1
|
Yeh CY, Aguirre K, Laveroni O, Kim S, Wang A, Liang B, Zhang X, Han LM, Valbuena R, Bassik MC, Kim YM, Plevritis SK, Snyder MP, Howitt BE, Jerby L. Mapping spatial organization and genetic cell-state regulators to target immune evasion in ovarian cancer. Nat Immunol 2024; 25:1943-1958. [PMID: 39179931 PMCID: PMC11436371 DOI: 10.1038/s41590-024-01943-5] [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: 10/04/2023] [Accepted: 07/25/2024] [Indexed: 08/26/2024]
Abstract
The drivers of immune evasion are not entirely clear, limiting the success of cancer immunotherapies. Here we applied single-cell spatial and perturbational transcriptomics to delineate immune evasion in high-grade serous tubo-ovarian cancer. To this end, we first mapped the spatial organization of high-grade serous tubo-ovarian cancer by profiling more than 2.5 million cells in situ in 130 tumors from 94 patients. This revealed a malignant cell state that reflects tumor genetics and is predictive of T cell and natural killer cell infiltration levels and response to immune checkpoint blockade. We then performed Perturb-seq screens and identified genetic perturbations-including knockout of PTPN1 and ACTR8-that trigger this malignant cell state. Finally, we show that these perturbations, as well as a PTPN1/PTPN2 inhibitor, sensitize ovarian cancer cells to T cell and natural killer cell cytotoxicity, as predicted. This study thus identifies ways to study and target immune evasion by linking genetic variation, cell-state regulators and spatial biology.
Collapse
Grants
- P30 CA124435 NCI NIH HHS
- U01 HG012069 NHGRI NIH HHS
- L.J. holds a Career Award at the Scientific Interface from the Burroughs Wellcome Fund (BWF) and a Liz Tilberis Early Career Award from the Ovarian Cancer Research Alliance (OCRA). This study was supported by the BWF (1019508.01; L.J.), National Human Genome Research Institute (NHGRI, U01HG012069; L.J.), OCRA (889076; L.J), Under One Umbrella, Stanford Women’s Cancer Center, Stanford Cancer Institute, a National Cancer Institute (NCI)-designated Comprehensive Cancer Center (251217; B.E.H., L.J.), as well as funds from the Departments of Genetics (L.J.) at Stanford University and from the Chan Zuckerberg Biohub (L.J.).
- This study was partially supported by the Stanford Women’s Cancer Center (251217; B.E.H., L.J.), and an NCI Center Support Grant (P30CA124435; B.E.H.), as well as funds from the Departments of Pathology (B.E.H.).
Collapse
Affiliation(s)
- Christine Yiwen Yeh
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Karmen Aguirre
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Olivia Laveroni
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Subin Kim
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Aihui Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooke Liang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoming Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lucy M Han
- Department of Pathology, California Pacific Medical Center, San Francisco, CA, USA
| | - Raeline Valbuena
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Young-Min Kim
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooke E Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Livnat Jerby
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
2
|
Xing L, Wang Z, Feng Y, Luo H, Dai G, Sang L, Zhang C, Qian J. The biological roles of CD47 in ovarian cancer progression. Cancer Immunol Immunother 2024; 73:145. [PMID: 38832992 PMCID: PMC11150368 DOI: 10.1007/s00262-024-03708-3] [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: 03/18/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
Ovarian cancer is one of the most lethal malignant tumors, characterized by high incidence and poor prognosis. Patients relapse occurred in 65-80% after initial treatment. To date, no effective treatment has been established for these patients. Recently, CD47 has been considered as a promising immunotherapy target. In this paper, we reviewed the biological roles of CD47 in ovarian cancer and summarized the related mechanisms. For most types of cancers, the CD47/Sirpα immune checkpoint has attracted the most attention in immunotherapy. Notably, CD47 monoclonal antibodies and related molecules are promising in the immunotherapy of ovarian cancer, and further research is needed. In the future, new immunotherapy regimens targeting CD47 can be applied to the clinical treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Linan Xing
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Zhao Wang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Yue Feng
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Haixia Luo
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Guijiang Dai
- Department of Comprehensive Office, The Second Affiliated Hospital of MuDanjiang Medical University, Mudanjiang, 157009, People's Republic of China
| | - Lin Sang
- Department of Obstetrics and Gynecology, People's Hospital of Anji, Huzhou, 310022, People's Republic of China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
3
|
Ma M, Wang C, Wu M, Gu S, Yang J, Zhang Y, Cheng S, Xu S, Zhang M, Wu Y, Zhao Y, Tian X, Voon DCC, Takahashi C, Sheng J, Wang Y. CSGALNACT2 restricts ovarian cancer migration and invasion by modulating MAPK/ERK pathway through DUSP1. Cell Oncol (Dordr) 2024; 47:897-915. [PMID: 38082211 PMCID: PMC11219422 DOI: 10.1007/s13402-023-00903-9] [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] [Accepted: 11/23/2023] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Ovarian cancer is one of the leading causes of cancer-related death among women. CSGALNACT2 is a vital Golgi transferase and is related to a variety of human diseases. However, its expression pattern and function in ovarian cancer remain uncertain. METHODS The Cancer Genome Atlas and GEPIA databases were used to assess the expression of CSGALNACT2 in ovarian cancer patients. RNA-seq, qRT-PCR, and IHC were used to verify the expression of CSGALNACT2 in ovarian cancer tissues. Then, in vivo and in vitro experiments were conducted to evaluate the role of CSGALNACT2 in the progression of ovarian cancer. RNA-seq and GSEA were used to reveal the potential biological function and oncogenic pathways of CSGALNACT2. RESULTS We demonstrated that the mRNA expression and protein level of CSGALNACT2 were significantly downregulated in ovarian cancer and ovarian cancer metastatic tissues. CSGALNACT2 can significantly inhibit the migration, invasion, and clonogenic growth of ovarian cancer in vitro and is progressively lost during ovarian cancer progression in vivo. CSGALNACT2 suppresses ovarian cancer migration and invasion via DUSP1 modulation of the MAPK/ERK pathway through RNA-seq, KEGG analysis, and Western blotting. Moreover, CSGALNACT2 expression was correlated with immune cell infiltration and had prognostic value in different immune cell-enriched or decreased ovarian cancer. In addition, patients with CSGALNACT2 downregulation are less likely to benefit from immunotherapy. CONCLUSION As an ovarian cancer suppressor gene, CSGALNACT2 inhibits the development of ovarian cancer, and it might be used as a prognostic biomarker in patients with ovarian cancer.
Collapse
Affiliation(s)
- Mingjun Ma
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Chao Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Meixuan Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sijia Gu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiani Yang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yue Zhang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shanshan Cheng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shilin Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongsong Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yaqian Zhao
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiu Tian
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | | | - Chiaki Takahashi
- Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Jindan Sheng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Yu Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
4
|
Deng M, Tang F, Chang X, Liu P, Ji X, Hao M, Wang Y, Yang R, Ma Q, Zhang Y, Miao J. Immunotherapy for Ovarian Cancer: Disappointing or Promising? Mol Pharm 2024; 21:454-466. [PMID: 38232985 DOI: 10.1021/acs.molpharmaceut.3c00986] [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] [Indexed: 01/19/2024]
Abstract
Ovarian cancer, one of the deadliest malignancies, lacks effective treatment, despite advancements in surgical techniques and chemotherapy. Thus, new therapeutic approaches are imperative to improving treatment outcomes. Immunotherapy, which has demonstrated considerable success in managing various cancers, has already found its place in clinical practice. This review aims to provide an overview of ovarian tumor immunotherapy, including its basics, key strategies, and clinical research data supporting its potential. In particular, this discussion highlights promising strategies such as checkpoint inhibitors, vaccines, and pericyte transfer, both individually and in combination. However, the advancement of new immunotherapies necessitates large controlled randomized trials, which will undoubtedly shape the future of ovarian cancer treatment.
Collapse
Affiliation(s)
- Mengqi Deng
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Fan Tang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Xiangyu Chang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Penglin Liu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Xuechao Ji
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Menglin Hao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Yixiao Wang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Ruiye Yang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Qingqing Ma
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
- Nanyuan Hospital of Fengtai District, Beijing 100006, China
| | - Yubo Zhang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Shandong 266011, China
| | - Jinwei Miao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| |
Collapse
|
5
|
Chen L, Gao W, Lin L, Sha C, Li T, Chen Q, Wei H, Yang M, Xing J, Zhang M, Zhao S, Xu W, Li Y, Long L, Zhu X. A methylation- and immune-related lncRNA signature to predict ovarian cancer outcome and uncover mechanisms of chemoresistance. J Ovarian Res 2023; 16:186. [PMID: 37674251 PMCID: PMC10483746 DOI: 10.1186/s13048-023-01260-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/13/2023] [Indexed: 09/08/2023] Open
Abstract
Tumor-associated lncRNAs regulated by epigenetic modification switches mediate immune escape and chemoresistance in ovarian cancer (OC). However, the underlying mechanisms and concrete targets have not been systematically elucidated. Here, we discovered that methylation modifications played a significant role in regulating immune cell infiltration and sensitizing OC to chemotherapy by modulating immune-related lncRNAs (irlncRNAs), which represent tumor immune status. Through deep analysis of the TCGA database, a prognostic risk model incorporating four methylation-related lncRNAs (mrlncRNAs) and irlncRNAs was constructed. Twenty-one mrlncRNA/irlncRNA pairs were identified that were significantly related to the overall survival (OS) of OC patients. Subsequently, we selected four lncRNAs to construct a risk signature predictive of OS and indicative of OC immune infiltration, and verified the robustness of the risk signature in an internal validation set. The risk score was an independent prognostic factor for OC prognosis, which was demonstrated via multifactorial Cox regression analysis and nomogram. Moreover, risk scores were negatively related to the expression of CD274, CTLA4, ICOS, LAG3, PDCD1, and PDCD1LG2 and negatively correlated with CD8+, CD4+, and Treg tumor-infiltrating immune cells. In addition, a high-risk score was associated with a higher IC50 value for cisplatin, which was associated with a significantly worse clinical outcome. Next, a competing endogenous RNA (ceRNA) network and a signaling pathway controlling the infiltration of CD8+ T cells were explored based on the lncRNA model, which suggested a potential therapeutic target for immunotherapy. Overall, this study constructed a prognostic model by pairing mrlncRNAs and irlncRNAs and revealed the critical role of the FTO/RP5-991G20.1/hsa-miR-1976/MEIS1 signaling pathway in regulating immune function and enhancing anticancer therapy.
Collapse
Affiliation(s)
- Lu Chen
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, Taixing People's Hospital, Taixing, Jiangsu, China
| | - Wujiang Gao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, Yangzhou First People's Hospital, Yangzhou, Jiangsu, China
| | - Li Lin
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Chunli Sha
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, The First People's Hospital of Nantong City, Nantong, Jiangsu, China
| | - Taoqiong Li
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Qi Chen
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Hong Wei
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Meiling Yang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
- Department of Gynaecology and Obstetrics, The First People's Hospital of Nantong City, Nantong, Jiangsu, China
| | - Jie Xing
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Mengxue Zhang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Shijie Zhao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China
| | - Wenlin Xu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China.
| | - Yuefeng Li
- Medical school, Jiangsu University, No. 301, Xuefu Road, Zhenjiang City, 212031, Jiangsu Province, China.
| | - Lulu Long
- Oncology Department, Affiliated People's Hospital of jiangsu university, No. 8, Dianli Road, Zhenjiang City, 212001, Jiangsu Province, China.
| | - Xiaolan Zhu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang City, 212001, Jiangsu Province, China.
| |
Collapse
|
6
|
Pawłowska A, Rekowska A, Kuryło W, Pańczyszyn A, Kotarski J, Wertel I. Current Understanding on Why Ovarian Cancer Is Resistant to Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:10859. [PMID: 37446039 DOI: 10.3390/ijms241310859] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The standard treatment of ovarian cancer (OC) patients, including debulking surgery and first-line chemotherapy, is unsatisfactory because of recurrent episodes in the majority (~70%) of patients with advanced OC. Clinical trials have shown only a modest (10-15%) response of OC individuals to treatment based on immune checkpoint inhibitors (ICIs). The resistance of OC to therapy is caused by various factors, including OC heterogeneity, low density of tumor-infiltrating lymphocytes (TILs), non-cellular and cellular interactions in the tumor microenvironment (TME), as well as a network of microRNA regulating immune checkpoint pathways. Moreover, ICIs are the most efficient in tumors that are marked by high microsatellite instability and high tumor mutation burden, which is rare among OC patients. The great challenge in ICI implementation is connected with distinguishing hyper-, pseudo-, and real progression of the disease. The understanding of the immunological, molecular, and genetic mechanisms of OC resistance is crucial to selecting the group of OC individuals in whom personalized treatment would be beneficial. In this review, we summarize current knowledge about the selected factors inducing OC resistance and discuss the future directions of ICI-based immunotherapy development for OC patients.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Department of Oncological Gynaecology and Gynaecology, Faculty of Medicine, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Anna Rekowska
- Students' Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Weronika Kuryło
- Students' Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Anna Pańczyszyn
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland
| | - Jan Kotarski
- Independent Laboratory of Cancer Diagnostics and Immunology, Department of Oncological Gynaecology and Gynaecology, Faculty of Medicine, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Department of Oncological Gynaecology and Gynaecology, Faculty of Medicine, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| |
Collapse
|
7
|
Mishra Y, Chattaraj A, Mishra V, Ranjan A, Tambuwala MM. Aptamers Versus Vascular Endothelial Growth Factor (VEGF): A New Battle against Ovarian Cancer. Pharmaceuticals (Basel) 2023; 16:849. [PMID: 37375796 DOI: 10.3390/ph16060849] [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: 04/10/2023] [Revised: 05/28/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer is one of the diseases that causes a high mortality as it involves unregulated and abnormal cell growth proliferation that can manifest in any body region. One of the typical ovarian cancer symptoms is damage to the female reproductive system. The death rate can be reduced through early detection of the ovarian cancer. Promising probes that can detect ovarian cancer are suitable aptamers. Aptamers, i.e., so-called chemical antibodies, have a strong affinity for the target biomarker and can typically be identified starting from a random library of oligonucleotides. Compared with other probes, ovarian cancer targeting using aptamers has demonstrated superior detection effectiveness. Various aptamers have been selected to detect the ovarian tumor biomarker, vascular endothelial growth factor (VEGF). The present review highlights the development of particular aptamers that target VEGF and detect ovarian cancer at its earliest stages. The therapeutic efficacy of aptamers in ovarian cancer treatment is also discussed.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, UK
| |
Collapse
|
8
|
Pawłowska A, Skiba W, Suszczyk D, Kuryło W, Jakubowicz-Gil J, Paduch R, Wertel I. The Dual Blockade of the TIGIT and PD-1/PD-L1 Pathway as a New Hope for Ovarian Cancer Patients. Cancers (Basel) 2022; 14:5757. [PMID: 36497240 PMCID: PMC9740841 DOI: 10.3390/cancers14235757] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The prognosis for ovarian cancer (OC) patients is poor and the five-year survival rate is only 47%. Immune checkpoints (ICPs) appear to be the potential targets in up-and-coming OC treatment. However, the response of OC patients to immunotherapy based on programmed cell death pathway (PD-1/PD-L1) inhibitors totals only 6-15%. The promising approach is a combined therapy, including other ICPs such as the T-cell immunoglobulin and ITIM domain/CD155/DNAX accessory molecule-1 (TIGIT/CD155/DNAM-1) axis. Preclinical studies in a murine model of colorectal cancer showed that the dual blockade of PD-1/PD-L1 and TIGIT led to remission in the whole studied group vs. the regression of the tumors with the blockade of a single pathway. The approach stimulates the effector activity of T cells and NK cells, and redirects the immune system activity against the tumor. The understanding of the synergistic action of the TIGIT and PD-1/PD-L1 blockade is, however, poor. Thus, the aim of this review is to summarize the current knowledge about the mode of action of the dual TIGIT and PD-1/PD-L1 blockade and its potential benefits for OC patients. Considering the positive impact of this combined therapy in malignancies, including lung and colorectal cancer, it appears to be a promising approach in OC treatment.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiktoria Skiba
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Weronika Kuryło
- Students’ Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
9
|
Dumitru A, Dobrica EC, Croitoru A, Cretoiu SM, Gaspar BS. Focus on PD-1/PD-L1 as a Therapeutic Target in Ovarian Cancer. Int J Mol Sci 2022; 23:ijms232012067. [PMID: 36292922 PMCID: PMC9603705 DOI: 10.3390/ijms232012067] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is considered one of the most aggressive and deadliest gynecological malignancies worldwide. Unfortunately, the therapeutic methods that are considered the gold standard at this moment are associated with frequent recurrences. Survival in ovarian cancer is associated with the presence of a high number of intra tumor infiltrating lymphocytes (TILs). Therefore, immunomodulation is considered to have an important role in cancer treatment, and immune checkpoint inhibitors may be useful for restoring T cell-mediated antitumor immunity. However, the data presented in the literature until now are not sufficient to allow for the identification and selection of patients who really respond to immunotherapy among those with ovarian cancer. Although there are some studies with favorable results, more prospective trials are needed in this sense. This review focuses on the current and future perspectives of PD-1/L1 blockade in ovarian cancer and analyzes the most important immune checkpoint inhibitors used, with the aim of achieving optimal clinical outcomes. Future studies and trials are needed to maximize the efficacy of immune checkpoint blockade therapy in ovarian cancer, as well as in all cancers, in general.
Collapse
Affiliation(s)
- Adrian Dumitru
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Pathology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Elena-Codruta Dobrica
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Dermatology, Elias University Hospital, 011461 Bucharest, Romania
| | - Adina Croitoru
- Department of Medical Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Oncology, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Correspondence:
| | - Bogdan Severus Gaspar
- Surgery Department, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
10
|
Fanale D, Corsini LR, Brando C, Cutaia S, Di Donna MC, Filorizzo C, Lisanti MC, Randazzo U, Magrin L, Romano R, Bazan Russo TD, Olive D, Vieni S, Pantuso G, Chiantera V, Russo A, Bazan V, Iovanna JL. Can circulating PD-1, PD-L1, BTN3A1, pan-BTN3As, BTN2A1 and BTLA levels enhance prognostic power of CA125 in patients with advanced high-grade serous ovarian cancer? Front Oncol 2022; 12:946319. [PMID: 36212445 PMCID: PMC9532861 DOI: 10.3389/fonc.2022.946319] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The most common subtype of ovarian cancer (OC) is the high-grade serous ovarian carcinoma (HGSOC), accounting for 70%–80% of all OC deaths. Although HGSOC is a potentially immunogenic tumor, clinical studies assessing the effectiveness of inhibitors of programmed death protein and its ligand (PD-1/PD-L1) in OC patients so far showed only response rates <15%. However, recent studies revealed an interesting prognostic role of plasma PD-1/PD-L1 and other circulating immunoregulatory molecules, such as the B- and T-lymphocyte attenuator (BTLA), butyrophilin sub-family 3A/CD277 receptors (BTN3A), and butyrophilin sub-family 2 member A1 (BTN2A1), in several solid tumors. Since evidence showed the prognostic relevance of pretreatment serum CA125 levels in OC, the aim of our study was to investigate if soluble forms of inhibitory immune checkpoints can enhance prognostic power of CA125 in advanced HGSOC women. Using specific ELISA tests, we examined the circulating PD-1, PD-L1, pan-BTN3As, BTN3A1, BTN2A1, and BTLA levels in 100 advanced HGSOC patients before treatment, correlating them with baseline serum CA125, age at diagnosis, body mass index (BMI), and peritoneal carcinomatosis. A multivariate analysis revealed that plasma BTN3A1 ≤4.75 ng/ml (HR, 1.94; 95% CI, 1.23–3.07; p=0.004), age at diagnosis ≤60 years (HR, 1.65; 95% CI, 1.05–2.59; p=0.03) and absence of peritoneal carcinomatosis (HR, 2.65; 95% CI, 1.66–4.22; p<0.0001) were independent prognostic factors for a longer progression-free survival (PFS) (≥30 months) in advanced HGSOC women. However, further two-factor multivariate analyses highlighted that baseline serum CA125 levels >401 U/ml and each soluble protein above respective concentration cutoff were covariates associated with shorter PFS (<30 months) and unfavorable clinical outcome, suggesting that contemporary measurement of both biomarkers than CA125 only could strengthen prognostic power of serum CA125 in predicting PFS of advanced HGSOC women. Plasma PD-L1, PD-1, BTN3A1, pan-sBTN3As, BTN2A1, or BTLA levels could be helpful biomarkers to increase prognostic value of CA125.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Sofia Cutaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | | | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Maria Chiara Lisanti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Raffaella Romano
- Department of Gynecologic Oncology, University of Palermo, Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1068, Centre National de la Recherche Scientifique Unité Mixte de Recherche (CNRS UMR) 7258 Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Salvatore Vieni
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Gianni Pantuso
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Vito Chiantera
- Department of Gynecologic Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
- *Correspondence: Viviana Bazan, ; Antonio Russo,
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
- *Correspondence: Viviana Bazan, ; Antonio Russo,
| | - Juan Lucio Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1068, Centre National de la Recherche Scientifique Unité Mixte de Recherche (CNRS UMR) 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
11
|
Fanale D, Dimino A, Pedone E, Brando C, Corsini LR, Filorizzo C, Fiorino A, Lisanti MC, Magrin L, Randazzo U, Bazan Russo TD, Russo A, Bazan V. Prognostic and Predictive Role of Tumor-Infiltrating Lymphocytes (TILs) in Ovarian Cancer. Cancers (Basel) 2022; 14:4344. [PMID: 36139508 PMCID: PMC9497073 DOI: 10.3390/cancers14184344] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decade, tumor-infiltrating lymphocytes (TILs) have been recognized as clinically relevant prognostic markers for improved survival, providing the immunological basis for the development of new therapeutic strategies and showing a significant prognostic and predictive role in several malignancies, including ovarian cancer (OC). In fact, many OCs show TILs whose typology and degree of infiltration have been shown to be strongly correlated with prognosis and survival. The OC histological subtype with the higher presence of TILs is the high-grade serous carcinoma (HGSC) followed by the endometrioid subtype, whereas mucinous and clear cell OCs seem to contain a lower percentage of TILs. The abundant presence of TILs in OC suggests an immunogenic potential for this tumor. Despite the high immunogenic potential, OC has been described as a highly immunosuppressive tumor with a high expression of PD1 by TILs. Although further studies are needed to better define their role in prognostic stratification and the therapeutic implication, intraepithelial TILs represent a relevant prognostic factor to take into account in OC. In this review, we will discuss the promising role of TILs as markers which are able to reflect the anticancer immune response, describing their potential capability to predict prognosis and therapy response in OC.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessandra Dimino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Maria Chiara Lisanti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
12
|
Wu W, Zhou S, Liu T, Liang D. Mitochondrial transcription factor B2 overexpression increases M2 macrophage infiltration via cytosolic mitochondrial DNA-stimulated Interleukin-6 secretion in ovarian cancer. Bioengineered 2022; 13:12211-12223. [PMID: 35577351 PMCID: PMC9275939 DOI: 10.1080/21655979.2022.2074615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mitochondrial transcription factor B2 (TFB2M) is a protein modulating both mitochondrial DNA (mtDNA) transcription and compacting. In this study, we explored the expression profile of TFB2M in ovarian cancer, its association with infiltration of tumor-associated macrophages (TAMs), and its influence on macrophage polarization. Serial sections of ovarian cancer tissue arrays were stained to detect TFB2M and CD163 expression. Epithelial ovarian cancer cell line OVISE and CAOV4 were used to assess the influence of TFB2M on IL-6 expression. THP-1 cells were utilized as an in vitro model for macrophage migration and polarization. Results showed that higher TFB2M expression is associated with poor survival in ovarian cancer patients. IHC staining confirmed a moderately positive correlation between TFB2M expression and the infiltration of CD163-positive cells in 68 primary ovarian cancer cases. TFB2M overexpression was associated with increased mtDNA outside the mitochondria and elevated IL-6 expression in ovarian cancer cells. When cytosolic mtDNA was selectively inhibited by DNase I, TFB2M-induced IL-6 upregulation was canceled. TFB2M overexpression could activate the nuclear factor kappa-B (NF-κB) signaling pathway via promoting nucleus entry of p65 and p-p65, which was abrogated by inhibiting cytosolic mtDNA, TLR9, or NF-κB signaling pathway. Conditioned medium from OIVSE cells with TFB2M overexpression could induce macrophage migration and M2 polarization. However, these inducing effects were abrogated by DNase I, TLR9 inhibitor, and anti-IL-6 R pretreatment. In conclusion, this study showed a novel role of TFB2M in the immunosuppressive tumor microenvironment. It promotes M2 macrophage infiltration via a cytosolic mtDNA/TLR9/NF-κB/IL-6 pathway in ovarian cancer.
Collapse
Affiliation(s)
- Weilu Wu
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Shijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tianmin Liu
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Dongni Liang
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
13
|
Tumor-reactive antibodies evolve from non-binding and autoreactive precursors. Cell 2022; 185:1208-1222.e21. [PMID: 35305314 DOI: 10.1016/j.cell.2022.02.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/20/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022]
Abstract
The tumor microenvironment hosts antibody-secreting cells (ASCs) associated with a favorable prognosis in several types of cancer. Patient-derived antibodies have diagnostic and therapeutic potential; yet, it remains unclear how antibodies gain autoreactivity and target tumors. Here, we found that somatic hypermutations (SHMs) promote antibody antitumor reactivity against surface autoantigens in high-grade serous ovarian carcinoma (HGSOC). Patient-derived tumor cells were frequently coated with IgGs. Intratumoral ASCs in HGSOC were both mutated and clonally expanded and produced tumor-reactive antibodies that targeted MMP14, which is abundantly expressed on the tumor cell surface. The reversion of monoclonal antibodies to their germline configuration revealed two types of classes: one dependent on SHMs for tumor binding and a second with germline-encoded autoreactivity. Thus, tumor-reactive autoantibodies are either naturally occurring or evolve through an antigen-driven selection process. These findings highlight the origin and potential applicability of autoantibodies directed at surface antigens for tumor targeting in cancer patients.
Collapse
|
14
|
Chung YM, Tsai WB, Khan PP, Ma J, Berek JS, Larrick JW, Hu MCT. FOXO3-dependent suppression of PD-L1 promotes anticancer immune responses via activation of natural killer cells. Am J Cancer Res 2022; 12:1241-1263. [PMID: 35411241 PMCID: PMC8984903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023] Open
Abstract
Boosting anticancer immunity by blocking immune checkpoints such as the programmed death-1 (PD-1) or its ligand (PD-L1) is a breakthrough anticancer therapy. However, many cancer patients do not respond well to immune checkpoint blockades (ICBs) alone. Here we show that low-dose pharmacological immunoactivators (e.g., SN38, topotecan, sorafenib, etc.) notably downregulate PD-L1 and upregulate FOXO3 expression in various human and murine cancer cell lines. In a mouse tumor model, low-dose SN38 treatment markedly suppresses tumor growth, reduces PD-L1 expression, and enhances FOXO3 expression in primary tumor specimens. SN38 therapy engages the tumor-infiltrating mouse NK1.1/CD49b/NKG2D-positive natural killer (NK) cells to attack tumor cells by inducing mouse IFN-γ and granzyme-B secretion in the tumor microenvironment (TME) in vivo. SN38 treatment also promotes tumor cell apoptosis in the TME. SN38 treatment significantly decreases STAT3-pY705 and IL-6 protein levels; FOXO3 is essential for SN38-mediated PD-L1 downregulation. Collectively, these findings may contribute to future translational or clinical investigations tackling difficult-to-treat cancers with immune-activating medicines or combined with ICB immunotherapy.
Collapse
Affiliation(s)
- Young Min Chung
- Panorama Institute of Molecular Medicine & Panorama Research InstituteSunnyvale, CA 94089, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stanford University School of MedicineStanford, CA 94305, USA
| | - Wen Bin Tsai
- Panorama Institute of Molecular Medicine & Panorama Research InstituteSunnyvale, CA 94089, USA
- Department of Genomic Medicine, University of Texas M. D. Anderson Cancer CenterHouston, TX 77030, USA
| | - Pragya P Khan
- Panorama Institute of Molecular Medicine & Panorama Research InstituteSunnyvale, CA 94089, USA
| | - Jessica Ma
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stanford University School of MedicineStanford, CA 94305, USA
| | - Jonathan S Berek
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stanford University School of MedicineStanford, CA 94305, USA
| | - James W Larrick
- Panorama Institute of Molecular Medicine & Panorama Research InstituteSunnyvale, CA 94089, USA
| | - Mickey C-T Hu
- Panorama Institute of Molecular Medicine & Panorama Research InstituteSunnyvale, CA 94089, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stanford University School of MedicineStanford, CA 94305, USA
| |
Collapse
|
15
|
Nikas IP, Lee C, Song MJ, Kim B, Ryu HS. Biomarkers expression among paired serous ovarian cancer primary lesions and their peritoneal cavity metastases in treatment-naïve patients: A single-center study. Cancer Med 2022; 11:2193-2203. [PMID: 35212471 PMCID: PMC9160817 DOI: 10.1002/cam4.4600] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/30/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022] Open
Abstract
Background High‐grade serous ovarian carcinoma (HGSOC), the most common histologic subtype of ovarian epithelial cancer, is associated with treatment resistance, enhanced recurrence rates, and poor prognosis. HGSOCs often metastasize to the peritoneal cavity, while fluid cytology examination could identify such metastases. This retrospective study aimed to identify potential biomarker discrepancies between paired HGSOC primary tissues and metastatic peritoneal fluid cytology samples, processed as cell blocks (CBs). Methods Twenty‐four pairs of formalin‐fixed, paraffin‐embedded primary tissues and metastatic CBs from an equal number of treatment‐naïve patients were used, and immunohistochemistry (IHC) for epidermal growth factor receptor (EGFR), human epidermal growth factor receptor, programmed cell death‐1 ligand 1 (PD‐L1), and CD147 was applied. Results 13/24 pairs showed discordant EGFR IHC results; in all these 13 patients, EGFR was positive (≥1+ membranous staining intensity found in at least 10% of the cancer cells) in the peritoneal, yet negative in the primary tissue samples. Notably, EGFR IHC was positive in 15/24 of the metastatic, whereas in just 2/24 of the primary HGSOC samples (p < 0.001). Although most PD‐L1 results were concordant, 5/24 and 6/24 pairs exhibited discordant results when stained with the E1L3N and 22C3 clones, respectively. Lastly, CD147 overexpression was found more often in the metastatic rather than the matched primary HGSOCs stained with CD147, though the difference was not significant. Conclusions Cytology from effusions could be considered for biomarker testing when present, even when tissue from the primary cancer is also available and adequately cellular, as it could provide additional information of potential clinical significance.
Collapse
Affiliation(s)
- Ilias P. Nikas
- School of Medicine, European University CyprusNicosiaCyprus
| | - Cheol Lee
- Department of Pathology, Seoul National University HospitalSeoulRepublic of Korea
| | - Min Ji Song
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University HospitalSeoulRepublic of Korea
| | - Bohyun Kim
- Department of Pathology, Seoul National University HospitalSeoulRepublic of Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University HospitalSeoulRepublic of Korea
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University HospitalSeoulRepublic of Korea
- Department of Pathology, Seoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
16
|
Asare-Werehene M, Tsuyoshi H, Zhang H, Salehi R, Chang CY, Carmona E, Librach CL, Mes-Masson AM, Chang CC, Burger D, Yoshida Y, Tsang BK. Plasma Gelsolin Confers Chemoresistance in Ovarian Cancer by Resetting the Relative Abundance and Function of Macrophage Subtypes. Cancers (Basel) 2022; 14:cancers14041039. [PMID: 35205790 PMCID: PMC8870487 DOI: 10.3390/cancers14041039] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/06/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Ovarian cancer is one of the deadliest female cancers with very poor survival, primarily due to late diagnosis, recurrence and chemoresistance. Although the over-expression of plasma gelsolin (pGSN) protects ovarian cancer cells from chemotherapy-induced death, its immunological role in the tumor microenvironment is less explored. Here, we demonstrate that pGSN over-expression downregulates the anti-tumor functions of M1 macrophages, an effect that contributes to chemoresistance and poor patient survival. This study demonstrates the novel inhibitory role of pGSN on tumor-infiltrated M1 macrophages and also offers new insights in maximizing the effectiveness of immunotherapy for ovarian cancer patients. Abstract Ovarian cancer (OVCA) is the most lethal gynaecological cancer with a 5-year survival rate less than 50%. Despite new therapeutic strategies, such as immune checkpoint blockers (ICBs), tumor recurrence and drug resistance remain key obstacles in achieving long-term therapeutic success. Therefore, there is an urgent need to understand the cellular mechanisms of immune dysregulation in chemoresistant OVCA in order to harness the host’s immune system to improve survival. The over-expression of plasma gelsolin (pGSN) mRNA is associated with a poorer prognosis in OVCA patients; however, its immuno-modulatory role has not been elucidated. In this study, for the first time, we report pGSN as an inhibitor of M1 macrophage anti-tumor functions in OVCA chemoresistance. Increased epithelial pGSN expression was associated with the loss of chemoresponsiveness and poor survival. While patients with increased M1 macrophage infiltration exhibited better survival due to nitric-oxide-induced ROS accumulation in OVCA cells, cohorts with poor survival had a higher infiltration of M2 macrophages. Interestingly, increased epithelial pGSN expression was significantly associated with the reduced survival benefits of infiltrated M1 macrophages, through apoptosis via increased caspase-3 activation and reduced production of iNOS and TNFα. Additionally, epithelial pGSN expression was an independent prognostic marker in predicting progression-free survival. These findings support our hypothesis that pGSN is a modulator of inflammation and confers chemoresistance in OVCA, in part by resetting the relative abundance and function of macrophage subtypes in the ovarian tumor microenvironment. Our findings raise the possibility that pGSN may be a potential therapeutic target for immune-mediated chemoresistance in OVCA.
Collapse
Affiliation(s)
- Meshach Asare-Werehene
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada; (M.A.-W.); (R.S.)
- Department of Cellular and Molecular Medicine & The Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
| | - Hideaki Tsuyoshi
- Department of Obstetrics and Gynecology, University of Fukui, Fukui 910-8507, Japan;
| | - Huilin Zhang
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- Department of Obstetrics and Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Reza Salehi
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada; (M.A.-W.); (R.S.)
- Department of Cellular and Molecular Medicine & The Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- CReATe Fertility Centre, 790 Bay Street, Suite 1100, Toronto, ON M5G 1N8, Canada;
| | - Chia-Yu Chang
- Department of Biological Science and Technology, Department of Electrophysics and Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; (C.-Y.C.); (C.-C.C.)
- Institute of Physics, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (E.C.); (A.-M.M.-M.)
| | - Clifford L. Librach
- CReATe Fertility Centre, 790 Bay Street, Suite 1100, Toronto, ON M5G 1N8, Canada;
- Departments of Obstetrics & Gynecology and Physiology, Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada; (E.C.); (A.-M.M.-M.)
| | - Chia-Ching Chang
- Department of Biological Science and Technology, Department of Electrophysics and Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; (C.-Y.C.); (C.-C.C.)
- Institute of Physics, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Dylan Burger
- Department of Cellular and Molecular Medicine & The Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, University of Fukui, Fukui 910-8507, Japan;
- Correspondence: (Y.Y.); (B.K.T.)
| | - Benjamin K. Tsang
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada; (M.A.-W.); (R.S.)
- Department of Cellular and Molecular Medicine & The Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- Correspondence: (Y.Y.); (B.K.T.)
| |
Collapse
|
17
|
Li X, Wang S, Mu W, Barry J, Han A, Carpenter RL, Jiang BH, Peiper SC, Mahoney MG, Aplin AE, Ren H, He J. Reactive oxygen species reprogram macrophages to suppress antitumor immune response through the exosomal miR-155-5p/PD-L1 pathway. J Exp Clin Cancer Res 2022; 41:41. [PMID: 35086548 PMCID: PMC8793215 DOI: 10.1186/s13046-022-02244-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022] Open
Abstract
Background Cancer cells have an imbalance in oxidation-reduction (redox) homeostasis. Understanding the precise mechanisms and the impact of the altered redox microenvironment on the immunologic reaction to tumors is limited. Methods We isolated exosomes from ovarian cancer cells through ultracentrifuge and characterized by Western-blots and Nanoparticle Tracking Analysis. 2D, 3D-coculture tumor model, and 3D live cell imaging were used to study the interactions between tumor cells, macrophages and CD3 T cells in vitro. The role of exosomal miR-155-5p in tumor growth was evaluated in xenograft nude mice models and immune-competent mice models. Flow cytometry and flow sorting were used to determine the expression levels of miR-155-5p and PD-L1 in ascites and splenic macrophages, and the percentages of CD3 T cells subpopulations. Results The elevation of reactive oxygen species (ROS) greatly downregulated exosomal miR-155-5p expression in tumor cells. Neutralization of ROS with N-acetyl-L-cysteine (NAC) increased the levels of miR-155-5p in tumor exosomes that were taken up by macrophages, leading to reduction of macrophage migration and tumor spheroid infiltration. We further found that programmed death ligand 1 (PD-L1) is a functional target of miR-155-5p. Co-culture of macrophages pre-treated with NAC-derived tumor exosomes or exosomal miR-155-5p with T-lymphocytes leading to an increased percentage of CD8+ T-lymphocyte and a decreased CD3+ T cell apoptosis through PD-L1 downregulation. Tumor growth in nude mice was delayed by treatment with NAC-derived tumor exosomes. Delivery of tumor exo-miR-155-5p in immune-intact mice suppressed ovarian cancer progression and macrophage infiltration, and activated CD8+ T cell function. It is of note that exo-miR-155-5p inhibited tumor growth more potently than the PD-L1 antibody, suggesting that in addition to PD-L1, other pathways may also be targeted by this approach. Conclusions Our findings demonstrate a novel mechanism, ROS-induced down-regulation of miR-155-5p, by which tumors modulate the microenvironment that favors tumor growth. Understanding of the negative impact of ROS on the tumor immune response will improve current therapeutic strategies. Targeting miR-155-5p can be an alternative approach to prevent formation of an immunosuppressive TME through downregulation of PD-L1 and other immunosuppressive factors. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02244-1.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA.,Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Shaomin Wang
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Wei Mu
- School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jennifer Barry
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Anna Han
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Richard L Carpenter
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Stephen C Peiper
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Hong Ren
- Department of Thoracic Surgery and Oncology, Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China.
| | - Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA.
| |
Collapse
|
18
|
Świderska J, Kozłowski M, Gaur M, Pius-Sadowska E, Kwiatkowski S, Machaliński B, Cymbaluk-Płoska A. Clinical Significance of BTLA, CD27, CD70, CD28 and CD80 as Diagnostic and Prognostic Markers in Ovarian Cancer. Diagnostics (Basel) 2022; 12:diagnostics12020251. [PMID: 35204342 PMCID: PMC8871082 DOI: 10.3390/diagnostics12020251] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/05/2022] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
It is very important to find new diagnostic and prognostic biomarkers. A total of 79 patients were enrolled in the study. The study group consisted of 37 patients with epithelial ovarian cancer, and the control group consisted of 42 patients with benign ovarian lesions. Five proteins involved in the immune response were studied: BTLA, CD27, CD70, CD28, CD80. The study material was serum and peritoneal fluid. The ROC curve was plotted, and the area under the curve was calculated to characterize the sensitivity and specificity of the studied parameters. Univariate and multivariate analyses were performed simultaneously using the Cox regression model. The cut-off level of CD27 was 120.6 pg/mL, with the sensitivity and specificity of 66 and 84% (p = 0.014). Unfavorable prognostic factors determined in serum were: CD27 (for PFS: HR 1.26, 95% CI 1.21–1.29, p = 0.047; for OS: HR 1.20, 95% CI 1.15–1.22, p = 0.014). Unfavorable prognostic factors determined in peritoneal fluid were: BTLA (for OS: HR 1.26, 95% CI 1.25–1.31, p = 0.033). We conclude that CD27 should be considered as a potential biomarker in the diagnosis of ovarian cancer. BTLA and CD27 are unfavorable prognostic factors for ovarian cancer.
Collapse
Affiliation(s)
- Janina Świderska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.Ś.); (M.G.); (A.C.-P.)
| | - Mateusz Kozłowski
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.Ś.); (M.G.); (A.C.-P.)
- Correspondence:
| | - Maria Gaur
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.Ś.); (M.G.); (A.C.-P.)
| | - Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (E.P.-S.); (B.M.)
| | - Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (E.P.-S.); (B.M.)
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.Ś.); (M.G.); (A.C.-P.)
| |
Collapse
|
19
|
Chung YM, Khan PP, Wang H, Tsai WB, Qiao Y, Yu B, Larrick JW, Hu MCT. Sensitizing tumors to anti-PD-1 therapy by promoting NK and CD8+ T cells via pharmacological activation of FOXO3. J Immunother Cancer 2021; 9:jitc-2021-002772. [PMID: 34887262 PMCID: PMC8663085 DOI: 10.1136/jitc-2021-002772] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Stimulating antitumor immunity by blocking programmed death-1 (PD-1) or its ligand (programmed death-ligand 1 (PD-L1) is a promising antitumor therapy. However, numerous patients respond poorly to PD-1/PD-L1 blockade. Unresponsiveness to immune-checkpoint blockade (ICB) can cast significant challenges to the therapeutic options for patients with hard-to-treat tumors. There is an unmet clinical need to establish new therapeutic approaches for mitigating ICB unresponsiveness in patients. In this study, we investigated the efficacy and role of low-dose antineoplastic agent SN-38 or metformin in sensitizing unresponsive tumors to respond to ICB therapy. METHODS We assessed the significant pathological relationships between PD-L1 and FOXO3 expression and between PD-L1 and c-Myc or STAT3 expression in patients with various tumors. We determined the efficacy of low-dose SN-38 or metformin in sensitizing unresponsive tumors to respond to anti-PD-1 therapy in a syngeneic tumor system. We deciphered novel therapeutic mechanisms underlying the SN-38 and anti-PD-1 therapy-mediated engagement of natural killer (NK) or CD8+ T cells to infiltrate tumors and boost antitumor immunity. RESULTS We showed that PD-L1 protein level was inversely associated with FOXO3 protein level in patients with ovarian, breast, and hepatocellular tumors. Low-dose SN-38 or metformin abrogated PD-L1 protein expression, promoted FOXO3 protein level, and significantly increased the animal survival rate in syngeneic mouse tumor models. SN-38 or metformin sensitized unresponsive tumors responding to anti-PD-1 therapy by engaging NK or CD8+ T cells to infiltrate the tumor microenvironment (TME) and secret interferon-γ and granzyme B to kill tumors. SN-38 suppressed the levels of c-Myc and STAT3 proteins, which controlled PD-L1 expression. FOXO3 was essential for SN38-mediated PD-L1 suppression. The expression of PD-L1 was compellingly linked to that of c-Myc or STAT3 in patients with the indicated tumors. CONCLUSION We show that SN-38 or metformin can boost antitumor immunity in the TME by inhibiting c-Myc and STAT3 through FOXO3 activation. These results may provide novel insight into ameliorating patient response to overarching immunotherapy for tumors.
Collapse
Affiliation(s)
- Young Min Chung
- Panorama Research Institute, Sunnyvale, California, USA.,Panorama Institute of Molecular Medicine, Sunnyvale, California, USA.,Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Pragya P Khan
- Panorama Research Institute, Sunnyvale, California, USA.,Panorama Institute of Molecular Medicine, Sunnyvale, California, USA
| | - Hong Wang
- Panorama Research Institute, Sunnyvale, California, USA
| | - Wen-Bin Tsai
- Panorama Research Institute, Sunnyvale, California, USA.,The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yanli Qiao
- Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Bo Yu
- Panorama Research Institute, Sunnyvale, California, USA
| | - James W Larrick
- Panorama Research Institute, Sunnyvale, California, USA.,Panorama Institute of Molecular Medicine, Sunnyvale, California, USA
| | - Mickey C-T Hu
- Panorama Research Institute, Sunnyvale, California, USA .,Panorama Institute of Molecular Medicine, Sunnyvale, California, USA.,Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
20
|
Multi-Marker Immunofluorescent Staining and PD-L1 Detection on Circulating Tumour Cells from Ovarian Cancer Patients. Cancers (Basel) 2021; 13:cancers13246225. [PMID: 34944844 PMCID: PMC8699768 DOI: 10.3390/cancers13246225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Circulating tumour cells (CTCs) have the potential to serve as a rich source of information for cancer diagnostic and therapeutic decisions. To fully exploit this minimally invasive diagnostic resource requires techniques that aid in enriching heterogenous populations of CTCs and markers to efficiently characterise these cells as tumour derived. In the present study we eva-luated the microfluidic enrichment of CTCs and a multi-marker staining methodology for the identification of heterogeneous CTCs in ovarian cancer (OC) patients and evaluation of PD-L1 expression. We showed, for the first time, the existence of hybrid CTCs with an epithelial/mesenchymal phenotype and their association with PD-L1 in OC. Incorporation of this method in future clinical trials can help predict immunotherapy responsiveness in OC patients. Abstract Detection of ovarian cancer (OC) circulating tumour cells (CTCs) is primarily based on targeting epithelial markers, thus failing to detect mesenchymal tumour cells. More importantly, the immune checkpoint inhibitor marker PD-L1 has not been demonstrated on CTCs from OC patients. An antibody staining protocol was developed and tested using SKOV-3 and OVCA432 OC cell lines. We targeted epithelial (cytokeratin (CK) and EpCAM), mesenchymal (vimentin), and OC-specific (PAX8) markers for detection of CTCs, and CD45/16 and CD31 were used for the exclusion of white blood and vascular endothelial cells, respectively. PD-L1 was used for CTC characterisation. CTCs were enriched using the Parsortix™ system from 16 OC patients. Results revealed the presence of CTCs in 10 (63%) cases. CTCs were heterogeneous, with 113/157 (72%) cells positive for CK/EpCAM (epithelial marker), 58/157 (37%) positive for vimentin (mesenchymal marker), and 17/157 (11%) for both (hybrid). PAX8 was only found in 11/157 (7%) CTCs. In addition, 62/157 (39%) CTCs were positive for PD-L1. Positivity for PD-L1 was significantly associated with the hybrid phenotype when compared with the epithelial (p = 0.007) and mesenchymal (p = 0.0009) expressing CTCs. Characterisation of CTC phenotypes in relation to clinical outcomes is needed to provide insight into the role that epithelial to mesenchymal plasticity plays in OC and its relationship with PD-L1.
Collapse
|
21
|
Ghoneum A, Almousa S, Warren B, Abdulfattah AY, Shu J, Abouelfadl H, Gonzalez D, Livingston C, Said N. Exploring the clinical value of tumor microenvironment in platinum-resistant ovarian cancer. Semin Cancer Biol 2021; 77:83-98. [PMID: 33476723 PMCID: PMC8286277 DOI: 10.1016/j.semcancer.2020.12.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/20/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Platinum resistance in epithelial ovarian cancer (OvCa) is rising at an alarming rate, with recurrence of chemo-resistant high grade serous OvCa (HGSC) in roughly 75 % of all patients. Additionally, HGSC has an abysmal five-year survival rate, standing at 39 % and 17 % for FIGO stages III and IV, respectively. Herein we review the crucial cellular interactions between HGSC cells and the cellular and non-cellular components of the unique peritoneal tumor microenvironment (TME). We highlight the role of the extracellular matrix (ECM), ascitic fluid as well as the mesothelial cells, tumor associated macrophages, neutrophils, adipocytes and fibroblasts in platinum-resistance. Moreover, we underscore the importance of other immune-cell players in conferring resistance, including natural killer cells, myeloid-derived suppressive cells (MDSCs) and T-regulatory cells. We show the clinical relevance of the key platinum-resistant markers and their correlation with the major pathways perturbed in OvCa. In parallel, we discuss the effect of immunotherapies in re-sensitizing platinum-resistant patients to platinum-based drugs. Through detailed analysis of platinum-resistance in HGSC, we hope to advance the development of more effective therapy options for this aggressive disease.
Collapse
Affiliation(s)
- Alia Ghoneum
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Sameh Almousa
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Bailey Warren
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Ammar Yasser Abdulfattah
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Alexandria University School of Medicine, Alexandria, Egypt
| | - Junjun Shu
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; The Third Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hebatullah Abouelfadl
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Department of Genetics, Animal Health Research Institute, Dokki, Egypt
| | - Daniela Gonzalez
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Christopher Livingston
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Neveen Said
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Departments of Urology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Comprehensive Cancer Center, Winston Salem, NC, 27157, USA.
| |
Collapse
|
22
|
Esmail S, Danter WR. Stem-cell based, machine learning approach for optimizing natural killer cell-based personalized immunotherapy for high-grade ovarian cancer. FEBS J 2021; 289:985-998. [PMID: 34582617 DOI: 10.1111/febs.16214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 12/18/2022]
Abstract
Advanced high-grade serous ovarian cancer continues to be a therapeutic challenge for those affected using the current therapeutic interventions. There is an increasing interest in personalized cancer immunotherapy using activated natural killer (NK) cells. NK cells account for approximately 15% of circulating white blood cells. They are also an important element of the tumor microenvironment (TME) and the body's immune response to cancers. In the present study, DeepNEU-C2Rx, a machine learning platform, was first used to create validated artificially induced pluripotent stem cell simulations. These simulations were then used to generate wild-type artificially induced NK cells (aiNK-WT) and TME simulations. Once validated, the aiNK-WT simulations were exposed to artificially induced high-grade serous ovarian cancer represented by aiOVCAR3. Cytolytic activity of aiNK was evaluated in presence and absence of aiOVCAR3 and data were compared with the literature for validation. The TME simulations suggested 26 factors that could be evaluated based on their ability to enhance aiNK-WT cytolytic activity in the presence of aiOVCAR3. The addition of programmed cell death-1 inhibitor leads to significant reinvigoration of aiNK cytolytic activity. The combination of programmed cell death-1 and glycogen synthase kinase 3 inhibitors showed further improvement. Further addition of ascitic fluid factor inhibitors leads to optimal aiNK activation. Our data showed that NK cell simulations could be used not only to pinpoint novel immunotherapeutic targets to reinvigorate the activity of NK cells against cancers, but also to predict the outcome of targeting tumors with specific genetic expression and mutation profiles.
Collapse
|
23
|
Dysfunctional activity of classical DNA end-joining renders acquired resistance to carboplatin in human ovarian cancer cells. Cancer Lett 2021; 520:267-280. [PMID: 34375710 DOI: 10.1016/j.canlet.2021.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/13/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the deadliest gynecological malignancy worldwide. Although chemotherapy is required as the most standard treatment strategy for ovarian cancer, the survival rates are very low, largely because of high incidence of recurrence due to resistance to conventional surgery and genotoxic chemotherapies. Carboplatin-resistant ovarian cancer cells were generated by continuous treatment over six months. Carboplatin-resistance induced morphological alterations and promoted the rates of proliferation and migration of SKOV3 compared to the parental cells. Interestingly, carboplatin-resistant SKOV3 showed the high levels of γH2AX foci formed at the basal level, and the levels of γH2AX foci remained even after the recovery time, suggesting that the DNA damage response and repair machinery were severely attenuated by carboplatin-resistance. Surprisingly, the expression levels of XRCC4, a critical factor in non-homologous end joining (NHEJ) DNA repair, were significantly decreased in carboplatin-resistant SKOV3 compared with those in non-resistant controls. Furthermore, restoration of NHEJ in carboplatin-resistant SKOV3 by suppression of ABCB1 and/or AR re-sensitized carboplatin-resistant cells to genotoxic stress and reduced their proliferation ability. Our findings suggest that attenuation of the NHEJ DNA repair machinery mediated by resistance to genotoxic stress might be a critical cause of chemoresistance in patients with ovarian cancer.
Collapse
|
24
|
An D, Banerjee S, Lee JM. Recent advancements of antiangiogenic combination therapies in ovarian cancer. Cancer Treat Rev 2021; 98:102224. [PMID: 34051628 PMCID: PMC8217312 DOI: 10.1016/j.ctrv.2021.102224] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is a deadly malignancy with a growing therapeutic armamentarium, though achieving sustained benefit in the clinic remains largely elusive. Through biomarker and genetic analysis, several pathways of resistance and sensitivity to commonly used therapeutics have been identified, expanding the potential of identifying unique drug combinations and indicating new directions for improving clinical outcomes. Here, we review the mechanisms of angiogenic response and antiangiogenic therapy in ovarian cancer, as well as the interactions it exhibits with the immune and DNA damage response pathways. We discuss results from clinical trials examining the combinations of antiangiogenics, PARP inhibitors, and immune checkpoint inhibitors are also discussed, as well as several ongoing trials.
Collapse
Affiliation(s)
- Daniel An
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
25
|
Eckert MA, Orozco C, Xiao J, Javellana M, Lengyel E. The Effects of Chemotherapeutics on the Ovarian Cancer Microenvironment. Cancers (Basel) 2021; 13:3136. [PMID: 34201616 PMCID: PMC8268261 DOI: 10.3390/cancers13133136] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/31/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is characterized by a complex and dynamic tumor microenvironment (TME) composed of cancer-associated fibroblasts (CAFs), immune cells, endothelial cells, and adipocytes. Although most approved therapies target cancer cells, a growing body of evidence suggests that chemotherapeutic agents have an important role in regulating the biology of the diverse cells that compose the TME. Understanding how non-transformed cells respond and adapt to established therapeutics is necessary to completely comprehend their action and develop novel therapeutics that interrupt undesired tumor-stroma interactions. Here, we review the effects of chemotherapeutic agents on normal cellular components of the host-derived TME focusing on CAFs. We concentrate on therapies used in the treatment of HGSOC and synthesize findings from studies focusing on other cancer types and benign tissues. Agents such as platinum derivatives, taxanes, and PARP inhibitors broadly affect the TME and promote or inhibit the pro-tumorigenic roles of CAFs by modifying the bidirectional cross-talk between tumor and stromal cells in the tumor organ. While most chemotherapy research focuses on cancer cells, these studies emphasize the need to consider all cell types within the tumor organ when evaluating chemotherapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (M.A.E.); (C.O.); (J.X.); (M.J.)
| |
Collapse
|
26
|
Walsh CS, Kamrava M, Rogatko A, Kim S, Li A, Cass I, Karlan B, Rimel BJ. Phase II trial of cisplatin, gemcitabine and pembrolizumab for platinum-resistant ovarian cancer. PLoS One 2021; 16:e0252665. [PMID: 34081738 PMCID: PMC8174738 DOI: 10.1371/journal.pone.0252665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To evaluate the combination of pembrolizumab, cisplatin and gemcitabine in recurrent platinum-resistant ovarian cancer. METHODS Patients received six cycles of chemotherapy with gemcitabine and cisplatin on day 1 and day 8 of a 21-day treatment cycle. Pembrolizumab was administered on day 1 of cycles 3-6 and as maintenance monotherapy in cycles 7-34. Palliative radiation to a non-target symptomatic lesion was allowed. The primary objective was overall response rate by RECIST 1.1 criteria. Secondary objectives included safety, progression-free survival, time to progression, duration of response and overall survival. RESULTS An interim analysis for futility was performed at 18 evaluable patients. Overall response rate was 60%, duration of response was 4.9 months and time to progression was 5.2 months. Progression-free survival at 6 and 12 months was 43% and 5%. Median progression-free survival was 6.2 months and median overall survival was 11.3 months. In all patients, CA125 levels reflected response and progression. There were no pseudoprogression events. After receiving palliative radiation during pembrolizumab maintenance, a patient with recurrent ovarian clear cell carcinoma had an exceptional and durable response that is ongoing for greater than 2 years. After consultation with the sponsor, based on the modest duration of response observed at the interim analysis for futility, the decision was made to close the trial to further accrual. CONCLUSIONS The addition of pembrolizumab to cisplatin and gemcitabine did not appear to provide benefit beyond chemotherapy alone in patients with recurrent platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Christine S. Walsh
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| | - Mitchell Kamrava
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Andre Rogatko
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sungjin Kim
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Andrew Li
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ilana Cass
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Beth Karlan
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Bobbie J. Rimel
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
27
|
Combination of gene set signatures correlates with response to nivolumab in platinum-resistant ovarian cancer. Sci Rep 2021; 11:11427. [PMID: 34075161 PMCID: PMC8169687 DOI: 10.1038/s41598-021-91012-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/12/2021] [Indexed: 01/01/2023] Open
Abstract
Based on our previous phase II clinical trial of anti-programmed death-1 (PD-1) antibody nivolumab for platinum-resistant ovarian cancer (n = 19, UMIN000005714), we aimed to identify the biomarkers predictive of response. Tumor gene expression was evaluated by proliferative, mesenchymal, differentiated, and immunoreactive gene signatures derived from high-grade serous carcinomas and a signature established prior for ovarian clear cell carcinoma. Resulting signature scores were statistically assessed with both univariate and multivariate approaches for correlation to clinical response. Analyses were performed to identify pathways differentially expressed by either the complete response (CR) or progressive disease (PD) patient groups. The clear cell gene signature was scored significantly higher in the CR group, and the proliferative gene signature had significantly higher scores in the PD group where nivolumab was not effective (respective p values 0.005 and 0.026). Combinations of gene signatures improved correlation with response, where a visual projection of immunoreactive, proliferative, and clear cell signatures differentiated clinical response. An applicable clinical response prediction formula was derived. Ovarian cancer-specific gene signatures and related pathway scores provide a robust preliminary indicator for ovarian cancer patients prior to anti-PD-1 therapy decisions.
Collapse
|
28
|
Falchook GS, Peeters M, Rottey S, Dirix LY, Obermannova R, Cohen JE, Perets R, Frommer RS, Bauer TM, Wang JS, Carvajal RD, Sabari J, Chapman S, Zhang W, Calderon B, Peterson DA. A phase 1a/1b trial of CSF-1R inhibitor LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid tumors. Invest New Drugs 2021; 39:1284-1297. [PMID: 33852104 DOI: 10.1007/s10637-021-01088-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/11/2021] [Indexed: 12/31/2022]
Abstract
Background LY3022855 is a recombinant, immunoglobulin, human monoclonal antibody targeting the colony-stimulating factor-1 receptor. This phase 1 trial determined the safety, pharmacokinetics, and antitumor activity of LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid cancers who had received standard anti-cancer treatments. Methods In Part A (dose-escalation), patients received intravenous (IV) LY3022855 25/50/75/100 mg once weekly (QW) combined with durvalumab 750 mg once every two weeks (Q2W) IV or LY3022855 50 or 100 mg QW IV with tremelimumab 75/225/750 mg once every four weeks. In Part B (dose-expansion), patients with non-small cell lung cancer (NSCLC) or ovarian cancer (OC) received recommended phase 2 dose (RP2D) of LY3022855 from Part A and durvalumab 750 mg Q2W. Results Seventy-two patients were enrolled (median age 61 years): Part A = 33, Part B = 39. In Part A, maximum tolerated dose was not reached, and LY3022855 100 mg QW and durvalumab 750 mg Q2W was the RP2D. Four dose-limiting equivalent toxicities occurred in two patients from OC cohort. In Part A, maximum concentration, area under the concentration-time curve, and serum concentration showed dose-dependent increase over two cycles of therapy. Overall rates of complete response, partial response, and disease control were 1.4%, 2.8%, and 33.3%. Treatment-emergent anti-drug antibodies were observed in 21.2% of patients. Conclusions LY3022855 combined with durvalumab or tremelimumab in patients with advanced NSCLC or OC had limited clinical activity, was well tolerated. The RP2D was LY3022855 100 mg QW with durvalumab 750 mg Q2W. ClinicalTrials.gov ID: NCT02718911 (Registration Date: May 3, 2011).
Collapse
Affiliation(s)
- Gerald S Falchook
- Drug Development Unit, Sarah Cannon Research Institute at HealthONE, 1800 Williams St Ste 300, Denver, CO, USA.
| | - Marc Peeters
- Department of Oncology, University Antwerpen, Universitair Ziekenhuis Antwerpen, Drie Eikenstraat, Edegem, Belgium
| | - Sylvie Rottey
- Drug Research Unit, Universitair Ziekenhuis Gent, Corneel Heymanslaan, Ghent, Belgium
| | - Luc Y Dirix
- Department of Medical Oncology, GZA Sint Augustinus, Oosterveldlaan, Wilrijk, Belgium
| | - Radka Obermannova
- Comprehensive Cancer Care Department - Faculty of Medicine, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Jonathan E Cohen
- Department of Oncology, Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem, Israel.,The Faculty of Medicine, The Wohl Institute for Translational Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Ruth Perets
- Department of Oncology, Clinical Research Institute at Rambam, Rambam Medical Center, Haifa, Israel.,Technion - Israel Institute of Technology, Haifa, Israel
| | - Ronnie Shapira Frommer
- The Ella Institute for Immuno-Oncology, Sheba Medical Center, Oncology Institute, Tel Hashomer, Ramat Gan, Israel
| | - Todd M Bauer
- Department of Medical Oncology, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | - Judy S Wang
- Hematologic Oncology, Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL, USA
| | - Richard D Carvajal
- Department of Medicine, Division of Hematology/Oncology, Columbia University, College of Physicians and Surgeons, New York, NY, USA
| | - Joshua Sabari
- Department of Medicine,Medical Oncology, NYU Langone Health, New York, NY, USA
| | | | - Wei Zhang
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | |
Collapse
|
29
|
Gao T, Mao Z, Li W, Pei R. Anti-PD-L1 DNA aptamer antagonizes the interaction of PD-1/PD-L1 with antitumor effect. J Mater Chem B 2021; 9:746-756. [PMID: 33319876 DOI: 10.1039/d0tb01668c] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor immune evasion enables cancer cells to escape destruction by the immune system, which causes poor prognosis and overall survival of some tumor patients. The binding of PD-L1 on tumor cells to PD-1 on T cells suppresses T cell function, and the axis is considered one of the major pathways mediating tumor cells to evade immune surveillance. The PD-L1 ligation of T cells has a profound inhibitory effect on the growth, cytokine secretion, and development of cytotoxicity. Aptamers, known as chemical antibodies, are single-stranded oligonucleotides with high affinity. In this work, we take a cell-SELEX with the engineered PD-L1-expressing cells as a target to obtain the aptamer, designated PL1, which specifically binds to PD-L1 with a Kd value of 95.73 nM, resulting in the inhibition of PD-1/PD-L1. The aptamer PL1 could restore the proliferation and IFN-γ rescue from the T cell inhibited by the PD-1/PD-L1 axis, and inhibit the growth of the CT26 colon carcinoma. The similar tumor inhibition efficacy and binding capacity of the aptamer PL1 as an antibody indicate that the aptamer PL1 can serve as an alternative therapeutic agent for cancer immunotherapy since the use of antibodies is often restricted by high cost, large size and poor tumor penetration.
Collapse
Affiliation(s)
- Tian Gao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | | | | | | |
Collapse
|
30
|
Zhang B, Nie X, Miao X, Wang S, Li J, Wang S. Development and verification of an immune-related gene pairs prognostic signature in ovarian cancer. J Cell Mol Med 2021; 25:2918-2930. [PMID: 33543590 PMCID: PMC7957197 DOI: 10.1111/jcmm.16327] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OV) is the most common gynaecological cancer worldwide. Immunotherapy has recently been proven to be an effective treatment strategy. The work here attempts to produce a prognostic immune-related gene pair (IRGP) signature to estimate OV patient survival. The Gene Expression Omnibus (GEO) and Cancer Genome Atlas (TCGA) databases provided the genetic expression profiles and clinical data of OV patients. Based on the InnateDB database and the least absolute shrinkage and selection operator (LASSO) regression model, we first identified a 17-IRGP signature associated with survival. The average area under the curve (AUC) values of the training, validation, and all TCGA sets were 0.869, 0.712, and 0.778, respectively. The 17-IRGP signature noticeably split patients into high- and low-risk groups with different prognostic outcomes. As suggested by a functional study, some biological pathways, including the Toll-like receptor and chemokine signalling pathways, were significantly negatively correlated with risk scores; however, pathways such as the p53 and apoptosis signalling pathways had a positive correlation. Moreover, tumour stage III, IV, grade G1/G2, and G3/G4 samples had significant differences in risk scores. In conclusion, an effective 17-IRGP signature was produced to predict prognostic outcomes in OV, providing new insights into immunological biomarkers.
Collapse
Affiliation(s)
- Bao Zhang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Xiaocui Nie
- Department of Obstetrics and GynecologyShenyang women's and children's hospitalShenyangChina
| | - Xinxin Miao
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Shuo Wang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Jing Li
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Shengke Wang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
31
|
Fritz I, Wagner P, Olsson H. Improved survival in several cancers with use of H 1-antihistamines desloratadine and loratadine. Transl Oncol 2021; 14:101029. [PMID: 33550204 PMCID: PMC7868613 DOI: 10.1016/j.tranon.2021.101029] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 01/21/2023] Open
Abstract
Improved cancer survival with use of antihistamines desloratadine and loratadine. Improved survival seen in tumors that respond to immune checkpoint therapy. A – potentially immunological – anti-tumor effect of desloratadine and loratadine.
Background We have previously shown an association with substantially improved survival in breast cancer and melanoma for desloratadine and loratadine users, and set out to find whether an improved survival can be seen in tumors with and without a known response to immune checkpoint therapy, such as anti-CTLA-4 or anti-PD-1. Methods We investigated survival and use of six common H1-antihistamines (cetirizine, clemastine, desloratadine, ebastine, fexofenadine and loratadine) in a nation-wide cohort of all 429,198 Swedish patients with ten types of immunogenic (gastric, colorectal/anal, pancreatic, lung, breast, prostate, kidney, and bladder cancer, melanoma and Hodgkin lymphoma) and six non-immunogenic (liver, uterine, ovarian, brain/CNS, and thyroid cancer and non-Hodgkin lymphoma) tumors diagnosed 2006–2017. Follow-up was until 2019–02–24. Findings Desloratadine use was associated with an improved survival for all immunogenic tumors, but not for the non-immunogenic ones. Loratadine use was associated with improved survival for some tumors. Use of the other antihistamines could not be shown to be consistently associated with improved survival to a statistically significant degree. Interpretation Our hypothesis is that our findings result from immune checkpoint inhibition, and we believe both desloratadine and loratadine should be tested in randomized clinical trials as treatment of immunogenic tumors, with priority given to trials of desloratadine as treatment of tumors with few therapy options and dismal prognoses, such as pancreatic cancer. If our results can be confirmed in a clinical setting, new, potentially curative, therapies could result for several tumors, including ones with dire prognoses and limited treatment options.
Collapse
Affiliation(s)
- Ildikó Fritz
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden.
| | - Philippe Wagner
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Håkan Olsson
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden; Department of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Batchu RB, Gruzdyn OV, Kolli BK, Dachepalli R, Umar PS, Rai SK, Singh N, Tavva PS, Weaver DW, Gruber SA. IL-10 Signaling in the Tumor Microenvironment of Ovarian Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:51-65. [PMID: 33559854 DOI: 10.1007/978-3-030-55617-4_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Unlike other malignancies, ovarian cancer (OC) creates a complex tumor microenvironment with distinctive peritoneal ascites consisting of a mixture of several immunosuppressive cells which impair the ability of the patient's immune system to fight the disease. The poor survival rates observed in advanced stage OC patients and the lack of effective conventional therapeutic options have been attributed in large part to the immature dendritic cells (DCs), IL-10 secreting regulatory T cells, tumor-associated macrophages, myeloid-derived suppressor cells, and cancer stem cells that secrete inhibitory cytokines. This review highlights the critical role played by the intraperitoneal presence of IL-10 in the generation of an immunosuppressive tumor microenvironment. Further, the effect of antibody neutralization of IL-10 on the efficacy of DC and chimeric antigen receptor T-cell vaccines will be discussed. Moreover, we will review the influence of IL-10 in the promotion of cancer stemness in concert with the NF-κB signaling pathway with regard to OC progression. Finally, understanding the role of IL-10 and its crosstalk with various cells in the ascitic fluid may contribute to the development of novel immunotherapeutic approaches with the potential to kill drug-resistant OC cells while minimizing toxic side effects.
Collapse
Affiliation(s)
- Ramesh B Batchu
- Wayne State University School of Medicine, Detroit, MI, USA. .,John D. Dingell VA Medical Center, Detroit, MI, USA.
| | - Oksana V Gruzdyn
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Bala K Kolli
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA.,Med Manor Organics Pvt. Ltd., Hyderabad, India
| | | | - Prem S Umar
- Med Manor Organics Pvt. Ltd., Hyderabad, India
| | | | | | | | | | - Scott A Gruber
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA
| |
Collapse
|
33
|
Inflammation and immunity in ovarian cancer. EJC Suppl 2020; 15:56-66. [PMID: 33240443 PMCID: PMC7569134 DOI: 10.1016/j.ejcsup.2019.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/15/2019] [Accepted: 12/27/2019] [Indexed: 12/30/2022] Open
Abstract
The standard first-line therapy for ovarian cancer is a combination of surgery and carboplatin/paclitaxel-based chemotherapy. Patients with longer survival and improved response to chemotherapy usually present T-cell inflamed tumours. The presence of tumour-infiltrating T cells (TILs) notably varies among the different subtypes of ovarian tumours, being highest in high-grade serous ovarian carcinoma, intermediate in endometrioid tumours, and lowest in low-grade serous, mucinous and clear cell tumours. Interestingly, the presence of TILs is often accompanied by a strong immunosuppressive tumour environment. A better understanding of the immune response against ovarian cancer and the tumour immune evasion mechanisms will enable improved prognostication, response prediction and immunotherapy of this disease. This article provides an overview of some ovarian cancer cell features relevant for antitumour response, such as tumour-associated antigens, including neoantigens, expression of inhibitory molecules, and other mechanisms of immune evasion. Moreover, we describe relevant immune cell types found in epithelial ovarian tumours, including T and B lymphocytes, regulatory T cells, natural killer cells, tumour-associated macrophages, myeloid-derived suppressor cells and neutrophils. We focus on how these components influence the burden of the tumour and the clinical outcome. The presence of spontaneous tumour-specific T lymphocytes and the existence of multiple immune evasion mechanisms in epithelial ovarian cancer (EOC) support the immunogenicity of this tumour. Tumour-infiltrating T lymphocytes (TILs) have been associated with disease outcome in EOC, indicating their clinical significance. The subtypes of EOC, mutations in TP53 and breast and ovarian cancer susceptibility protein 1/2 and the immune expression signature are factors associated to TIL density in EOC. The tumour microenvironment in EOC consists of a dynamic and complex network of soluble factors, inhibitory receptors and immunosuppressive cells.
Collapse
|
34
|
Wei Y, Ou T, Lu Y, Wu G, Long Y, Pan X, Yao D. Classification of ovarian cancer associated with BRCA1 mutations, immune checkpoints, and tumor microenvironment based on immunogenomic profiling. PeerJ 2020; 8:e10414. [PMID: 33282564 PMCID: PMC7694562 DOI: 10.7717/peerj.10414] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/02/2020] [Indexed: 01/10/2023] Open
Abstract
Background Ovarian cancer is a highly fatal gynecological malignancy and new, more effective treatments are needed. Immunotherapy is gaining attention from researchers worldwide, although it has not proven to be consistently effective in the treatment of ovarian cancer. We studied the immune landscape of ovarian cancer patients to improve the efficacy of immunotherapy as a treatment option. Methods We obtained expression profiles, somatic mutation data, and clinical information from The Cancer Genome Atlas. Ovarian cancer was classified based on 29 immune-associated gene sets, which represented different immune cell types, functions, and pathways. Single-sample gene set enrichment (ssGSEA) was used to quantify the activity or enrichment levels of the gene sets in ovarian cancer, and the unsupervised machine learning method was used sort the classifications. Our classifications were validated using Gene Expression Omnibus datasets. Results We divided ovarian cancer into three subtypes according to the ssGSEA score: subtype 1 (low immunity), subtype 2 (median immunity), and subtype 3 (high immunity). Most tumor-infiltrating immune cells and immune checkpoint molecules were upgraded in subtype 3 compared with those in the other subtypes. The tumor mutation burden (TMB) was not significantly different among the three subtypes. However, patients with BRCA1 mutations were consistently detected in subtype 3. Furthermore, most immune signature pathways were hyperactivated in subtype 3, including T and B cell receptor signaling pathways, PD-L1 expression and PD-1 checkpoint pathway the NF-κB signaling pathway, Th17 cell differentiation and interleukin-17 signaling pathways, and the TNF signaling pathway. Conclusion Ovarian cancer subtypes that are based on immune biosignatures may contribute to the development of novel therapeutic treatment strategies for ovarian cancer.
Collapse
Affiliation(s)
- Yousheng Wei
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Tingyu Ou
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yan Lu
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Guangteng Wu
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Ying Long
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xinbin Pan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Desheng Yao
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| |
Collapse
|
35
|
Modulation of Immune Infiltration of Ovarian Cancer Tumor Microenvironment by Specific Subpopulations of Fibroblasts. Cancers (Basel) 2020; 12:cancers12113184. [PMID: 33138184 PMCID: PMC7692816 DOI: 10.3390/cancers12113184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor immune infiltration plays a key role in the progression of solid tumors, including ovarian cancer, and immunotherapies are rapidly emerging as effective treatment modalities. However, the role of cancer-associated fibroblasts (CAFs), a predominant stromal constituent, in determining the tumor-immune microenvironment and modulating efficacy of immunotherapies remains poorly understood. We have conducted an extensive bioinformatic analysis of our and other publicly available ovarian cancer datasets (GSE137237, GSE132289 and GSE71340), to determine the correlation of fibroblast subtypes within the tumor microenvironment (TME) with the characteristics of tumor-immune infiltration. We identified (1) four functional modules of CAFs in ovarian cancer that are associated with the TME and metastasis of ovarian cancer, (2) immune-suppressive function of the collagen 1,3,5-expressing CAFs in primary ovarian cancer and omental metastases, and (3) consistent positive correlations between the functional modules of CAFs with anti-immune response genes and negative correlation with pro-immune response genes. Our study identifies a specific fibroblast subtype, fibroblast functional module (FFM)2, in the ovarian cancer tumor microenvironment that can potentially modulate a tumor-promoting immune microenvironment, which may be detrimental toward the effectiveness of ovarian cancer immunotherapies.
Collapse
|
36
|
Mei KC, Liao YP, Jiang J, Chiang M, Khazaieli M, Liu X, Wang X, Liu Q, Chang CH, Zhang X, Li J, Ji Y, Melano B, Telesca D, Xia T, Meng H, Nel AE. Liposomal Delivery of Mitoxantrone and a Cholesteryl Indoximod Prodrug Provides Effective Chemo-immunotherapy in Multiple Solid Tumors. ACS NANO 2020; 14:13343-13366. [PMID: 32940463 PMCID: PMC8023019 DOI: 10.1021/acsnano.0c05194] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We developed a custom-designed liposome carrier for codelivery of a potent immunogenic cell death (ICD) stimulus plus an inhibitor of the indoleamine 2,3-dioxygenase (IDO-1) pathway to establish a chemo-immunotherapy approach for solid tumors in syngeneic mice. The carrier was constructed by remote import of the anthraquinone chemotherapeutic agent, mitoxantrone (MTO), into the liposomes, which were further endowed with a cholesterol-conjugated indoximod (IND) prodrug in the lipid bilayer. For proof-of-principle testing, we used IV injection of the MTO/IND liposome in a CT26 colon cancer model to demonstrate the generation of a robust immune response, characterized by the appearance of ICD markers (CRT and HMGB-1) as well as evidence of cytotoxic cancer cell death, mediated by perforin and granzyme B. Noteworthy, the cytotoxic effects involved natural killer (NK) cell, which suggests a different type of ICD response. The immunotherapy response was significantly augmented by codelivery of the IND prodrug, which induced additional CRT expression, reduced number of Foxp3+ Treg, and increased perforin release, in addition to extending animal survival beyond the effect of an MTO-only liposome. The outcome reflects the improved pharmacokinetics of MTO delivery to the cancer site by the carrier. In light of the success in the CT26 model, we also assessed the platform efficacy in further breast cancer (EMT6 and 4T1) and renal cancer (RENCA) models, which overexpress IDO-1. Encapsulated MTO delivery was highly effective for inducing chemo-immunotherapy responses, with NK participation, in all tumor models. Moreover, the growth inhibitory effect of MTO was enhanced by IND codelivery in EMT6 and 4T1 tumors. All considered, our data support the use of encapsulated MTO delivery for chemo-immunotherapy, with the possibility to boost the immune response by codelivery of an IDO-1 pathway inhibitor.
Collapse
Affiliation(s)
- Kuo-Ching Mei
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Michelle Chiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Mercedeh Khazaieli
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Liu
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Qi Liu
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Chong Hyun Chang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiao Zhang
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Juan Li
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Ying Ji
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Brenda Melano
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Donatello Telesca
- Department of Biostatistics, University of California, Los Angeles, California, 90095, United States
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Huan Meng
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, 90095, United States
| | - Andre E. Nel
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, 90095, United States
| |
Collapse
|
37
|
Zagorulya M, Duong E, Spranger S. Impact of anatomic site on antigen-presenting cells in cancer. J Immunother Cancer 2020; 8:e001204. [PMID: 33020244 PMCID: PMC7537336 DOI: 10.1136/jitc-2020-001204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 12/24/2022] Open
Abstract
Checkpoint blockade immunotherapy (CBT) can induce long-term clinical benefits in patients with advanced cancer; however, response rates to CBT vary by cancer type. Cancers of the skin, lung, and kidney are largely responsive to CBT, while cancers of the pancreas, ovary, breast, and metastatic lesions to the liver respond poorly. The impact of tissue-resident immune cells on antitumor immunity is an emerging area of investigation. Recent evidence indicates that antitumor immune responses and efficacy of CBT depend on the tissue site of the tumor lesion. As myeloid cells are predominantly tissue-resident and can shape tumor-reactive T cell responses, it is conceivable that tissue-specific differences in their function underlie the tissue-site-dependent variability in CBT responses. Understanding the roles of tissue-specific myeloid cells in antitumor immunity can open new avenues for treatment design. In this review, we discuss the roles of tissue-specific antigen-presenting cells (APCs) in governing antitumor immune responses, with a particular focus on the contributions of tissue-specific dendritic cells. Using the framework of the Cancer-Immunity Cycle, we examine the contributions of tissue-specific APC in CBT-sensitive and CBT-resistant carcinomas, highlight how these cells can be therapeutically modulated, and identify gaps in knowledge that remain to be addressed.
Collapse
Affiliation(s)
- Maria Zagorulya
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ellen Duong
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Stefani Spranger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
38
|
Understanding and addressing barriers to successful adenovirus-based virotherapy for ovarian cancer. Cancer Gene Ther 2020; 28:375-389. [PMID: 32951021 DOI: 10.1038/s41417-020-00227-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/09/2020] [Indexed: 01/17/2023]
Abstract
Ovarian cancer is the leading cause of death among women with gynecological cancer, with an overall 5-year survival rate below 50% due to a lack of specific symptoms, late stage at time of diagnosis and a high rate of recurrence after standard therapy. A better understanding of heterogeneity, genetic mutations, biological behavior and immunosuppression in the tumor microenvironment have allowed the development of more effective therapies based on anti-angiogenic treatments, PARP and immune checkpoint inhibitors, adoptive cell therapies and oncolytic vectors. Oncolytic adenoviruses are commonly used platforms in cancer gene therapy that selectively replicate in tumor cells and at the same time are able to stimulate the immune system. In addition, they can be genetically modified to enhance their potency and overcome physical and immunological barriers. In this review we highlight the challenges of adenovirus-based oncolytic therapies targeting ovarian cancer and outline recent advances to improve their potential in combination with immunotherapies.
Collapse
|
39
|
James NE, Woodman M, DiSilvestro PA, Ribeiro JR. The Perfect Combination: Enhancing Patient Response to PD-1-Based Therapies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E2150. [PMID: 32756436 PMCID: PMC7466102 DOI: 10.3390/cancers12082150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, with an overall 5-year survival of only 47%. As the development of novel targeted therapies is drastically necessary in order to improve patient survival, current EOC clinical trials have heavily focused on immunotherapeutic approaches, centered upon programmed cell death 1 (PD-1) inhibitors. While PD-1 monotherapies have only exhibited modest responses for patients, it has been theorized that in order to enhance EOC patient response to immunotherapy, combinatorial regimens must be investigated. In this review, unique challenges to EOC PD-1 response will be discussed, along with a comprehensive description of both preclinical and clinical studies evaluating PD-1-based combinatorial therapies. Promising aspects of PD-1-based combinatorial approaches are highlighted, while also discussing specific preclinical and clinical areas of research that need to be addressed, in order to optimize EOC patient immunotherapy response.
Collapse
Affiliation(s)
- Nicole E. James
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Morgan Woodman
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Paul A. DiSilvestro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| | - Jennifer R. Ribeiro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| |
Collapse
|
40
|
Xu Q, Deng B, Li M, Chen Y, Zhuan L. circRNA-UBAP2 promotes the proliferation and inhibits apoptosis of ovarian cancer though miR-382-5p/PRPF8 axis. J Ovarian Res 2020; 13:81. [PMID: 32690086 PMCID: PMC7372761 DOI: 10.1186/s13048-020-00685-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/13/2020] [Indexed: 01/08/2023] Open
Abstract
Objective circular RNAs (circRNAs) have been reported to be essential regulators of multiple malignant cancers. However, the functions of circRNAs in ovarian cancer need to be further explored. The aim of our study is to explore the role of circRNA-UBAP2 in ovarian cancer and its mechanism. Results circRNA-UBAP2 was upregulated in ovarian cancer tissues and cell lines. Knockdown of circRNA-UBAP2 inhibited cell proliferation and promoted cell apoptosis, but circRNA-UBAP2 overexpressed got opposite results. In addition, circRNA-UBAP2 targeted miR-382-5p and downregulated its expression, PRPF8 was a target gene of miR-382-5p. Furthemore, circRNA-UBAP2/miR-382-5p/PRPF8 axis affected the proliferation, apoptosis and cell cycle of ovarian cancer through the mechanism of competing endogenous RNAs (ceRNA). Conclusion circRNA-UBAP2 acted as a ceRNA to sponged miR-382-5p, increased the expression level of PRPF8, and prompted proliferation and inhibited apoptosis in ovarian cancer cells.
Collapse
Affiliation(s)
- Qin Xu
- Department of Reproductive Medicine, Reproductive Medical Centre, The First People's Hospital of Yunnan Province, No. 157 Jin Bi Road, Kunming, People's Republic of China
| | - Bo Deng
- Department of Reproductive Medicine, Reproductive Medical Centre, The First People's Hospital of Yunnan Province, No. 157 Jin Bi Road, Kunming, People's Republic of China
| | - Manlin Li
- Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Yang Chen
- Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Li Zhuan
- Department of Reproductive Medicine, Reproductive Medical Centre, The First People's Hospital of Yunnan Province, No. 157 Jin Bi Road, Kunming, People's Republic of China.
| |
Collapse
|
41
|
Volpin V, Michels T, Sorrentino A, Menevse AN, Knoll G, Ditz M, Milenkovic VM, Chen CY, Rathinasamy A, Griewank K, Boutros M, Haferkamp S, Berneburg M, Wetzel CH, Seckinger A, Hose D, Goldschmidt H, Ehrenschwender M, Witzens-Harig M, Szoor A, Vereb G, Khandelwal N, Beckhove P. CAMK1D Triggers Immune Resistance of Human Tumor Cells Refractory to Anti-PD-L1 Treatment. Cancer Immunol Res 2020; 8:1163-1179. [PMID: 32665263 DOI: 10.1158/2326-6066.cir-19-0608] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/16/2020] [Accepted: 07/09/2020] [Indexed: 11/16/2022]
Abstract
The success of cancer immunotherapy is limited by resistance to immune checkpoint blockade. We therefore conducted a genetic screen to identify genes that mediated resistance against CTLs in anti-PD-L1 treatment-refractory human tumors. Using PD-L1-positive multiple myeloma cells cocultured with tumor-reactive bone marrow-infiltrating CTL as a model, we identified calcium/calmodulin-dependent protein kinase 1D (CAMK1D) as a key modulator of tumor-intrinsic immune resistance. CAMK1D was coexpressed with PD-L1 in anti-PD-L1/PD-1 treatment-refractory cancer types and correlated with poor prognosis in these tumors. CAMK1D was activated by CTL through Fas-receptor stimulation, which led to CAMK1D binding to and phosphorylating caspase-3, -6, and -7, inhibiting their activation and function. Consistently, CAMK1D mediated immune resistance of murine colorectal cancer cells in vivo The pharmacologic inhibition of CAMK1D, on the other hand, restored the sensitivity toward Fas-ligand treatment in multiple myeloma and uveal melanoma cells in vitro Thus, rapid inhibition of the terminal apoptotic cascade by CAMK1D expressed in anti-PD-L1-refractory tumors via T-cell recognition may have contributed to tumor immune resistance.
Collapse
Affiliation(s)
- Valentina Volpin
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany.,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany
| | - Tillmann Michels
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany.,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany.,iOmx Therapeutics AG, Martinsried/Munich, Germany
| | - Antonio Sorrentino
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany.,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany
| | - Ayse N Menevse
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Madlen Ditz
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, Regensburg, Germany
| | - Chih-Yeh Chen
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Anchana Rathinasamy
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium, Essen, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signalling and Functional Genomics, Heidelberg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, Molecular Neurosciences, University of Regensburg, Regensburg, Germany
| | - Anja Seckinger
- Labor für Myelomforschung, Medizinische Klinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Dirk Hose
- Department of Hematology and Immunology, Myeloma Center Brussels, Jette, Belgium
| | - Hartmut Goldschmidt
- Department of Internal Medicine V and National Center of Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Mathias Witzens-Harig
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Arpad Szoor
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI), University Regensburg, Regensburg, Germany. .,German Cancer Research Center (DKFZ), Translational Immunology, Heidelberg, Germany.,Department of Hematology, Oncology, Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
42
|
Zeng XY, Yuan J, Wang C, Zeng D, Yong JH, Jiang XY, Lan H, Xiao SS. circCELSR1 facilitates ovarian cancer proliferation and metastasis by sponging miR-598 to activate BRD4 signals. Mol Med 2020; 26:70. [PMID: 32640974 PMCID: PMC7346459 DOI: 10.1186/s10020-020-00194-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/22/2020] [Indexed: 02/08/2023] Open
Abstract
Background Ovarian cancer is one of the most common gynecologic cancers and has high mortality rate due to the lack of early diagnosis method and efficient therapeutic agents. circCELSR1 is up-regulated in ovarian cancer, but its role and mechanisms in ovarian cancer are unclear. Methods Gene expression of circCELSR1, miR-598 and BRD4 in ovarian cells was examined by qRT-PCR. Protein level was determined by Western blotting. Bioinformatic analysis and luciferase assay determined the molecular binding among circCELSR1, miR-598 and BRD4 3′ UTR. Cell proliferation, migration, invasion and apoptosis were determined by colony formation, wound healing assay, transwell assay and flow cytometry analysis, respectively. An abdominal cavity metastasis nude mice model was used to determine the in vivo function of circCELSR1. Results circCELSR1 and BRD4 were promoted, but miR-598 was suppressed in various ovarian cancer cells. circCELSR1 bound to miR-598 and promoted expression of its downstream target BRD4. Knockdown of circCELSR1 suppressed proliferation, migration, invasion and epithelial-mesenchymal transition (EMT), but promoted apoptosis in ovarian cancer cells, and these effects were reversed by miR-598 inhibition or BRD4 overexpression. circCELSR1 inhibition decreased the expression of BRD4 and its downstream proliferation/migration related genes by targeting miR-598. Furthermore, knockdown of circCELSR1 suppressed ovarian cancer growth and metastasis in nude mice. Conclusion Knockdown of circCELSR1 inhibited BRD4-mediated proliferation/migration related signaling via sponging miR-598, thereby repressing ovarian cancer progression. This study provides a new regulatory mechanism of ovarian cancer may facilitate the development of therapeutic agents for ovarian cancer.
Collapse
Affiliation(s)
- Xiang-Yang Zeng
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Jing Yuan
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Chen Wang
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Da Zeng
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Jia-Hui Yong
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Xiao-Yan Jiang
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Hua Lan
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China
| | - Song-Shu Xiao
- Department of Gynecology, the Third Xiangya Hospital of Central South University, No.138 Tongzipo Road, Changsha, 410013, Hunan Province, PR China.
| |
Collapse
|
43
|
Asare-Werehene M, Communal L, Carmona E, Han Y, Song YS, Burger D, Mes-Masson AM, Tsang BK. Plasma Gelsolin Inhibits CD8 + T-cell Function and Regulates Glutathione Production to Confer Chemoresistance in Ovarian Cancer. Cancer Res 2020; 80:3959-3971. [PMID: 32641415 DOI: 10.1158/0008-5472.can-20-0788] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/28/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
Abstract
Although initial treatment of ovarian cancer is successful, tumors typically relapse and become resistant to treatment. Because of poor infiltration of effector T cells, patients are mostly unresponsive to immunotherapy. Plasma gelsolin (pGSN) is transported by exosomes (small extracellular vesicle, sEV) and plays a key role in ovarian cancer chemoresistance, yet little is known about its role in immunosurveillance. Here, we report the immunomodulatory roles of sEV-pGSN in ovarian cancer chemoresistance. In chemosensitive conditions, secretion of sEV-pGSN was low, allowing for optimal CD8+ T-cell function. This resulted in increased T-cell secretion of IFNγ, which reduced intracellular glutathione (GSH) production and sensitized chemosensitive cells to cis-diaminedichloroplatinum (CDDP)-induced apoptosis. In chemoresistant conditions, increased secretion of sEV-pGSN by ovarian cancer cells induced apoptosis in CD8+ T cells. IFNγ secretion was therefore reduced, resulting in high GSH production and resistance to CDDP-induced death in ovarian cancer cells. These findings support our hypothesis that sEV-pGSN attenuates immunosurveillance and regulates GSH biosynthesis, a phenomenon that contributes to chemoresistance in ovarian cancer. SIGNIFICANCE: These findings provide new insight into pGSN-mediated immune cell dysfunction in ovarian cancer chemoresistance and demonstrate how this dysfunction can be exploited to enhance immunotherapy.
Collapse
Affiliation(s)
- Meshach Asare-Werehene
- Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Laudine Communal
- Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Euridice Carmona
- Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Youngjin Han
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Sang Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Dylan Burger
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Benjamin K Tsang
- Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, Ontario, Canada. .,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
44
|
Okła K, Rajtak A, Czerwonka A, Bobiński M, Wawruszak A, Tarkowski R, Bednarek W, Szumiło J, Kotarski J. Accumulation of blood-circulating PD-L1-expressing M-MDSCs and monocytes/macrophages in pretreatment ovarian cancer patients is associated with soluble PD-L1. J Transl Med 2020; 18:220. [PMID: 32487171 PMCID: PMC7268341 DOI: 10.1186/s12967-020-02389-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Previous studies have shown clinical relevance of programmed death-ligand 1 (PD-L1) and soluble PD-L1 (sPD-L1) in human cancers. However, still contradictory results exist. Our aim was evaluation of PD-L1-expressing monocytic myeloid-derived suppressor cells (M-MDSCs), monocytes/macrophages (MO/MA), tumour cells (TC) and immune/inflammatory cells (IC) as well as investigation of the sPD-L1 in ovarian cancer (OC) patients. METHODS The group of 74 pretreatment women were enrollment to the study. The expression of PD-L1 on M-MDSCS and MO/MA was assessed by flow cytometry. The profile of sPD-L1 was examined with ELISA. The expression of PD-L1 in mononuclear cells (MCs) was analyzed using real time PCR. PD-L1 immunohistochemical analysis was prepared on TC and IC. An in silico validation of prognostic significance of PD-L1 mRNA expression was performed based microarray datasets. RESULTS OC patients had significantly higher frequency of MO/MA versus M-MDSC in the blood, ascites and tumour (each p < 0.0001). In contrast, PD-L1 expression was higher on M-MDSCs versus MO/MA in the blood and ascites (each p < 0.0001), but not in the tumour (p > 0.05). Significantly higher accumulation of blood-circulating M-MDSC, MO/MA, PD-L1+M-MDSC, PD-L1+MO/MA and sPD-L1 was observed in patients versus control (p < 0.001, p < 0.05, p < 0.001, p < 0.001 and p < 0.0001, respectively). Accumulation of these factors was clinicopathologic-independent (p > 0.05). The expression of PD-L1 was significantly higher on IC versus TC (p < 0.0001) and was clinicopathologic-independent (p > 0.05) except higher level of PD-L1+TC in the endometrioid versus mucinous tumours. Interestingly, blood-circulating sPD-L1 positively correlated with PD-L1+M-MDSCs (p = 0.03) and PD-L1+MO/MA (p = 0.02) in the blood but not with these cells in the ascites and tumours nor with PD-L1+TC/IC (each p > 0.05). PD-L1 and sPD-L1 were not predictors of overall survival (OS; each p > 0.05). Further validation revealed no association between PD-L1 mRNA expression and OS in large independent OC patient cohort (n = 655, p > 0.05). CONCLUSIONS Although PD-L1 may not be a prognostic factor for OC, our study demonstrated impaired immunity manifested by up-regulation of PD-L1/sPD-L1. Furthermore, there was a positive association between PD-L1+ myeloid cells and sPD-L1 in the blood, suggesting that sPD-L1 may be a noninvasive surrogate marker for PD-L1+myeloid cells immunomonitoring in OC. Overall, these data should be under consideration during future clinical studies/trials.
Collapse
Affiliation(s)
- Karolina Okła
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, 20-081, Lublin, Poland.
| | - Alicja Rajtak
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, 20-081, Lublin, Poland
| | - Arkadiusz Czerwonka
- Department of Virology and Immunology, Maria Curie-Sklodowska University, 20-031, Lublin, Poland
| | - Marcin Bobiński
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, 20-081, Lublin, Poland
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-081, Lublin, Poland
| | - Rafał Tarkowski
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, 20-081, Lublin, Poland
| | - Wiesława Bednarek
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, 20-081, Lublin, Poland
| | - Justyna Szumiło
- Department of Clinical Pathomorphology, Medical University of Lublin, 20-090, Lublin, Poland
| | - Jan Kotarski
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, 20-081, Lublin, Poland
| |
Collapse
|
45
|
Leong L, Tan HL, Cua S, Yong KSM, Chen Q, Choo A. Preclinical Activity of Embryonic Annexin A2-Specific Chimeric Antigen Receptor T Cells Against Ovarian Cancer. Int J Mol Sci 2020; 21:ijms21020381. [PMID: 31936170 PMCID: PMC7013580 DOI: 10.3390/ijms21020381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/30/2019] [Accepted: 01/06/2020] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptors (CARs) have found clinical success in B cell malignancies, but a dearth of potential targets limits their wider clinical application, especially in solid tumours. Here, we describe the development of an anti-annexin A2 CAR, CAR(2448), derived from an antibody found to have activity against epithelial ovarian cancer cell lines. The spacer length of CAR(2448) was optimised based on in vitro cytotoxic activity against ovarian cancer (OC) cell lines via a real-time cytotoxicity assay. The longer spacer CAR(2448)L T cells exhibit significant effector activity, inducing inflammatory cytokine release and cytotoxicity against OC cell lines. Furthermore, CAR(2448)L-BBz T cells induced enhanced survival in an in vivo OC xenograft model and reduced tumour volume by 76.6%. Our preclinical studies of CAR(2448) suggest its potential for the unmet need of novel strategies for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Leonard Leong
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
- NUS Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, Singapore 119077, Singapore
| | - Heng Liang Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
| | - Simeon Cua
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
| | - Kylie Su Mei Yong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore (NUS), Singapore 117575, Singapore
- Correspondence:
| |
Collapse
|