1
|
Zhang T, Yang J, Li S, Shi X, Yang J. Ovarian squamous cell carcinoma: clinicopathological features, prognosis and immunotherapy outcomes. J Gynecol Oncol 2025; 36:36.e54. [PMID: 39791511 DOI: 10.3802/jgo.2025.36.e54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/21/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVE To explore the characteristics and survival outcomes of ovarian squamous cell carcinoma (SCC) and the treatment effectiveness of immune checkpoint inhibitors (ICIs). METHODS Patients diagnosed with ovarian SCC at Peking Union Medical College Hospital between January 2000 and September 2023 were included. Overall survival (OS) and progression-free survival (PFS) were analyzed using the Kaplan-Meier method. Univariate and multivariate analysis of OS were performed using the Cox proportional hazards model. RESULTS A total of 42 patients were included, with a median age of 51.5 years (range, 23-74). The majority had SCC arising from teratomas (54.8%), followed by endometriosis (14.3%) and Brenner's tumor (2.4%). Patients undergoing molecular testing exhibited a median tumor mutation burden (TMB) of 10.00 mutations/Mb (range, 7.28-46.86), predominantly featuring PIK3CA mutations. Thirty-eight patients (90.5%) received adjuvant chemotherapy. The median OS was 42.0 months, with the 1- and 5-year OS rates were 73.7% and 48.7%, respectively. And the median PFS was 26.9 months, with the 1- and 5-year PFS rates were 57.5% and 43.8%, respectively. Five patients underwent first-line postoperative adjuvant therapy combining ICIs with chemotherapy. During the 9.5 to 25.1 months follow-up, 4 patients showed no evidence of disease, while 1 relapsed and received treatment. Late-stage disease and younger age at diagnosis were associated with worse survival outcomes. CONCLUSION The prognosis for ovarian SCC remains unfavorable. The stage and age were prognostic predictors for survival. ICIs may be beneficial for patients with ovarian SCC, particularly those with a high TMB.
Collapse
Affiliation(s)
- Tianyu Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Jie Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China.
| | - Sijian Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Xiaohua Shi
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| |
Collapse
|
2
|
Luo J, Gao YX, He Q, Wang WX. Immunotherapy in young adults with mature cystic ovarian Teratoma-Derived squamous cell carcinoma: a case series. J Ovarian Res 2025; 18:100. [PMID: 40369679 PMCID: PMC12076871 DOI: 10.1186/s13048-025-01681-8] [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: 12/20/2024] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Mature cystic teratomas (MCTs), also known as dermoid cysts, are common benign germ cell tumors. These predominantly benign tumors can become malignant, particularly in postmenopausal women; the overall transformation rate of MCTs is 1-2%, accounting for approximately 80% squamous cell carcinoma of cases. Malignant transformation remains exceptionally rare in young adults. CASE PRESENTATION Herein, we report two cases of squamous cell carcinoma arising from MCTs in a 25-year-old patient, staged FIGO 2018 IIIA1 and IC2 according to the International Federation of Gynecology and Obstetrics (FIGO) 2018 classification. The results of histopathological examination confirmed squamous cell carcinoma arising from the teratomas. Immunohistochemical staining revealed programmed death-ligand 1 expression. Both patients underwent fertility-sparing surgeries and subsequently received adjuvant chemotherapy in combination with programmed death receptor 1 (PD-1) inhibitor immunotherapy. A complete clinical response was ultimately achieved. Patients 1 and 2 were monitored for 29 and 20 months after operation, respectively. A complete response was maintained in Patients1 and 2 for 14 and 16 months, respectively, as of the last follow-up. CONCLUSION Squamous cell carcinoma arising from MCTs is exceedingly rare in young adults and exhibits nonspecific clinical manifestations and imaging characteristics, hindering preoperative diagnosis. These tumors display highly aggressive biological behavior and are typically associated with a poor prognosis and a high postoperative recurrence rate. Immunotherapy intervention at diverse different time points for patients at different stages can increase their survival period.
Collapse
Affiliation(s)
- Jing Luo
- Department of Obstetrics and Gynecology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical College, Xinxiang, 453000, Henan, China
| | - Yu-Xia Gao
- Department of Obstetrics and Gynecology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical College, Xinxiang, 453000, Henan, China
| | - Qian He
- Department of Obstetrics and Gynecology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical College, Xinxiang, 453000, Henan, China
| | - Wen-Xiang Wang
- Department of Obstetrics and Gynecology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical College, Xinxiang, 453000, Henan, China.
| |
Collapse
|
3
|
Zhang C, Jiao S, Zeng D, Jiang W, Wang R, Zheng B, Wang M, Wang S, Gui X. IL-11/IL-11R signal inhibition by 9MW3811 remodels immune tumor microenvironment and enhances anti-tumor efficacy of PD-1 blockade. NPJ Precis Oncol 2025; 9:138. [PMID: 40355638 PMCID: PMC12069607 DOI: 10.1038/s41698-025-00913-w] [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: 06/27/2024] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
Recent studies have uncovered evidences for pro-tumorigenic activities attributed to IL-11, prompting a renewed focus on therapeutic strategies targeting IL-11 signaling for anti-tumor treatment. Here, we introduce 9MW3811, a monoclonal antibody designed to neutralize IL-11 effectively. By disrupting the IL-11/IL-11Rα/gp130 complex, 9MW3811 inhibits JAK/STAT3 signaling, significantly reducing tumor growth in diverse mouse models. More importantly, 9MW3811 synergizes with anti-PD-1 therapy, even in PD-1 non-responsive models like CT26. Single-cell RNA-seq analysis reveals that 9MW3811 remodels the tumor microenvironment by enhancing CD8+ T cell infiltration and reversing T cell exhaustion via upregulated XCL1 and downregulated CCL7, boosting anti-tumor cytotoxicity. Furthermore, 9MW3811 counteracts PD-1-induced T cell exhaustion, with anti-PD-1 antibodies effectively mitigating PD-1 upregulation post-9MW3811 treatment. These compelling findings support ongoing clinical trials of 9MW3811, aiming to translate these preclinical insights into therapeutic benefits for cancer patients.
Collapse
Affiliation(s)
- Chang Zhang
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
- Beijing Kohnoor Science & Technology Co. Ltd., 102206, Beijing, China
| | - Shasha Jiao
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
- Beijing Kohnoor Science & Technology Co. Ltd., 102206, Beijing, China
| | - Dadi Zeng
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
- Beijing Kohnoor Science & Technology Co. Ltd., 102206, Beijing, China
| | - Wen Jiang
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
| | - Rongjuan Wang
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
- Beijing Kohnoor Science & Technology Co. Ltd., 102206, Beijing, China
| | - Bin Zheng
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
| | - Min Wang
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China
| | - Shuang Wang
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China.
- Beijing Kohnoor Science & Technology Co. Ltd., 102206, Beijing, China.
| | - Xun Gui
- Mabwell (Shanghai) Bioscience Co. Ltd., 201210, Shanghai, China.
| |
Collapse
|
4
|
Yoshida K, Yokoi A, Suzuki H, Tamauchi S, Kitagawa M, Inami E, Nakayama J, Mori Y, Okamoto K, Suzuki Y, Yoshida H, Kato T, Kajiyama H, Yamamoto Y. Single-Nucleus RNA Sequencing and Spatial Transcriptomics for Squamous Cell Carcinoma Arising From Ovarian Mature Teratoma. Cancer Sci 2025; 116:1339-1351. [PMID: 39948701 PMCID: PMC12044660 DOI: 10.1111/cas.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 05/02/2025] Open
Abstract
Squamous cell carcinoma arising from mature teratoma (SCC-MT) is a rare ovarian malignancy. The detailed molecular pathology of SCC-MT is not well understood. Moreover, the prognosis of the patients remains poor because no standard treatment has been established. In this study, we performed single-nucleus RNA sequencing and spatial transcriptomics using clinical SCC-MT samples to identify novel therapeutic candidates. snRNA-seq revealed three epithelial cell clusters, of which one was significantly associated with epidermis and keratinocyte development. Moreover, spatial transcriptomics revealed that the epithelial-mesenchymal transition was significantly inhibited, and the MYC and E2F targets were significantly activated in cancer spots on specimen sections. We focused on KLF5, which was one of the upregulated genes in cancer cells, and performed a functional analysis using NOSCC-1, a cell line derived from an SCC-MT. KLF5 downregulation significantly decreased cell proliferation and increased apoptosis. Furthermore, we previously identified miR-145-5p as a downregulated miRNA in SCC-MT. We demonstrated that miR-145-5p overexpression attenuated cell proliferation and decreased KLF5 expression. In conclusion, through multi-omics analyses, we identified unique gene expression profiles of SCC-MT and determined a role for KLF5 in SCC-MT development. Therefore, KLF5-related factors may be novel therapeutic targets, and further studies are needed to improve the diagnosis and treatment of SCC-MT.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Akira Yokoi
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
| | - Hironori Suzuki
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Satoshi Tamauchi
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Masami Kitagawa
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Eri Inami
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Jun Nakayama
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
- Department of Oncogenesis and Growth RegulationResearch Institute, Osaka International Cancer InstituteOsakaJapan
| | - Yutaro Mori
- Advanced Comprehensive Research OrganizationTeikyo UniversityTokyoJapan
| | - Koji Okamoto
- Advanced Comprehensive Research OrganizationTeikyo UniversityTokyoJapan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoJapan
| | - Hiroshi Yoshida
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
| | - Tomoyasu Kato
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Hiroaki Kajiyama
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yusuke Yamamoto
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| |
Collapse
|
5
|
Maurac Pašalić I, Sabol M, Prtenjača E, Puvačić Solomun L, Pavić M. Mature cystic teratoma and their malignant transformation. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109641. [PMID: 40348473 DOI: 10.1016/j.ejso.2025.109641] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 05/14/2025]
Abstract
Mature cystic teratomas (MCT) are benign germ cells tumors which are fairly common and are often diagnosed incidentally. In most cases they don't present a significant risk to survival, but in some rare cases they can progress to a malignant phenotype. In such cases the treatment must be adjusted to the new conditions and tailored to suit the malignant phenotype. In this review, we aim to describe the main clinical and biological features of various types of recorded malignant transformations and/or complications, as well as stress their differences in clinical behavior and therapeutic options. The management of benign vs. malignant MCT differs greatly and a specific subtype of transformation should be taken into consideration when designing therapy, as it can greatly improve the survival and quality of life.
Collapse
Affiliation(s)
- Ivana Maurac Pašalić
- School of Medicine, University of Zagreb, Zagreb, Croatia; Department of Gynecology and Obstetrics, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia.
| | | | | | - Mato Pavić
- School of Medicine, University of Zagreb, Zagreb, Croatia; Department of Gynecology and Obstetrics, University Hospital Centre Zagreb, Zagreb, Croatia.
| |
Collapse
|
6
|
Brown ME, Thirawatananond P, Peters LD, Kern EJ, Vijay S, Sachs LK, Posgai AL, Brusko MA, Shapiro MR, Mathews CE, Bacher R, Brusko TM. Inhibition of CD226 co-stimulation suppresses diabetes development in the NOD mouse by augmenting regulatory T cells and diminishing effector T cell function. Diabetologia 2025; 68:397-418. [PMID: 39636437 PMCID: PMC11732877 DOI: 10.1007/s00125-024-06329-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/10/2024] [Indexed: 12/07/2024]
Abstract
AIMS/HYPOTHESIS Immunotherapeutics targeting T cells are crucial for inhibiting autoimmune disease progression proximal to disease onset in type 1 diabetes. There is an outstanding need to augment the durability and effectiveness of T cell targeting therapies by directly restraining proinflammatory T cell subsets, while simultaneously augmenting regulatory T cell (Treg) activity. Here, we present a novel strategy for preventing diabetes incidence in the NOD mouse model using a blocking monoclonal antibody targeting the type 1 diabetes risk-associated T cell co-stimulatory receptor, CD226. METHODS Female NOD mice were treated with anti-CD226 at 7-8 weeks of age and then monitored for diabetes incidence and therapeutic mechanism of action. RESULTS Compared with isotype-treated controls, anti-CD226-treated NOD mice showed reduced insulitis severity (0.84-fold, p=0.0002) at 12 weeks and decreased disease incidence (HR 0.41, p=0.015) at 30 weeks. Flow cytometric analysis performed 5 weeks post treatment demonstrated reduced proliferation of conventional CD4+ T cells (0.87-fold, p=0.030) and CD8+ (0.78-fold, p=0.0018) effector memory T cells in spleens of anti-CD226-treated mice. Phenotyping of pancreatic Tregs revealed increased CD25 expression (2.05-fold, p=0.0073) and signal transducer and activator of transcription 5 (STAT5) phosphorylation (1.39-fold, p=0.0007) following anti-CD226, with splenic Tregs displaying augmented suppression of CD4+ responder T cells (Tresps) (1.49-fold, p=0.0008, 1:2 Treg:Tresp) in vitro. Anti-CD226-treated mice exhibited reduced frequencies of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-reactive CD8+ T cells in the pancreas, using both ex vivo tetramer staining (0.50-fold, p=0.0317) and single-cell T cell receptor sequencing (0.61-fold, p=0.022) approaches. 51Cr-release assays demonstrated reduced cell-mediated lysis of beta cells (0.61-fold, p<0.0001, 1:1 effector:target) by anti-CD226-treated autoreactive cytotoxic T lymphocytes. CONCLUSIONS/INTERPRETATION CD226 blockade reduces T cell cytotoxicity and improves Treg function, representing a targeted and rational approach for restoring immune regulation in type 1 diabetes.
Collapse
MESH Headings
- Animals
- Mice, Inbred NOD
- Mice
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/drug effects
- Female
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- T Lineage-Specific Activation Antigen 1
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
Collapse
Affiliation(s)
- Matthew E Brown
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Puchong Thirawatananond
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Leeana D Peters
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Elizabeth J Kern
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sonali Vijay
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lindsey K Sachs
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Amanda L Posgai
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Maigan A Brusko
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Melanie R Shapiro
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Clayton E Mathews
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rhonda Bacher
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - Todd M Brusko
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Jiang G, Wang Z, Cheng Z, Wang W, Lu S, Zhang Z, Anene CA, Khan F, Chen Y, Bailey E, Xu H, Dong Y, Chen P, Zhang Z, Gao D, Wang Z, Miao J, Xue X, Wang P, Zhang L, Gangeswaran R, Liu P, Chard Dunmall LS, Li J, Guo Y, Dong J, Lemoine NR, Li W, Wang J, Wang Y. The integrated molecular and histological analysis defines subtypes of esophageal squamous cell carcinoma. Nat Commun 2024; 15:8988. [PMID: 39419971 PMCID: PMC11487165 DOI: 10.1038/s41467-024-53164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is highly heterogeneous. Our understanding of full molecular and immune landscape of ESCC remains limited, hindering the development of personalised therapeutic strategies. To address this, we perform genomic-transcriptomic characterizations and AI-aided histopathological image analysis of 120 Chinese ESCC patients. Here we show that ESCC can be categorized into differentiated, metabolic, immunogenic and stemness subtypes based on bulk and single-cell RNA-seq, each exhibiting specific molecular and histopathological features based on an amalgamated deep-learning model. The stemness subgroup with signature genes, such as WFDC2, SFRP1, LGR6 and VWA2, has the poorest prognosis and is associated with downregulated immune activities, a high frequency of EP300 mutation/activation, functional mutation enrichment in Wnt signalling and the highest level of intratumoural heterogeneity. The immune profiling by transcriptomics and immunohistochemistry reveals ESCC cells overexpress natural killer cell markers XCL1 and CD160 as immune evasion. Strikingly, XCL1 expression also affects the sensitivity of ESCC cells to common chemotherapy drugs. This study opens avenues for ESCC treatment and provides a valuable public resource to better understand ESCC.
Collapse
Affiliation(s)
- Guozhong Jiang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Zhizhong Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, People's Republic of China
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Shuangshuang Lu
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Zifang Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Chinedu A Anene
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Centre for Biomedical Science Research, Leeds Beckett University, Leeds, LS1 3HE, UK
| | - Faraz Khan
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Yue Chen
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Emma Bailey
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Huisha Xu
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Yunshu Dong
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Peinan Chen
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, People's Republic of China
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Zhongxian Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Dongling Gao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Zhimin Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Jinxin Miao
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Xia Xue
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Pengju Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Rathi Gangeswaran
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Peng Liu
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Junkuo Li
- Department of Molecular Pathology, Anyang Cancer Hospital, Anyang City, 455000, Henan Province, People's Republic of China
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, People's Republic of China
| | - Jianzeng Dong
- Department of Cardiology, Centre for Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chao Yang District, Beijing, 100029, People's Republic of China
| | - Nicholas R Lemoine
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
| | - Jun Wang
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom.
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
8
|
Brown ME, Thirawatananond P, Peters LD, Kern EJ, Vijay S, Sachs LK, Posgai AL, Brusko MA, Shapiro MR, Mathews CE, Bacher R, Brusko TM. Inhibition of CD226 Co-Stimulation Suppresses Diabetes Development in the NOD Mouse by Augmenting Tregs and Diminishing Effector T Cell Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603756. [PMID: 39071293 PMCID: PMC11275941 DOI: 10.1101/2024.07.16.603756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Aims/hypothesis Immunotherapeutics targeting T cells are crucial for inhibiting autoimmune disease progression proximal to disease onset in type 1 diabetes. A growing number of T cell-directed therapeutics have demonstrated partial therapeutic efficacy, with anti-CD3 (α-CD3) representing the only regulatory agency-approved drug capable of slowing disease progression through a mechanism involving the induction of partial T cell exhaustion. There is an outstanding need to augment the durability and effectiveness of T cell targeting by directly restraining proinflammatory T helper type 1 (Th1) and type 1 cytotoxic CD8+ T cell (Tc1) subsets, while simultaneously augmenting regulatory T cell (Treg) activity. Here, we present a novel strategy for reducing diabetes incidence in the NOD mouse model using a blocking monoclonal antibody targeting the type 1 diabetes-risk associated T cell co-stimulatory receptor, CD226. Methods Female NOD mice were treated with anti-CD226 between 7-8 weeks of age and then monitored for diabetes incidence and therapeutic mechanism of action. Results Compared to isotype-treated controls, anti-CD226 treated NOD mice showed reduced insulitis severity at 12 weeks and decreased disease incidence at 30 weeks. Flow cytometric analysis performed five weeks post-treatment demonstrated reduced proliferation of CD4+ and CD8+ effector memory T cells in spleens of anti-CD226 treated mice. Phenotyping of pancreatic Tregs revealed increased CD25 expression and STAT5 phosphorylation following anti-CD226, with splenic Tregs displaying augmented suppression of CD4+ T cell responders in vitro. Anti-CD226 treated mice exhibited reduced frequencies of islet-specific glucose-6-phosphatase catalytic subunit related protein (IGRP)-reactive CD8+ T cells in the pancreas, using both ex vivo tetramer staining and single-cell T cell receptor sequencing (scTCR-seq) approaches. 51Cr-release assays demonstrated reduced cell-mediated lysis of beta-cells by anti-CD226-treated autoreactive cytotoxic T lymphocytes. Conclusions/interpretation CD226 blockade reduces T cell cytotoxicity and improves Treg function, representing a targeted and rational approach for restoring immune regulation in type 1 diabetes.
Collapse
Affiliation(s)
- Matthew E. Brown
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Puchong Thirawatananond
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Leeana D. Peters
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Elizabeth J. Kern
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Sonali Vijay
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Lindsey K. Sachs
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Amanda L. Posgai
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Maigan A. Brusko
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Melanie R. Shapiro
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Clayton E. Mathews
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Rhonda Bacher
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610
| | - Todd M. Brusko
- Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
9
|
Lokhande L, Nilsson D, de Matos Rodrigues J, Hassan M, Olsson LM, Pyl PT, Vasquez L, Porwit A, Gerdtsson AS, Jerkeman M, Ek S. Quantification and Profiling of Early and Late Differentiation Stage T Cells in Mantle Cell Lymphoma Reveals Immunotherapeutic Targets in Subsets of Patients. Cancers (Basel) 2024; 16:2289. [PMID: 39001353 PMCID: PMC11240320 DOI: 10.3390/cancers16132289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
With the aim to advance the understanding of immune regulation in MCL and to identify targetable T-cell subsets, we set out to combine image analysis and spatial omic technology focused on both early and late differentiation stages of T cells. MCL patient tissue (n = 102) was explored using image analysis and GeoMx spatial omics profiling of 69 proteins and 1812 mRNAs. Tumor cells, T helper (TH) cells and cytotoxic (TC) cells of early (CD57-) and late (CD57+) differentiation stage were analyzed. An image analysis workflow was developed based on fine-tuned Cellpose models for cell segmentation and classification. TC and CD57+ subsets of T cells were enriched in tumor-rich compared to tumor-sparse regions. Tumor-sparse regions had a higher expression of several key immune suppressive proteins, tentatively controlling T-cell expansion in regions close to the tumor. We revealed that T cells in late differentiation stages (CD57+) are enriched among MCL infiltrating T cells and are predictive of an increased expression of immune suppressive markers. CD47, IDO1 and CTLA-4 were identified as potential targets for patients with T-cell-rich MCL TIME, while GITR might be a feasible target for MCL patients with sparse T-cell infiltration. In subgroups of patients with a high degree of CD57+ TC-cell infiltration, several immune checkpoint inhibitors, including TIGIT, PD-L1 and LAG3 were increased, emphasizing the immune-suppressive features of this highly differentiated T-cell subset not previously described in MCL.
Collapse
Affiliation(s)
- Lavanya Lokhande
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | - Daniel Nilsson
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | | | - May Hassan
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | - Lina M. Olsson
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | - Paul-Theodor Pyl
- Department of Laboratory Medicine, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, 221 00 Lund, Sweden
| | - Louella Vasquez
- Department of Laboratory Medicine, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, 221 00 Lund, Sweden
| | - Anna Porwit
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, 221 00 Lund, Sweden
| | | | - Mats Jerkeman
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, 221 00 Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
10
|
Gan D, Zhu Y, Lu X, Li J. SCIPAC: quantitative estimation of cell-phenotype associations. Genome Biol 2024; 25:119. [PMID: 38741183 PMCID: PMC11089691 DOI: 10.1186/s13059-024-03263-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
Numerous algorithms have been proposed to identify cell types in single-cell RNA sequencing data, yet a fundamental problem remains: determining associations between cells and phenotypes such as cancer. We develop SCIPAC, the first algorithm that quantitatively estimates the association between each cell in single-cell data and a phenotype. SCIPAC also provides a p-value for each association and applies to data with virtually any type of phenotype. We demonstrate SCIPAC's accuracy in simulated data. On four real cancerous or noncancerous datasets, insights from SCIPAC help interpret the data and generate new hypotheses. SCIPAC requires minimum tuning and is computationally very fast.
Collapse
Affiliation(s)
- Dailin Gan
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, 46556, IN, USA
| | - Yini Zhu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, 46556, IN, USA
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, 46556, IN, USA
- Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, 46202, IN, USA
| | - Jun Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, 46556, IN, USA.
| |
Collapse
|
11
|
Jia F, Sun S, Li J, Wang W, Huang H, Hu X, Pan S, Chen W, Shen L, Yao Y, Zheng S, Chen H, Xia W, Yuan H, Zhou J, Yu X, Zhang T, Zhang B, Huang J, Ni C. Neoadjuvant chemotherapy-induced remodeling of human hormonal receptor-positive breast cancer revealed by single-cell RNA sequencing. Cancer Lett 2024; 585:216656. [PMID: 38266804 DOI: 10.1016/j.canlet.2024.216656] [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: 09/19/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
Hormone receptor-positive breast cancer (HR+ BC) is known to be relatively insensitive to chemotherapy, and since chemotherapy has remained the major neoadjuvant therapy for HR+ BC, the undetermined mechanism of chemoresistance and how chemotherapy reshapes the immune microenvironment need to be explored by high-throughput technology. By using single-cell RNA sequencing and multiplexed immunofluorescence staining analysis of HR+ BC samples (paired pre- and post-neoadjuvant chemotherapy (NAC)), the levels of previously unrecognized immune cell subsets, including CD8+ T cells with pronounced expression of T-cell development (LMNA) and cytotoxicity (FGFBP2) markers, CD4+ T cells characterized by proliferation marker (ATP1B3) expression and macrophages characterized by CD52 expression, were found to be increased post-NAC, which were predictive of chemosensitivity and their antitumor function was also validated with in vitro experiments. In terms of immune checkpoint expression of CD8+ T cells, we found their changes were inconsistent post-NAC, that LAG3, VSIR were decreased, and PDCD1, HAVCR2, CTLA4, KLRC1 and BTLA were increased. In addition, we have identified novel genomic and transcriptional patterns of chemoresistant cancer cells, both innate and acquired, and have confirmed their prognostic value with TCGA cohorts. By shedding light on the ecosystem of HR+ BC reshaped by chemotherapy, our results uncover valuable candidates for predicting chemosensitivity and overcoming chemoresistance in HR+ BC.
Collapse
Affiliation(s)
- Fang Jia
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Shanshan Sun
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Jiaxin Li
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Wenwen Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huanhuan Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Xiaoxiao Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Pan
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Yao
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwei Zheng
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Xia
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Hongjun Yuan
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jun Zhou
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuyan Yu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Zhang
- Department of Radiotherapy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Zhang
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Chao Ni
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China.
| |
Collapse
|
12
|
Tamura R, Kushiya N, Yamaguchi M, Nishikawa N, Motoyama T, Kawasaki T, Kikuchi A. Refractory ovarian squamous cell carcinoma arising from a seromucinous borderline tumor with squamous overgrowth: A case report. Gynecol Oncol Rep 2024; 51:101323. [PMID: 38273936 PMCID: PMC10808907 DOI: 10.1016/j.gore.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024] Open
Abstract
Ovarian squamous cell carcinoma (SCC) is rare, and most cases arise from ovarian teratomas. Herein, we present a case of ovarian SCC arising from an ovarian seromucinous borderline tumor (SMBT) with squamous overgrowth. A 71-year-old woman an underwent emergency laparotomy due to the rupture of a right ovarian tumor suspected to be a borderline or malignant tumor. We performed a total abdominal hysterectomy, bilateral salpingo-oophorectomy, and partial omentectomy. The postoperative diagnosis was stage IC3 ovarian SCC arising from the SMBT with a squamous overgrowth. Subsequently, she underwent six cycles of combination therapy comprising paclitaxel and carboplatin. Two months after the last chemotherapy treatment, she presented with back pain. A CT scan showed a 14 mm pelvic tumor affecting the ureter, leading to right hydronephrosis. The patient underwent tumor resection and ureteroureterostomy. The pathological diagnosis was keratinizing SCC, representing ovarian cancer recurrence. Eight months after the removal of the recurrent tumor, we found a 35 mm recurrent pelvic tumor causing right hydronephrosis. Additionally, a 20 mm pleural dissemination was identified. Comprehensive genome profiling of recurrent tumor revealed genomic abnormalities in TP53, ARID1A, PTEN, PIK3R1, and CDKN2A/2B. Regarding immunotherapy biomarkers, the microsatellite instability test result was negative, the tumor mutation burden was low, and PD-L1 was highly expressed. The patient was referred to another hospital for participation in an immunotherapy clinical trial for ovarian SCC. This case indicates that refractory ovarian SCC can arise from SMBT. Further evaluation of additional cases is required to identify the molecular biological characteristics of ovarian SCC.
Collapse
Affiliation(s)
- Ryo Tamura
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Naohisa Kushiya
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Masayuki Yamaguchi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | | | - Teiichi Motoyama
- Department of Pathology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Takashi Kawasaki
- Department of Pathology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Akira Kikuchi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| |
Collapse
|
13
|
Zhao H, Lin Z, Zhang Y, Liu J, Chen Q. Investigating the Heterogeneity of Immune Cells in Triple-Negative Breast Cancer at the Single-Cell Level before and after Paclitaxel Chemotherapy. Int J Mol Sci 2023; 24:14188. [PMID: 37762493 PMCID: PMC10532302 DOI: 10.3390/ijms241814188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Despite the numerous treatments for triple-negative breast cancer (TNBC), chemotherapy is still one of the most effective methods. However, the impact of chemotherapy on immune cells is not yet clear. Therefore, this study aims to explore the different roles of immune cells and their relationship with treatment outcomes in the tumor and blood before and after paclitaxel therapy. We analyzed the single-cell sequencing data of immune cells in tumors and blood before and after paclitaxel treatment. We confirmed a high correlation between T cells, innate lymphoid cells (ILCs), and therapeutic efficacy. The differences in T cells were analyzed related to therapeutic outcomes before and after paclitaxel treatment. In the effective treatment group, post-treatment tumor-infiltrating CD8+ T cells were associated with elevated inflammation, cytokines, and Toll-like-receptor-related gene expression, which were expected to enhance anti-tumor capabilities in tumor immune cells. Moreover, we found that the expression of immune-checkpoint-related genes is also correlated with treatment outcomes. In addition, an ILC subgroup, b_ILC1-XCL1, in which the corresponding marker gene XCL1 was highly expressed, was mainly present in the effective treatment group and was also associated with higher patient survival rates. Overall, we found differences in gene expression in T cells across different groups and a correlation between the expression of immune checkpoint genes in T cells, the b_ILC1-XCL1 subgroup, and patient prognosis.
Collapse
Affiliation(s)
- Heng Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou 350117, China; (H.Z.); (Z.L.); (Y.Z.); (J.L.)
| | - Zhang Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou 350117, China; (H.Z.); (Z.L.); (Y.Z.); (J.L.)
- College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yangfan Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou 350117, China; (H.Z.); (Z.L.); (Y.Z.); (J.L.)
| | - Jingjing Liu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou 350117, China; (H.Z.); (Z.L.); (Y.Z.); (J.L.)
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, College Town, Fuzhou 350117, China; (H.Z.); (Z.L.); (Y.Z.); (J.L.)
| |
Collapse
|
14
|
Ji X, Zhai P, Yang H, Wang H, Wang X. Recurrent squamous cell carcinoma arising in ovary mature cystic teratoma: A case report. Medicine (Baltimore) 2023; 102:e34734. [PMID: 37565860 PMCID: PMC10419499 DOI: 10.1097/md.0000000000034734] [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: 05/26/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
RATIONALE Malignant transformation of mature cystic teratoma is very rare, of which squamous cell carcinoma (SCC) is the most common type. Prognosis of SCC arising in mature cystic teratoma of the ovary is very poor. Our experience may provide new ideas for the treatment of this disease. PATIENT CONCERNS The patient was a 56-year-old woman and was admitted for a lower abdominal pain. She underwent a laparoscopic surgery with 4 cycles of chemotherapy and had achieved a complete response; 10 months after the completion of initial treatment, her cancer relapsed. She underwent a cytoreductive surgery with concurrent chemoradiotherapy and has achieved a complete response again. DIAGNOSES This patient was initially diagnosed with ovarian cancer (stage IIIB) arising from malignant transformation of mature teratoma; 10 months after the completion of initial treatment, she was diagnosed with recurrent ovarian cancer. INTERVENTIONS This patient was initially treated with laparoscopic bilateral salpingo-oophorectomy. After histopathological confirmation that she had ovarian cancer, she underwent laparoscopic total hysterectomy and omentectomy with 4 cycles of chemotherapy. After her ovarian cancer recurred, she underwent open cytoreductive surgery and concurrent chemoradiotherapy. OUTCOMES The patient achieved complete response after both initial and relapsed treatment. LESSONS Optimal cytoreduction and concurrent chemoradiotherapy may be an option to improve the prognosis of patients with recurrent SCC arising in ovary mature cystic teratoma.
Collapse
Affiliation(s)
- Xuechao Ji
- Department of Obstetrics and Gynecology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Peiling Zhai
- Department of Obstetrics and Gynecology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Hanchao Yang
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xinbo Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
15
|
Sato Y, Yano M, Eto S, Takano K, Nasu K. Metastasis from follicular lymphoma to an ovarian mature teratoma: a case report of tumor-to-tumor metastasis. J Ovarian Res 2023; 16:106. [PMID: 37254134 DOI: 10.1186/s13048-023-01188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/16/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Tumor-to-tumor metastasis (TTM) is a rare but well-established phenomenon where histologically distinct tumors metastasize within each other. Here we report the first "known" case of follicular lymphoma that metastasized and extended to a mature ovarian teratoma. CASE PRESENTATION A 59-year-old Japanese postmenopausal woman visited our hospital for a detailed examination of an ovarian tumor. Clinical imaging suggested it to be either teratoma-associated ovarian cancer with multiple lymph node metastases, or tumor-to-tumor metastasis from malignant lymphoma to ovarian teratoma. A bilateral adnexectomy and retroperitoneal lymph node biopsy were performed. Lined with squamous epithelium, the cyst constituted a mature ovarian teratoma, and the solid part showed diffuse proliferation of abnormal lymphoid cells. Immunohistochemically, the abnormal lymphoid cells were negative for CD5, MUM1, and CyclinD1, and positive for CD10, CD20, CD21, BCL2, and BCL6. Genetic analysis using G-banding and fluorescence in situ hybridization identified a translocation of t(14;18) (q32;q21), and we diagnosed tumor-to-tumor metastasis from nodal follicular lymphoma to mature ovarian teratoma. Twelve months after surgery, the patient showed no progression without adjuvant therapy. CONCLUSIONS The present case suggests that molecular approaches are useful in the diagnosis of TTM in mature ovarian teratomas when morphologic and immunohistochemical findings alone are insufficient for diagnoses.
Collapse
Affiliation(s)
- Yusuke Sato
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Oita, Japan
| | - Mitsutake Yano
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Oita, Japan.
| | - Satoshi Eto
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kuniko Takano
- Department of Medical Oncology and Hematology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kaei Nasu
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Oita, Japan
- Division of Obstetrics and Gynecology, Support System for Community Medicine, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
16
|
Tamura R, Yamaguchi M, Kitani Y, Nishikawa N, Kawasaki T, Kikuchi A. Successful treatment of advanced squamous cell carcinoma arising from mature cystic teratoma of the ovary with homologous recombination deficiency: A case report. Int J Surg Case Rep 2023; 107:108329. [PMID: 37216734 DOI: 10.1016/j.ijscr.2023.108329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023] Open
Abstract
INTRODUCTION AND IMPORTANCE Squamous cell carcinoma (SCC) arising from mature cystic teratoma of the ovary (MCT-SCC) has a poor prognosis at advanced stages. Although the relationship between homologous recombination deficiency (HRD) and platinum-based chemotherapy sensitivity or poly (ADP ribose) polymerase (PARP) inhibitor efficacy in epithelial ovarian cancer has been demonstrated in clinical trials, the significance of HRD status in MCT-SCC has not previously been described. CASE PRESENTATION A 73-year-old woman underwent emergency laparotomy due to ovarian tumor rupture. The ovarian tumor was strongly adherent to the surrounding pelvic organs and could not be completely resected. The postoperative diagnosis was stage IIIB MCT-SCC (pT3bNXM0) of the left ovary. After surgery, we conducted the myChoice CDx. The genomic instability (GI) score of 87 was remarkably high, and there was no BRCA1/2 pathogenic mutation. After six courses of combination therapy with paclitaxel and carboplatin, the residual tumors had shrunk by 73 %. We performed interval debulking surgery (IDS), and the residual tumors were completely resected. Subsequently, the patient underwent two courses of the combination of paclitaxel, carboplatin, and bevacizumab, followed by maintenance therapy with olaparib and bevacizumab. Twelve months after IDS, no recurrence has been observed. CLINICAL DISCUSSION The present case suggests that there are some HRD cases among MCT-SCC patients and that IDS and maintenance therapy with PARP inhibitors may be effective in such cases, as in epithelial ovarian cancer. CONCLUSION Although the frequency of HRD-positive status in MCT-SCC remains unknown, HRD testing may provide appropriate treatment options for advanced MCT-SCC.
Collapse
Affiliation(s)
- Ryo Tamura
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan; Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Japan.
| | - Masayuki Yamaguchi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yohei Kitani
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | | | - Takashi Kawasaki
- Department of Pathology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Akira Kikuchi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| |
Collapse
|
17
|
Mayeur S, Lhermitte B, Gantzer J, Molitor A, Stemmelen T, Meyer S, Kolmer A, Kurtz JE, Bahram S, Carapito R. Genomic profiling of a metastatic anaplastic melanocytic neuroectodermal tumor arising from a mature thymic teratoma as part of a mediastinal germ cell tumor. Cold Spring Harb Mol Case Stud 2023; 9:mcs.a006257. [PMID: 37160315 DOI: 10.1101/mcs.a006257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/11/2023] [Indexed: 05/11/2023] Open
Abstract
Following chemotherapy, a mediastinal germ cell tumor can lead to a mature teratoma that is composed of tissues derived from all three germ layers. Although teratoma is usually curable, in rare cases it can give rise to various somatic tumors and exceptionally it undergoes melanocytic neuroectodermal tumor (MNT) transformation, a process that is not well-described. We report a patient with a postchemotherapy thymic teratoma associated with an MNT component who, 10 years later, additionally presented a vertebral metastasis corresponding to an anaplastic MNT. Using exome sequencing of the mature teratoma, the MNT and its metastatic vertebral anaplastic MNT components, we identified 19 somatic mutations shared by at least two components. Six mutations were common to all three components, and three of them were located in the known cancer-related genes KRAS (p.E63K), TP53 (p.P222X), and POLQ (p.S447P). Gene set enrichment analysis revealed that the melanoma tumorigenesis pathway was enriched in mutated genes including the four major driver genes KRAS, TP53, ERBB4, and KDR, indicating that these genes may be involved in the development of the anaplastic MNT transformation of the teratoma. To our knowledge, this is the first molecular study realized on MNT. Understanding the clinicopathological and molecular characteristics of these tumors is essential to better understand their development and to improve therapeutics.
Collapse
Affiliation(s)
- Sylvain Mayeur
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
- Department of Pathology, Strasbourg University Hospitals, Strasbourg 67200, France
| | - Benoit Lhermitte
- Department of Pathology, Strasbourg University Hospitals, Strasbourg 67200, France
| | - Justine Gantzer
- Pôle d'oncologie médico-chirurgicale et d'hématologie, ICANS-Europe, Strasbourg 67200, France
| | - Anne Molitor
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Tristan Stemmelen
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Sébastien Meyer
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Aline Kolmer
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Jean-Emmanuel Kurtz
- Pôle d'oncologie médico-chirurgicale et d'hématologie, ICANS-Europe, Strasbourg 67200, France
| | - Seiamak Bahram
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France;
- Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg 67091, France
| | - Raphael Carapito
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France;
- Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg 67091, France
| |
Collapse
|
18
|
Tamura R, Nakaoka H, Yachida N, Ueda H, Ishiguro T, Motoyama T, Inoue I, Enomoto T, Yoshihara K. Spatial genomic diversity associated with APOBEC mutagenesis in squamous cell carcinoma arising from ovarian teratoma. Cancer Sci 2023; 114:2145-2157. [PMID: 36762791 PMCID: PMC10154883 DOI: 10.1111/cas.15754] [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] [Received: 11/16/2022] [Revised: 01/28/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Although the gross and microscopic features of squamous cell carcinoma arising from ovarian mature cystic teratoma (MCT-SCC) vary from case to case, the spatial spreading of genomic alterations within the tumor remains unclear. To clarify the spatial genomic diversity in MCT-SCCs, we performed whole-exome sequencing by collecting 16 samples from histologically different parts of two MCT-SCCs. Both cases showed histological diversity within the tumors (case 1: nonkeratinizing and keratinizing SCC and case 2: nonkeratinizing SCC and anaplastic carcinoma) and had different somatic mutation profiles by histological findings. Mutation signature analysis revealed a significantly enriched apolipoprotein B mRNA editing enzyme catalytic subunit (APOBEC) signature at all sites. Intriguingly, the spread of genomic alterations within the tumor and the clonal evolution patterns from nonmalignant epithelium to cancer sites differed between cases. TP53 mutation and copy number alterations were widespread at all sites, including the nonmalignant epithelium, in case 1. Keratinizing and nonkeratinizing SCCs were differentiated by the occurrence of unique somatic mutations from a common ancestral clone. In contrast, the nonmalignant epithelium showed almost no somatic mutations in case 2. TP53 mutation and the copy number alteration similarities were observed only in nonkeratinizing SCC samples. Nonkeratinizing SCC and anaplastic carcinoma shared almost no somatic mutations, suggesting that each locally and independently arose in the MCT. We demonstrated that two MCT-SCCs with different histologic findings were highly heterogeneous tumors with clearly different clones associated with APOBEC-mediated mutagenesis, suggesting the importance of evaluating intratumor histological and genetic heterogeneity among multiple sites of MCT-SCC.
Collapse
Affiliation(s)
- Ryo Tamura
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Tokyo, Japan
| | - Nozomi Yachida
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Haruka Ueda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuya Ishiguro
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Teiichi Motoyama
- Department of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ituro Inoue
- Division of Human Genetics, National Institute of Genetics, Mishima, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
19
|
Yoshimura K, Yamanoi K, Kanai M, Okunomiya A, Sagae Y, Sunada M, Taki M, Ukita M, Chigusa Y, Horie A, Yamaguchi K, Hamanishi J, Minamiguchi S, Yamamoto N, Muto M, Mandai M. Nivolumab for malignant transformation of ovarian mature cystic teratoma. Gynecol Oncol Rep 2022; 44:101115. [PMID: 36560959 PMCID: PMC9764164 DOI: 10.1016/j.gore.2022.101115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Mature cystic teratoma of the ovary (MCT) occasionally undergoes malignant transformation (MT) that is resistant to chemotherapy and has a poor prognosis. We experienced a case of clinically aggressive MCT-MT that invades surrounding organs and tissues. Although tumor was resected entirely, a rapid tumor recurrence occurred during postoperative chemotherapy (paclitaxel + ifosfamide + cisplatin). The results of comprehensive genomic profiling test performed early in the postoperative period showed a high tumor mutational burden of 23 mutations/Mb. Treatment with nivolumab monotherapy has promptly been initiated and has been very successful for more than one year.
Collapse
Affiliation(s)
- Kayoko Yoshimura
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan,Corresponding author.
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Asuka Okunomiya
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Yusuke Sagae
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Masumi Sunada
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Masayo Ukita
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto Univerisy, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto City, Kyoto 606-8507, Japan
| |
Collapse
|
20
|
Cocuz IG, Cocuz ME, Repanovici A, Sabău AH, Niculescu R, Tinca AC, Vunvulea V, Budin CE, Szoke AR, Popelea MC, Moraru R, Cotoi TC, Cotoi OS. Scientific Research Directions on the Histopathology and Immunohistochemistry of the Cutaneous Squamous Cell Carcinoma: A Scientometric Study. Medicina (B Aires) 2022; 58:medicina58101449. [PMID: 36295609 PMCID: PMC9611311 DOI: 10.3390/medicina58101449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/08/2022] [Accepted: 10/12/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction: Cutaneous squamous cell carcinoma (cSCC) is one of the most frequently occurring types of cancer in humans. Scientometric research is an innovative method for analyzing the research trends in various domains, with great implications in the field of medicine. Materials and Methods: We searched the Web of Science database with the following established query terms: “Squamous cell carcinoma”, “skin”, and “immunohistochemistry”. After applying the inclusion and exclusion criteria, a total of 76 articles were selected. The present study aims to analyze, based on the frequency of use of keywords with scientometric algorithms and map-based distributions, the trends of the research concerning cSCCs in 2017–2022. Results: A graphical representation based on 11 scientometric maps presented the division of the keywords into seven clusters, from which seven categories of research interest were defined. The clusters represent a multidisciplinary approach to the diagnosis and treatment of cSCCs, cancer diagnostics, patient outcomes, histopathological importance, management of cSCCs, role of progression, and adequate treatment of and importance of immunohistochemistry for cSCCs. The distribution of the citations shows the importance of the available research on cSCCs by analyzing the first five most-cited articles included in our study in direct concordance with the seven defined clusters. Conclusion: The scientometric research method reveals the interest of research in the multidisciplinary approach used to obtain the best outcomes for the patient, including a targeted investigation, as well as diagnostic and treatment options. The trends in the research reveal that histopathological diagnostics and immunohistochemistry, combined with molecular techniques, are the most important tools used to establish a personalized diagnosis, thus increasing the quality of life and life expectancy for patients with cSCCs.
Collapse
Affiliation(s)
- Iuliu Gabriel Cocuz
- Doctoral School of Medicine and Pharmacy, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | - Maria Elena Cocuz
- Fundamental Prophylactic and Clinical Disciplines Department, Faculty of Medicine, Transilvania University of Brasov, 500003 Brașov, Romania
- Clinical Infectious Diseases Hospital of Brasov, 500174 Brasov, Romania
- Correspondence:
| | - Angela Repanovici
- Faculty of Product Design and Environment, Transilvania University of Brasov, 500036 Brasov, Romania
| | - Adrian-Horațiu Sabău
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | - Raluca Niculescu
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | - Andreea-Cătălina Tinca
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | - Vlad Vunvulea
- Anatomy and Embryology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | - Corina Eugenia Budin
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | - Andreea Raluca Szoke
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| | | | - Raluca Moraru
- Anatomy and Embryology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
- Department of Plastic Surgery, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Titiana Cornelia Cotoi
- Pharmaceutical Technique Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
- Pharmacy No. 2, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Ovidiu Simion Cotoi
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
- Pathophysiology Department, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade” of Targu Mures, 540142 Targu Mures, Romania
| |
Collapse
|
21
|
Wang Y, Zhang L, Shi G, Liu M, Zhao W, Zhang Y, Wang Y, Zhang N. Effects of Inflammatory Response Genes on the Immune Microenvironment in Colorectal Cancer. Front Genet 2022; 13:886949. [PMID: 35464849 PMCID: PMC9032353 DOI: 10.3389/fgene.2022.886949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The close relationship between colorectal cancer and inflammation has been widely reported. However, the relationship between colorectal cancer and inflammation at the genetic level is not fully understood.Method: From a genetic perspective, this study explored the relationship between inflammation-related genes and the immune microenvironment in colorectal cancer. We identified prognostic genes, namely CX3CL1, CCL22, SERPINE1, LTB4R, XCL1, GAL, TIMP1, ADIPOQ, and CRH, by using univariate and multivariate regression analyses. A risk scoring model for inflammatory response was established, and patients in The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database were divided into two groups: high risk group and low risk group.Results: The analysis showed that the prognosis of the two groups was significantly different, and the low-risk group had a higher survival rate and longer survival time. Pathways related to apoptosis, inflammatory response, and hypoxia were significantly enriched as shown via Gene Set Enrichment Analysis (GSEA). Activated dendritic cell infiltration was found in both the TCGA and GEO databases, and the CCL21 gene played a significant role in the process of activated dendritic cell infiltration. CCL21 gene was also positively correlated with inflammatory response, and the gene expression and risk score were significantly different between the two groups.Conclusion: In summary, inflammatory response has a direct impact on patients with colorectal cancer in the prognosis and immune infiltration and further research studies on the inflammatory response can help in advancing the development of immunotherapy for colorectal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ying Wang
- *Correspondence: Ying Wang, ; Nan Zhang,
| | - Nan Zhang
- *Correspondence: Ying Wang, ; Nan Zhang,
| |
Collapse
|
22
|
Song XC, Wang YX, Yu M, Cao DY, Yang JX. Case Report: Management of Recurrent Ovarian Squamous Cell Carcinoma With PD-1 Inhibitor. Front Oncol 2022; 12:789228. [PMID: 35356229 PMCID: PMC8959438 DOI: 10.3389/fonc.2022.789228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Malignant transformations, such as ovarian squamous cell carcinoma (SCC) in ovarian mature cystic teratoma (OMCT), are rare tumors. The management of recurrent disease is still a challenge, and the gene mutations involved remain unclear. We herein report a recurrent case of ovarian SCC with a PIK3CA gene variation and immunohistochemical staining of programmed death-ligand 1 (PD-L1) >10%. This patient achieved clinical remission after platinum-based effective chemotherapy and programmed death 1 (PD-1) immunotherapy.
Collapse
Affiliation(s)
- Xiao-Chen Song
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetrics & Gynecologic Diseases, Beijing, China
| | - Yong-Xue Wang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetrics & Gynecologic Diseases, Beijing, China
| | - Mei Yu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetrics & Gynecologic Diseases, Beijing, China
| | - Dong-Yan Cao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetrics & Gynecologic Diseases, Beijing, China
| | - Jia-Xin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetrics & Gynecologic Diseases, Beijing, China
| |
Collapse
|
23
|
Characterizing cell interactions at scale with made-to-order droplet ensembles (MODEs). Proc Natl Acad Sci U S A 2022; 119:2110867119. [PMID: 35074872 PMCID: PMC8812558 DOI: 10.1073/pnas.2110867119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 01/22/2023] Open
Abstract
The study of specific cell–cell interactions at scale would be a significant advancement in single-cell biology with clear utility in immuno-oncology. Our development of Droplet Assembly provides a tool for such studies by extending the benefits of single-cell droplet microfluidics to high-order cell analyses. This technology allows for the construction, sorting, and downstream processing of cell–cell interactions and is compatible with single-cell genomic readouts. Cell–cell interactions are important to numerous biological systems, including tissue microenvironments, the immune system, and cancer. However, current methods for studying cell combinations and interactions are limited in scalability, allowing just hundreds to thousands of multicell assays per experiment; this limited throughput makes it difficult to characterize interactions at biologically relevant scales. Here, we describe a paradigm in cell interaction profiling that allows accurate grouping of cells and characterization of their interactions for tens to hundreds of thousands of combinations. Our approach leverages high-throughput droplet microfluidics to construct multicellular combinations in a deterministic process that allows inclusion of programmed reagent mixtures and beads. The combination droplets are compatible with common manipulation and measurement techniques, including imaging, barcode-based genomics, and sorting. We demonstrate the approach by using it to enrich for chimeric antigen receptor (CAR)-T cells that activate upon incubation with target cells, a bottleneck in the therapeutic T cell engineering pipeline. The speed and control of our approach should enable valuable cell interaction studies.
Collapse
|
24
|
Shen H, Li Y, Feng M, Shen X, Wu D, Zhang C, Yang Y, Yang M, Hu J, Liu J, Wang W, Zhang Q, Song F, Yang J, Chen K, Li X. Miscell: An efficient self-supervised learning approach for dissecting single-cell transcriptome. iScience 2021; 24:103200. [PMID: 34712916 PMCID: PMC8529514 DOI: 10.1016/j.isci.2021.103200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/05/2021] [Accepted: 09/28/2021] [Indexed: 01/22/2023] Open
Abstract
We developed Miscell, a self-supervised learning approach with deep neural network as latent feature encoder for mining information from single-cell transcriptomes. We demonstrated the capability of Miscell with canonical single-cell analysis tasks including delineation of single-cell clusters and identification of cluster-specific marker genes. We evaluated Miscell along with three state-of-the-art methods on three heterogeneous datasets. Miscell achieved at least comparable or better performance than the other methods by significant margin on a variety of clustering metrics such as adjusted rand index, normalized mutual information, and V-measure score. Miscell can identify cell-type specific markers by quantifying the influence of genes on cell clusters via deep learning approach.
Collapse
Affiliation(s)
- Hongru Shen
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Yang Li
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Mengyao Feng
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Xilin Shen
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Dan Wu
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yichen Yang
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Meng Yang
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Jiani Hu
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Jilei Liu
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Wei Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Qiang Zhang
- Department of Maxillofacial and Otorhinolaryngology Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| | - Xiangchun Li
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin 300060, China
| |
Collapse
|
25
|
Tamura R, Yoshihara K, Matsuo K, Yachida N, Miyoshi A, Takahashi K, Sugino K, Yamaguchi M, Mori Y, Suda K, Ishiguro T, Okuda S, Motoyama T, Nakaoka H, Kikuchi A, Ueda Y, Inoue I, Enomoto T. Proposing a molecular classification associated with hypercoagulation in ovarian clear cell carcinoma. Gynecol Oncol 2021; 163:327-333. [PMID: 34452748 DOI: 10.1016/j.ygyno.2021.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND Although ovarian clear cell carcinoma (CCC) is associated with high incidence of thromboembolism, the clinicopathological and biological significance of hypercoagulable status in CCC remains unclear. MATERIALS AND METHODS We retrospectively analyzed pretreatment D-dimer levels, thromboembolic status, and clinical outcome of 125 CCCs in the discovery set and 143 CCCs in two other independent validation sets. Next, we performed RNA sequencing of 93 CCCs and compared coagulation-related gene profiles with 2492 pan-cancer data. We investigated differences in molecular characteristics of CCC subclasses based on coagulation status. RESULTS In the discovery dataset, D-dimer elevation above the normal range was significantly associated with shorter progression-free and overall survival, irrespective to thromboembolic status. Multivariate analysis identified D-dimer elevation and clinical stage as an independent prognostic factors. We confirmed the prognostic significance of D-dimer elevation in the validation sets. Tissue factor and IL6, which are considered key elements of cancer-induced hypercoagulation, were highly expressed in CCC than in other cancers regardless of D-dimer level. Higher activity of various oncogenic pathways was observed in CCC with compared to without D-dimer elevation. Moreover, hierarchical cluster analysis divided 57 CCCs with D-dimer elevation into immunologically hot and cold tumor subtypes. Hot tumors were characterized by enrichment of T-cell inflamed phenotype, inflammation, the epithelial-mesenchymal transition, and high serum levels of CRP, and cold tumors by enrichment of cell cycle and MYC pathways. CONCLUSIONS CCC represents hypercoagulable disease and elevate D-dimer is a prognostic factor for decreased survival in CCC. D-dimer high CCC has distinct molecular characteristics into the inflammatory-driven pathway (hot tumor) and the immune-suppressive pathway (cold tumor). Treatment implication of our proposed molecular classification merits further investigation.
Collapse
Affiliation(s)
- Ryo Tamura
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Koji Matsuo
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Nozomi Yachida
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ai Miyoshi
- Department of Obstetrics and Gynecology, Osaka University School of Medicine, Suita, Japan
| | - Kotaro Takahashi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Kentaro Sugino
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Manako Yamaguchi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaro Mori
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuaki Suda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuya Ishiguro
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Japan; Medical AI Center, Niigata University School of Medicine, Niigata, Japan
| | - Teiichi Motoyama
- Department of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Tokyo, Japan
| | - Akira Kikuchi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University School of Medicine, Suita, Japan
| | - Ituro Inoue
- Human Genetics Laboratory, National Institute of Genetics, Mishima, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
26
|
Wu M, Bennett JA, Reid P, Fleming GF, Kurnit KC. Successful treatment of squamous cell carcinoma arising from a presumed ovarian mature cystic teratoma with pembrolizumab. Gynecol Oncol Rep 2021; 37:100837. [PMID: 34368414 PMCID: PMC8326349 DOI: 10.1016/j.gore.2021.100837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/16/2021] [Indexed: 11/25/2022] Open
Abstract
Metastatic SCC arising from the ovary is rare, and the optimal treatment is unknown. Pembrolizumab successfully treated a patient with metastatic SCC. Patients on pembrolizumab should be monitored for immune-related adverse events.
Collapse
Affiliation(s)
- Meng Wu
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Jennifer A Bennett
- Department of Pathology, University of Chicago, Chicago, IL, United States
| | - Pankti Reid
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, United States
| | - Gini F Fleming
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, United States
| | - Katherine C Kurnit
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
27
|
Tamauchi S, Suzuki S, Xuboya C, Yoshihara M, Yoshida K, Ikeda Y, Yoshikawa N, Kajiyama H, Kikkawa F. Establishment of a patient-derived xenograft model and cell line of malignant transformation of mature cystic teratoma of the ovary. J Obstet Gynaecol Res 2020; 47:713-719. [PMID: 33300248 DOI: 10.1111/jog.14596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/09/2020] [Accepted: 11/29/2020] [Indexed: 11/29/2022]
Abstract
AIM Malignant transformation of mature cystic teratoma (MTMCT) of the ovary is a rare gynecological malignancy and commonly arises in women older than 50 years of age. The most common histological type of MTMCT is squamous cell carcinoma (SCC), and the prognosis is extremely poor. Patient-derived xenograft (PDX) models are promising animal models for preclinical drug screening. Here, we report the generation of a new PDX model of MTMCT, and a new cell line established from the tumors of PDX model animals. METHODS Tumor tissue was obtained from a 32-year-old patient with MTMCT. To generate PDX, NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ) mice, a strain of super-immunodeficient mice, were used. Tumor-bearing mice were sacrificed, followed by the collection of these tumors and re-transplantation into new NSG mice (in vivo passage). Tumor samples were also cultured in vitro. Adherent cells were continuously cultured and passaged, a cell line was established. RESULTS In the primary PDX mouse, tumor engraftment was confirmed 30 days after tumor implantation. After three times in vivo passage, we confirmed that the cryopreserved tumors could be engrafted even when transplanted into BALB/c nude mice. Using the tumor tissue at the time of the first in vivo passage, a new cell line NOSCC1 was established. PDX tumors and cell-line derived xenograft tumors exhibited similar morphology of SCC. CONCLUSION We established a new PDX model of MTMCT and a new cell line of it, which may be important tools for the development of new therapies and the elucidation of the carcinogenic mechanisms of MTMCT.
Collapse
Affiliation(s)
- Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Suzuki
- Department of Gynecologic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Chang Xuboya
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
28
|
MYBL2 amplification in breast cancer: Molecular mechanisms and therapeutic potential. Biochim Biophys Acta Rev Cancer 2020; 1874:188407. [DOI: 10.1016/j.bbcan.2020.188407] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 02/08/2023]
|
29
|
Ciechanowska A, Popiolek-Barczyk K, Ciapała K, Pawlik K, Oggioni M, Mercurio D, de Simoni MG, Mika J. Traumatic brain injury in mice induces changes in the expression of the XCL1/XCR1 and XCL1/ITGA9 axes. Pharmacol Rep 2020; 72:1579-1592. [PMID: 33185818 PMCID: PMC7704520 DOI: 10.1007/s43440-020-00187-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 11/25/2022]
Abstract
Background Every year, millions of people suffer from various forms of traumatic brain injury (TBI), and new approaches with therapeutic potential are required. Although chemokines are known to be involved in brain injury, the importance of X-C motif chemokine ligand 1 (XCL1) and its receptors, X-C motif chemokine receptor 1 (XCR1) and alpha-9 integrin (ITGA9), in the progression of TBI remain unknown. Methods Using RT-qPCR/Western blot/ELISA techniques, changes in the mRNA/protein levels of XCL1 and its two receptors, in brain areas at different time points were measured in a mouse model of TBI. Moreover, their cellular origin and possible changes in expression were evaluated in primary glial cell cultures. Results Studies revealed the spatiotemporal upregulation of the mRNA expression of XCL1, XCR1 and ITGA9 in all the examined brain areas (cortex, thalamus, and hippocampus) and at most of the evaluated stages after brain injury (24 h; 4, 7 days; 2, 5 weeks), except for ITGA9 in the thalamus. Moreover, changes in XCL1 protein levels occurred in all the studied brain structures; the strongest upregulation was observed 24 h after trauma. Our in vitro experiments proved that primary murine microglial and astroglial cells expressed XCR1 and ITGA9, however they seemed not to be a main source of XCL1. Conclusions These findings indicate that the XCL1/XCR1 and XCL1/ITGA9 axes may participate in the development of TBI. The XCL1 can be considered as one of the triggers of secondary injury, therefore XCR1 and ITGA9 may be important targets for pharmacological intervention after traumatic brain injury. Graphic abstract ![]()
Collapse
Affiliation(s)
- Agata Ciechanowska
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31-343, Kraków, Poland
| | - Katarzyna Popiolek-Barczyk
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31-343, Kraków, Poland
| | - Katarzyna Ciapała
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31-343, Kraków, Poland
| | - Katarzyna Pawlik
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31-343, Kraków, Poland
| | - Marco Oggioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri, 2, 20156, Milan, Italy
| | - Domenico Mercurio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri, 2, 20156, Milan, Italy
| | - Maria-Grazia de Simoni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri, 2, 20156, Milan, Italy
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31-343, Kraków, Poland.
| |
Collapse
|
30
|
Yachida N, Yoshihara K, Suda K, Nakaoka H, Ueda H, Sugino K, Yamaguchi M, Mori Y, Yamawaki K, Tamura R, Ishiguro T, Isobe M, Motoyama T, Inoue I, Enomoto T. ARID1A protein expression is retained in ovarian endometriosis with ARID1A loss-of-function mutations: implication for the two-hit hypothesis. Sci Rep 2020; 10:14260. [PMID: 32868822 PMCID: PMC7459315 DOI: 10.1038/s41598-020-71273-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022] Open
Abstract
ARID1A loss-of-function mutation accompanied by a loss of ARID1A protein expression is considered one of the most important driver events in endometriosis-associated ovarian cancer. Although our recent genomic study clarified that ARID1A loss-of-function mutations were detected in 13% of ovarian endometriosis, an association between the ARID1A mutation status and ARID1A protein expression in ovarian endometriosis remains unclear. We performed immunohistochemical staining for ARID1A in 78 ovarian endometriosis samples and 99 clear cell carcinoma samples. We revealed that not only 70 endometriosis samples without ARID1A mutations but also eight endometriosis samples with ARID1A loss-of-function mutations retained ARID1A protein expression. On the other hand, most of clear cell carcinomas with ARID1A loss-of-function mutations showed a loss of ARID1A protein expression. In particular, clear cell carcinoma samples which harbor multiple ARID1A loss-of-function mutations or both a single ARID1A loss-of-function mutation and ARID1A allelic imbalance lost ARID1A protein expression. However, ARID1A protein expression was retained in seven clear cell carcinomas with ARID1A loss-of-function mutations. These results suggest that a single ARID1A loss-of-function mutation is insufficient for ARID1A loss in ovarian endometriosis and some clear cell carcinoma. Further driver events may be needed for the malignant transformation of ovarian endometriosis with ARID1A loss-of-function mutations.
Collapse
Affiliation(s)
- Nozomi Yachida
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan.
| | - Kazuaki Suda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Hirofumi Nakaoka
- Human Genetics Laboratory, National Institute of Genetics, Mishima, 411-8540, Japan.,Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Haruka Ueda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Kentaro Sugino
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Manako Yamaguchi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Yutaro Mori
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Kaoru Yamawaki
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Ryo Tamura
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Tatsuya Ishiguro
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Masanori Isobe
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| | - Teiichi Motoyama
- Department of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Ituro Inoue
- Human Genetics Laboratory, National Institute of Genetics, Mishima, 411-8540, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ward, Niigata, 951-8510, Japan
| |
Collapse
|
31
|
Fukase M, Ohta T, Watanabe N, Suzuki Y, Seino M, Sudo T, Nagase S. Squamous cell carcinoma arising from a mature cystic teratoma of the ovary: Successful treatment with carboplatin, paclitaxel, and bevacizumab. Gynecol Oncol Rep 2020; 34:100632. [PMID: 32964091 PMCID: PMC7490979 DOI: 10.1016/j.gore.2020.100632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/13/2020] [Accepted: 08/23/2020] [Indexed: 11/29/2022] Open
Abstract
Squamous cell carcinoma arising from a mature cystic teratoma of the ovary is rare. The optimal treatment for this disease is unclear. We report a successfully treating case of SCC arising from MCT with carboplatin, paclitaxel, and bevacizumab. Bevacizumab might have been one of the treatment options for patients with advanced-stage of this disease.
Ovarian mature cystic teratomas (MCT) are usually benign. However, squamous cell carcinoma (SCC) develops in 0.17–2% of MCT. Because of its low frequency, the optimal treatment for this disease is unclear. We present a case of SCC arising from MCT. Our patient was a 30-year-old nulliparous woman complaining of right lower abdominal pain, who was referred to our hospital for a pelvic solid mass with suspected malignant ovarian tumor. A diagnosis of SCC arising from MCT was suspected based on the elevation in SCC antigen and the imaging. After surgery to remove the tumor, the diagnosis was confirmed based on the pathology. Although the surgery by radical cytoreduction was optimal, the tumor showed early recurrence. Therefore, we administrated combination chemotherapy, consisting of carboplatin, paclitaxel, and bevacizumab, followed by maintenance therapy with bevacizumab. Response to the chemotherapy regimen was complete and the patient was alive with no evidence of recurrence for 45 months after starting the initial surgery. We discuss the clinical characteristics of the malignant transformation of MCT and the treatment of SCC arising from MCT.
Collapse
Affiliation(s)
- Mika Fukase
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| | - Tsuyoshi Ohta
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| | - Norikazu Watanabe
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| | - Yuriko Suzuki
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| | - Manabu Seino
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| | - Takeshi Sudo
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| | - Satoru Nagase
- Department of Obstetrics Gynecology, Faculty of Medicine, Yamagata University, Iida-nishi, 2-2-2, Yamagata 990-9585, Japan
| |
Collapse
|
32
|
Zhang C, Ma T. Poorer prognosis of ovarian squamous cell carcinoma than serous carcinoma: a propensity score matching analysis based on the SEER database. J Ovarian Res 2020; 13:75. [PMID: 32611433 PMCID: PMC7329546 DOI: 10.1186/s13048-020-00675-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/18/2020] [Indexed: 01/16/2023] Open
Abstract
Background Ovarian squamous cell carcinoma (SCC) is a rare cancer with possible poor survival, however no direct evidence supports this viewpoint and the independent prognostic factors are controversial. Patients with ovarian SCC and serous carcinoma (SC) who were diagnosed between 2004 and 2016 were selected using the recent released SEER database. Propensity score matching was used to balance the characteristics of the two groups. The difference of survival between patients with ovarian SCC and SC was explored using Kaplan-Meier method. Cox regression analyses were performed to further identify the independent prognostic factors among patients with ovarian SCC. Results Of 15,286 patients (15,106 SC cases and 180 SCC cases), 304 were identified in the matched cohort (200 SC cases and 104 SCC cases). The overall survival and cause-specific survival for patients with SCC were significantly poorer (Plog-rank < 0.001). The median survival time was 21 months for SCC and 95 months for SC. Patients who underwent bilateral salpingo-oophorectomy with hysterectomy and omentectomy seemed to have a better outcome. In multivariate analysis, older age at diagnosis, larger tumor size, bilateral and FIGO stage IV malignancy were the independent risk factors for poor disease outcome. Conclusions The prognosis of ovarian SCC is worse than ovarian SC. Older age at diagnosis, advanced disease stage, larger tumor size and bilateral malignancy are the independent risk factors for poor survival.
Collapse
Affiliation(s)
- Cheng Zhang
- Anhui Provincial Cancer Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tai Ma
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
33
|
Zhang H, Qin C, Gan H, Guo X, Zhang L. Construction of an Immunogenomic Risk Score for Prognostication in Colon Cancer. Front Genet 2020; 11:499. [PMID: 32508884 PMCID: PMC7253627 DOI: 10.3389/fgene.2020.00499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 01/18/2023] Open
Abstract
Immune-related genes (IRGs) play regulatory roles in the immune system and are involved in the initiation and progression of colon cancer. This study aimed to develop an immunogenomic risk score for predicting survival outcomes among colon cancer patients. We analyzed the expressions of IRGs in colon specimens and discovered 484 differentially expressed IRGs when we compared specimens from colon cancer and adjacent normal tissue. Univariate Cox regression analyses were performed to identify 26 IRGs that were associated with survival. A Cox proportional hazards model with a lasso penalty identified five optimal IRGs for constructing the immunogenomic risk score (CD1B, XCL1, PLCG2, NGF, and OXTR). The risk score had good performance in predicting overall survival among patients with colon cancer and was correlated with the amount of tumor-infiltrating immune cells. Our findings suggest that the immunogenomic risk score may be useful for prognostication in colon cancer cases. Furthermore, the five IRGs included in the risk score might be useful targets for investigating the initiation of colon cancer and designing personalized treatments.
Collapse
Affiliation(s)
- Han Zhang
- First Clinical Medical College, Chongqing Medical University, Chongqing, China.,Department of Digestive Oncology, Three Gorges Hospital, Chongqing University, Chongqing, China
| | - Chuan Qin
- Department of Gastrointestinal Surgery, Three Gorges Hospital, Chongqing University, Chongqing, China
| | - Hua Gan
- First Clinical Medical College, Chongqing Medical University, Chongqing, China
| | - Xiong Guo
- First Clinical Medical College, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Digestive Oncology, Three Gorges Hospital, Chongqing University, Chongqing, China
| |
Collapse
|