1
|
Liu Y, Dong K, Yao Y, Lu B, Wang L, Ji G, Zhang H, Zhao Z, Yang X, Huang R, Zhou W, Pan X, Cui X. Construction and validation of renal cell carcinoma tumor cell differentiation-related prognostic classification (RCC-TCDC): an integrated bioinformatic analysis and clinical study. Ann Med 2025; 57:2490830. [PMID: 40248945 PMCID: PMC12010653 DOI: 10.1080/07853890.2025.2490830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a heterogeneous malignancy with diverse gene expression patterns, molecular landscapes, and differentiation characteristics of tumor cells. It is imperative to develop molecular RCC classification based on tumor cell differentiation for precise risk stratification and personalized therapy. METHODS We obtained scRNA-seq profiles from GSE159115 and bulk RNA-seq profiles from TCGA-KIRC cohort. We then performed scRNA-seq cluster analysis, monocle2 pseudotime analysis, and prognostic analysis to obtain tumor cell differentiation-related prognostic genes (TCDGs). Subsequently, we conducted consensus clustering to construct the RCC tumor cell differentiation-related prognostic classification (RCC-TCDC) and implemented prognostic and multi-omics analyses. Moreover, we utilized Lasso regression to help develop a multivariable prognostic model. In addition, we performed correlation analysis and Cmap algorithm for regulatory network establishment and candidate inhibitor prediction. We eventually included 370 kidney neoplasm patients in Xinhua cohort to undergo immunohistochemical staining and scoring for classification and comprehensive statistical analyses, including Chi-square tests, Kaplan-Meier survival analyses, and multivariable Cox regression analysis . RESULTS 32 TCDGs were identifiedand RCC-TCDC was constructed to classify TCGA-KIRC patients into RCC-low differentiation (RCC-LD) (S100A11+ SH3BGRL3+, high risk), RCC-moderate differentiation (TSPAN7+, medium risk), and RCC-high differentiation (RCC-HD) (AQP1+ NPR3+, low risk). Notably, RCC-LD was validated as anindependent risk factor for both OS (p = 0.015, HR = 14.0, 95%CI = 1.67-117.8) and PFS (p = 0.010, HR = 4.0, 95%CI = 1.39-11.7) of RCC patients in Xinhua cohort, taking RCC-HD as reference. CONCLUSIONS We constructed and validated a robust molecular classification system, RCC-TCDC, elucidating three distinct RCC subtypes.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo Ji
- Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wang Zhou
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Li H, Zheng L, Zhang X, Yu X, Zhong G, Chen X, Chen X, Chen L. SH3 domain‑binding glutamic acid‑rich protein‑like 3 is associated with hyperglycemia and a poor outcome in Epstein‑Barr virus‑negative gastric carcinoma. Oncol Lett 2025; 29:8. [PMID: 39492939 PMCID: PMC11526421 DOI: 10.3892/ol.2024.14754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/03/2024] [Indexed: 11/05/2024] Open
Abstract
SH3 domain-binding glutamic acid-rich protein-like 3 (SH3BGRL3) is involved in several human cancers. However, its relationship with gastric cancer (GC) remains elusive. Multiple online bioinformatic tools were used to evaluate the messenger (m)RNA expression levels of SH3BGRL3 in GC using data from The Cancer Genome Atlas and Gene Expression Omnibus databases. Reverse transcription-quantitative PCR and tissue microarray-based immunohistochemistry were performed to assess SH3BGRL3 expression in relation to clinicopathological parameters and outcomes in patients with GC. Significant differentially expressed genes (DEGs) of SH3BGRL3 were enriched and visualized. Furthermore, associations between the expression of SH3BGRL3 and the infiltration of immune cells were explored. SH3BGRL3 exhibited aberrant expression in tumor tissues compared with adjacent normal tissues at the mRNA and protein expression levels, especially in Epstein-Barr virus-negative GC (EBVnGC). Higher SH3BGRL3 expression was significantly associated with increasing tumor-node-metastasis staging, tumor budding, perineural invasion, EGFR expression, and a notably higher preoperative blood glucose concentration in clinical specimens. Multivariate analysis revealed that higher SH3BGRL3 expression was an independent adverse prognostic factor for the overall survival of patients with EBVnGC (hazard ratio, 1.666; P=0.018). Furthermore, the stratified analysis revealed that the SH3BGRL3 phenotype could help to refine prognosis in patients. The C-index of the nomogram was 0.740 when combining SH3BGRL3 with other clinicopathological parameters, which indicated a good model for clinical follow-up decisions. Gene functional enrichment analysis also revealed that the DEGs of SH3BGRL3 were mainly enriched in regulating ATP metabolism, ATP synthesis, oxidative phosphorylation and the electron transport chain in GC. Moreover, a higher SH3BGRL3 expression was significantly positively correlated with the infiltrating macrophages in GC. In conclusion, SH3BGRL3 is upregulated in GC, particularly in EBVnGC. Higher SH3BGRL3 expression is closely associated with hyperglycemia and poor outcomes in patients with EBVnGC, suggesting its potential as a biomarker and prognostic predictor.
Collapse
Affiliation(s)
- Houqiang Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Lanqing Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Nursing Department, Fujian Provincial Hospital, Fuzhou, Fujian 35001, P.R. China
| | - Xia Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xunbin Yu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Guodong Zhong
- Department of Pathology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian 350003, P.R. China
| | - Xiaoyan Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xin Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Linying Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
3
|
Yang H, Wang H, He Y, Yang Y, Thompson EW, Xia D, Burke LJ, Cao L, Hooper JD, Roberts MS, Crawford DHG, Liang X. Identification and characterization of TM4SF1 + tumor self-seeded cells. Cell Rep 2024; 43:114512. [PMID: 39003738 DOI: 10.1016/j.celrep.2024.114512] [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: 09/11/2023] [Revised: 04/30/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
Tumor self-seeding is a process whereby circulating tumor cells (CTCs) recolonize the primary tumor, which promotes tumor growth, angiogenesis, and invasion. However, the detailed nature and functions of tumor self-seeded cells (TSCs) have not been well defined due to challenges in tracking and isolating TSCs. Here, we report an accurate animal model using photoconvertible tagging to recapitulate the spontaneous process of tumor self-seeding and identify TSCs as a subpopulation of primary tumor cells with enhanced invasiveness and survival. We demonstrate transmembrane-4-L-six-family-1 (TM4SF1) as a marker of TSCs, which promotes migration, invasion, and anchorage-independent survival in cancer cells. By analyzing single-cell RNA sequencing datasets, we identify a potential TSC population with a metastatic profile in patients with cancer, which is detectable in early-stage disease and expands during cancer progression. In summary, we establish a framework to study TSCs and identify emerging cell targets with diagnostic, prognostic, or therapeutic potential in cancers.
Collapse
Affiliation(s)
- Haotian Yang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Haolu Wang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Yaowu He
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Yang Yang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Erik W Thompson
- School of Biomedical Sciences, Queensland University of Technology and Translational Research Institute, Brisbane, QLD 4000, Australia
| | - Di Xia
- Genome Innovation Hub, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Leslie J Burke
- Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Lu Cao
- Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - John D Hooper
- Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Michael S Roberts
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Darrell H G Crawford
- Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Xiaowen Liang
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; Gallipoli Medical Research, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia.
| |
Collapse
|
4
|
Yang X, Wang Y, Rong S, An J, Lan X, Yin B, Sun Y, Wang P, Tan B, Xuan Y, Xie S, Su Z, Li Y. Gene SH3BGRL3 regulates acute myeloid leukemia progression through circRNA_0010984 based on competitive endogenous RNA mechanism. Front Cell Dev Biol 2023; 11:1173491. [PMID: 37397256 PMCID: PMC10313326 DOI: 10.3389/fcell.2023.1173491] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction: Acute myeloid leukemia (AML) is a malignant proliferative disease affecting the bone marrow hematopoietic system and has a poor long-term outcome. Exploring genes that affect the malignant proliferation of AML cells can facilitate the accurate diagnosis and treatment of AML. Studies have confirmed that circular RNA (circRNA) is positively correlated with its linear gene expression. Therefore, by exploring the effect of SH3BGRL3 on the malignant proliferation of leukemia, we further studied the role of circRNA produced by its exon cyclization in the occurrence and development of tumors. Methods: Genes with protein-coding function obtained from the TCGA database. we detected the expression of SH3BGRL3 and circRNA_0010984 by real-time quantitative polymerase chain reaction (qRT-PCR). We synthesized plasmid vectors and carried out cell experiments, including cell proliferation, cell cycle and cell differentiation by cell transfection. We also studied the transfection plasmid vector (PLVX-SHRNA2-PURO) combined with a drug (daunorubicin) to observe the therapeutic effect. The miR-375 binding site of circRNA_0010984 was queried using the circinteractome databases, and the relationship was validated by RNA immunoprecipitation and Dual-luciferase reporter assay. Finally, a protein-protein interaction network was constructed with a STRING database. GO and KEGG functional enrichment identified mRNA-related functions and signaling pathways regulated by miR-375. Results: We identified the related gene SH3BGRL3 in AML and explored the circRNA_0010984 produced by its cyclization. It has a certain effect on the disease progression. In addition, we verified the function of circRNA_0010984. We found that circSH3BGRL3 knockdown specifically inhibited the proliferation of AML cell lines and blocked the cell cycle. We then discussed the related molecular biological mechanisms. CircSH3BGRL3 acts as an endogenous sponge for miR-375 to isolate miR-375 and inhibits its activity, increases the expression of its target YAP1, and ultimately activates the Hippo signaling pathway involved in malignant tumor proliferation. Discussion: We found that SH3BGRL3 and circRNA_0010984 are important to AML. circRNA_0010984 was significantly up-regulated in AML and promoted cell proliferation by regulating miR-375 through molecular sponge action.
Collapse
Affiliation(s)
- Xiancong Yang
- Department of Clinical Laboratory, The Second Medical College of Binzhou Medical University, Yantai, China
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Yaoyao Wang
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Simin Rong
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Jiayue An
- Department of Clinical Laboratory, The Second Medical College of Binzhou Medical University, Yantai, China
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Xiaoxu Lan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Baohui Yin
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yunxiao Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Pingyu Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Boyu Tan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Ye Xuan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Shuyang Xie
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Zhenguo Su
- Department of Clinical Laboratory, The Second Medical College of Binzhou Medical University, Yantai, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| |
Collapse
|
5
|
Cao Y, Li Y, Liu R, Zhou J, Wang K. Preclinical and Basic Research Strategies for Overcoming Resistance to Targeted Therapies in HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15092568. [PMID: 37174034 PMCID: PMC10177527 DOI: 10.3390/cancers15092568] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/16/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
The amplification of epidermal growth factor receptor 2 (HER2) is associated with a poor prognosis and HER2 gene is overexpressed in approximately 15-30% of breast cancers. In HER2-positive breast cancer patients, HER2-targeted therapies improved clinical outcomes and survival rates. However, drug resistance to anti-HER2 drugs is almost unavoidable, leaving some patients with an unmet need for better prognoses. Therefore, exploring strategies to delay or revert drug resistance is urgent. In recent years, new targets and regimens have emerged continuously. This review discusses the fundamental mechanisms of drug resistance in the targeted therapies of HER2-positive breast cancer and summarizes recent research progress in this field, including preclinical and basic research studies.
Collapse
Affiliation(s)
- Yi Cao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| | - Yunjin Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| | - Ruijie Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pathology, School of Basic Medical science, Central South University, Changsha 410008, China
| |
Collapse
|
6
|
Heo H, Kim JH, Lim HJ, Kim JH, Kim M, Koh J, Im JY, Kim BK, Won M, Park JH, Shin YJ, Yun MR, Cho BC, Kim YS, Kim SY, Kim M. DNA methylome and single-cell transcriptome analyses reveal CDA as a potential druggable target for ALK inhibitor-resistant lung cancer therapy. Exp Mol Med 2022; 54:1236-1249. [PMID: 35999456 PMCID: PMC9440127 DOI: 10.1038/s12276-022-00836-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/23/2022] [Accepted: 06/20/2022] [Indexed: 11/08/2022] Open
Abstract
Acquired resistance to inhibitors of anaplastic lymphoma kinase (ALK) is a major clinical challenge for ALK fusion-positive non-small-cell lung cancer (NSCLC). In the absence of secondary ALK mutations, epigenetic reprogramming is one of the main mechanisms of drug resistance, as it leads to phenotype switching that occurs during the epithelial-to-mesenchymal transition (EMT). Although drug-induced epigenetic reprogramming is believed to alter the sensitivity of cancer cells to anticancer treatments, there is still much to learn about overcoming drug resistance. In this study, we used an in vitro model of ceritinib-resistant NSCLC and employed genome-wide DNA methylation analysis in combination with single-cell (sc) RNA-seq to identify cytidine deaminase (CDA), a pyrimidine salvage pathway enzyme, as a candidate drug target. CDA was hypomethylated and upregulated in ceritinib-resistant cells. CDA-overexpressing cells were rarely but definitively detected in the naïve cell population by scRNA-seq, and their abundance was increased in the acquired-resistance population. Knockdown of CDA had antiproliferative effects on resistant cells and reversed the EMT phenotype. Treatment with epigenome-related nucleosides such as 5-formyl-2'-deoxycytidine selectively ablated CDA-overexpressing resistant cells via accumulation of DNA damage. Collectively, our data suggest that targeting CDA metabolism using epigenome-related nucleosides represents a potential new therapeutic strategy for overcoming ALK inhibitor resistance in NSCLC.
Collapse
Affiliation(s)
- Haejeong Heo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jong-Hwan Kim
- Korea Bioinformation Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Hyun Jung Lim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jeong-Hwan Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Joo-Young Im
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Bo-Kyung Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Misun Won
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Ji-Hwan Park
- Korea Bioinformation Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Yang-Ji Shin
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Mi Ran Yun
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Byoung Chul Cho
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong Sung Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Functional Genomics Institute, PDXen Biosystems Co., Daejeon, 34129, Republic of Korea
| | - Seon-Young Kim
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
- Korea Bioinformation Center, KRIBB, Daejeon, 34141, Republic of Korea.
| | - Mirang Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|