1
|
Yang J, Wu B, Li G, Zhang C, Xie Y, Kong W, Zeng Z. Landscape of epithelial cell subpopulations in the human esophageal squamous cell carcinoma microenvironment. Heliyon 2024; 10:e38091. [PMID: 39391485 PMCID: PMC11466536 DOI: 10.1016/j.heliyon.2024.e38091] [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: 07/09/2024] [Revised: 08/19/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Aims We sought to reveal the landscape of epithelial cell subpopulations in the human esophageal squamous cell carcinoma microenvironment and investigate their parts on esophageal squamous carcinoma (ESCC) development. Background Epithelial cells play an important role in the occurrence and development of ESCC through multiple mechanisms. While the landscape of epithelial cell subpopulations in ESCC, remains unclear. Objective Exploring the role of epithelial cell subpopulations in ESCC progression. Methods Seurat R package was used for single-cell RNA sequencing (scRNA-seq) data filtering, dimensionality reduction, clustering and differentially expressed genes analysis. Cellmarker database was adopted for cell cluster annotation. Functional enrichment analysis was carried out by Gene Ontology (GO) analysis. InferCNV package was conducted for copy number variation (CNV) of epithelial cell subpopulations in all chromosomal regions. Pseudotime trajectory analysis was implemented for exploring differentiation trajectory of epithelial cells subgroups during the cancer progression. CellChat analysis was used for probing the interactions between epithelial cells and NK/T cells. cellular experiments were performed using Quantitative Real-Time Polymerase Chain Reaction (RT-qPCR), Wound-Healing Assay and transwell. Results 11 major cell subpopulations were identified in ESCC and adjunct tissues. Further reclassification of epithelial cells uncovered 4 subpopulations. Enrichment analysis revealed that highly expressed genes in 4 epithelial cell subpopulations were related to cell proliferation, immune response and angiogenesis. CNV analysis found that UBD + epithelial cells and GAS2L3+ epithelial cells had a higher proportion of CNV. Cell differentiation trajectories disclosed that KRT6C+ and GSTA1+ epithelial cells were in an intermediate state of differentiation, while UBD+ and GAS2L3+ epithelial cells are in an end state of differentiation during ESCC progression. Finally, we found that four epithelial cell subpopulations all inhibited NK/T cells through NECTIN2-TIGIT and CLEC2B-KLRB1. Low ATF3 and DDIT3 mRNA expression inhibited ESCC cell migration and invasion. Conclusion Here, we obtained a through epithelial cell atlas of ESCC at single-cell resolution, explored the role of epithelial cell in ESCC progression, and unveiled immunosuppressive signals to NK/T cells in promoting ESCC. Our findings expand the comprehension of epithelial cells and offer a theoretical guidance for future anti-epithelial cell treatment of ESCC.
Collapse
Affiliation(s)
- Jingrong Yang
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Bo Wu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
- Department of Emergency, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
| | - Guo Li
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Chenxi Zhang
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Yongwei Xie
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Wencui Kong
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
| | - Zhiyong Zeng
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| |
Collapse
|
2
|
Liu K, Han H, Xiong K, Zhai S, Yang X, Yu X, Chen B, Liu M, Dong Q, Meng H, Gu Y. Single-cell landscape of intratumoral heterogeneity and tumor microenvironment remolding in pre-nodal metastases of breast cancer. J Transl Med 2024; 22:804. [PMID: 39210391 PMCID: PMC11363495 DOI: 10.1186/s12967-024-05625-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The metastasis of cancer cells is influenced by both their intrinsic characteristics and the tumor microenvironment (TME). However, the molecular mechanisms underlying pre-nodal metastases of breast cancer remain unclear. METHODS We integrated a total of 216,963 cells from 54 samples across 6 single-cell datasets to profile the cellular landscape differences between primary tumors and pre-nodal metastases. RESULTS We revealed three distinct metastatic epithelial cell subtypes (Epi1, Epi2 and Epi3), which exhibited different metastatic mechanisms. Specifically, the marker gene KCNK15 of the Epi1 subtype exhibited increased gene expression along the cell differentiation trajectory and was specifically regulated by the transcription factor ASCL1. In the Epi3 subtype, we highlighted NR2F1 as a regulator targeting the marker gene MUCL1. Additionally, we found that the Epi2 and Epi3 subtypes shared some regulons, such as ZEB1 and NR2C1. Similarly, we identified specific subtypes of stromal and immune cells in the TME, and discovered that vascular cancer-associated fibroblasts might promote capillary formation through CXCL9+ macrophages in pre-nodal metastases. All three subtypes of metastatic epithelial cells were associated with poor prognosis. CONCLUSIONS In summary, this study dissects the intratumoral heterogeneity and remodeling of the TME in pre-nodal metastases of breast cancer, providing novel insights into the mechanisms underlying breast cancer metastasis.
Collapse
Affiliation(s)
- Kaidong Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Huiming Han
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kai Xiong
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Songmei Zhai
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiuqi Yang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinmiao Yu
- Department of Human Anatomy, Harbin Medical University, Harbin, China
| | - Bo Chen
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingyue Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Qi Dong
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Li J, Sun Y, Zhi X, Sun Y, Abudousalamu Z, Lin Q, Li B, Yao L, Chen M. Unraveling the molecular mechanisms of lymph node metastasis in ovarian cancer: focus on MEOX1. J Ovarian Res 2024; 17:61. [PMID: 38486335 PMCID: PMC10938838 DOI: 10.1186/s13048-024-01384-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) is a major factor contributing to the high mortality rate of ovarian cancer, making the treatment of this disease challenging. However, the molecular mechanism underlying LNM in ovarian cancer is still not well understood, posing a significant obstacle to overcome. RESULTS Through data mining from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, we have identified MEOX1 as a specific gene associated with LNM in ovarian cancer. The expression of MEOX1 was found to be relatively high in serous ovarian adenocarcinoma, and its higher expression were associated with increased tumor grade and poorer clinical prognosis for ovarian cancer patients. Bioinformatics analysis revealed that MEOX1 exhibited the highest mRNA levels among all cancer types in ovarian cancer tissues and cell lines. Furthermore, gene set enrichment analysis (GSEA) and pathway analysis demonstrated that MEOX1 was involved in various LNM-related biological activities, such as lymphangiogenesis, lymphatic vessel formation during metastasis, epithelial-mesenchymal transition (EMT), G2/M checkpoint, degradation of extracellular matrix, and collagen formation. Additionally, the expression of MEOX1 was positively correlated with the expression of numerous prolymphangiogenic factors in ovarian cancer. To validate our findings, we conducted experiments using clinical tissue specimens and cell lines, which confirmed that MEOX1 was highly expressed in high-grade serous ovarian cancer (HGSOC) tissues and various ovarian cancer cell lines (A2780, SKOV3, HO8910, and OVCAR5) compared to normal ovarian tissues and normal ovarian epithelial cell line IOSE-80, respectively. Notably, we observed a higher protein level of MEOX1 in tumor tissues of LNM-positive HGSOC compared to LNM-negative HGSOC. Moreover, our fundamental experiments demonstrated that suppression of MEOX1 led to inhibitory effects on ovarian cancer cell proliferation and EMT, while overexpression of MEOX1 enhanced the proliferation and EMT capacities of ovarian cancer cells. CONCLUSIONS The results of our study indicate that MEOX1 plays a role in the lymph node metastasis of ovarian cancer by regulating multiple biological activities, including the proliferation and EMT of ovarian cancer, lymphangiogenesis, and ECM remodeling. Our findings suggest that MEOX1 could serve as a potential biomarker for the diagnosis and treatment of ovarian cancer with LNM.
Collapse
Affiliation(s)
- Jiajia Li
- Department of Gynecology Oncology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Yihua Sun
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yating Sun
- Department of Gynecology Oncology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Zulimire Abudousalamu
- Department of Gynecology Oncology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Qianhan Lin
- Department of Gynecology Oncology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Bin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Liangqing Yao
- Department of Gynecology Oncology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| | - Mo Chen
- Department of Gynecology Oncology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
4
|
Seferbekova Z, Lomakin A, Yates LR, Gerstung M. Spatial biology of cancer evolution. Nat Rev Genet 2022; 24:295-313. [PMID: 36494509 DOI: 10.1038/s41576-022-00553-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Abstract
The natural history of cancers can be understood through the lens of evolution given that the driving forces of cancer development are mutation and selection of fitter clones. Cancer growth and progression are spatial processes that involve the breakdown of normal tissue organization, invasion and metastasis. For these reasons, spatial patterns are an integral part of histological tumour grading and staging as they measure the progression from normal to malignant disease. Furthermore, tumour cells are part of an ecosystem of tumour cells and their surrounding tumour microenvironment. A range of new spatial genomic, transcriptomic and proteomic technologies offers new avenues for the study of cancer evolution with great molecular and spatial detail. These methods enable precise characterizations of the tumour microenvironment, cellular interactions therein and micro-anatomical structures. In conjunction with spatial genomics, it emerges that tumours and microenvironments co-evolve, which helps explain observable patterns of heterogeneity and offers new routes for therapeutic interventions.
Collapse
|
5
|
Aurora Kinase A and Bcl-xL Inhibition Suppresses Metastasis in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms231710053. [PMID: 36077449 PMCID: PMC9456092 DOI: 10.3390/ijms231710053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 12/02/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease that accounts for 10–15% of all breast cancer cases. Within TNBC, the treatment of basal B is the most challenging due to its highly invasive potential, and thus treatments to suppress metastasis formation in this subgroup are urgently needed. However, the mechanisms underlying the metastatic ability of TNBC remain unclear. In the present study, we investigated the role of Aurora A and Bcl-xL in regulating basal B cell invasion. We found gene amplification and elevated protein expression in the basal B cells, which also showed increased invasiveness in vitro, compared to basal A cells. Chemical inhibition of Aurora A with alisertib and siRNA-mediated knockdown of BCL2L1 decreased the number of invading cells compared to non-treated cells in basal B cell lines. The analysis of the correlation between AURKA and BCL2L1 expression in TNBC and patient survival revealed significantly decreased relapse-free survival (n = 534, p = 0.012) and distant metastasis-free survival (n = 424, p = 0.017) in patients with primary tumors exhibiting a high combined expression of AURKA and BCL2L1. Together, our findings suggest that high levels of Aurora A and Bcl-xL promote metastasis, and inhibition of these proteins may suppress metastasis and improve patient survival in basal B TNBC.
Collapse
|
6
|
Muhammad A, Forcados GE, Katsayal BS, Bako RS, Aminu S, Sadiq IZ, Abubakar MB, Yusuf AP, Malami I, Faruk M, Ibrahim S, Pase PA, Ahmed S, Abubakar IB, Abubakar M, Yates C. Potential epigenetic modifications implicated in triple- to quadruple-negative breast cancer transition: a review. Epigenomics 2022; 14:711-726. [PMID: 35473304 DOI: 10.2217/epi-2022-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Current research on triple-negative breast cancer (TNBC) has resulted in delineation into the quadruple-negative breast cancer (QNBC) subgroup. Epigenetic modifications such as DNA methylation, histone posttranslational modifications and associated changes in chromatin architecture have been implicated in breast cancer pathogenesis. Herein, the authors highlight genes with observed epigenetic modifications that are associated with more aggressive TNBC/QNBC pathogenesis and possible interventions. Advanced literature searches were done on PubMed/MEDLINE, Scopus and Google Scholar. The results suggest that nine epigenetically altered genes/differentially expressed proteins in addition to the downregulated androgen receptor are associated with TNBC aggressiveness and could be implicated in the TNBC to QNBC transition. Thus, restoring the normal expression of these genes via epigenetic reprogramming could be therapeutically beneficial to TNBC and QNBC patients.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria.,Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | | | - Babangida Sanusi Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Rabiatu Suleiman Bako
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Suleiman Aminu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Murtala Bello Abubakar
- Department of Physiology, Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Sokoto State, Nigeria.,Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Sokoto State, Nigeria
| | | | - Ibrahim Malami
- Department of Pharmacognosy & Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Nigeria.,Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Sokoto State, Nigeria
| | - Mohammed Faruk
- Department of Pathology, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Sani Ibrahim
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Peter Abur Pase
- Department of Surgery, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Saad Ahmed
- Department of Pathology, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Ibrahim Babangida Abubakar
- Deparment of Biochemistry, Kebbi State University of Science & Technology, PMB 1144, Aliero, Kebbi State, Nigeria
| | - Murtala Abubakar
- Department of Pathology, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Clayton Yates
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
7
|
Li X, Dowling EK, Yan G, Dereli Z, Bozorgui B, Imanirad P, Elnaggar JH, Luna A, Menter DG, Pilié PG, Yap TA, Kopetz S, Sander C, Korkut A. Precision combination therapies based on recurrent oncogenic co-alterations. Cancer Discov 2022; 12:1542-1559. [PMID: 35412613 PMCID: PMC9524464 DOI: 10.1158/2159-8290.cd-21-0832] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/28/2021] [Accepted: 03/23/2022] [Indexed: 11/16/2022]
Abstract
Cancer cells depend on multiple driver alterations whose oncogenic effects can be suppressed by drug combinations. Here, we provide a comprehensive resource of precision combination therapies tailored to oncogenic co-alterations that are recurrent across patient cohorts. To generate the resource, we developed Recurrent Features Leveraged for Combination Therapy (REFLECT), which integrates machine learning and cancer informatics algorithms. Using multi-omic data, the method maps recurrent co-alteration signatures in patient cohorts to combination therapies. We validated the REFLECT pipeline using data from patient-derived xenografts, in vitro drug screens, and a combination therapy clinical trial. These validations demonstrate that REFLECT-selected combination therapies have significantly improved efficacy, synergy, and survival outcomes. In patient cohorts with immunotherapy response markers, DNA repair aberrations, and HER2 activation, we have identified therapeutically actionable and recurrent co-alteration signatures. REFLECT provides a resource and framework to design combination therapies tailored to tumor cohorts in data-driven clinical trials and pre-clinical studies.
Collapse
Affiliation(s)
- Xubin Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Gonghong Yan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zeynep Dereli
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Behnaz Bozorgui
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Parisa Imanirad
- Department of Systems Biology, and The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jacob H. Elnaggar
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Augustin Luna
- cBio Center, Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - David G. Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Patrick G. Pilié
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy A. Yap
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chris Sander
- cBio Center, Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Corresponding Author: Anil Korkut, Bioinformatics & Comp Biology, Phone: 718-300-0666, , 1515 Holcombe Blvd., Houston, Texas 77030-4009
| |
Collapse
|
8
|
Clinical and genomic analyses of neuroendocrine neoplasms of the breast. Mod Pathol 2022; 35:495-505. [PMID: 34728787 DOI: 10.1038/s41379-021-00965-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023]
Abstract
Breast neuroendocrine neoplasms (NENs) constitute a rare histologic subtype that includes both neuroendocrine tumors (NETs) and neuroendocrine carcinomas (NECs). In this study, we aimed to gain insight into the clinical and molecular characteristics of NENs of the breast. NEN and paired distant normal fresh tissues and clinicopathological data were obtained from 17 patients with NENs, and clinicopathological data were collected from 755 patients with invasive breast carcinomas of no special type (IBCs-NST). We compared the clinicopathological characteristics of NENs and IBCs-NST and performed whole-exome sequencing (WES) of both NEN and paired normal tissues. Compared with the IBC-NST patients, the NEN patients had a higher mean age, lower clinical stage, and lower pathological nodal (pN) stage (P < 0.001, P < 0.001, and P = 0.017, respectively). The most frequently mutated gene in NENs was KMT2C (3/17, 17.6%). NENs had copy number variations (CNVs) of 8q, 11q, and 17q amplification and 17q and 11q deletion and harbored the following specific genes related to tumorigenesis: (i) suppressor genes with loss of heterozygosity (LOH) such as ACE (2/17, 11.8%); (ii) tumor driver genes such as GATA3 (2/17, 11.8%); and (iii) susceptibility genes such as MAP3K4 (17/17, 100%) and PDE4DIP (17/17, 100%). The oncogenic/likely oncogenic mutations of NETs in PI3K pathway genes (50.0%, 18.2%; P < 0.001) and MAPK signaling pathway genes (83.3%, 18.2%; P = 0.035) affected higher proportions than those of NECs. In conclusion, this study provides certain clinical and molecular evidence supporting NENs as a distinct subtype of breast cancer and provides some potential molecular features for distinguishing NETs from NECs.
Collapse
|
9
|
Zhu Z, Wang W, Lin F, Jordan T, Li G, Silverman S, Qiu S, Joy AA, Chen C, Hockley DL, Zhang X, Zhou Q, Postovit LM, Zhang X, Hou Y, Mackey JR, Li B, Wong GKS. Genome profiles of pathologist-defined cell clusters by multiregional LCM and G&T-seq in one triple-negative breast cancer patient. CELL REPORTS MEDICINE 2021; 2:100404. [PMID: 34755126 PMCID: PMC8561166 DOI: 10.1016/j.xcrm.2021.100404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/30/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023]
Abstract
Pathological examination is the gold standard for cancer diagnosis, and breast tumor cells are often found in clusters. We report a case study on one triple-negative breast cancer (TNBC) patient, analyzing tumor development, metastasis, and prognosis with simultaneous DNA and RNA sequencing of pathologist-defined cell clusters from multiregional frozen sections. The cell clusters are isolated by laser capture microdissection (LCM) from primary tumor tissue, lymphatic vessels, and axillary lymph nodes. Data are reported for a total of 97 cell clusters. A combination of tumor cell-cluster clonality and phylogeny reveals 3 evolutionarily distinct pathways for this patient, each associated with a unique mRNA signature, and each correlated with disparate survival outcomes. Hub gene analysis indicates that extensive downregulation of ribosomal protein mRNA is a potential marker of poor prognosis in breast cancer. Pathologically diverse cell clusters share genomic and transcriptomic profiles Transcriptome-defined clones are more complex than genome-defined clones Three distinct pathways were inferred, each with disparate survival outcomes Lower expression of ribosomal proteins may be an indicator of poor prognosis
Collapse
Affiliation(s)
- Zhongyi Zhu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiwei Wang
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada.,Geneis, Bldg A, 5 Guangshun North Street, Beijing 100102, China
| | - Feng Lin
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Tracy Jordan
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Guibo Li
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Sveta Silverman
- Department of Pathology and Laboratory Medicine, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Si Qiu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Anil Abraham Joy
- Division of Medical Oncology, Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada
| | - Chao Chen
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Deanna L Hockley
- Division of Medical Oncology, Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada
| | - Xi Zhang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Qing Zhou
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Lynne M Postovit
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Xiuqing Zhang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Yong Hou
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - John R Mackey
- Division of Medical Oncology, Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada
| | - Bo Li
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Gane Ka-Shu Wong
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| |
Collapse
|
10
|
Single-cell RNA sequencing reveals cell heterogeneity and transcriptome profile of breast cancer lymph node metastasis. Oncogenesis 2021; 10:66. [PMID: 34611125 PMCID: PMC8492772 DOI: 10.1038/s41389-021-00355-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Molecular mechanisms underlying breast cancer lymph node metastasis remain unclear. Using single-cell sequencing, we investigated the transcriptome profile of 96,796 single cells from 15 paired samples of primary tumors and axillary lymph nodes. We identified nine cancer cell subclusters including CD44 + / ALDH2 + /ALDH6A1 + breast cancer stem cells (BCSCs), which had a copy-number variants profile similar to that of normal breast tissue. Importantly, BCSCs existed only in primary tumors and evolved into metastatic clusters infiltrating into lymph nodes. Furthermore, transcriptome data suggested that NECTIN2-TIGIT-mediated interactions between metastatic breast cancer cells and tumor microenvironment (TME) cells, which promoted immune escape and lymph node metastasis. This study is the first to delineate the transcriptome profile of breast cancer lymph node metastasis using single-cell RNA sequencing. Our findings offer novel insights into the mechanisms underlying breast cancer metastasis and have implications in developing novel therapies to inhibit the initiation of breast cancer metastasis.
Collapse
|
11
|
Wu Q, Zhang W, Wang Y, Min Q, Zhang H, Dong D, Zhan Q. MAGE-C3 promotes cancer metastasis by inducing epithelial-mesenchymal transition and immunosuppression in esophageal squamous cell carcinoma. Cancer Commun (Lond) 2021; 41:1354-1372. [PMID: 34347390 PMCID: PMC8696229 DOI: 10.1002/cac2.12203] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 01/10/2023] Open
Abstract
Background Evading immune surveillance is necessary for tumor metastasis. Thus, there is an urgent need to better understand the interaction between metastasis and mechanisms of tumor immune evasion. In this study, we aimed to clarify a novel mechanism that link tumor metastasis and immunosuppression in the development of esophageal squamous cell carcinoma (ESCC). Methods The expression of melanoma‐associated antigen C3 (MAGE‐C3) was detected using immunohistochemistry. Transwell assays were used to evaluate the migration and invasion ability of esophageal squamous cell carcinoma (ESCC) cells. Metastasis assays in mice were used to evaluate metastatic ability in vivo. Lymphocyte‐mediated cytotoxicity assays were performed to visualize the immune suppression function on tumor cells. RNA sequencing was performed to identify differentially expressed genes between MAGE‐C3 overexpressing ESCC cells and control cells. Gene ontology (GO) enrichment analyses was performed to identify the most altered pathways influenced by MAGE‐C3. The activation of the interferon‐γ (IFN‐γ) pathway was analyzed using Western blotting, GAS luciferase reporter assays, immunofluorescence, and flow cytometry. The role of MAGE‐C3 in the IFN‐γ pathway was determined by Western blotting and immunoprecipitation. Furthermore, immunohistochemistry and flow cytometry analysis monitored the changes of infiltrated T cell populations in murine lung metastases. Results MAGE‐C3 was overexpressed in ESCC tissues. High expression of MAGE‐C3 had a significant association with the risk of lymphatic metastasis and poor survival in patients with ESCC. Functional experiments revealed that MAGE‐C3 promoted tumor metastasis by activating the epithelial‐mesenchymal transition (EMT). MAGE‐C3 repressed antitumor immunity and regulated cytokine secretion of T cells, implying an immunosuppressive function. Mechanistically, MAGE‐C3 facilitated IFN‐γ signaling and upregulated programmed cell death ligand 1 (PDL1) by binding with IFN‐γ receptor 1 (IFNGR1) and strengthening the interaction between IFNGR1 and signal transducer and activator of transcription 1 (STAT1). Interestingly, MAGE‐C3 displayed higher tumorigenesis in immune‐competent mice than in immune‐deficient nude mice, confirming the immunosuppressive role of MAGE‐C3. Furthermore, mice bearing MAGE‐C3‐overexpressing tumors showed worse survival and more lung metastases with decreased CD8+ infiltrated T cells and increased programmed cell death 1 (PD‐1)+CD8+ infiltrated T cells. Conclusion MAGE‐C3 enhances tumor metastasis through promoting EMT and protecting tumors from immune surveillance, and could be a potential prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China.,Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100142, P. R. China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China.,Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100142, P. R. China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Hongyue Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Dezuo Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China.,Shenzhen Bay Laboratory, Institute of Cancer Research, Shenzhen, Guangdong, 518107, P. R. China.,Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100142, P. R. China
| |
Collapse
|
12
|
Chen SC, Yu CC, Chang HK, Lin YC, Lo YF, Shen SC, Kuo WL, Tsai HP, Chou HH, Chu CH, Shen WC, Wu RC, Ueng SH, Huang YT. Discrepancy of Breast and Axillary Pathologic Complete Response and Outcomes in Different Subtypes of Node-positive Breast Cancer after Neoadjuvant Chemotherapy. J Cancer 2021; 12:5365-5374. [PMID: 34335953 PMCID: PMC8317533 DOI: 10.7150/jca.62830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/24/2021] [Indexed: 11/05/2022] Open
Abstract
Few studies have analyzed the discrepancy between breast pathologic complete response (B-pCR) and axillary node pCR (N-pCR) rates and their impact on survival outcomes in different intrinsic subtypes of early breast cancer after neoadjuvant chemotherapy (NAC). We retrospectively reviewed B-pCR, N-pCR, and total (breast and axillary node) pCR (T-pCR) after NAC to assess the discrepancy and outcomes between 2005 and 2017. A total of 968 patients diagnosed with cT1-4c, N1-2, and M0 breast cancer were enrolled in the study. The median age was 49 years and the median follow-up time was 45 months. Of these patients, 213 achieved T-pCR, 31 achieved B-pCR with axillary node pathologic non-complete response (N-non pCR), 245 achieved N-pCR with breast pathologic non-complete response (B-non pCR), and 479 achieved total (breast and axillary node) pathologic non-complete response (T-non pCR) after NAC. The highest B-pCR and N-pCR rates were found in the hormone receptor-negative, human epidermal growth factor receptor 2-positive HR(-)HER2(+) subtype, while the lowest B-pCR rate was found in the HR(+)HER2(-) subtype. The N-pCR rate was correlated to the B-pCR rate (P<0.001), but was higher than the B-pCR rate in all subtypes. The 5-year overall survival (OS) rates for patients with T-pCR, B-pCR, and N-pCR were 91.2%, 91.7%, and 91.9%, respectively. For non-pCR, non-pCR, and non-pCR, the 5-year OS rates were 73.6%, 78.9%, and 74.7%, respectively (P<0.0001). B-non pCR patients had a lower risk of recurrence than T-non pCR or N-non-pCR patients, although there were no differences in OS among them. In conclusion, the N-pCR rate was higher than the B-pCR rate after NAC in all intrinsic subtypes, and N-non pCR or T-non pCR patients had the worst outcomes.
Collapse
Affiliation(s)
- Shin-Cheh Chen
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chi-Chang Yu
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hsien-Kun Chang
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Chang Lin
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Feng Lo
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shih-Che Shen
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Lin Kuo
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hsiu-Pei Tsai
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hsu-Huan Chou
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chia-Hui Chu
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Chi Shen
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ren-Chin Wu
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shir-Hwa Ueng
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Ting Huang
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University Medical College, Taoyuan, Taiwan
| |
Collapse
|
13
|
Pariyar M, Johns A, Thorne RF, Scott RJ, Avery-Kiejda KA. Copy number variation in triple negative breast cancer samples associated with lymph node metastasis. Neoplasia 2021; 23:743-753. [PMID: 34225099 PMCID: PMC8259224 DOI: 10.1016/j.neo.2021.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a highly metastatic and aggressive subtype of breast cancer and cases presenting with lymph node involvement have worse outcomes. This study aimed to determine the regions of copy number variation (CNV) associated with lymph node metastasis in TNBC patients. CNV analyses were performed in a study cohort of 23 invasive ductal carcinomas (IDCs), 12 lymph node metastases (LNmets), and 7 normal adjacent tissues (NATs); as well as in an independent cohort containing 70 TNBC IDCs and the same 7 NATs. CNV-associated genes were analyzed using GO-enrichment and Pathway analysis. The prognostic role for genes showing CNV-based changes in messenger RNA expression was determined using the Kaplan-Meier plotter database. For the IDCs, there were a number of variations that were common in both the study and independent cohorts in the amplified regions of 1q, 8q, 19 (p and q), 2p, 5p and the deleted regions in 8p followed by 5q, and 19p. The most frequently amplified regions in the LNmets of the study cohort were 4q28.3, 2p, 3q24, 1q21.2, 10p, 12p11.1, 8q, 20p11.22-20p11.21, 21q22.13, 6p22.1 and the most frequently deleted regions were in 1p36.23, 4q21.1 and 5q. A total of 686 (441 amplified and 245 deleted) genes were associated with LNmets. The LNmet-associated genes were highly enriched for “regulation of complement activation,” “regulation of protein activation cascade,” “regulation of humoral immune response,” “oxytocin signalling pathway,” and “TRAIL binding” pathways. Moreover, 6/686 LNmet-associated genes showed CNV-based changes in their mRNA expression of which, high expression of ASPM and KIF14 was significantly associated with worse relapse-free survival. This study has identified several CNV regions in TNBC that could play a major role in metastasis to the lymph node.
Collapse
Affiliation(s)
- Mamta Pariyar
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Andrea Johns
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Rick F Thorne
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Kelly A Avery-Kiejda
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
14
|
Wu SL, Zhang X, Chang M, Huang C, Qian J, Li Q, Yuan F, Sun L, Yu X, Cui X, Jiang J, Cui M, Liu Y, Wu HW, Liang ZY, Wang X, Niu Y, Tong WM, Jin F. Genome-wide 5-hydroxymethylcytosine Profiling Analysis Identifies MAP7D1 as A Novel Regulator of Lymph Node Metastasis in Breast Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:64-79. [PMID: 33716151 PMCID: PMC8498923 DOI: 10.1016/j.gpb.2019.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/07/2019] [Accepted: 05/31/2019] [Indexed: 11/28/2022]
Abstract
Although DNA 5-hydroxymethylcytosine (5hmC) is recognized as an important epigenetic mark in cancer, its precise role in lymph node metastasis remains elusive. In this study, we investigated how 5hmC associates with lymph node metastasis in breast cancer. Accompanying with high expression of TET1 and TET2 proteins, large numbers of genes in the metastasis-positive primary tumors exhibit higher 5hmC levels than those in the metastasis-negative primary tumors. In contrast, the TET protein expression and DNA 5hmC decrease significantly within the metastatic lesions in the lymph nodes compared to those in their matched primary tumors. Through genome-wide analysis of 8 sets of primary tumors, we identified 100 high-confidence metastasis-associated 5hmC signatures, and it is found that increased levels of DNA 5hmC and gene expression of MAP7D1 associate with high risk of lymph node metastasis. Furthermore, we demonstrate that MAP7D1, regulated by TET1, promotes tumor growth and metastasis. In conclusion, the dynamic 5hmC profiles during lymph node metastasis suggest a link between DNA 5hmC and lymph node metastasis. Meanwhile, the role of MAP7D1 in breast cancer progression suggests that the metastasis-associated 5hmC signatures are potential biomarkers to predict the risk for lymph node metastasis, which may serve as diagnostic and therapeutic targets for metastatic breast cancer.
Collapse
Affiliation(s)
- Shuang-Ling Wu
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China; Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xiaoyi Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Center for Bioinformatics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Mengqi Chang
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Changcai Huang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Center for Bioinformatics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Jun Qian
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Center for Bioinformatics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Qing Li
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Fang Yuan
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry, Peking University, Beijing 100871, China
| | - Lihong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College. Beijing 100005, China
| | - Xinmiao Yu
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China
| | - Xinmiao Cui
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China
| | - Jiayi Jiang
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China
| | - Mengyao Cui
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China
| | - Ye Liu
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China
| | - Huan-Wen Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences. Beijing 100005, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences. Beijing 100005, China
| | - Xiaoyue Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Center for Bioinformatics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yamei Niu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China.
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Molecular Pathology Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China; Center for Experimental Animal Research, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College. Beijing 100005, China.
| | - Feng Jin
- Department of Surgical Oncology and Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
15
|
Tang YJ, Huang J, Tsushima H, Ban GI, Zhang H, Oristian KM, Puviindran V, Williams N, Ding X, Ou J, Jung SH, Lee CL, Jiao Y, Chen BJ, Kirsch DG, Alman BA. Tracing Tumor Evolution in Sarcoma Reveals Clonal Origin of Advanced Metastasis. Cell Rep 2020; 28:2837-2850.e5. [PMID: 31509746 PMCID: PMC6750751 DOI: 10.1016/j.celrep.2019.08.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/17/2019] [Accepted: 08/05/2019] [Indexed: 11/03/2022] Open
Abstract
Cellular heterogeneity is frequently observed in cancer, but the biological significance of heterogeneous tumor clones is not well defined. Using multicolor reporters and CRISPR-Cas9 barcoding, we trace clonal dynamics in a mouse model of sarcoma. We show that primary tumor growth is associated with a reduction in clonal heterogeneity. Local recurrence of tumors following surgery or radiation therapy is driven by multiple clones. In contrast, advanced metastasis to the lungs is driven by clonal selection of a single metastatic clone (MC). Using RNA sequencing (RNA-seq) and in vivo assays, we identify candidate suppressors of metastasis, namely, Rasd1, Reck, and Aldh1a2. These genes are downregulated in MCs of the primary tumors prior to the formation of metastases. Overexpression of these suppressors of metastasis impair the ability of sarcoma cells to colonize the lungs. Overall, this study reveals clonal dynamics during each step of tumor progression, from initiation to growth, recurrence, and distant metastasis.
Collapse
Affiliation(s)
- Yuning J Tang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jianguo Huang
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Hidetoshi Tsushima
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Ga I Ban
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Hongyuan Zhang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Kristianne M Oristian
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Vijitha Puviindran
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Nerissa Williams
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Xiruo Ding
- Department of Biostatics and Bioinformatics, Duke University, Durham, NC, USA
| | - Jianhong Ou
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA
| | - Sin-Ho Jung
- Department of Biostatics and Bioinformatics, Duke University, Durham, NC, USA
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Yiqun Jiao
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Benny J Chen
- Duke Cancer Institute, Duke University, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA.
| | - Benjamin A Alman
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University, Durham, NC, USA; Regeneration Next Initiative, Duke University, Durham, NC, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
16
|
To B, Isaac D, Andrechek ER. Studying Lymphatic Metastasis in Breast Cancer: Current Models, Strategies, and Clinical Perspectives. J Mammary Gland Biol Neoplasia 2020; 25:191-203. [PMID: 33034778 DOI: 10.1007/s10911-020-09460-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/24/2020] [Indexed: 03/23/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women and the second most common cause of cancer-related deaths in the United States. Although early detection has significantly decreased breast cancer mortality, patients diagnosed with distant metastasis still have a very poor prognosis. The most common site that breast cancer spreads to are local lymph nodes. Therefore, the presence of lymph node metastasis remains one of most important prognostic factors in breast cancer patients. Given its significant clinical implications, increased efforts have been dedicated to better understand the molecular mechanism governing lymph node metastasis in breast cancer. The identification of lymphatic-specific biomarkers, including podoplanin and LYVE-1, has propelled the field of lymphatic metastasis forward. In addition, several animal models such as cell line-derived xenografts, patient-derived xenografts, and spontaneous tumor models have been developed to recreate the process of lymphatic metastasis. Moreover, the incorporation of various -omic platforms have provided further insight into the genetic drivers facilitating lymphatic metastasis, as well as potential biomarkers and therapeutic targets. Here, we highlight various models of lymphatic metastasis, their potential pitfalls, and other tools available to study lymphatic metastasis including imaging modalities and -omic studies.
Collapse
Affiliation(s)
- Briana To
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Daniel Isaac
- Division of Hematology and Oncology, MSU Breslin Cancer Center, Lansing, MI, USA
| | - Eran R Andrechek
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
17
|
Zhou J, Sanchez-Vega F, Caso R, Tan KS, Brandt WS, Jones GD, Yan S, Adusumilli PS, Bott M, Huang J, Isbell JM, Sihag S, Molena D, Rusch VW, Chatila WK, Rekhtman N, Yang F, Ladanyi M, Solit DB, Berger MF, Schultz N, Jones DR. Analysis of Tumor Genomic Pathway Alterations Using Broad-Panel Next-Generation Sequencing in Surgically Resected Lung Adenocarcinoma. Clin Cancer Res 2019; 25:7475-7484. [PMID: 31455678 DOI: 10.1158/1078-0432.ccr-19-1651] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/22/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE The majority of broad-panel tumor genomic profiling has used a gene-centric approach, although much of that data is unused in clinical decision making. We hypothesized that a pathway-centric approach using next-generation sequencing (NGS), combined with conventional clinicopathologic features, may better predict disease-free survival (DFS) in early stage lung adenocarcinoma. EXPERIMENTAL DESIGN Utilizing our prospectively maintained database, we analyzed 492 patients with primary, untreated, completely surgically resected lung adenocarcinoma. Ten canonical pathways were analyzed using broad-panel NGS. The correlations of DFS and number (and type) of pathway (NPA) were analyzed using the Kaplan-Meier method and log-rank test. Associations between altered pathways and clinicopathologic variables, as well as identification of actionable therapeutic strategies were explored. RESULTS Median NPA for the cohort was two (range, 0-5). Smoking status, solid morphologic appearance on preoperative CT, maximal standardized uptake value, pathologic tumor size, aggressive histologic subtype, lymphovascular invasion, visceral pleural invasion, and positive lymph nodes were significantly associated with NPA (P < 0.05). Of 543 actionable genetic alterations identified, 455 (84%) were within the RTK/RAS pathway. A total of 86 tumors had actionable therapeutic genomic alterations in >1 pathway. On multivariable analysis, higher NPA was significantly associated with worse DFS (HR, 1.31; P = 0.014). CONCLUSIONS NPA and specific pathway alterations are associated with clinicopathologic features in patients with surgically resected lung adenocarcinoma. Cell cycle, Hippo, TGFβ, and p53 pathway alterations are associated with poor DFS. Finally, NPA is an independent risk factor for poor DFS in our cohort.See related commentary by Blakely, p. 7269.
Collapse
Affiliation(s)
- Jian Zhou
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Thoracic Department, Peking University People's Hospital, Beijing, China
| | - Francisco Sanchez-Vega
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raul Caso
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kay See Tan
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Whitney S Brandt
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gregory D Jones
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shi Yan
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasad S Adusumilli
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew Bott
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James Huang
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James M Isbell
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Smita Sihag
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valerie W Rusch
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Walid K Chatila
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fan Yang
- Thoracic Department, Peking University People's Hospital, Beijing, China
| | - Marc Ladanyi
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B Solit
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R Jones
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York. .,Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
18
|
Jia B, Zhao X, Wang Y, Wang J, Wang Y, Yang Y. Prognostic roles of MAGE family members in breast cancer based on KM-Plotter Data. Oncol Lett 2019; 18:3501-3516. [PMID: 31516568 PMCID: PMC6733005 DOI: 10.3892/ol.2019.10722] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 06/19/2019] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the second leading cause of cancer-associated mortality among women worldwide, and the prevalence and mortality rates associated with this disease are high in Western countries. The melanoma-associated antigen (MAGE) family proteins are well-known tumor-specific antigens; this family includes >60 proteins that serve an important part in cell cycle withdrawal, neuronal differentiation and apoptosis. The aim of the present study was to identify a biomarker within the MAGE family that is specific for breast cancer. In the present study, the prognostic role of MAGE mRNA expression was investigated in patients with breast cancer using the Kaplan-Meier plotter database. The prognostic value of MAGE members in the different intrinsic subtypes of breast cancer was further investigated, as well as the clinicopathological features of the disease. The results of the present study indicated that patients with breast cancer that had high mRNA expression levels of MAGEA5, MAGEA8, MAGEB4 and MAGEB6 had an improved relapse-free survival, whereas those with high mRNA expression levels of MAGEB18 and MAGED4 did not. These results suggested that MAGEA5, MAGEA8, MAGEB4 and MAGEB6 may have roles as tumor suppressors in the occurrence and development of breast cancer, whereas MAGEB18 and MAGED4 may possess carcinogenic potential. MAGED2, MAGED3 and MAGEF1 had different effects depending on the type of breast cancer. In particular, high MAGEC3 mRNA expression was associated with worse RFS in lymph node-positive breast cancer, but with improved RFS in lymph node-negative breast cancer. In patients with wild-type TP53 and patients with different pathological grades of breast cancer, MAGEE2, MAGEH1 and MAGEL2 were more worthy of attention as potential prognostic factors. The results of the present study may help to elucidate the role of MAGE family members in the development of breast cancer, and may promote further research that identifies MAGE-targeting reagents for the treatment of breast cancer.
Collapse
Affiliation(s)
- Binghan Jia
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co., Ltd., Beijing 100730, P.R. China
| | - Xiaoling Zhao
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co., Ltd., Beijing 100730, P.R. China
| | - Yao Wang
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co., Ltd., Beijing 100730, P.R. China
| | - Jinlong Wang
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co., Ltd., Beijing 100730, P.R. China
| | - Yingying Wang
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co., Ltd., Beijing 100730, P.R. China
| | - Yuemei Yang
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co., Ltd., Beijing 100730, P.R. China
| |
Collapse
|
19
|
Xie G, Yang H, Ma D, Sun Y, Chen H, Hu X, Jiang YZ, Shao ZM. Integration of whole-genome sequencing and functional screening identifies a prognostic signature for lung metastasis in triple-negative breast cancer. Int J Cancer 2019; 145:2850-2860. [PMID: 30977117 DOI: 10.1002/ijc.32329] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/26/2019] [Accepted: 04/08/2019] [Indexed: 12/24/2022]
Abstract
Lung metastasis is one of the leading causes of death for triple-negative breast cancer (TNBC). We sought to characterize the genetic alterations underlying TNBC lung metastases by integrating whole-genome sequencing and functional screening. Furthermore, we aimed to develop a metastasis-related gene signature for TNBC patients to improve risk stratification. In this prospective observational study, we first conducted whole-genome sequencing of paired primary tumor and lung metastasis from one TNBC patient to identify potential genetic driver alterations. An in vivo gain-of-function screening using an amplified open reading frame library was then employed to screen candidate genes promoting lung metastasis. Finally, we applied Cox proportional hazard regression modeling to develop a prognostic gene signature from 14 candidate genes in TNBC. Compared to the primary tumor, copy number amplifications of chromosomes 3q and 8q were identified in the lung metastasis. We discovered an enrichment of 14 genes from chromosomes 3q and 8q in mouse lung metastases model. We further developed and validated a four-gene signature (ENY2, KCNK9, TNFRSF11B and KCNMB2) that predicts recurrence-free survival and lung metastasis in TNBC. Our data also demonstrated that upregulated expression of ENY2 could promote invasion and lung metastasis of TNBC cells both in vitro and in vivo. In conclusion, our study reveals functional genes with copy number amplifications among chromosome 3q and 8q in lung metastasis of TNBC. And we develop a functional gene signature that can effectively stratify patients into low- and high-risk subgroups of recurrence, helping frame personalized treatments for TNBC.
Collapse
Affiliation(s)
- Guangdong Xie
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haiyuan Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yihua Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haiquan Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin Hu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Dong W, Nicolson NG, Choi J, Barbieri AL, Kunstman JW, Abou Azar S, Knight J, Bilguvar K, Mane SM, Lifton RP, Korah R, Carling T. Clonal evolution analysis of paired anaplastic and well-differentiated thyroid carcinomas reveals shared common ancestor. Genes Chromosomes Cancer 2018; 57:645-652. [PMID: 30136351 DOI: 10.1002/gcc.22678] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/12/2018] [Accepted: 08/03/2018] [Indexed: 02/04/2023] Open
Abstract
Foci of papillary or follicular thyroid carcinoma are frequently noted in thyroidectomy specimens of anaplastic thyroid carcinoma (ATC). However, whether ATCs evolve from these co-existing well-differentiated thyroid carcinomas (WDTCs) has not been well-understood. To investigate the progression of ATC in patients with co-existing WDTCs, five ATC tumors with co-existing WDTCs and matching normal tissues were whole-exome sequenced. After mapping the somatic alteration landscape, evolutionary lineages were constructed by sub-clone analysis. Though each tumor harbored at least some unique private mutations, all five ATCs demonstrated numerous overlapping mutations with matched WDTCs. Clonal analysis further demonstrated that each ATC/WDTC pair shared a common ancestor, with some pairs diverging early in their evolution and others in which the ATC seems to arise directly from a sub-clone of the WDTC. Though the precise lineal relationship remains ambiguous, based on the genetic relationship, our study clearly suggests a shared origin of ATC and WDTC.
Collapse
Affiliation(s)
- Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Norman G Nicolson
- Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, New Haven, Connecticut
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Andrea L Barbieri
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - John W Kunstman
- Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, New Haven, Connecticut
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Sara Abou Azar
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - James Knight
- Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut
| | - Kaya Bilguvar
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut
| | - Shrikant M Mane
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Reju Korah
- Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, New Haven, Connecticut
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Tobias Carling
- Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, New Haven, Connecticut
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
21
|
Peintinger F, Reitsamer R, Smidt ML, Kühn T, Liedtke C. Lymph Nodes in Breast Cancer - What Can We Learn from Translational Research? Breast Care (Basel) 2018; 13:342-347. [PMID: 30498419 PMCID: PMC6257149 DOI: 10.1159/000492435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Clinical observations about lack of survival benefit after extensive axillary surgery and biological discordance between primary breast tumors and axillary lymph nodes raise the question of the actual metastatic potential of axillary nodal disease. The exploration of intratumoral heterogeneity and detection of genomic differences between the primary and lymph nodes indicate some similarity between the number of mutations in synchronous axillary node metastases and those in the primary lesion, suggesting a favorable prognosis. The hematogenous route of metastasis needs to be considered in findings of different subclones between nodal and distant metastases. Modern tools such as whole-genome sequencing applied in multiple tumor areas may guide more precisely the extent of axillary surgery.
Collapse
Affiliation(s)
- Florentia Peintinger
- Institute of Pathology, Medical University Graz, Graz, Austria
- Department of Gynecology and Obstetrics, General Hospital Leoben, Leoben, Austria
| | - Roland Reitsamer
- Department of Senology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Marjolein L. Smidt
- Department of Surgical Oncology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Thorsten Kühn
- Department of Gynecology and Obstetrics, Hospital Esslingen, Esslingen, Germany
| | - Cornelia Liedtke
- Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| |
Collapse
|