Richmond, BC V6X 2W8, Canada; (J.-F.H.); (R.A.B.)
| | - Paramjit S. Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Correspondence: ; Tel.: +1-204-258-1230
| | - Bram Ramjiawan
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
4
|
Nagano T, Tachihara M, Nishimura Y. Molecular Mechanisms and Targeted Therapies Including Immunotherapy for Non-Small Cell Lung Cancer. Curr Cancer Drug Targets 2020; 19:595-630. [PMID: 30526458 DOI: 10.2174/1568009619666181210114559] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/21/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
Abstract
Lung cancer is the leading cause of cancer death worldwide. Molecular targeted therapy has greatly advanced the field of treatment for non-small cell lung cancer (NSCLC), which accounts for the majority of lung cancers. Indeed, gefitinib, which was the first molecular targeted therapeutic agent, has actually doubled the survival time of NSCLC patients. Vigorous efforts of clinicians and researchers have revealed that lung cancer develops through the activating mutations of many driver genes including the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1), v-Raf murine sarcoma viral oncogene homolog B (BRAF), and rearranged during transfection (RET) genes. Although ALK, ROS1, and RET are rare genetic abnormalities, corresponding tyrosine kinase inhibitors (TKIs) can exert dramatic therapeutic effects. In addition to anticancer drugs targeting driver genes, bevacizumab specifically binds to human vascular endothelial growth factor (VEGF) and blocks the VEGF signaling pathway. The VEGF signal blockade suppresses angiogenesis in tumor tissues and inhibits tumor growth. In this review, we also explore immunotherapy, which is a promising new NSCLC treatment approach. In general, antitumor immune responses are suppressed in cancer patients, and cancer cells escape from the immune surveillance mechanism. Immune checkpoint inhibitors (ICIs) are antibodies that target the primary escape mechanisms, immune checkpoints. Patients who respond to ICIs are reported to experience longlasting therapeutic effects. A wide range of clinical approaches, including combination therapy involving chemotherapy or radiation plus adjuvant therapy, are being developed.
Collapse
Affiliation(s)
- Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
5
|
Yao J, Xue X, Qu D, Westphalen CB, Ge Y, Zhang L, Li M, Gao T, Chandrakesan P, Vega KJ, Peng J, An G, Weygant N. Reverse engineering a predictive signature characterized by proliferation, DNA damage, and immune escape from stage I lung adenocarcinoma recurrence. Acta Biochim Biophys Sin (Shanghai) 2020; 52:638-653. [PMID: 32395755 DOI: 10.1093/abbs/gmaa036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/14/2020] [Indexed: 12/24/2022] Open
Abstract
Identifying early-stage cancer patients at risk for progression is a major goal of biomarker research. This report describes a novel 19-gene signature (19-GCS) that predicts stage I lung adenocarcinoma (LAC) recurrence and response to therapy and performs comparably in pancreatic adenocarcinoma (PAC), which shares LAC molecular traits. Kaplan-Meier, Cox regression, and cross-validation analyses were used to build the signature from training, test, and validation sets comprising 831 stage I LAC transcriptomes from multiple independent data sets. A statistical analysis was performed using the R language. Pathway and gene set enrichment were used to identify underlying mechanisms. 19-GCS strongly predicts overall survival and recurrence-free survival in stage I LAC (P=0.002 and P<0.001, respectively) and in stage I-II PAC (P<0.0001 and P<0.0005, respectively). A multivariate cox regression analysis demonstrated the independence of 19-GCS from significant clinical factors. Pathway analyses revealed that 19-GCS high-risk LAC and PAC tumors are characterized by increased proliferation, enhanced stemness, DNA repair deficiency, and compromised MHC class I and II antigen presentation along with decreased immune infiltration. Importantly, high-risk LAC patients do not appear to benefit from adjuvant cisplatin while PAC patients derive additional benefit from FOLFIRINOX compared with gemcitabine-based regimens. When validated prospectively, this proof-of-concept biomarker may contribute to tailoring treatment, recurrence reduction, and survival improvements in early-stage lung and pancreatic cancers.
Collapse
Affiliation(s)
- Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xinying Xue
- Department of Respiratory and Critical Care Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Dongfeng Qu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, 73103, OK, USA
- Stephenson Cancer Center, Oklahoma City, 73104, OK, USA
| | - C Benedikt Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, Ludwig Maximilian University of Munich, 81377, Munich, Germany
| | - Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Liyang Zhang
- Xiangya Hospital, Central South University, Changsha 410008, China
| | - Manyu Li
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Tianbo Gao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Parthasarathy Chandrakesan
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, 73103, OK, USA
- Stephenson Cancer Center, Oklahoma City, 73104, OK, USA
| | - Kenneth J Vega
- Division of Gastroenterology and Hepatology, Augusta University, Augusta, 30912, GA, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou 350122, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou 350122, China
| |
Collapse
|
6
|
Kim HC, Choi CM. Current Status of Immunotherapy for Lung Cancer and Future Perspectives. Tuberc Respir Dis (Seoul) 2020; 83:14-19. [PMID: 31905428 PMCID: PMC6953488 DOI: 10.4046/trd.2019.0039] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/02/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Lung cancer remains the most common cause of cancer-related deaths worldwide. Although there are many possible treatments, including targeted therapies such as epidermal growth factor receptor tyrosine kinase inhibitors and anaplastic lymphoma kinase inhibitors, new therapeutic strategies are needed to improve clinical outcomes. Immunotherapy through the use of immune checkpoint inhibitors has provided one of the most important breakthroughs in the management of solid tumors, including lung cancers, and has shown promising results in numerous clinical trials. This review will present the current status of immunotherapy for lung cancer and future perspectives on these treatments.
Collapse
Affiliation(s)
- Ho Cheol Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chang Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
7
|
Hou Y, Guo H, Guo Y, Zhang Y, Han H. [Preliminary Study on the Biological Markers for I-IIb Stage Non-small Cell Lung Cancer Based on a Serum-peptidomics]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:20-25. [PMID: 30674389 PMCID: PMC6348161 DOI: 10.3779/j.issn.1009-3419.2019.01.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
背景与目的 非小细胞肺癌是发病率最高的一种肺恶性肿瘤,早发现、早诊断、早治疗是其治疗原则,因其发病隐匿,缺乏早期筛查的特异性标志物,多数患者就诊时已处于晚期阶段,导致其5年生存率较低,预后不理想。探索一种敏感的生物学标志物,是目前肺癌诊疗的重点。本文基于差异多肽组学技术,旨在筛选Ⅰ期-Ⅱb期非小细胞肺癌的血清生物标志物。 方法 利用纳升超高效液相色谱联合四级杆轨道阱质谱技术,比较和筛选正常健康人群、肺部良性病变患者、非小细胞肺癌患者血清中的差异多肽片段,通过对目标肽段的鉴别和定量分析,探寻非小细胞肺癌早期诊断的肿瘤生物标志物。 结果 通过与人类蛋白质组数据库进行比对,共鉴定到545个多肽,分别来自118个蛋白。将各组样本中肽段的谱图数进行比较,共筛选出201个差异多肽,经定量分析发现峰面积的变异系数(coefficient of variation, CV)≤30%的目标肽段共计7个,其中在各组间呈趋势变化的有2条肽段,分别是肺癌组中表达下调的肽段QGAKIPKPEASFSPR和肺癌组中表达上调的肽段CDDYRLC,它们分别来自于中间α球蛋白抑制因子H4蛋白(ITIH4)和基质γ-羧基谷氨酸蛋白(MGP)。 结论 将血清多肽组学用于筛选肿瘤标志物的研究,可为非小细胞肺癌的早期诊断提供新线索,其中ITIH4与MGP蛋白水解的特异性肽段是潜在早期非小细胞肺癌患者的血清标志物。
Collapse
Affiliation(s)
- Yuelong Hou
- Department of Thoracic Surgery, Third Central Hospital of Tianjin; Tianjin Institute of Hepatobiliary Disease; Tianjin Key Laboratory of Artificial Cell; Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Hongqi Guo
- Department of Thoracic Surgery, Third Central Hospital of Tianjin; Tianjin Institute of Hepatobiliary Disease; Tianjin Key Laboratory of Artificial Cell; Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Yongkuan Guo
- Department of Thoracic Surgery, Third Central Hospital of Tianjin; Tianjin Institute of Hepatobiliary Disease; Tianjin Key Laboratory of Artificial Cell; Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Yukun Zhang
- Department of Thoracic Surgery, Third Central Hospital of Tianjin; Tianjin Institute of Hepatobiliary Disease; Tianjin Key Laboratory of Artificial Cell; Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Hongli Han
- Department of Thoracic Surgery, Third Central Hospital of Tianjin; Tianjin Institute of Hepatobiliary Disease; Tianjin Key Laboratory of Artificial Cell; Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| |
Collapse
|