1
|
Jin YP, Li GW, Xu QQ, Wang XL. ZEB1 promotes the immune escape of ovarian cancer through the MCSF-CCL18 axis. Cancer Cell Int 2025; 25:95. [PMID: 40089803 PMCID: PMC11909986 DOI: 10.1186/s12935-025-03724-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
This study aimed to determine the molecular mechanisms underlying immune escape in ovarian cancer. Samples of ovarian cancer were used to explore the regulatory pathways involved in the malignant phenotype. Tumor cell models with different levels of factor expression were constructed via transfection, and their regulation was determined through investigation of protein expressions. Moreover, our study aimed to investigate the effects of M2 polarization and TAMs aggregation on the apoptosis of CD8 + T-cells, and determine their regulatory axis. Results revealed ZEB1 may promote CCL18 expression via upregulation of MCSF concentration. Notably, high CCL18 expression levels were associated with the aggregation of M2-TAMs and the apoptosis of CD8 + T-cells. In addition, results of the present study demonstrated that the proliferation and invasion of ovarian cancer cells with high expression levels of proteins associated with ZEB1 signal pathway were increased. At the same time the growth rate of tumors in mice was reduced following ZEB1 knockdown, and the volume/weight of tumors were markedly decreased both in vitro and in vivo. Moreover, our results revealed that the aggregation of M2-TAMs and the apoptosis of CD8 + T-cells were significantly decreased in tumor cells following ZEB1 knockdown. Thus, these results verified that ZEB1 may promote the M2 polarization of TAMs via the MCSF axis, leading to the increased secretion of CCL18. Moreover, the MCSF axis may mediate immune escape through the induction of CD8 + T-cell apoptosis, ultimately promoting the malignant phenotype in ovarian cancer cells.
Collapse
Affiliation(s)
- Yan-Ping Jin
- School of Nursing, Jiangsu Health Vocational College, Nanjing, 211899, Jiangsu, China
| | - Guo-Wei Li
- School of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing, 210038, Jiangsu, China.
| | - Qian-Qian Xu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital North), Suzhou, 215008, China
| | - Xiao-Lan Wang
- Department of Obstetrics and Gynecology, Zhongda Hospital Jiangbei Branch, School of Medicine, Southeast University, Nanjing, 210048, Jiangsu, China
| |
Collapse
|
2
|
Pounds R, Croft W, Pearce H, Hossain T, Singh K, Balega J, Jeevan DN, Sundar S, Kehoe S, Yap J, Moss P, Zuo J. The emergence of DNAM-1 as the facilitator of NK cell-mediated killing in ovarian cancer. Front Immunol 2025; 15:1477781. [PMID: 39835114 PMCID: PMC11743932 DOI: 10.3389/fimmu.2024.1477781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Ovarian cancer (OC) is the sixth most common malignancy in women and the poor 5-year survival emphasises the need for novel therapies. NK cells play an important role in the control of malignant disease but the nature of tumour-infiltrating and peripheral NK cells in OC remains unclear. Methods Using flow cytometric analysis, we studied the phenotype and function of NK cells in blood, primary tumour and metastatic tissue in 80 women with OC. The cell type contexture of metastatic OC tissue was explored utilising scRNAseq analysis, with a focus on portraying an immunogenic tumour microenvironment and determining the characteristics of a dysfunctional NK cell population. Results The proportion of peripheral NK cells was markedly elevated with a highly activated profile and increased cytotoxicity. In contrast, NK cell numbers in primary tumour and metastasis were substantially reduced, with downregulation of activatory receptors together with elevated PD-1 expression. scRNA-Seq identified 5 NK cell subpopulations along with increased exhausted and immature NK cells within tumour tissue compared to normal tissue. These features were attenuated following chemotherapy where higher levels of activated and cytotoxic NK cells associated with improved disease-free survival. Correlation of NK cell phenotype with clinical outcomes revealed high levels of DNAM-1 expression on tissue-localised and peripheral NK cells to be associated with reduced survival. Expression of PVR, the DNAM-1 ligand, was significantly increased on tumours and DNAM-1 mediated NK cell lysis of primary tumour tissue was observed in vitro. Discussion These findings reveal profound modulation of the tumour tissue and systemic profile of NK cells which likely contributes to the high rates of local progression and metastasis seen with OC. Immunotherapeutic approaches that overcome local immune suppression and enhance DNAM-1-targeted lysis of OC offer the potential to improve disease control.
Collapse
Affiliation(s)
- Rachel Pounds
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, United Kingdom
| | - Wayne Croft
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tasnia Hossain
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Kavita Singh
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, United Kingdom
| | - Janos Balega
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, United Kingdom
| | - David N. Jeevan
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sudha Sundar
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, United Kingdom
| | - Sean Kehoe
- Oxford Gynaecological Cancer Centre, Churchill Hospital, Oxford University Hospitals Foundation Trust, Oxford, United Kingdom
| | - Jason Yap
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Somri-Gannam L, Meisel-Sharon S, Hantisteanu S, Bar-Noy T, Sigal E, Groisman G, Hallak M, Werner H, Bruchim I. IGF1R inhibition and PD-1 blockade improve anti-tumor immune response in epithelial ovarian cancer. Front Oncol 2024; 14:1410447. [PMID: 39450263 PMCID: PMC11499063 DOI: 10.3389/fonc.2024.1410447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/10/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction The insulin-like growth factor (IGF) system plays a key role in regulating growth and invasiveness in epithelial ovarian cancer (EOC) and is considered a promising therapeutic target. EOC is an immunosuppressive disease, although there are limited data about the involvement of the IGF1R system in the anti-tumor immune response in the EOC microenvironment. Methods In the current study, we hypothesized that IGF 1 receptor (IGF1R) involvement in the maturation of dendritic cells (DC) with the co-inhibition of IGF1R and PD-1 would affect the EOC microenvironment. Results We found that DC pretreated with IGF1R inhibitor resulted in fewer EOC cells. Moreover, in vivo experiments conducted with an EOC mouse model, with anti-PD-1/IGF1R combined, resulted in lower tumor weight compared to individual treatments. Additionally, anti-PD-1/IGF1R treatment increased DC by 34% compared with AEW-541 and 40% with anti-PD-1. The combined treatment increased CD8+ T-cell levels compared to AEW-541 alone. RNA-seq data analysis indicated that anti-PD-1/IGF1R led to a more potent immune response, as reflected by altered gene expression levels related to anti-tumor immune response, compared with either treatment alone. Discussion These findings provide novel evidence that IGF1R axis inhibition combined with PD-1 blockade may be an effective therapeutic strategy for selected EOC patient populations.
Collapse
Affiliation(s)
- Lina Somri-Gannam
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shilhav Meisel-Sharon
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Shay Hantisteanu
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Tomer Bar-Noy
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Emiliya Sigal
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Gabriel Groisman
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Mordechai Hallak
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Bruchim
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| |
Collapse
|
4
|
Abounar SA, El-Nikhely NA, Turkowski K, Savai R, Saeed H. CRISPR/Cas-Mediated Knockdown of PD-L1 and KRAS in Lung Cancer Cells. Int J Mol Sci 2024; 25:9086. [PMID: 39201772 PMCID: PMC11354560 DOI: 10.3390/ijms25169086] [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: 06/07/2024] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Cancer cells can escape death and surveillance by the host immune system in various ways. Programmed cell death ligand 1 (PD-L1) is a transmembrane protein that is expressed by most cell types, including cancer cells, and can provide an inhibitory signal to its receptor PD-1, which is expressed on the surface of activated T cells, impairing the immune response. PD-L1/PD-1-mediated immune evasion is observed in several KRAS-mutated cancers. In the current study, we used the CRISPR/Cas9 system to knock down PD-L1 and KRAS in adenocarcinoma lung cells (A549 and H1975). Knockdown of PD-L1 was validated by qPCR and coculture with lymphocytes. The cells were functionally analyzed for cell cycle, migration and apoptosis. In addition, the effects of PD-L1 and KRAS downregulation on chemotherapy sensitivity and expression of inflammatory markers were investigated. Suppression of PD-L1 and KRAS led to a slowdown of the cell cycle in the G0/G1 phase and reduced migration, increased sensitivity to chemotherapy and triggered apoptosis of cancer cells. In addition, the conditioned medium of the modulated cells significantly affected the native cancer cells and reduced their viability and drug resistance. Our study suggests that dual silencing of PD-L1 and KRAS by CRISPR/Cas9 may be a promising therapeutic approach for the treatment of lung cancer.
Collapse
Affiliation(s)
- Summer A. Abounar
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt; (S.A.A.); (H.S.)
| | - Nefertiti A. El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt; (S.A.A.); (H.S.)
- Program of Molecular Biotechnology, Faculty of Advanced Basic Sciences, Alamein International University, New Alamein City, Marsa Matrouh 5060310, Egypt
| | - Kati Turkowski
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, 35390 Giessen, Germany
| | - Rajkumar Savai
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, 35390 Giessen, Germany
| | - Hesham Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt; (S.A.A.); (H.S.)
| |
Collapse
|
5
|
Kment J, Newsted D, Young S, Vermeulen MC, Laight BJ, Greer PA, Lan Y, Craig AW. Blockade of TGF-β and PD-L1 by bintrafusp alfa promotes survival in preclinical ovarian cancer models by promoting T effector and NK cell responses. Br J Cancer 2024; 130:2003-2015. [PMID: 38622286 PMCID: PMC11183086 DOI: 10.1038/s41416-024-02677-9] [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] [Received: 10/03/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Failure of immunotherapy in high-grade serous ovarian cancer (HGSC) may be due to high levels of transforming growth factor-β (TGF-β) in ascites or tumour immune microenvironment (TIME). Here, we test whether coordinated blockade of TGF-β and PD-L1 with bintrafusp alfa (BA) can provoke anti-tumour immune responses in preclinical HGSC models. METHODS BA is a first-in-class bifunctional inhibitor of TGF-β and PD-L1, and was tested for effects on overall survival and altered TIME in syngeneic HGSC models. RESULTS Using a mouse ID8-derived HGSC syngeneic model with IFNγ-inducible PD-L1 expression, BA treatments significantly reduced ascites development and tumour burden. BA treatments depleted TGF-β and VEGF in ascites, and skewed the TIME towards cytotoxicity compared to control. In the BR5 HGSC syngeneic model, BA treatments increased tumour-infiltrating CD8 T cells with effector memory and cytotoxic markers, as well as cytolytic NK cells. Extended BA treatments in the BR5 model produced ∼50% BA-cured mice that were protected from re-challenge. These BA-cured mice had increased peritoneal T-effector memory and NK cells compared to controls. CONCLUSIONS Our preclinical studies of BA in advanced ovarian cancer models support further testing of BA as an improved immunotherapy option for patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Jacob Kment
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Daniel Newsted
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Stephanie Young
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Michael C Vermeulen
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Brian J Laight
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Peter A Greer
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Yan Lan
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Andrew W Craig
- Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
6
|
Delanghe JR, Delrue C, Speeckaert R, Speeckaert MM. Unlocking the link between haptoglobin polymorphism and noninfectious human diseases: insights and implications. Crit Rev Clin Lab Sci 2024; 61:275-297. [PMID: 38013410 DOI: 10.1080/10408363.2023.2285929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
Haptoglobin (Hp) is a polymorphic protein that was initially described as a hemoglobin (Hb)-binding protein. The major functions of Hp are to scavenge Hb, prevent iron loss, and prevent heme-based oxidation. Hp regulates angiogenesis, nitric oxide homeostasis, immune responses, and prostaglandin synthesis. Genetic polymorphisms in the Hp gene give rise to different phenotypes, including Hp 1-1, Hp 2-1, and Hp 2-2. Extensive research has been conducted to investigate the association between Hp polymorphisms and several medical conditions including cardiovascular disease, inflammatory bowel disease, cancer, transplantation, and hemoglobinopathies. Generally, the Hp 2-2 phenotype is associated with increased disease risk and poor outcomes. Over the years, the Hp 2 allele has spread under genetic pressures. Individuals with the Hp 2-2 phenotype generally exhibit lower levels of CD163 expression in macrophages. The decreased expression of CD163 may be associated with the poor antioxidant capacity in the serum of subjects carrying the Hp 2-2 phenotype. However, the Hp 1-1 phenotype may confer protection in some cases. The Hp1 allele has strong antioxidant, anti-inflammatory, and immunomodulatory properties. It is important to note that the benefits of the Hp1 allele may vary depending on genetic and environmental factors as well as the specific disease or condition under consideration. Therefore, the Hp1 allele may not necessarily confer advantages in all situations, and its effects may be context-dependent. This review highlights the current understanding of the role of Hp polymorphisms in cardiovascular disease, inflammatory bowel disease, cancer, transplantation, hemoglobinopathies, and polyuria.
Collapse
Affiliation(s)
- Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
- Research Foundation-Flanders (FWO), Brussels, Belgium
| |
Collapse
|
7
|
Said SA, de Hullu JA, van der Aa MA, Walraven JEW, Bekkers RLM, Slangen BFM, Pickkers P, van Altena AM. Impact of Sepsis on the Oncologic Outcomes of Advanced Epithelial Ovarian Cancer Patients: A Multicenter Observational Study. Cancers (Basel) 2023; 15:4642. [PMID: 37760610 PMCID: PMC10526225 DOI: 10.3390/cancers15184642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE The sepsis-induced inflammatory response may potentially affect malignant cells. Recently, a case of spontaneous regression of a histologically confirmed International Federation of Gynecology and Obstetrics (FIGO) stage IIIC epithelial ovarian cancer (EOC) following sepsis was reported. The aim of our study was to assess the impact of sepsis on the oncologic outcomes of advanced-stage EOC patients. METHODS Gynecologic oncologic patients admitted to the Intensive Care Unit of three oncologic centers between 2006 and 2019 were identified and patients who experienced sepsis following advanced-stage EOC diagnosis were selected. Survival outcomes were compared with advanced-stage EOC patients from the Netherlands Cancer Registry (NCR). To correct for case-mix differences, propensity score matching using 1:3 nearest neighbor matching was conducted after which survival analyses were repeated. RESULTS A total of 18 of 215 patients with advanced-stage EOC experienced sepsis. Sepsis patients had similar distributions of patient, tumor, and treatment characteristics to 3988 patients from the NCR cohort. A total of 3 of 18 patients died from the complications of sepsis. While the remaining patients initially responded to treatment, 14/15 patients relapsed. The median (IQR) overall survival was 31 (24-44) and 35 (20-60) months for the sepsis and unmatched NCR cohort (p = 0.56), respectively. The median (IQR) progression-free survival was 16 (11-21) and 16 (11-27) months (p = 0.90), respectively. Survival outcomes did not differ following propensity matching (overall survival of 31 (24-44) vs. 36 (20-56) months, p = 0.40; progression-free survival of 16 (11-21) and 16 (12-21) months, p = 0.72). CONCLUSION In this observational study, the occurrence of sepsis did not affect the oncologic and survival outcomes of advanced-stage EOC patients.
Collapse
Affiliation(s)
- Sherin A. Said
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 DT Utrecht, The Netherlands;
| | - Joanne A. de Hullu
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
| | - Maaike A. van der Aa
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 DT Utrecht, The Netherlands;
| | - Janneke E. W. Walraven
- Department of Medical Oncology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands
| | - Ruud L. M. Bekkers
- Department of Obstetrics and Gynecology, Catharina Hospital, 5623 EJ Eindhoven, The Netherlands
- GROW–School for Oncology and Reproduction, University of Maastricht, 6229 GT Maastricht, The Netherlands
| | - Brigitte F. M. Slangen
- GROW–School for Oncology and Reproduction, University of Maastricht, 6229 GT Maastricht, The Netherlands
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands;
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
| |
Collapse
|
8
|
Zhao L, Chen X, Wu H, He Q, Ding L, Yang B. Strategies to synergize PD-1/PD-L1 targeted cancer immunotherapies to enhance antitumor responses in ovarian cancer. Biochem Pharmacol 2023; 215:115724. [PMID: 37524205 DOI: 10.1016/j.bcp.2023.115724] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Anti-programmed cell death 1/programmed cell death ligand 1 (anti-PD-1/PD-L1) antibodies have developed rapidly but exhibited modest activity in ovarian cancer (OC), achieving a clinical response rate ranging from 5.9% to 19%. Current evidence indicate that the establishment of an integrated cancer-immunity cycle is a prerequisite for anti-PD-1/PD-L1 antibodies. Any impairment in this cycle, including lack of cancer antigens release, impaired antigen-presenting, decreased T cell priming and activation, less T cells that are trafficked or infiltrated in tumor microenvironment (TME), and low tumor recognition and killings, will lead to decreased infiltrated cytotoxic T cells to tumor bed and treatment failure. Therefore, combinatorial strategies aiming to modify cancer-immunity cycle and reprogram tumor immune microenvironment are of great interest. By far, various strategies have been studied to enhance responsiveness to PD-1/PD-L1 inhibitors in OC. Platinum-based chemotherapy increases neoantigens release; poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) improve the function of antigen-presenting cells and promote the trafficking of T cells into tumors; epigenetic drugs help to complete the immune cycle by affecting multiple steps; immunotherapies like anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies reactivate T cells, and other treatment strategies like radiotherapy helps to increase the expression of tumor antigens. In this review, we will summarize the preclinical studies by analyzing their contribution in modifying the cancer immunity cycle and remodeling tumor environment, and we will also summarize recent progress in clinical trials and discuss some perspectives to improve these treatment strategies.
Collapse
Affiliation(s)
- Lin Zhao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xi Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
9
|
Shakiba Y, Vorobyev PO, Mahmoud M, Hamad A, Kochetkov DV, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recombinant Strains of Oncolytic Vaccinia Virus for Cancer Immunotherapy. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:823-841. [PMID: 37748878 DOI: 10.1134/s000629792306010x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 09/27/2023]
Abstract
Cancer virotherapy is an alternative therapeutic approach based on the viruses that selectively infect and kill tumor cells. Vaccinia virus (VV) is a member of the Poxviridae, a family of enveloped viruses with a large linear double-stranded DNA genome. The proven safety of the VV strains as well as considerable transgene capacity of the viral genome, make VV an excellent platform for creating recombinant oncolytic viruses for cancer therapy. Furthermore, various genetic modifications can increase tumor selectivity and therapeutic efficacy of VV by arming it with the immune-modulatory genes or proapoptotic molecules, boosting the host immune system, and increasing cross-priming recognition of the tumor cells by T-cells or NK cells. In this review, we summarized the data on bioengineering approaches to develop recombinant VV strains for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Yasmin Shakiba
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Pavel O Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Marah Mahmoud
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Azzam Hamad
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Dmitriy V Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Gaukhar M Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Federal Research Clinical Center for Specialized Medical Care and Medical Technologies, Federal Medical-Biological Agency (FMBA), Moscow, 115682, Russia
- Federal Center of Brain Research and Neurotechnologies of the FMBA of Russia, Moscow, 117513, Russia
| | - Vladimir P Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Federal Research Clinical Center for Specialized Medical Care and Medical Technologies, Federal Medical-Biological Agency (FMBA), Moscow, 115682, Russia
- Federal Center of Brain Research and Neurotechnologies of the FMBA of Russia, Moscow, 117513, Russia
| | - Peter M Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Anastasia V Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
10
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
11
|
Li C, Deng T, Cao J, Zhou Y, Luo X, Feng Y, Huang H, Liu J. Identifying ITGB2 as a Potential Prognostic Biomarker in Ovarian Cancer. Diagnostics (Basel) 2023; 13:diagnostics13061169. [PMID: 36980477 PMCID: PMC10047357 DOI: 10.3390/diagnostics13061169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Epithelial ovarian cancer is by far the most lethal gynecological malignancy. The exploration of promising immunomarkers to predict prognosis in ovarian cancer patients remains challenging. In our research, we carried out an integrated bioinformatic analysis of genome expressions and their immune characteristics in the ovarian cancer microenvironment with validation in different experiments. We filtrated 332 differentially expressed genes with 10 upregulated hub genes from the Gene Expression Omnibus database. These genes were closely related to ovarian tumorigenesis. Subsequently, the survival and immune infiltration analysis demonstrated that the upregulation of five candidate genes, ITGB2, VEGFA, CLDN4, OCLN, and SPP1, were correlated with an unfavorable clinical outcome and increased immune cell infiltration in ovarian cancer. Of these genes, ITGB2 tended to be the gene most correlated with various immune cell infiltrations and had a strong correlation with significant M2 macrophages infiltration (r = 0.707, p = 4.71 × 10-39), while it had a moderate correlation with CD4+/CD8+ T cells and B cells. This characteristic explains why the high expression of ITGB2 was accompanied by immune activation but did not reverse carcinogenesis. Additionally, we confirmed that ITGB2 was over-expressed in ovarian cancer tissues and was mainly located in cytoplasm, detected by Western blotting and the immunohistochemical method. In summary, ITGB2 may serve as a prognostic immunomarker for ovarian cancer patients.
Collapse
Affiliation(s)
- Chanyuan Li
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Ting Deng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Junya Cao
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yun Zhou
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiaolin Luo
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yanling Feng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - He Huang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Jihong Liu
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
12
|
Extracellular vesicles derived from M2-polarized tumor-associated macrophages promote immune escape in ovarian cancer through NEAT1/miR-101-3p/ZEB1/PD-L1 axis. Cancer Immunol Immunother 2023; 72:743-758. [PMID: 36319716 DOI: 10.1007/s00262-022-03305-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022]
Abstract
Evidence has been presented demonstrating that CD8+ T cells confer anti-cancer effects, which offers a promising approach to enhance immunotherapy. M2-polarized tumor-associated macrophages (TAMs) could transfer RNA to cancer cells by secreting extracellular vesicles (EVs) and stimulate immune escape of cancer cells. Thus, the current study aimed at exploring how EVs derived from M2-polarized TAMs (M2-TAMs) affected the proliferation of ovarian cancer (OC) cells and apoptosis of CD8+ T cells. M2-TAMs were observed in OC tissues, which promoted proliferation of OC cells and CD8+ T cell apoptosis by secreting EVs. OC-associated differentially expressed gene NEAT1 was screened by bioinformatics analysis. The in vitro and in vivo effects of TAM-EVs-NEAT1 and its regulatory mechanism were assessed using gain- and loss-of-function assays in co-culture systems of TAMs-derived EVs, OC cells, and CD8+ T cells and in tumor-bearing mice. NEAT1 was highly expressed in M2-derived EVs and OC cells co-cultured with M2-derived EVs. NEAT1 sponged miR-101-3p to increase ZEB1 and PD-L1 expression. In vitro and in vivo assays confirmed the tumor-supporting effects of NEAT1 delivered by M2-derived EVs on OC cell proliferation and CD8+ T cell apoptosis as well as tumor growth. Collectively, M2-derived EVs containing NEAT1 exerted a tumor-promoting role in OC via the miR-101-3p/ZEB1/PD-L1 axis.
Collapse
|
13
|
Chen C, Zhang L, Ruan Z. GATA3 Encapsulated by Tumor-Associated Macrophage-Derived Extracellular Vesicles Promotes Immune Escape and Chemotherapy Resistance of Ovarian Cancer Cells by Upregulating the CD24/Siglec-10 Axis. Mol Pharm 2023; 20:971-986. [PMID: 36547230 DOI: 10.1021/acs.molpharmaceut.2c00557] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor-associated macrophages (TAMs) possess great potential in the development of ovarian cancer (OC). Aberrant GATA-binding protein-3 (GATA3) expression has been found in TAM-derived extracellular vesicles (EVs). This study is intended to investigate the regulatory mechanism of TAM-derived EVs, expressing GATA3 in immune escape and chemotherapy resistance of OC cells. In silico analysis was employed to identify differentially expressed genes. The expression of GATA3, CD24, and sialic acid-binding igg-like lectin 10 (Siglec-10) in OC tissues and cells was characterized, with their correlation verified. OC cells were co-cultured with TAM-derived EVs and CD8+T cells. The functional significance of GATA3/CD24/Siglec-10 in immune escape and chemotherapy resistance of OC cells was assayed by the gain and loss of function experiments. In vivo experiments were also performed for further validation. High expressions of GATA3, CD24, and Siglec-10 were observed in OC tissues and cells. GATA3 could be transferred by TAM-derived EVs into OC cells, which facilitated immune escape and resistance to cisplatin of OC cells. GATA3 up-regulated CD24 to increase Siglec-10 expression. The in vivo assay confirmed the promoting effect of GATA3 delivered by TAM-derived EVs on OC through activation of the CD24/Siglec-10 axis. Collectively, TAM-derived EVs harboring GATA3 played a tumor-promoting role in immune escape and chemotherapy resistance of OC cells via the CD24/Siglec-10 axis.
Collapse
Affiliation(s)
- Chao Chen
- Department of Obstetrics and Gynecology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, P.R. China
| | - Li Zhang
- Department of Obstetrics and Gynecology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, P.R. China
| | - Zhengyi Ruan
- Department of Obstetrics and Gynecology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, P.R. China
| |
Collapse
|
14
|
Israelsson P, Björk E, Nagaev I, Nagaeva O, Lundin E, Mincheva-Nilsson L, Ottander U. NKG2D-mediated cytotoxicity improves after primary surgery for high-grade serous ovarian cancer. Am J Reprod Immunol 2023; 89:e13647. [PMID: 36335434 PMCID: PMC10077899 DOI: 10.1111/aji.13647] [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: 04/12/2022] [Revised: 09/28/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022] Open
Abstract
PROBLEM Tumors compromise the patients' immune system to promote their own survival. We have previously reported that HGSC exosomes play a central role, downregulating NKG2D cytotoxicity. Primary surgery's effect on tumor exosomes and NKG2D cytotoxicity in HGSC patients has not been studied before. The overall objective of this study was to explore the effect of surgery on the exosome-induced impairment of NKG2D cytotoxicity in HGSC. METHOD OF STUDY Paired pre- and post-operative blood samples were subjected to cell and exosome analyses regarding the NKG2D receptor and ligands, and NKG2D-mediated cytotoxicity. Lymphocytes were phenotyped by immunoflow cytometry. Exosomes, isolated by ultracentrifugation, and characterized by nanoparticle tracking analysis, transmission and immune electron microscopy and western blot were used in functional cytotoxic experiments. HGSC explant culture-derived exosomes, previously studied by us, were used for comparison. RESULTS HGSC exosomes from patients' sera downregulated NKG2D-mediated cytotoxicity in NK cells of healthy donors. In a subgroup of subjects, NKG2D expression on CTLs and NK cells was upregulated after surgery, correlating to a decrease in the concentration of exosomes in postoperative sera. An overall significantly improved NKG2D-mediated cytotoxic response of the HGSC patients' own NK cells in postoperative compared to preoperative samples was noted. CONCLUSIONS Surgical removal of the primary tumor has a beneficial effect, relieving the exosome-mediated suppression of NKG2D cytotoxicity in HGSC patients, thus boostering their ability to combat cancer.
Collapse
Affiliation(s)
- Pernilla Israelsson
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden.,Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden.,Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Emma Björk
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden.,Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
| | - Ivan Nagaev
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
| | - Olga Nagaeva
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
| | - Eva Lundin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Lucia Mincheva-Nilsson
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
| | - Ulrika Ottander
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| |
Collapse
|
15
|
Yang Q, Bae G, Nadiradze G, Castagna A, Berezhnoy G, Zizmare L, Kulkarni A, Singh Y, Weinreich FJ, Kommoss S, Reymond MA, Trautwein C. Acidic ascites inhibits ovarian cancer cell proliferation and correlates with the metabolomic, lipidomic and inflammatory phenotype of human patients. J Transl Med 2022; 20:581. [PMID: 36503580 PMCID: PMC9743551 DOI: 10.1186/s12967-022-03763-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/05/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The poor prognosis of ovarian cancer patients is strongly related to peritoneal metastasis with the production of malignant ascites. However, it remains largely unclear how ascites in the peritoneal cavity influences tumor metabolism and recurrence. This study is an explorative approach aimed at for a deeper molecular and physical-chemical characterization of malignant ascites and to investigate their effect on in vitro ovarian cancer cell proliferation. METHODS This study included 10 malignant ascites specimens from patients undergoing ovarian cancer resection. Ascites samples were deeply phenotyped by 1H-NMR based metabolomics, blood-gas analyzer based gas flow analysis and flow cytomertry based a 13-plex cytokine panel. Characteristics of tumor cells were investigated in a 3D spheroid model by SEM and metabolic activity, adhesion, anti-apoptosis, migratory ability evaluated by MTT assay, adhesion assay, flowcytometry and scratch assay. The effect of different pH values was assessed by adding 10% malignant ascites to the test samples. RESULTS The overall extracellular (peritoneal) environment was alkaline, with pH of ascites at stage II-III = 7.51 ± 0.16, and stage IV = 7.78 ± 0.16. Ovarian cancer spheroids grew rapidly in a slightly alkaline environment. Decreasing pH of the cell culture medium suppressed tumor features, metabolic activity, adhesion, anti-apoptosis, and migratory ability. However, 10% ascites could prevent tumor cells from being affected by acidic pH. Metabolomics analysis identified stage IV patients had significantly higher concentrations of alanine, isoleucine, phenylalanine, and glutamine than stage II-III patients, while stage II-III patients had significantly higher concentrations of 3-hydroxybutyrate. pH was positively correlated with acetate, and acetate positively correlated with lipid compounds. IL-8 was positively correlated with lipid metabolites and acetate. Glutathione and carnitine were negatively correlated with cytokines IL-6 and chemokines (IL-8 & MCP-1). CONCLUSION Alkaline malignant ascites facilitated ovarian cancer progression. Additionally, deep ascites phenotyping by metabolomics and cytokine investigations allows for a refined stratification of ovarian cancer patients. These findings contribute to the understanding of ascites pathology in ovarian cancer.
Collapse
Affiliation(s)
- Qianlu Yang
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany
| | - Gyuntae Bae
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Giorgi Nadiradze
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Department of General and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Arianna Castagna
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Department of General and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Laimdota Zizmare
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Aditi Kulkarni
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Yogesh Singh
- grid.411544.10000 0001 0196 8249Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Research Institute of Women’s Health, Women’s Hospital, University Hospital Tübingen, Tübingen, Germany
| | - Frank J. Weinreich
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany
| | - Stefan Kommoss
- grid.411544.10000 0001 0196 8249Research Institute of Women’s Health, Women’s Hospital, University Hospital Tübingen, Tübingen, Germany
| | - Marc A. Reymond
- National Center for Pleura and Peritoneum, NCT South-West Germany, Tübingen, Germany ,grid.411544.10000 0001 0196 8249Department of General and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Trautwein
- grid.411544.10000 0001 0196 8249Present Address: Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Lawler J. Counter Regulation of Tumor Angiogenesis by Vascular Endothelial Growth Factor and Thrombospondin-1. Semin Cancer Biol 2022; 86:126-135. [PMID: 36191900 DOI: 10.1016/j.semcancer.2022.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 10/31/2022]
Abstract
Considerable progress has been made in our understanding of the process of angiogenesis in the context of normal and tumor tissue over the last fifty years. Angiogenesis, like most physiological processes, is carefully controlled by dynamic and opposing effects of positive factors, such as vascular endothelial growth factor (VEGF), and negative factors, such as thrombospondin-1. In most cases, the progression of a small mass of cancerous cells to a life-threatening tumor depends upon the initiation of angiogenesis and involves the dysregulation of the angiogenic balance. Whereas our newfound appreciation for the role of angiogenesis in cancer has opened up new avenues for treatment, the success of these treatments, which have focused almost exclusively on antagonizing the VEGF pathway, has been limited to date. It is anticipated that this situation will improve as more therapeutics that target other pathways are developed, more strategies for combination therapies are advanced, more detailed stratification of patient populations occurs, and a better understanding of resistance to anti-angiogenic therapy is gained.
Collapse
Affiliation(s)
- Jack Lawler
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, The Center for Vascular Biology Research, 99 Brookline Ave, Boston MA 02215, United States.
| |
Collapse
|
17
|
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: 22] [Impact Index Per Article: 7.3] [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
|
18
|
Impact of Activation of EGFL7 within Microenvironment of High Grade Ovarian Serous Carcinoma on Infiltration of CD4+ and CD8+ Lymphocytes. Medicina (B Aires) 2022; 58:medicina58050588. [PMID: 35630004 PMCID: PMC9144271 DOI: 10.3390/medicina58050588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 11/23/2022] Open
Abstract
Background: It has been demonstrated that Egfl7 promotes tumor cell escape from immunity by downregulating the activation of tumor blood vessels. Aim: to analyze mRNA expression of EGFL7 within the tumor microenvironment of high-grade ovarian serous carcinoma and its association with a number of intraepithelial CD4+/CD8+ lymphocytes and ICAM-1 expression. Methods: qPCR analysis of EGFL7 mRNA in cancer cells and adjacent stromal endothelium microdissected from formalin-fixed paraffin-embedded tumors of 59 high-grade ovarian serous carcinoma patients, was performed. Infiltration of intraepithelial lymphocytes (CD4+/CD8+) and expression of ICAM-1 were evaluated by immunohistochemistry and compared between tumors with different statuses of EGFL7 expression. Results: EGFL7 was expressed in cancer cells (9/59, 15.25%), endothelium (8/59, 13.56%), or both cancer cells and adjacent endothelium (4/59, 6.78%). ICAM-1 was expressed on cancer cells (47/59, 79.66%), stromal endothelium (46/59, 77.97%), or both epithelium and endothelium (40 of 59, 67.8%). EGFL7-positivity of cancer cells and endothelium was associated with lower intraepithelial inflow of CD4+ (p = 0.022 and p = 0.029, respectively) and CD8+ lymphocytes (p = 0.004 and p = 0.031, respectively) but impact neither epithelial nor endothelial ICAM-1 expression (p = 0.098 and p = 0.119, respectively). The patients’ median follow-up was 23.83 months (range 1.07–78.07). Lack of prognostic significance of EGFL7-status and ICAM-1 expression was notified. Conclusion: EGFL7 is activated in the cancer cells as frequently as in the endothelium of human high-grade ovarian serous carcinoma. Activation of EGFL7 in cancer cells and/or endothelial cells could negatively impact diapedesis regardless of localization.
Collapse
|
19
|
Dholakia J, Scalise CB, Katre AA, Goldsberry WN, Meza-Perez S, Randall TD, Norian LA, Novak L, Arend RC. Sequential modulation of the Wnt/β-catenin signaling pathway enhances tumor-intrinsic MHC I expression and tumor clearance. Gynecol Oncol 2021; 164:170-180. [PMID: 34844776 DOI: 10.1016/j.ygyno.2021.09.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Progress in immunotherapy use for gynecologic malignancies is hampered by poor tumor antigenicity and weak T cell infiltration of the tumor microenvironment (TME). Wnt/β-catenin pathway modulation demonstrated patient benefit in clinical trials as well as enhanced immune cell recruitment in preclinical studies. The purpose of this study was to characterize the pathways by which Wnt/β-catenin modulation facilitates a more immunotherapy-favorable TME. METHODS Human tumor samples and in vivo patient-derived xenograft and syngeneic murine models were administered Wnt/β-catenin modulating agents DKN-01 and CGX-1321 individually or in sequence. Analytical methods included immunohistochemistry, flow cytometry, multiplex cytokine/chemokine array, and RNA sequencing. RESULTS DKK1 blockade via DKN-01 increased HLA/MHC expression in human and murine tissues, correlating with heightened expression of known MHC I regulators: NFkB, IL-1, LPS, and IFNy. PORCN inhibition via CGX-1321 increased production of T cell chemoattractant CXCL10, providing a mechanism for observed increases in intra-tumoral T cells. Diverse leukocyte recruitment was noted with elevations in B cells and macrophages, with increased tumor expression of population-specific chemokines. Sequential DKK1 blockade and PORCN inhibition decreased tumor burden as evidenced by reduced omental weights. CONCLUSIONS Wnt/β-catenin pathway modulation increases MHC I expression and promotes tumor leukocytic infiltration, facilitating a pro-immune TME associated with decreased tumor burden. This intervention overcomes common tumor immune-evasion mechanisms and may render ovarian tumors susceptible to immunotherapy.
Collapse
Affiliation(s)
- Jhalak Dholakia
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Carly B Scalise
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Ashwini A Katre
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Whitney N Goldsberry
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Selene Meza-Perez
- University of Alabama at Birmingham, Division of Immunology & Rheumatology, Birmingham, AL, United States of America
| | - Troy D Randall
- University of Alabama at Birmingham, Division of Immunology & Rheumatology, Birmingham, AL, United States of America; University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, United States of America
| | - Lyse A Norian
- University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, United States of America; University of Alabama at Birmingham, Department of Nutrition Sciences, Birmingham, AL, United States of America
| | - Lea Novak
- University of Alabama at Birmingham, Department of Anatomic Pathology, Birmingham, AL, United States of America
| | - Rebecca C Arend
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America; University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, United States of America.
| |
Collapse
|
20
|
Fucikova J, Coosemans A, Orsulic S, Cibula D, Vergote I, Galluzzi L, Spisek R. Immunological configuration of ovarian carcinoma: features and impact on disease outcome. J Immunother Cancer 2021; 9:jitc-2021-002873. [PMID: 34645669 PMCID: PMC8515436 DOI: 10.1136/jitc-2021-002873] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian carcinoma (EOC) is a relatively rare malignancy but is the fifth-leading cause of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant disease to the peritoneum. At odds with other neoplasms, EOC is virtually insensitive to immune checkpoint inhibitors, correlating with a tumor microenvironment that exhibits poor infiltration by immune cells and active immunosuppression. Here, we comparatively summarize the humoral and cellular features of primary and metastatic EOC, comparatively analyze their impact on disease outcome, and propose measures to alter them in support of treatment sensitivity and superior patient survival.
Collapse
Affiliation(s)
- Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sandra Orsulic
- UCLA David Geffen School of Medicine and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ignace Vergote
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven, Belgium
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
21
|
Sun L, Kees T, Almeida AS, Liu B, He XY, Ng D, Han X, Spector DL, McNeish IA, Gimotty P, Adams S, Egeblad M. Activating a collaborative innate-adaptive immune response to control metastasis. Cancer Cell 2021; 39:1361-1374.e9. [PMID: 34478639 PMCID: PMC8981964 DOI: 10.1016/j.ccell.2021.08.005] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 06/01/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages (TAMs) promote metastasis and inhibit T cells, but macrophages can be polarized to kill cancer cells. Macrophage polarization could thus be a strategy for controlling cancer. We show that macrophages from metastatic pleural effusions of breast cancer patients can be polarized to kill cancer cells with monophosphoryl lipid A (MPLA) and interferon (IFN) γ. MPLA + IFNγ injected intratumorally or intraperitoneally reduces primary tumor growth and metastasis in breast cancer mouse models, suppresses metastasis, and enhances chemotherapy response in an ovarian cancer model. Both macrophages and T cells are critical for the treatment's anti-metastatic effects. MPLA + IFNγ stimulates type I IFN signaling, reprograms CD206+ TAMs to inducible NO synthase (iNOS)+ macrophages, and activates cytotoxic T cells through macrophage-secreted interleukin-12 (IL-12) and tumor necrosis factor alpha (TNFα). MPLA and IFNγ are used individually in clinical practice and together represent a previously unexplored approach for engaging a systemic anti-tumor immune response.
Collapse
Affiliation(s)
- Lijuan Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Tim Kees
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Bodu Liu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David Ng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xiao Han
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
| | - David L Spector
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Iain A McNeish
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Phyllis Gimotty
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA 19104-6021, USA
| | - Sylvia Adams
- Perlmutter Cancer Center, New York University, New York, NY 10016, USA
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
22
|
A HER2 Tri-Specific NK Cell Engager Mediates Efficient Targeting of Human Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13163994. [PMID: 34439149 PMCID: PMC8394622 DOI: 10.3390/cancers13163994] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary HER2 is a marker known to be over-expressed on breast cancer, rendering it one of the most useful solid tumor targets for antibody-based therapies. Despite expression on ovarian cancer, results targeting HER2 in this setting have been disappointing, thus requiring more aggressive approaches. Natural killer (NK) cells are known as principal mediators of cancer cell killing, but cancer cells find ways to deter them. We devised a tri-specific biological drug containing antibody fragments that simultaneously binds NK cells and cancer cells and at the same time delivers a natural cytokine signal that triggers robust NK cell expansion. In vitro studies show the drug augments NK cell killing of a number of HER2-positive human cell lines, while enhancing NK cell activation and proliferation. Studies in mice engrafted with human ovarian cancer showed the drug has anti-tumor efficacy, clearly demonstrating its ability to bolster NK cells in their ability to contain tumor cell growth. Abstract Clinical studies validated antibodies directed against HER2, trastuzumab, and pertuzumab, as useful methodology to target breast cancer cases where HER2 is expressed. The hope was that HER2 targeting using these antibodies in ovarian cancer patients would prove useful as well, but clinical studies have shown lackluster results in this setting, indicating a need for a more comprehensive approach. Immunotherapy approaches stimulating the innate immune system show great promise, although enhancing natural killer (NK) function is not an established mainstream immunotherapy. This study focused on a new nanobody platform technology in which the bispecific antibody was altered to incorporate a cytokine. Herein we describe bioengineered CAM1615HER2 consisting of a camelid VHH antibody fragment recognizing CD16 and a single chain variable fragment (scFv) recognizing HER2 cross-linked by the human interleukin-15 (IL-15) cytokine. This tri-specific killer engager (TriKETM) showed in vitro prowess in its ability to kill ovarian cancer human cell lines. In addition, we demonstrated its efficacy in inducing potent anti-cancer effects in an in vivo xenograft model of human ovarian cancer engrafting both cancer cells and human NK cells. While previous approaches with trastuzumab and pertuzumab faltered in ovarian cancer, the hope is incorporating targeting and cytokine priming within the same molecule will enhance efficacy in this setting.
Collapse
|
23
|
Tonetti CR, de Souza-Araújo CN, Yoshida A, da Silva RF, Alves PCM, Mazzola TN, Derchain S, Fernandes LGR, Guimarães F. Ovarian Cancer-Associated Ascites Have High Proportions of Cytokine-Responsive CD56bright NK Cells. Cells 2021; 10:cells10071702. [PMID: 34359872 PMCID: PMC8306021 DOI: 10.3390/cells10071702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy, with serous histotype as the most prevalent epithelial ovarian cancer (EOC). Peritoneal ascites is a frequent comorbidity in advanced EOC. EOC-associated ascites provide a reliable sampling source for studying lymphocytes directly from tumor environment. Herein, we carried out flow cytometry-based analysis to readdress issues on NK and T lymphocyte subsets in women with advanced EOC, additionally evaluating phenotypic modulation of their intracellular pathways involved in interleukin (IL)-2 and IL-15 signaling. Results depicted ascites as an inflammatory and immunosuppressive environment, presenting significantly (p < 0.0001) higher amounts of IL-6 and IL-10 than in the patients' blood, as well as significantly (p < 0.05) increased expression of checkpoint inhibitory receptors (programmed death protein-1, PD-1) and ectonucleotidase (CD39) on T lymphocytes. However, NK lymphocytes from EOC-associated ascites showed higher (p < 0.05) pS6 phosphorylation compared with NK from blood. Additionally, in vitro treatment of lymphocytes with IL-2 or IL-15 elicited significantly (p < 0.001) phosphorylation of the STAT5 protein in NK, CD3 and CD8 lymphocytes, both from blood and ascites. EOC-associated ascites had a significantly (p < 0.0001) higher proportion of NK CD56bright lymphocytes than blood, which, in addition, were more responsive (p < 0.05) to stimulation by IL-2 than CD56dim NK. EOC-associated ascites allow studies on lymphocyte phenotype modulation in the tumor environment, where inflammatory profile contrasts with the presence of immunosuppressive elements and development of cellular self-regulating mechanisms.
Collapse
Affiliation(s)
- Cláudia Rodrigues Tonetti
- School of Medicine Sciences, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (C.R.T.); (C.N.d.S.-A.); (R.F.d.S.); (S.D.); (L.G.R.F.)
| | - Caroline Natânia de Souza-Araújo
- School of Medicine Sciences, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (C.R.T.); (C.N.d.S.-A.); (R.F.d.S.); (S.D.); (L.G.R.F.)
| | - Adriana Yoshida
- Centro de Atenção Integral à Saúde da Mulher (CAISM), Women’s Hospital José Aristodemo Pinotti, University of Campinas, Rua Alexander Fleming-101, Campinas CEP 13083-881, SP, Brazil;
| | - Rodrigo Fernandes da Silva
- School of Medicine Sciences, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (C.R.T.); (C.N.d.S.-A.); (R.F.d.S.); (S.D.); (L.G.R.F.)
| | - Paulo César Martins Alves
- Center for Investigation in Pediatrics, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (P.C.M.A.); (T.N.M.)
| | - Taís Nitsch Mazzola
- Center for Investigation in Pediatrics, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (P.C.M.A.); (T.N.M.)
| | - Sophie Derchain
- School of Medicine Sciences, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (C.R.T.); (C.N.d.S.-A.); (R.F.d.S.); (S.D.); (L.G.R.F.)
- Centro de Atenção Integral à Saúde da Mulher (CAISM), Women’s Hospital José Aristodemo Pinotti, University of Campinas, Rua Alexander Fleming-101, Campinas CEP 13083-881, SP, Brazil;
| | - Luís Gustavo Romani Fernandes
- School of Medicine Sciences, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (C.R.T.); (C.N.d.S.-A.); (R.F.d.S.); (S.D.); (L.G.R.F.)
| | - Fernando Guimarães
- School of Medicine Sciences, University of Campinas, Rua Tessália Vieira de Camargo-126, Campinas CEP 13083-887, SP, Brazil; (C.R.T.); (C.N.d.S.-A.); (R.F.d.S.); (S.D.); (L.G.R.F.)
- Centro de Atenção Integral à Saúde da Mulher (CAISM), Women’s Hospital José Aristodemo Pinotti, University of Campinas, Rua Alexander Fleming-101, Campinas CEP 13083-881, SP, Brazil;
- Correspondence: ; Tel.: +55-(19)-35219462
| |
Collapse
|
24
|
Xu QR, Tang J, Liao HY, Yu BT, He XY, Zheng YZ, Liu S. Long non-coding RNA MEG3 mediates the miR-149-3p/FOXP3 axis by reducing p53 ubiquitination to exert a suppressive effect on regulatory T cell differentiation and immune escape in esophageal cancer. J Transl Med 2021; 19:264. [PMID: 34140005 PMCID: PMC8212454 DOI: 10.1186/s12967-021-02907-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/24/2021] [Indexed: 01/27/2023] Open
Abstract
Background Long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3) has been implicated in the progression of esophageal cancer (EC). However, the specific mechanism of the involvement of MEG3 in EC development in relation to the regulation of immune escape remains uncertain. Thus, the aim of the current study was to investigate the effect of MEG3 on EC via microRNA-149-3p (miR-149-3p). Methods Gain- and loss-of-function experiments were initially performed in EC cells in addition to the establishment of a 4-nitroquinoline 1-oxide-induced EC mouse model aimed at evaluating the respective roles of forkhead box P3 (FOXP3), MEG3, miR-149-3p, mouse double minute 2 homolog (MDM2) and p53 in T cell differentiation and immune escape observed in EC. Results EC tissues were found to exhibit upregulated FOXP3 and MDM2 while MEG3, p53 and miR-149-3p were all downregulated. FOXP3 was confirmed to be a target gene of miR-149-3p with our data suggesting it reduced p53 ubiquitination and degradation by means of inhibiting MDM2. P53 was enriched in the promoter of miR-149-3p to upregulate miR-149-3p. The overexpression of MEG3, p53 or miR-149-3p or silencing FOXP3 was associated with a decline in CD25+FOXP3+CD4+ T cells, IL-10+CD4+ T cells and IL-4+CD4+ T cells in spleen tissues, IL-4, and IL-10 levels as well as C-myc, N-myc and Ki-67 expression in EC mice. Conclusion Collectively, MEG3 decreased FOXP3 expression and resulted in repressed regulatory T cell differentiation and immune escape in EC mice by upregulating miR-149-3p via MDM2-mediated p53. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02907-1.
Collapse
Affiliation(s)
- Qi-Rong Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Jian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Hong-Ying Liao
- Department of Thoracic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen UniversityMedical University, No. 26, Erheng Road, Yuancun, Tianhe District, Guangzhou, 510655, Guangdong Province, P. R. China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xiang-Yuan He
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Yu-Zhen Zheng
- Department of Thoracic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen UniversityMedical University, No. 26, Erheng Road, Yuancun, Tianhe District, Guangzhou, 510655, Guangdong Province, P. R. China.
| | - Sheng Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
25
|
Pujade-Lauraine E, Fujiwara K, Ledermann JA, Oza AM, Kristeleit R, Ray-Coquard IL, Richardson GE, Sessa C, Yonemori K, Banerjee S, Leary A, Tinker AV, Jung KH, Madry R, Park SY, Anderson CK, Zohren F, Stewart RA, Wei C, Dychter SS, Monk BJ. Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN Ovarian 200): an open-label, three-arm, randomised, phase 3 study. Lancet Oncol 2021; 22:1034-1046. [PMID: 34143970 DOI: 10.1016/s1470-2045(21)00216-3] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Most patients with ovarian cancer will relapse after receiving frontline platinum-based chemotherapy and eventually develop platinum-resistant or platinum-refractory disease. We report results of avelumab alone or avelumab plus pegylated liposomal doxorubicin (PLD) compared with PLD alone in patients with platinum-resistant or platinum-refractory ovarian cancer. METHODS JAVELIN Ovarian 200 was an open-label, parallel-group, three-arm, randomised, phase 3 trial, done at 149 hospitals and cancer treatment centres in 24 countries. Eligible patients were aged 18 years or older with epithelial ovarian, fallopian tube, or peritoneal cancer (maximum of three previous lines for platinum-sensitive disease, none for platinum-resistant disease) and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1:1) via interactive response technology to avelumab (10 mg/kg intravenously every 2 weeks), avelumab plus PLD (40 mg/m2 intravenously every 4 weeks), or PLD and stratified by disease platinum status, number of previous anticancer regimens, and bulky disease. Primary endpoints were progression-free survival by blinded independent central review and overall survival in all randomly assigned patients, with the objective to show whether avelumab alone or avelumab plus PLD is superior to PLD. Safety was assessed in all patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT02580058. The trial is no longer enrolling patients and this is the final analysis of both primary endpoints. FINDINGS Between Jan 5, 2016, and May 16, 2017, 566 patients were enrolled and randomly assigned (combination n=188; PLD n=190, avelumab n=188). At data cutoff (Sept 19, 2018), median duration of follow-up for overall survival was 18·4 months (IQR 15·6-21·9) for the combination group, 17·4 months (15·2-21·3) for the PLD group, and 18·2 months (15·8-21·2) for the avelumab group. Median progression-free survival by blinded independent central review was 3·7 months (95% CI 3·3-5·1) in the combination group, 3·5 months (2·1-4·0) in the PLD group, and 1·9 months (1·8-1·9) in the avelumab group (combination vs PLD: stratified HR 0·78 [repeated 93·1% CI 0·59-1·24], one-sided p=0·030; avelumab vs PLD: 1·68 [1·32-2·60], one-sided p>0·99). Median overall survival was 15·7 months (95% CI 12·7-18·7) in the combination group, 13·1 months (11·8-15·5) in the PLD group, and 11·8 months (8·9-14·1) in the avelumab group (combination vs PLD: stratified HR 0·89 [repeated 88·85% CI 0·74-1·24], one-sided p=0·21; avelumab vs PLD: 1·14 [0·95-1·58], one-sided p=0·83]). The most common grade 3 or worse treatment-related adverse events were palmar-plantar erythrodysesthesia syndrome (18 [10%] in the combination group vs nine [5%] in the PLD group vs none in the avelumab group), rash (11 [6%] vs three [2%] vs none), fatigue (ten [5%] vs three [2%] vs none), stomatitis (ten [5%] vs five [3%] vs none), anaemia (six [3%] vs nine [5%] vs three [2%]), neutropenia (nine [5%] vs nine [5%] vs none), and neutrophil count decreased (eight [5%] vs seven [4%] vs none). Serious treatment-related adverse events occurred in 32 (18%) patients in the combination group, 19 (11%) in the PLD group, and 14 (7%) in the avelumab group. Treatment-related adverse events resulted in death in one patient each in the PLD group (sepsis) and avelumab group (intestinal obstruction). INTERPRETATION Neither avelumab plus PLD nor avelumab alone significantly improved progression-free survival or overall survival versus PLD. These results provide insights for patient selection in future studies of immune checkpoint inhibitors in platinum-resistant or platinum-refractory ovarian cancer. FUNDING Pfizer and Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
| | - Keiichi Fujiwara
- Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Jonathan A Ledermann
- University College London Cancer Institute, London, UK; University College London Hospitals, London, UK
| | - Amit M Oza
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rebecca Kristeleit
- University College London Cancer Institute, London, UK; University College London Hospitals, London, UK
| | - Isabelle-Laure Ray-Coquard
- Centre Léon Bérard, Service de Cancérologie Médicale, Université Claude Bernard Lyon 1, Lyon, France; Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Paris, France
| | - Gary E Richardson
- Cabrini Hospital, Department of Medical Oncology, Malvern, VIC, Australia
| | - Cristiana Sessa
- Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Susana Banerjee
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Alexandra Leary
- Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, South Korea
| | - Radoslaw Madry
- Poznan University of Medical Sciences, Department of Oncology, Poznan, Poland
| | - Sang-Yoon Park
- Center for Uterine Cancer, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, South Korea
| | | | | | | | - Caimiao Wei
- Pfizer, Global Biometrics and Data Management, Groton, CT, USA
| | | | - Bradley J Monk
- Arizona Oncology (US Oncology Network), University of Arizona College of Medicine, Creighton University School of Medicine, Phoenix, AZ, USA
| |
Collapse
|
26
|
Niu N, Shen W, Zhong Y, Bast RC, Jazaeri A, Sood AK, Liu J. Expression of B7-H4 and IDO1 is associated with drug resistance and poor prognosis in high-grade serous ovarian carcinomas. Hum Pathol 2021; 113:20-27. [PMID: 33887301 DOI: 10.1016/j.humpath.2021.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/27/2022]
Abstract
High-grade serous ovarian carcinoma (HGSC) is the most lethal gynecologic malignancy. While immune checkpoint inhibitors against PD-L1 and CTLA-4 have shown significant effects in multiple tumor types, the response rate to single-agent immune checkpoint inhibitors is low in HGSC. Alternative biomarkers and targets must be identified to guide patient selection and new therapeutic strategies in HGSC. Here, we aim to investigate the clinical significance of novel immune modulators, including B7-H4, IDO1, Tim3, IL6, and IL-8, in patients with HGSC. A total of 48 patients with HGSCs, comprising 24 cases that were sensitive and 24 that were resistant to standard paclitaxel and carboplatin chemotherapy, were selected for our initial analysis. A NanoString assay including 33 immune-related genes was used to compare the expression of different immune regulatory molecules in the sensitive and resistant groups. Differentially expressed proteins were verified using multiplex immunohistochemical staining on tissue arrays of 202 patients with HGSCs who underwent primary surgery at MDACC. We analyzed the expression levels of immune checkpoints and compared expression profiles with clinicopathologic features including response, progression-free survival, and overall survival. HGSC tumors resistant to therapy expressed higher levels of B7-H4 (69.3%), IDO1 (71.8%), Tim3 (89.1%), and inflammatory factors IL-6 and IL-8, and expressed higher Tim3 in stromal components. High expression of B7-H4 and IDO1 was associated with significantly lower overall survival and progression-free survival. B7-H4 and IDO1 were co-expressed in 49.1% of studied cases. A panel of immunomodulatory proteins including B7-H4, IDO1, Tim3, IL-6, and IL-8 are expressed at high levels in HGSCs. These modulators represent novel targets to enhance immunotherapy in patients with HGSCs.
Collapse
Affiliation(s)
- Na Niu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiwei Shen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, 710038, China
| | - Yanping Zhong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Jain S, Annett SL, Morgan MP, Robson T. The Cancer Stem Cell Niche in Ovarian Cancer and Its Impact on Immune Surveillance. Int J Mol Sci 2021; 22:4091. [PMID: 33920983 PMCID: PMC8071330 DOI: 10.3390/ijms22084091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is an aggressive gynaecological cancer with extremely poor prognosis, due to late diagnosis as well as the development of chemoresistance after first-line therapy. Research advances have found stem-like cells present in ovarian tumours, which exist in a dynamic niche and persist through therapy. The stem cell niche interacts extensively with the immune and non-immune components of the tumour microenvironment. Significant pathways associated with the cancer stem cell niche have been identified which interfere with the immune component of the tumour microenvironment, leading to immune surveillance evasion, dysfunction and suppression. This review aims to summarise current evidence-based knowledge on the cancer stem cell niche within the ovarian cancer tumour microenvironment and its effect on immune surveillance. Furthermore, the review seeks to understand the clinical consequences of this dynamic interaction by highlighting current therapies which target these processes.
Collapse
Affiliation(s)
| | | | | | - Tracy Robson
- School of Pharmacy and Biomolecular Science, RCSI University of Medicine and Health Sciences, 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (S.J.); (S.L.A.); (M.P.M.)
| |
Collapse
|
28
|
Zou X, Zhao Y, Liang X, Wang H, Zhu Y, Shao Q. Double Insurance for OC: miRNA-Mediated Platinum Resistance and Immune Escape. Front Immunol 2021; 12:641937. [PMID: 33868274 PMCID: PMC8047328 DOI: 10.3389/fimmu.2021.641937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer (OC) is still the leading cause of death among all gynecological malignancies, despite the recent progress in cancer therapy. Immune escape and drug resistance, especially platinum-based chemotherapy, are significant factors causing disease progression, recurrence and poor prognosis in OC patients. MicroRNAs(miRNAs) are small noncoding RNAs, regulating gene expression at the transcriptional level. Accumulating evidence have indicated their crucial roles in platinum resistance. Importantly, they also act as mediators of tumor immune escape/evasion. In this review, we summarize the recent study of miRNAs involved in platinum resistance of OC and systematically analyses miRNAs involved in the regulation of OC immune escape. Further understanding of miRNAs roles and their possible mechanisms in platinum resistance and tumor escape may open new avenues for improving OC therapy.
Collapse
Affiliation(s)
- Xueqin Zou
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China.,Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yangjing Zhao
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China.,Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiuting Liang
- Department of Obstetrics and Gynecology, Xuzhou Hospital Affiliated to Jiangsu University, Xuzhou, China
| | - Hui Wang
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China.,Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yanling Zhu
- Department of Obstetrics and Gynecology, Xuzhou Hospital Affiliated to Jiangsu University, Xuzhou, China
| | - Qixiang Shao
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China.,Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China.,Jiangsu College of Nursing, School of Medical Science and Laboratory Medicine, Huai'an, China
| |
Collapse
|
29
|
Macciò A, Oppi S, Madeddu C. COVID-19 and cytokine storm syndrome: can what we know about interleukin-6 in ovarian cancer be applied? J Ovarian Res 2021; 14:28. [PMID: 33550983 PMCID: PMC7868172 DOI: 10.1186/s13048-021-00772-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Improving early diagnosis along with timely and effective treatment of COVID-19 are urgently needed. However, at present, the mechanisms underlying disease spread and development, defined prognosis, and immune status of patients with COVID-19 remain to be determined. Patients with severe disease state exhibit a hyperinflammatory response associated with cytokine storm syndrome, hypercoagulability, and depressed cell-mediated immunity. These clinical manifestations, sharing similar pathogenesis, have been well-studied in patients with advanced ovarian cancer. The present review suggests treatment approaches for COVID-19 based on strategies used against ovarian cancer, which shares similar immunopathology and associated coagulation disorders.The chronicization of the hyperinflammatory cytokine storm in patients with severe COVID-19 highlights a defective resistance phase that leads to aspecific chronic inflammation, associated with oxidative stress, which impairs specific T-cell response, induces tissue and endothelial damage, and thrombosis associated with systemic effects that lead to severe multi-organ failure and death. These events are similar to those observed in advanced ovarian cancer which share similar pathogenesis mediated primarily by Interleukin-6, which is, as well demonstrated in ovarian cancer, the key cytokine driving the immunopathology, related systemic symptoms, and patient prognosis.Consistent with findings in other disease models with similar immunopathology, such as advanced ovarian cancer, treatment of severe COVID-19 infection should target inflammation, oxidative stress, coagulation disorders, and immunodepression to improve patient outcome. Correctly identifying disease stages, based on available laboratory data, and developing a specific protocol for each phase is essential for effective treatment.
Collapse
Affiliation(s)
- Antonio Macciò
- Department of Gynecologic Oncology, Businco Hospital, "Azienda di Rilievo Nazionale ad Alta Specializzazione G. Brotzu", Via Jenner, 09100, Cagliari, Italy.
| | - Sara Oppi
- Hematology and Transplant Center, Businco Hospital, "Azienda di Rilievo Nazionale ad Alta Specializzazione G. Brotzu", Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
30
|
Glucose metabolism characteristics and TLR8-mediated metabolic control of CD4 + Treg cells in ovarian cancer cells microenvironment. Cell Death Dis 2021; 12:22. [PMID: 33414414 PMCID: PMC7790820 DOI: 10.1038/s41419-020-03272-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022]
Abstract
Immunotherapy is expected to become the most promising new treatment for ovarian cancer owing to its immunogenicity. However, immunosuppression in the tumor microenvironment is a major obstacle to the efficacy of tumor therapy. Studies have found different metabolism ways of regulatory T cells (Tregs) in the cancer environment may be related to the immunosuppression and Toll-like receptor 8 (TLR8) can reverse the suppression function of Tregs. But it is still unclear that if the TLR8-mediated function reversal is associated with the change of glucose metabolism of Tregs. It was found that the positive expression rates of Glut1, HIF-1α, and Ki67 in CD4+ Treg cells of OC were significantly higher than that in benign ovarian tumor and HC, and also significantly higher than that in CD4+ Teffs of OC. What’s more, compared with CD4+ Teff group, CD4+ Tregs highly expressed seven genes and three proteins related to glucose metabolism and had higher levels of glucose uptake and glycolysis. After activating TLR8 signal of CD4+ Tregs, the proliferation level of naive CD4+ T cells was higher than that of the control group. At the same time, the expression levels of eight genes and five proteins related to glucose metabolism in CD4+ Treg cells with TLR8 activated were decreased and levels of glucose uptake and glycolysis were also lower. Furthermore, TLR8 signaling also downregulated the mTOR pathway in CD4+ Tregs. CD4+ Tregs pretreated with 2-deoxy-d-Glucose (2-DG) and galloflavin also attenuated the inhibition of Teffs proliferation. Although CD4+ Tregs pretreated with 2-DG and galloflavin before activating TLR8 signal had no significant difference compared with the group only treated with inhibitors, which suggested TLR8-mediated reversal of CD4+ Treg cells inhibitory function in ovarian cancer cells co-cultured microenvironment had a causal relationship with glucose metabolism.
Collapse
|
31
|
Yang J, Hong S, Zhang X, Liu J, Wang Y, Wang Z, Gao L, Hong L. Tumor Immune Microenvironment Related Gene-Based Model to Predict Prognosis and Response to Compounds in Ovarian Cancer. Front Oncol 2021; 11:807410. [PMID: 34966691 PMCID: PMC8710702 DOI: 10.3389/fonc.2021.807410] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/25/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The tumor immune microenvironment (TIME) has been recognized to be an imperative factor facilitating the acquisition of many cancer-related hallmarks and is a critical target for targeted biological therapy. This research intended to construct a risk score model premised on TIME-associated genes for prediction of survival and identification of potential drugs for ovarian cancer (OC) patients. METHODS AND RESULTS The stromal and immune scores were computed utilizing the ESTIMATE algorithm in OC patient samples from The Cancer Genome Atlas (TCGA) database. Weighted gene co-expression network and differentially expressed genes analyses were utilized to detect stromal-and immune-related genes. The Least Absolute Shrinkage and Selection Operator (LASSO)-Cox regression was utilized for additional gene selection. The genes that were selected were utilized as the input for a stepwise regression to construct a TIME-related risk score (TIMErisk), which was then validated in Gene Expression Omnibus (GEO) database. For the evaluation of the protein expression levels of TIME regulators, the Human Protein Atlas (HPA) dataset was utilized, and for their biological functions, the TIMER and CIBERSORT algorithm, immunoreactivity, and Immune Cell Abundance Identifier (ImmuCellAI) were used. Possible OC medications were forecasted utilizing the Genomics of Drug Sensitivity in Cancer (GDSC) database and connectivity map (CMap). TIMErisk was developed based on ALPK2, CPA3, PTGER3, CTHRC1, PLA2G2D, CXCL11, and ZNF683. High TIMErisk was recognized as a poor factor for survival in the GEO and TCGA databases; subgroup analysis with FIGO stage, grade, lymphatic and venous invasion, debulking, and tumor site also indicated similar results. Functional immune cells corresponded to more incisive immune reactions, including secretion of chemokines and interleukins, natural killer cell cytotoxicity, TNF signaling pathway, and infiltration of activated NK cells, eosinophils, and neutrophils in patients with low TIMErisk. Several small molecular medications which may enhance the prognosis of patients in the TIMErisk subgroup were identified. Lastly, an enhanced predictive performance nomogram was constructed by compounding TIMErisk with the FIGO stage and debulking. CONCLUSION These findings may offer a valuable indicator for clinical stratification management and personalized therapeutic options for OC patients and may be a foundation for future mechanistic research of their association.
Collapse
|
32
|
McCaw TR, Goel N, Brooke DJ, Katre AA, Londoño AI, Smith HJ, Randall TD, Arend RC. Class I histone deacetylase inhibition promotes CD8 T cell activation in ovarian cancer. Cancer Med 2020; 10:709-717. [PMID: 33369199 PMCID: PMC7877343 DOI: 10.1002/cam4.3337] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Patients with epithelial ovarian cancer (EOC) typically present with late‐stage disease, posing a significant challenge to treatment. Although taxane and platinum‐based chemotherapy plus surgical debulking are initially effective, EOC is marked by frequent recurrence with resistant disease. Immunotherapy represents an appealing treatment paradigm given the ability of immune cells to engage metastatic sites and impede recurrence; however, response rates to checkpoint blockade in ovarian cancer have been disappointing. Here, we tested whether class I HDAC inhibition can promote anti‐tumor T cell responses in a spontaneous and nonspontaneous murine model of EOC. Methods We used the spontaneous Tg‐MISIIR‐Tag and nonspontaneous ID8 models of murine ovarian cancer to test this hypothesis. Whole tumor transcriptional changes were assessed using the nCounter PanCancer Mouse Immune Profiling Panel. Changes in select protein expression of regulatory and effector T cells were measured by flow cytometry. Results We found that treatment with the class I HDAC inhibitor entinostat upregulated pathways and genes associated with CD8 T cell cytotoxic function, while downregulating myeloid derived suppressor cell chemoattractants. Suppressive capacity of regulatory T cells within tumors and associated ascites was significantly reduced, reversing the CD8‐Treg ratio. Conclusions Our findings suggest class I HDAC inhibition can promote activation of intratumoral CD8 T cells, potentially by compromising suppressive networks within the EOC tumor microenvironment. In this manner, class I HDAC inhibition might render advanced‐stage EOC susceptible to immunotherapeutic treatment modalities.
Collapse
Affiliation(s)
- Tyler R McCaw
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nidhi Goel
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dewey J Brooke
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashwini A Katre
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Angelina I Londoño
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Haller J Smith
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rebecca C Arend
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
33
|
Roane BM, Meza-Perez S, Katre AA, Goldsberry WN, Randall TD, Norian LA, Birrer MJ, Arend RC. Neutralization of TGFβ Improves Tumor Immunity and Reduces Tumor Progression in Ovarian Carcinoma. Mol Cancer Ther 2020; 20:602-611. [PMID: 33323456 DOI: 10.1158/1535-7163.mct-20-0412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/17/2020] [Accepted: 12/08/2020] [Indexed: 01/10/2023]
Abstract
The immunosuppressive effects of TGFβ promotes tumor progression and diminishes response to therapy. In this study, we used ID8-p53-/- tumors as a murine model of high-grade serous ovarian cancer. An mAb targeting all three TGFβ ligands was used to neutralize TGFβ. Ascites and omentum were collected and changes in T-cell response were measured using flow. Treatment with anti-TGFβ therapy every other day following injection of tumor cells resulted in decreased ascites volume (4.1 mL vs. 0.7 mL; P < 0.001) and improved the CD8:Treg ratio (0.37 vs. 2.5; P = 0.02) compared with untreated mice. A single dose of therapy prior to tumor challenge resulted in a similar reduction of ascites volume (2.7 vs. 0.67 mL; P = 0.002) and increased CD8:Tregs ratio (0.36 vs. 1.49; P = 0.007), while also significantly reducing omental weight (114.9 mg vs. 93.4 mg; P = 0.017). Beginning treatment before inoculation with tumor cells and continuing for 6 weeks, we observe similar changes and prolonged overall survival (median 70 days vs. 57.5 days). TGFβ neutralization results in favorable changes to the T-cell response within the tumor microenvironment, leading to decreased tumor progression in ovarian cancer. The utilization of anti-TGFβ therapy may be an option for management in patients with ovarian cancer to improve clinical outcomes and warrants further investigation.
Collapse
Affiliation(s)
- Brandon M Roane
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Selene Meza-Perez
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ashwini A Katre
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Whitney N Goldsberry
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama
| | - Lyse A Norian
- Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael J Birrer
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Rebecca C Arend
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama. .,Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama
| |
Collapse
|
34
|
Maas RJ, Hoogstad-van Evert JS, Van der Meer JM, Mekers V, Rezaeifard S, Korman AJ, de Jonge PK, Cany J, Woestenenk R, Schaap NP, Massuger LF, Jansen JH, Hobo W, Dolstra H. TIGIT blockade enhances functionality of peritoneal NK cells with altered expression of DNAM-1/TIGIT/CD96 checkpoint molecules in ovarian cancer. Oncoimmunology 2020; 9:1843247. [PMID: 33224630 PMCID: PMC7657585 DOI: 10.1080/2162402x.2020.1843247] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Advanced ovarian cancer (OC) patients have a poor 5-year survival of only 28%, emphasizing the medical need for improved therapies. Adjuvant immunotherapy could be an attractive approach since OC is an immunogenic disease and the presence of tumor-infiltrating lymphocytes has shown to positively correlate with patient survival. Among these infiltrating lymphocytes are natural killer (NK) cells, key players involved in tumor targeting, initiated by signaling via activating and inhibitory receptors. Here, we investigated the role of the DNAM-1/TIGIT/CD96 axis in the anti-tumor response of NK cells toward OC. Ascites-derived NK cells from advanced OC patients showed lower expression of activating receptor DNAM-1 compared to healthy donor peripheral blood NK cells, while inhibitory receptor TIGIT and CD96 expression was equal or higher, respectively. This shift to a more inhibitory phenotype could also be induced in vitro by co-culturing healthy donor NK cells with OC tumor spheroids, and in vivo on intraperitoneally infused NK cells in SKOV-3 OC bearing NOD/SCID-IL2Rγnull (NSG) mice. Interestingly, TIGIT blockade enhanced degranulation and interferon gamma (IFNγ) production of healthy donor CD56dim NK cells in response to OC tumor cells, especially when DNAM-1/CD155 interactions were in place. Importantly, TIGIT blockade boosted functional responsiveness of CD56dim NK cells of OC patients with a baseline reactivity against SKOV-3 cells. Overall, our data show for the first time that checkpoint molecules TIGIT/DNAM-1/CD96 play an important role in NK cell responsiveness against OC, and provides rationale for incorporating TIGIT interference in NK cell-based immunotherapy in OC patients.
Collapse
Affiliation(s)
- Ralph Ja Maas
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janneke S Hoogstad-van Evert
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jolien Mr Van der Meer
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vera Mekers
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Somayeh Rezaeifard
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alan J Korman
- Bristol-Myers Squibb, Redwood City, CA, USA.,AK Vir Biotechnology, San Francisco, CA, USA
| | - Paul Kjd de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeannette Cany
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob Woestenenk
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicolaas Pm Schaap
- Department of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Leon F Massuger
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joop H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
35
|
Chang C, Chen YP, Medeiros LJ, Chen TY, Chang KC. Higher infiltration of intratumoral CD25+ FOXP3+ lymphocytes correlates with a favorable prognosis in patients with diffuse large B-cell lymphoma. Leuk Lymphoma 2020; 62:76-85. [PMID: 32962457 DOI: 10.1080/10428194.2020.1817438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T-cells (Tregs) play an important role in cancer immunity but their prognostic impact is controversial in diffuse large B-cell lymphoma (DLBCL). Intratumoral Tregs in DLBCL (n = 70) were evaluated by double-stained CD25 and FOXP3 lymphocytes in formalin-fixed paraffin-embedded tissues, and correlated with clinicopathologic features. We found that increased numbers of intratumoral FOXP3+ lymphocytes (>2.4/HPF) and CD25 + FOXP3+ lymphocytes (>0.8/HPF) are favorable prognosticators (p = .004 and p < .001, respectively) in DLBCL patients, along with age <70 years, stage I-II disease, normal serum LDH level and low IPI scores (p < .001, .002, .002, and <.001, respectively). On multivariate analyses, a higher number of CD25 + FOXP3+ lymphocytes retained prognostic significance (p = .040). Interestingly, higher Treg infiltration correlated with increased infiltration by cytotoxic T-lymphocytes (γ = 0.294, p = .038) and nodal location (γ = 0.390, p = .004), but not with infiltration by CD123+ plasmacytoid dendritic cells, which were reported to induce Tregs with immune tolerance. Therefore, congruent with literature meta-analyses, higher intratumoral CD25 + FOXP3+ lymphocytes have a beneficial impact on DLBCL.
Collapse
Affiliation(s)
- Chen Chang
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Ping Chen
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tsai-Yun Chen
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Kung-Chao Chang
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
36
|
The influence of secreted factors and extracellular vesicles in ovarian cancer metastasis. EJC Suppl 2020; 15:38-48. [PMID: 33240441 PMCID: PMC7573474 DOI: 10.1016/j.ejcsup.2019.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/02/2019] [Accepted: 09/15/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer cells mainly metastasise within the peritoneal cavity, the lethal consequence of tumour progression in this cancer type. Classically, changes in tumour cells, such as epithelial to mesenchymal transition, involve the down-regulatinon of E-cadherin, activation of extracellular proteases and integrin-mediated adhesion. However, our current understanding of ovarian tumour progression suggests the implication of both intrinsic and extrinsic factors. It has been proposed that ovarian cancer metastases are a consequence of the crosstalk between cancer cells and the tumour microenvironment by soluble factors and extracellular vesicles. Characterisation of the alterations in both the tumour cells and the surrounding microenvironment has emerged as a new research field to understand ovarian cancer metastasis. In this mini review, we will summarise the most recent findings, focusing our attention on the role of secreted factors and extracellular vesicles in ovarian cancer metastasis. During ovarian cancer metastasis, tumour cells metastasise in the mesothelium as primarily ‘soil’ for ovarian cancer ‘seeds’. Soluble factors and extracellular vesicles secreted by tumor cells are involved in the generation of the pre-metastatic niche. Cancer-associated fibroblasts (CAFs) represent the majority of stromal cells in various types of human carcinoma, including ovarian cancer. Analysis of early metastasis to the omentum indicates that ovarian cancer cells rely on the interaction with immune cells such as macrophages. Liquid biopsy analyses in ovarian cancer may help to define novel biomarkers improving patient survival and reduce lethality.
Collapse
|
37
|
Profound Functional Suppression of Tumor-Infiltrating T-Cells in Ovarian Cancer Patients Can Be Reversed Using PD-1-Blocking Antibodies or DARPin® Proteins. J Immunol Res 2020; 2020:7375947. [PMID: 32832572 PMCID: PMC7424497 DOI: 10.1155/2020/7375947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/30/2020] [Indexed: 12/29/2022] Open
Abstract
PD-1/PD-L1 blockade has revolutionized the field of immunooncology. Despite the relative success, the response rate to anti-PD-1 therapy requires further improvements. Our aim was to explore the enhancement of T-cell function by using novel PD-1-blocking proteins and compare with clinically approved monoclonal antibodies (mAbs). We isolated T-cells from the ascites and tumor of 17 patients with advanced epithelial ovarian cancer (EOC) and analyzed the effects using the mAbs nivolumab and pembrolizumab and two novel engineered ankyrin repeat proteins (DARPin® proteins). PD-1 blockade with either mAb or DARPin® molecule significantly increased the release of IFN-γ, granzyme B, IL-2, and TNF-α, demonstrating successful reinvigoration. The monovalent DARPin® protein was less effective compared to its bivalent equivalent, demonstrating that bivalency brings an additional benefit to PD-1 blockade. Overall, we found a higher fold increase of lymphokine secretion in response to the PD-1 blockade by tumor-derived T-cells; however, the absolute amounts were significantly lower compared to the release from ascites-derived T-cells. Our results demonstrate that PD-1 blockade can only partially reinvigorate functionally suppressed T-cells from EOC patients. This warrants further investigation preferably in combination with other therapeutics. The study provides an early pilot proof-of-concept for the potential use of DARPin® proteins as eligible alternative scaffold proteins to block PD-1.
Collapse
|
38
|
Yousefzadeh Y, Hallaj S, Baghi Moornani M, Asghary A, Azizi G, Hojjat-Farsangi M, Ghalamfarsa G, Jadidi-Niaragh F. Tumor associated macrophages in the molecular pathogenesis of ovarian cancer. Int Immunopharmacol 2020; 84:106471. [PMID: 32305830 DOI: 10.1016/j.intimp.2020.106471] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/23/2022]
Abstract
The tumor microenvironment is a critical factor that enhances cancer progression, drug resistance, and failure of therapeutic approaches. Several cellular and non-cellular factors are involved in cancer promotion. Among the several cell populations in the tumor microenvironment, macrophages, as one of the most abundant innate immune cells within the tumor milieu, have attracted extensive attention among several researchers because of their critical role in innate pathophysiology of multiple disorders, as well as ovarian cancer. High plasticity and consequent high ability to adapt to environmental alternations by adjusting their cellular metabolism and immunological phenotype is the notable characteristic of macrophages. Therefore, the critical function of tumor-associated macrophages in ovarian cancer is highlighted in the growing body of recent studies. In this article, we will comprehensively focus on significant impacts of the macrophages on ovarian cancer progression, by discussing the role of macrophages as one of the fundamental immune cells present in tumor milieu, in metabolic reprogramming of transformed cells, and involvement of these cells in the ovarian cancer initiation, progression, invasion, and angiogenesis. Moreover, we will summarise recent studies evaluating the effects of targeting macrophages in ovarian cancer.
Collapse
Affiliation(s)
- Yousef Yousefzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahin Hallaj
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Baghi Moornani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Asghary
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
39
|
Th17 Cells and IL-17 As Novel Immune Targets in Ovarian Cancer Therapy. JOURNAL OF ONCOLOGY 2020; 2020:8797683. [PMID: 32148497 PMCID: PMC7054820 DOI: 10.1155/2020/8797683] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/29/2020] [Indexed: 01/12/2023]
Abstract
Ovarian cancer (OC) is usually diagnosed at an advanced stage and is related with poor prognosis. Despite numerous studies, the pathogenesis of OC is still unknown. Recent studies indicate the role of the immune system in the development and spread of OC. The identification of factors and mechanisms involved in that process and their modulation is crucial for creating effective antitumor therapy. We investigated the potential role of Th17 cells in OC patients (n = 71) by analyzing the frequencies of Th17 cells in three different environments, i.e., peripheral blood (PB), peritoneal fluid (PF), and tissue (Th17 infiltrating cells), and the concentration of IL-17A in plasma and PF of patients in terms of their clinical and prognostic significance. Th17 cells were analyzed by flow cytometry as a percentage of CD4+ lymphocytes that expressed intracellular expression of IL-17A. The level of IL-17A in plasma and PF were determined by ELISA. Our results showed accumulation of Th17 cells among tumor-infiltrating CD4+ lymphocytes (p < 0.001 in relation to PB). Moreover, the percentage of Th17 cells in both PB and PF of OC patients was significantly lower than that in benign tumors group (n = 35). There were no significant differences in the percentage of Th17 cells in PB, PF, and tissue in relation to clinicopathological characteristics of OC patients and survival. The lower percentage of Th17 cells in the PB and PF of OC patients may promote evasion of host immune response by cancer cells. The concentration of IL-17A in plasma of OC patients was higher (p < 0.0001) than that in both benign tumors and control group (n = 10). The PF IL-17A level in OC patients was higher (p < 0.0001) than that in women with benign ovarian tumors, indicating its synthesis in OC microenvironment. Higher IL-17A level in PF is correlated with longer (median: 36.5 vs. 27 months) survival of OC patients.
Collapse
|
40
|
Chandra A, Pius C, Nabeel M, Nair M, Vishwanatha JK, Ahmad S, Basha R. Ovarian cancer: Current status and strategies for improving therapeutic outcomes. Cancer Med 2019; 8:7018-7031. [PMID: 31560828 PMCID: PMC6853829 DOI: 10.1002/cam4.2560] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 12/14/2022] Open
Abstract
Of all the gynecologic tumors, ovarian cancer (OC) is known to be the deadliest. Advanced‐stages of OC are linked with high morbidity and low survival rates despite the immense amount of research in the field. Shortage of promising screening tools for early‐stage detection is one of the major challenges linked with the poor survival rate for patients with OC. In OC, therapeutic management is used with multidisciplinary approaches that includes debulking surgery, chemotherapy, and (rarely) radiotherapy. Recently, there is an increasing interest in using immunomodulation for treating OC. Relapse rates are high in this malignancy and averages around every 2‐years. Further treatments after the relapse are more intense, increasing the toxicity, resistance to chemotherapy drugs, and financial burden to patients with poor quality‐of‐life. A procedure that has been studied to help reduce the morbidity rate involves pre‐sensitizing cancer cells with standard therapy in order to produce optimal results with minimum dosage. Utilizing such an approach, platinum‐based agents are effective due to their increased response to platinum‐based chemotherapy in relapsed cases. These chemo‐drugs also help address the issue of drug resistance. After conducting an extensive search with available literature and the resources for clinical trials, information is precisely documented on current research, biomarkers, options for treatment and clinical trials. Several schemes for enhancing the therapeutic responses for OC are discussed systematically in this review with an attempt in summarizing the recent developments in this exciting field of translational/clinical research.
Collapse
Affiliation(s)
- Ashwin Chandra
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - Cima Pius
- Miami Medical School, Miami, FL, USA
| | - Madiha Nabeel
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | - Maya Nair
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | - Jamboor K Vishwanatha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | | | - Riyaz Basha
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA.,Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
41
|
Nersesian S, Glazebrook H, Toulany J, Grantham SR, Boudreau JE. Naturally Killing the Silent Killer: NK Cell-Based Immunotherapy for Ovarian Cancer. Front Immunol 2019; 10:1782. [PMID: 31456796 PMCID: PMC6699519 DOI: 10.3389/fimmu.2019.01782] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer (OC) is diagnosed in ~22,000 women in the US each year and kills 14,000 of them. Often, patients are not diagnosed until the later stages of disease, when treatment options are limited, highlighting the urgent need for new and improved therapies for precise cancer control. An individual's immune function and interaction with tumor cells can be prognostic of the response to cancer treatment. Current emerging therapies for OC include immunotherapies, which use antibodies or drive T cell-mediated cancer recognition and elimination. In OC, these have been limited by adverse side effects and tumor characteristics including inter- and intra-tumoral heterogeneity, lack of targetable antigens, loss of tumor human leukocyte antigen expression, high levels of immunosuppressive factors, and insufficient immune cell trafficking. Natural killer (NK) cells may be ideal as primary or collateral effectors to these nascent immunotherapies. NK cells exhibit multiple functions that combat immune escape and tumor relapse: they kill targets and elicit inflammation through antigen-independent pathways and detect loss of HLA as a signal for activation. NK cells are efficient mediators of tumor immune surveillance and control, suppressed by the tumor microenvironment and rescued by immune checkpoint blockade. NK cells are regulated by a variety of activating and inhibitory receptors and already known to be central effectors across an array of existing therapies. In this article, we highlight interactions between NK cells and OC and their potential to change the immunosuppressive tumor microenvironment and participate in durable immune control of OC.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.,Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Haley Glazebrook
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jay Toulany
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.,Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Stephanie R Grantham
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jeanette E Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.,Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
42
|
Lee JM, Botesteanu DA, Tomita Y, Yuno A, Lee MJ, Kohn EC, Annunziata CM, Matulonis U, MacDonald LA, Nair JR, Macneill KM, Trepel JB. Patients with BRCA mutated ovarian cancer may have fewer circulating MDSC and more peripheral CD8 + T cells compared with women with BRCA wild-type disease during the early disease course. Oncol Lett 2019; 18:3914-3924. [PMID: 31516602 DOI: 10.3892/ol.2019.10731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022] Open
Abstract
Immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) are associated with immunologic tolerance and poor prognosis in ovarian cancer (OvCa). We hypothesized that women with germline BRCA1 and BRCA2 mutation-associated (gBRCAm) OvCa would have fewer circulating immunosuppressive immune cells compared to those with BRCA wild-type (BRCAwt) disease during their early disease course (<5 years post-diagnosis) where gBRCAm is a favorable prognostic factor. We collected and viably froze peripheral blood mononuclear cells (PBMCs) from patients with recurrent OvCa olaparib clinical trials (NCT01445418/NCT01237067). Immune subset analyses were performed using flow cytometry for Tregs, exhausted CD8+ T cells, monocytes and MDSCs. Functional marker expression, including cytotoxic T lymphocyte-associated protein 4 (CTLA-4), T cell immunoglobulin and mucin domain 3 (TIM-3) and programmed cell death protein 1 (PD-1) was evaluated. Data were analyzed using FlowJo. Pretreatment PBMCs were collected from 41 patients (16 gBRCAm/25 BRCAwt). The percentage of MDSCs among viable CD45+ PBMC was lower in gBRCAm OvCa compared with BRCAwt OvCa (median 0.565 vs. 0.93%, P=0.0086) but this difference was not seen in those women >5 years post-diagnosis. CD8+ T cells among viable CD45+ PBMCs and CTLA-4+/CD8+ T cells were higher in gBRCAm carriers than patients with BRCAwt, in particular for those <5 years post-diagnosis (median 20.4 vs. 9.78%, P=0.031 and median MFI 0.19 vs. 0.22, P=0.0074, respectively). TIM-3 expression on Tregs was associated with poor progression-free survival, independent of gBRCAm status (P<0.001). Our pilot data suggested that patients with gBRCAm OvCa may have fewer circulating MDSCs but higher CD8+ T cells in PBMCs during their early disease course. This may contribute to the observed survival benefit for these women in their first post-diagnosis decade.
Collapse
Affiliation(s)
- Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Dana-Adriana Botesteanu
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yusuke Tomita
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Akira Yuno
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Elise C Kohn
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ursula Matulonis
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Lauren A MacDonald
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Jayakumar R Nair
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Kimberley M Macneill
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Jane B Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat Commun 2019; 10:3000. [PMID: 31278254 PMCID: PMC6611910 DOI: 10.1038/s41467-019-10979-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
Tumor-driven immune suppression is a major barrier to successful immunotherapy in ovarian carcinomas (OvCa). Among various mechanisms responsible for immune suppression, arginase-1 (ARG1)-carrying small extracellular vesicles (EVs) emerge as important contributors to tumor growth and tumor escape from the host immune system. Here, we report that small EVs found in the ascites and plasma of OvCa patients contain ARG1. EVs suppress proliferation of CD4+ and CD8+ T-cells in vitro and in vivo in OvCa mouse models. In mice, ARG1-containing EVs are transported to draining lymph nodes, taken up by dendritic cells and inhibit antigen-specific T-cell proliferation. Increased expression of ARG1 in mouse OvCa cells is associated with accelerated tumor progression that can be blocked by an arginase inhibitor. Altogether, our studies show that tumor cells use EVs as vehicles to carry over long distances and deliver to immune cells a metabolic checkpoint molecule – ARG1, mitigating anti-tumor immune responses. Cancer cells employ a variety of ways to escape the immune system. Here, the authors show that ovarian cancer cells produce small extracellular vescicles containing arginase 1 that are taken up by dendritic cells in the draining lymph nodes, resulting in inhibition of antigen-specific T-cell proliferation.
Collapse
|
44
|
Peptide-mediated delivery of therapeutic mRNA in ovarian cancer. Eur J Pharm Biopharm 2019; 141:180-190. [PMID: 31103743 DOI: 10.1016/j.ejpb.2019.05.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy in the developed world. In spite of intensive research, the mortality has hardly decreased over the past twenty years. This necessitates the exploration of novel therapeutic modalities. Transient protein expression through delivery of mRNA is emerging as a highly promising option. In contrast to gene therapy there is no risk of integration into the genome. Here, we explore the expression of mRNA in models of ovarian cancer of increasing complexity. The cell-penetrating peptide (CPP) PepFect 14 (PF14) was used to formulate CPP-mRNA nanoparticles. Efficient expression of a reporter protein was achieved in two-dimensional tissue cultures and in three-dimensional cancer cell spheroids. PF14 nanoparticles greatly outperformed a lipid-based transfection agent in vivo, leading to expression in various cell types of tumor associated tissue. Protein expression was restricted to the peritoneal cavity. Messenger RNA expression across different cell types was confirmed in primary ovarian cancer explants. As ovarian cancer is confined to the peritoneal cavity in most cases, the results create the basis for applications in which the tumor microenvironment is transiently modified through protein expression.
Collapse
|
45
|
Roane BM, Arend RC, Birrer MJ. Review: Targeting the Transforming Growth Factor-Beta Pathway in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11050668. [PMID: 31091744 PMCID: PMC6562901 DOI: 10.3390/cancers11050668] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 02/07/2023] Open
Abstract
Despite extensive efforts, there has been limited progress in optimizing treatment of ovarian cancer patients. The vast majority of patients experience recurrence within a few years despite a high response rate to upfront therapy. The minimal improvement in overall survival of ovarian cancer patients in recent decades has directed research towards identifying specific biomarkers that serve both as prognostic factors and targets for therapy. Transforming Growth Factor-β (TGF-β) is a superfamily of proteins that have been well studied and implicated in a wide variety of cellular processes, both in normal physiologic development and malignant cellular growth. Hypersignaling via the TGF-β pathway is associated with increased tumor dissemination through various processes including immune evasion, promotion of angiogenesis, and increased epithelial to mesenchymal transformation. This pathway has been studied in various malignancies, including ovarian cancer. As targeted therapy has become increasingly prominent in drug development and clinical research, biomarkers such as TGF-β are being studied to improve outcomes in the ovarian cancer patient population. This review article discusses the role of TGF-β in ovarian cancer progression, the mechanisms of TGF-β signaling, and the targeted therapies aimed at the TGF-β pathway that are currently being studied.
Collapse
Affiliation(s)
- Brandon M Roane
- Department of Obstetrics and Gynecology-Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Rebecca C Arend
- Department of Obstetrics and Gynecology-Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Michael J Birrer
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
46
|
Yahata T, Mizoguchi M, Kimura A, Orimo T, Toujima S, Kuninaka Y, Nosaka M, Ishida Y, Sasaki I, Fukuda-Ohta Y, Hemmi H, Iwahashi N, Noguchi T, Kaisho T, Kondo T, Ino K. Programmed cell death ligand 1 disruption by clustered regularly interspaced short palindromic repeats/Cas9-genome editing promotes antitumor immunity and suppresses ovarian cancer progression. Cancer Sci 2019; 110:1279-1292. [PMID: 30702189 PMCID: PMC6447841 DOI: 10.1111/cas.13958] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/15/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death ligand 1 (PD‐L1) on tumor cells suppresses anti‐tumor immunity and has an unfavorable prognostic impact in ovarian cancer patients. We herein report the pathophysiological and therapeutic impacts of PD‐L1 disruption in ovarian cancer. PD‐L1 was genetically disrupted in the murine ovarian cancer cell line ID8 using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9‐mediated genome editing. PD‐L1 knockout (KO) and control ovarian cancer cells were intraperitoneally inoculated into syngeneic mice, and survival and tumor dissemination were evaluated. Survival times were significantly longer in the PD‐L1‐KO ID8‐inoculated groups than in their control groups, and its therapeutic benefit was enhanced in combination with the cisplatin treatment. Tumor weights and ascites volumes were significantly lower in the PD‐L1‐KO ID8 groups than in their control groups. Immunohistochemical and immunofluorescence analyses showed that intratumoral CD4+ T cells, CD8+ T cells, NK cells and CD11c+ M1 macrophages were significantly increased, whereas regulatory T cells were significantly decreased in the PD‐L1‐KO ID8 groups compared with those in their control groups. The intratumoral mRNA expression of interferon‐γ, tumor‐necrosis factor‐α, interleukin (IL)‐2, IL‐12a, CXCL9 and CXCL10 was significantly stronger, while that of IL‐10, vascular endothelial growth factor, CXCL1 and CXCL2 was significantly weaker in the PD‐L1‐KO ID8 groups. These results indicate that CRISPR/Cas9‐mediated PD‐L1 disruption on tumor cells promotes anti‐tumor immunity by increasing tumor‐infiltrating lymphocytes and modulating cytokine/chemokine profiles within the tumor microenvironment, thereby suppressing ovarian cancer progression. These results suggest that PD‐L1‐targeted therapy by genome editing may be a novel therapeutic strategy for ovarian cancer.
Collapse
Affiliation(s)
- Tamaki Yahata
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Mika Mizoguchi
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Orimo
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Saori Toujima
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Izumi Sasaki
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuri Fukuda-Ohta
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroaki Hemmi
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naoyuki Iwahashi
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Tomoko Noguchi
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuhiko Ino
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
47
|
Wefers C, Duiveman-de Boer T, Yigit R, Zusterzeel PLM, van Altena AM, Massuger LFAG, De Vries IJM. Survival of Ovarian Cancer Patients Is Independent of the Presence of DC and T Cell Subsets in Ascites. Front Immunol 2019; 9:3156. [PMID: 30687337 PMCID: PMC6336918 DOI: 10.3389/fimmu.2018.03156] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/21/2018] [Indexed: 01/05/2023] Open
Abstract
Ascites is a prominent feature of ovarian cancer and could serve as liquid biopsy to assess the immune status of patients. Tumor-infiltrating T lymphocytes are correlated with improved survival in ovarian cancer. To investigate whether immune cells in ascites are associated with patient outcome, we analyzed the amount of dendritic cell (DC) and T cell subsets in ascites from ovarian cancer patients diagnosed with high-grade serous cancer (HGSC). Ascites was collected from 62 HGSC patients prior to chemotherapy. Clinicopathological, histological and follow-up data from patients were collected. Ascites-derived immune cells were isolated using density-gradient centrifugation. The presence of myeloid DCs (BDCA-1+, BDCA-3+, CD16+), pDCs (CD123+BDCA-2+), and T cells (CD4+, CD8+) was analyzed using flow cytometry. Complete cytoreduction, response to primary treatment and chemosensitivity were associated with improved patient outcome. In contrast, immune cells in ascites did not significantly correlate with patient survival. However, we observed a trend toward improved outcome for patients having low percentages of CD4+ T cells. Furthermore, we assessed the expression of co-stimulatory and co-inhibitory molecules on T cells and non-immune cells in 10 ascites samples. PD-1 was expressed by 30% of ascites-derived T cells and PD-L1 by 50% of non-immune cells. However, the percentage of DC and T cell subsets in ascites was not directly correlated to the survival of HGSC patients.
Collapse
Affiliation(s)
- Christina Wefers
- Department of Tumor Immunology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
- Department of Obstetrics and Gynecology, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Tjitske Duiveman-de Boer
- Department of Tumor Immunology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Refika Yigit
- Department of Tumor Immunology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
- Department of Obstetrics and Gynecology, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Petra L. M. Zusterzeel
- Department of Obstetrics and Gynecology, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Leon F. A. G. Massuger
- Department of Obstetrics and Gynecology, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - I. Jolanda M. De Vries
- Department of Tumor Immunology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
48
|
Roelofsen T, Wefers C, Gorris MAJ, Textor JC, Massuger LFAG, de Vries IJM, van Altena AM. Spontaneous Regression of Ovarian Carcinoma After Septic Peritonitis; A Unique Case Report. Front Oncol 2018; 8:562. [PMID: 30555799 PMCID: PMC6281979 DOI: 10.3389/fonc.2018.00562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/12/2018] [Indexed: 11/23/2022] Open
Abstract
Despite advances in therapy, ovarian cancer remains the most lethal gynecological malignancy and prognosis has not substantially improved over the past 3 decades. Immunotherapy is a promising new treatment option. However, the immunosuppressive cancer microenvironment must be overcome for immunotherapy to be successful. Here, we present a unique case of spontaneous regression of ovarian carcinoma after septic peritonitis. A 79-year-old woman was diagnosed with stage IIIc ovarian cancer. The omental cake biopsy was complicated by sepsis. Although the patient recovered, her physical condition did not allow further treatment for her ovarian cancer. After 6 months, spontaneous regression of the tumor was observed during surgery. Analysis of the immune infiltrate in the tissues showed a shift from a pro-tumorigenic to an anti-tumorigenic immune response after sepsis. Strong activation of the immune system during sepsis overruled the immunosuppressive tumor microenvironment and allowed for a potent anti-tumor immune response. More understanding of immunological responses in cases with cancer and septic peritonitis might be crucial to identify potential new targets for immunotherapy.
Collapse
Affiliation(s)
- Thijs Roelofsen
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Christina Wefers
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Johannes C. Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Leon F. A. G. Massuger
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
49
|
Gao Q, Xiang SD, Wilson K, Madondo M, Stephens AN, Plebanski M. Sperm Protein 17 Expression by Murine Epithelial Ovarian Cancer Cells and Its Impact on Tumor Progression. Cancers (Basel) 2018; 10:cancers10080276. [PMID: 30127274 PMCID: PMC6115966 DOI: 10.3390/cancers10080276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The cancer testis antigen sperm protein 17 (Sp17) is a promising antigenic target in epithelial ovarian cancer (EOC) vaccine development. However, its role in ovarian cancer is unclear. We isolated and expanded Sp17+ and Sp17− clones from the murine EOC cell line ID8, and compared their in-vitro cell growth characteristics and in-vivo tumorigenicity. We also examined the potential co-expression of molecules that may influence cancer cell survival and interaction with immune cells. These include stimulatory and immunosuppressive molecules, such as major histocompatibility class I molecules (MHC I), MHC II, cytotoxic T lymphocyte associated antigen-4 (CTLA-4), CD73, CD39, tumor necrosis factor receptor II (TNFRII), signal transducer and activator of transcription 3 (STAT3) and programmed death-ligand 1 (PD-L1). Whilst the presence of Sp17 was not correlated with the ID8 cell proliferation/growth capacity in vitro, it was critical to enable progressive tumor formation in vivo. Flow cytometry revealed that Sp17+ ID8 cells displayed higher expression of both STAT3 and PD-L1, whilst MHC II expression was lower. Moreover, Sp17high (PD-L1+MHCII−) cell populations showed significantly enhanced resistance to Paclitaxel-induced cell death in vitro compared to Sp17low (PD-L1−MHCII+) cells, which was associated in turn with increased STAT3 expression. Together, the data support Sp17 as a factor associated with in-vivo tumor progression and chemo-resistance, validating it as a suitable target for vaccine development.
Collapse
Affiliation(s)
- Qian Gao
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Sue D Xiang
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Kirsty Wilson
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Mutsa Madondo
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Andrew N Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
50
|
McCloskey CW, Rodriguez GM, Galpin KJC, Vanderhyden BC. Ovarian Cancer Immunotherapy: Preclinical Models and Emerging Therapeutics. Cancers (Basel) 2018; 10:cancers10080244. [PMID: 30049987 PMCID: PMC6115831 DOI: 10.3390/cancers10080244] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has emerged as one of the most promising approaches for ovarian cancer treatment. The tumor microenvironment (TME) is a key factor to consider when stimulating antitumoral responses as it consists largely of tumor promoting immunosuppressive cell types that attenuate antitumor immunity. As our understanding of the determinants of the TME composition grows, we have begun to appreciate the need to address both inter- and intra-tumor heterogeneity, mutation/neoantigen burden, immune landscape, and stromal cell contributions. The majority of immunotherapy studies in ovarian cancer have been performed using the well-characterized murine ID8 ovarian carcinoma model. Numerous other animal models of ovarian cancer exist, but have been underutilized because of their narrow initial characterizations in this context. Here, we describe animal models that may be untapped resources for the immunotherapy field because of their shared genomic alterations and histopathology with human ovarian cancer. We also shed light on the strengths and limitations of these models, and the knowledge gaps that need to be addressed to enhance the utility of preclinical models for testing novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Curtis W McCloskey
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| | - Galaxia M Rodriguez
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| | - Kristianne J C Galpin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|