1
|
Wei C, Chen G, Chen K, Fang S, He H. Immune alterations and overexpression of CTCF in endometrial carcinoma: insights from molecular subtyping. Cancer Cell Int 2024; 24:392. [PMID: 39623397 PMCID: PMC11613940 DOI: 10.1186/s12935-024-03576-y] [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: 06/24/2024] [Accepted: 11/14/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Endometrial cancer (EC) is a prevalent epithelial malignancy originating in the female endometrium, and its global incidence has been on the rise over the past decade. Despite significant scientific progress has been achieved recently, the genetic factors underlying EC pathogenesis remain poorly understood, warranting further investigation. METHODS We employed transcriptomic datasets from the Genomic Data Commons database to extract variable and clinical data. Quantile normalization and log2 transformations were applied to obtain a gene expression matrix for the sample cohort. Various assays, such as quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, immunohistochemistry (IHC), wound healing assay, transwell assay, and TUNEL assay, were employed in the study to examine the involvement of CTCF in EC cell biology. Additionally, in vivo experiments were conducted using a subcutaneous transplantation tumor model in athymic nude mice. The potential mechanism of action of CTCF was also illustrated by identifying differentially expressed genes (DEGs) and functions after interfering with CTCF gene expression through the GSPAdb online database. RESULTS After categorizing 543 samples into cohorts with high and low ImmuneScores, we discovered 1025 genes that were differentially expressed, including 745 genes that were up-regulated and 280 genes that were down-regulated in the high scores group compared to the low scores group. Tumor mutation burden (TMB) analysis highlighted 11 genes with the highest mutation frequency. Furthermore, 16 immune checkpoints and 50 immune regulatory factors exhibited differential expression. Among these, CTCF was up-regulated in EC. We found that CTCF knockdown could diminish EC's invasive ability and metastatic potential while enhancing apoptosis. In vivo experiments corroborated that CTCF knockdown could reduce tumor growth. The GSPAdb online database identified differential expression pathways mainly enriched in cellular metabolism as well as some intracellular classical signaling pathways after interfering with CTCF gene expression. In addition, we identified potential downstream regulators of CTCF through protein interaction networks. CONCLUSION This study unveiled comprehensive molecular characteristics and DEGs in EC, emphasizing the up-regulation of CTCF in EC. Our findings collectively suggest that CTCF represents a promising therapeutic target, and our gene molecular typing model offers a novel approach for prognostic evaluation in EC.
Collapse
Affiliation(s)
- Caiping Wei
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Guowei Chen
- Department of Gynecology, Liuzhou Worker's Hospital, Liuzhou, Guangxi, China
| | - Kun Chen
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Shuang Fang
- Department of Gynecology, Liuzhou Worker's Hospital, Liuzhou, Guangxi, China
| | - Hongying He
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China.
- Key Laboratory of Medical Molecular Diagnostics of Liuzhou, Key Laboratory for Nucleic Acid Molecular Diagnosis and Application of Guangxi Health and Wellness Commission, Liuzhou, Guangxi, China.
- Department of Gynecology, GuangZhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou, Guangxi, China.
| |
Collapse
|
2
|
Morgan JE, Jaferi N, Shonibare Z, Huang GS. ARID1A in Gynecologic Precancers and Cancers. Reprod Sci 2024; 31:2150-2162. [PMID: 38740655 DOI: 10.1007/s43032-024-01585-w] [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: 01/05/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
The highest frequency of genetic alterations in the tumor suppressor ARID1A occurs in malignancies of the female reproductive tract. The prevalence of ARID1A alterations in gynecologic precancers and cancers is summarized from the literature, and the putative mechanisms of tumor suppressive action examined both in benign/precursor lesions including endometriosis and atypical hyperplasia and in malignancies of the ovary, uterus, cervix and vagina. ARID1A alterations in gynecologic cancers are usually loss-of-function mutations, resulting in diminished or absent protein expression. ARID1A deficiency results in pleiotropic downstream effects related not only to its role in transcriptional regulation as a SWI/SNF complex subunit, but also related to the functions of ARID1A in DNA replication and repair, immune modulation, cell cycle progression, endoplasmic reticulum (ER) stress and oxidative stress. The most promising actionable signaling pathway interactions and therapeutic vulnerabilities of ARID1A mutated cancers are presented with a critical review of the currently available experimental and clinical evidence. The role of ARID1A in response to chemotherapeutic agents, radiation therapy and immunotherapy is also addressed. In summary, the multi-faceted role of ARID1A mutation in precancer and cancer is examined through a clinical lens focused on development of novel preventive and therapeutic interventions for gynecological cancers.
Collapse
Affiliation(s)
- Jaida E Morgan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Nishah Jaferi
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Zainab Shonibare
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Gloria S Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA.
- Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology, Yale School of Medicine, Yale Cancer Center, Yale University, PO Box 208063, New Haven, CT, 06520-8063, USA.
| |
Collapse
|
3
|
Gatius S, Matias Guiu X, Davidson B. Molecular features for timely cancer diagnosis and treatment - tumors of the ovary, fallopian tube and endometrium. Virchows Arch 2024; 484:339-351. [PMID: 38099957 DOI: 10.1007/s00428-023-03710-7] [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: 07/07/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 03/19/2024]
Abstract
Gynecologic pathology has moved, within only a few years, from being a diagnostic area devoid of molecular testing into a diagnostic discipline in which such analyses are becoming routine. The direct relevance of molecular characterization to the choice of treatment of patients with carcinomas originating in both the uterus and adnexae makes it likely that such testing will only expand along with our understanding of the molecular make-up of these tumors. As a consequence, gynecologic pathologists have become an integral part of patient management, rather than lab personnel providing external services.In parallel, molecular testing is expanding as a tool for diagnosing rare tumors affecting these organs, including soft tissue tumors, sex cord-stromal tumors and germ cell tumors, as well as other rare entities. Increased knowledge in this area bears directly on the ability to diagnose these tumors in a reproducible manner, as well as recognize and consult on genetic diseases. Hopefully, despite the inherent difficulty in studying rare cancers, it will also translate into new therapeutic options for the malignant ones among these rare cancers.
Collapse
Affiliation(s)
- Sonia Gatius
- Department of Pathology, Hospital Universitari Arnau de Vilanova de Lleida, Universitat de Lleida, IRBLleida, CIBERONC, Lleida, Spain.
| | - Xavier Matias Guiu
- Department of Pathology, Hospital Universitari Arnau de Vilanova de Lleida, Universitat de Lleida, IRBLleida, CIBERONC, Lleida, Spain
- Department of Pathology, Hospital Universitari de Bellvitge, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Ben Davidson
- Department of Pathology, Oslo University Hospital Montebello, Norwegian Radium Hospital, N-0310, Oslo, Norway.
- University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316, Oslo, Norway.
| |
Collapse
|
4
|
Wen Y, Ye S, Li Z, Zhang X, Liu C, Wu Y, Zheng R, Xu C, Tian J, Shu L, Yan Q, Ai F, Ma J. HDAC6 inhibitor ACY-1215 enhances STAT1 acetylation to block PD-L1 for colorectal cancer immunotherapy. Cancer Immunol Immunother 2024; 73:7. [PMID: 38231305 PMCID: PMC10794344 DOI: 10.1007/s00262-023-03624-y] [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: 11/01/2023] [Accepted: 12/27/2023] [Indexed: 01/18/2024]
Abstract
The search for effective combination therapy with immune checkpoint inhibitors (ICI) has become important for cancer patients who do not respond to the ICI well. Histone deacetylases (HDACs) inhibitors have attracted wide attention as anti-tumor agents. ACY-1215 is a selective inhibitor of HDAC6, which can inhibit the growth of a variety of tumor. We previously revealed that HDAC family is highly expressed in colorectal cancer specimens and mouse models. In this study, ACY-1215 was combined with anti-PD1 to treat tumor-bearing mice associated with colorectal cancer. ACY-1215 combined with anti-PD1 effectively inhibited the colorectal tumor growth. The expression of PD-L1 in tumor of mice were inhibited by ACY-1215 and anti-PD1 combination treatment, whereas some biomarkers reflecting T cell activation were upregulated. In a co-culture system of T cells and tumor cells, ACY-1215 helped T cells to kill tumor cells. Mechanically, HDAC6 enhanced the acetylation of STAT1 and inhibited the phosphorylation of STAT1, thus preventing STAT1 from entering the nucleus to activate PD-L1 transcription. This study reveals a novel regulatory mechanism of HDAC6 on non-histone substrates, especially on protein acetylation. HDAC6 inhibitors may be of great significance in tumor immunotherapy and related combination strategies.
Collapse
Affiliation(s)
- Yuqing Wen
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Shuyu Ye
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengshuo Li
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Xiaoyue Zhang
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Can Liu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Yangge Wu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Run Zheng
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Chenxiao Xu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Junrui Tian
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Lanjun Shu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China.
| | - Jian Ma
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan, China.
| |
Collapse
|
5
|
Peng J, Xie F, Qin P, Liu Y, Niu H, Sun J, Xue H, Zhao Q, Liu J, Wu J. Recent development of selective inhibitors targeting the HDAC6 as anti-cancer drugs: Structure, function and design. Bioorg Chem 2023; 138:106622. [PMID: 37244230 DOI: 10.1016/j.bioorg.2023.106622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
HDAC6, a member of the histone deacetylase family, mainly is a cytosolic protein and regulates cell growth by acting on non-histone substrates, such as α -tubulin, cortactin, heat shock protein HSP90, programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1), that are closely related to the proliferation, invasion, immune escape and angiogenesis of cancer tissues. The approved drugs targeting the HDACs are all pan-inhibitors and have many side effects due to their lack of selectivity. Therefore, development of selective inhibitors of HDAC6 has attracted much attention in the field of cancer therapy. In this review, we will summarize the relationship between HDAC6 and cancer, and discuss the design strategies of HDAC6 inhibitors for cancer treatment in recent years.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Fei Xie
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Pengxia Qin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yujing Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoqian Niu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jie Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoyu Xue
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Qianlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingqian Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingde Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
6
|
Jiang YK, Shuai YJ, Ding HM, Zhang H, Huang C, Wang L, Sun JY, Wei WJ, Xiao XY, Jiang GS. ARID1A Inactivation Increases Expression of circ0008399 and Promotes Cisplatin Resistance in Bladder Cancer. Curr Med Sci 2023; 43:560-571. [PMID: 37142816 DOI: 10.1007/s11596-023-2731-8] [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: 01/02/2023] [Accepted: 02/20/2023] [Indexed: 05/06/2023]
Abstract
OBJECTIVE Cisplatin (CDDP)-based chemotherapy is a first-line, drug regimen for muscle-invasive bladder cancer (BC) and metastatic bladder cancer. Clinically, resistance to CDDP restricts the clinical benefit of some bladder cancer patients. AT-rich interaction domain 1A (ARID1A) gene mutation occurs frequently in bladder cancer; however, the role of CDDP sensitivity in BC has not been studied. METHODS We established ARID1A knockout BC cell lines using CRISPR/Cas9 technology. IC50 determination, flow cytometry analysis of apoptosis, and tumor xenograft assays were performed to verify changes in the CDDP sensitivity of BC cells losing ARID1A. qRT-PCR, Western blotting, RNA interference, bioinformatic analysis, and ChIP-qPCR analysis were performed to further explore the potential mechanism of ARID1A inactivation in CDDP sensitivity in BC. RESULTS It was found that ARID1A inactivation was associated with CDDP resistance in BC cells. Mechanically, loss of ARID1A promoted the expression of eukaryotic translation initiation factor 4A3 (EIF4A3) through epigenetic regulation. Increased expression of EIF4A3 promoted the expression of hsa_circ_0008399 (circ0008399), a novel circular RNA (circRNA) identified in our previous study, which, to some extent, showed that ARID1A deletion caused CDDP resistance through the inhibitory effect of circ0008399 on the apoptosis of BC cells. Importantly, EIF4A3-IN-2 specifically inhibited the activity of EIF4A3 to reduce circ0008399 production and restored the sensitivity of ARID1A inactivated BC cells to CDDP. CONCLUSION Our research deepens the understanding of the mechanisms of CDDP resistance in BC and elucidates a potential strategy to improve the efficacy of CDDP in BC patients with ARID1A deletion through combination therapy targeting EIF4A3.
Collapse
Affiliation(s)
- Yang-Kai Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu-Jun Shuai
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua-Min Ding
- Department of Urology, Jingshan Union Hospital of Huazhong University of Science and Technology (People's Hospital of Jingshan), Jingshan, 431899, China
| | - Hui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia-Yin Sun
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wen-Jie Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xing-Yuan Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Guo-Song Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
Sun D, Zhao H, Zhou H, Tao J, Li T, Zhu J, Hou H. ARID1A deficiency associated with MMR deficiency and a high abundance of tumor-infiltrating lymphocytes predicts a good prognosis of endometrial carcinoma. Transl Oncol 2023; 33:101685. [PMID: 37137217 PMCID: PMC10176062 DOI: 10.1016/j.tranon.2023.101685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND ARID1A alterations have been detected in 40% of endometrial carcinomas (ECs) and are associated with loss of its expression. The role of ARID1A in tumorigenesis and development is complex, and the prognostic role in EC remains controversial. Hence, it is of great significance to confirm the role of ARID1A in EC. METHODS A total of 549 EC patients (cohort A) from TCGA were evaluated to explore the prognostic role of ARID1A. NGS was performed for 13 EC patients (cohort B), and expression of ARID1A, CD3, CD8 and mismatch repair (MMR) proteins in 52 patients (cohort C) from our center was determined by immunohistochemistry (IHC). The Kaplan-Meier method was used to perform survival analyses. RESULTS ARID1A alterations were detected in 32% of EC patients and correlated with good disease-free survival (DFS, P = 0.004) and overall survival (OS, P = 0.0353). ARID1A alterations were found to co-occur with MMR-related gene mutations and correlated with higher PD-L1 expression. Patients concomitantly harboring ARID1A alterations and MMR-related gene mutations had the best prognosis (DFS: P = 0.0488; OS: P = 0.0024). A cohort from our center showed that ARID1A deficiency was an independent prognostic factor and predicted longer recurrence-free survival (P = 0.0476). ARID1A loss was associated with a tendency toward MSI-H (P = 0.0060). ARID1A alterations and expression loss were associated with a higher abundance of CD3+ (P = 0.0406) and CD8+ (P = 0.0387) T cells. CONCLUSION ARID1A alterations and expression loss are tightly associated with MMR deficiency and a high abundance of tumor-infiltrating lymphocytes, which might contribute to the good prognosis of EC.
Collapse
Affiliation(s)
- Dantong Sun
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Han Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, 266000, Qingdao, China
| | - Hai Zhou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Junyan Tao
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Tianjun Li
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Jingjuan Zhu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Helei Hou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
8
|
Jo H, Shim K, Jeoung D. Targeting HDAC6 to Overcome Autophagy-Promoted Anti-Cancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23179592. [PMID: 36076996 PMCID: PMC9455701 DOI: 10.3390/ijms23179592] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Histone deacetylases (HDACs) regulate gene expression through the epigenetic modification of chromatin structure. HDAC6, unlike many other HDACs, is present in the cytoplasm. Its deacetylates non-histone proteins and plays diverse roles in cancer cell initiation, proliferation, autophagy, and anti-cancer drug resistance. The development of HDAC6-specific inhibitors has been relatively successful. Mechanisms of HDAC6-promoted anti-cancer drug resistance, cancer cell proliferation, and autophagy are discussed. The relationship between autophagy and anti-cancer drug resistance is discussed. The effects of combination therapy, which includes HDAC6 inhibitors, on the sensitivity of cancer cells to chemotherapeutics and immune checkpoint blockade are presented. A summary of clinical trials involving HDAC6-specific inhibitors is also presented. This review presents HDAC6 as a valuable target for developing anti-cancer drugs.
Collapse
|
9
|
Li J, Yu M, Fu S, Liu D, Tan Y. Role of Selective Histone Deacetylase 6 Inhibitor ACY-1215 in Cancer and Other Human Diseases. Front Pharmacol 2022; 13:907981. [PMID: 35652048 PMCID: PMC9149003 DOI: 10.3389/fphar.2022.907981] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 01/03/2023] Open
Abstract
The deacetylation process regulated by histone deacetylases (HDACs) plays an important role in human health and diseases. HDAC6 belongs to the Class IIb of HDACs family, which mainly modifies non-histone proteins located in the cytoplasm. HDAC6 plays a key role in tumors, neurological diseases, and inflammatory diseases. Therefore, targeting HDAC6 has become a promising treatment strategy in recent years. ACY-1215 is the first orally available highly selective HDAC6 inhibitor, and its efficacy and therapeutic effects are being continuously verified. This review summarizes the research progress of ACY-1215 in cancer and other human diseases, as well as the underlying mechanism, in order to guide the future clinical trials of ACY-1215 and more in-depth mechanism researches.
Collapse
Affiliation(s)
- Jianglei Li
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Meihong Yu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Shifeng Fu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Yuyong Tan
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| |
Collapse
|