1
|
Sun R, Li S, Ye W, Lu Y. Development of a prognostic model based on lysosome-related genes for ovarian cancer: insights into tumor microenvironment, mutation patterns, and personalized treatment strategies. Cancer Cell Int 2024; 24:419. [PMID: 39702158 DOI: 10.1186/s12935-024-03586-w] [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: 01/15/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is often associated with an unfavorable prognosis. Given the crucial involvement of lysosomes in tumor advancement, lysosome-related genes (LRGs) hold promise as potential therapeutic targets. METHODS To identify differentially expressed lysosome-related genes (DE-LRGs), we performed a matching analysis between differentially expressed genes (DEGs) in OC and the pool of LRGs. Genes with prognostic significance were analyzed using multiple regression analyses to construct a prognostic risk signature. The model's efficacy was validated through survival analysis in various cohorts. We further explored the model's correlation with clinical attributes, tumor microenvironment (TME), mutational patterns, and drug sensitivity. The quantitative real-time polymerase chain reaction (qRT-PCR) validated gene expression in OC cells. RESULTS A 10-gene prognostic risk signature was established. Survival analysis confirmed its predictive accuracy across cohorts. The signature served as an independent prognostic element for OC. The high-risk and low-risk groups demonstrated notable disparities in terms of immune infiltration patterns, mutational characteristics, and sensitivity to therapeutic agents. The qRT-PCR results corroborated and validated the findings obtained from the bioinformatic analyses. CONCLUSIONS We devised a 10-LRG prognostic model linked to TME, offering insights for tailored OC treatments.
Collapse
Affiliation(s)
- Ran Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Siyi Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Wanlu Ye
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Yanming Lu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
2
|
Kefas J, Flynn M. Unlocking the potential of immunotherapy in platinum-resistant ovarian cancer: rationale, challenges, and novel strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:39. [PMID: 39534871 PMCID: PMC11555186 DOI: 10.20517/cdr.2024.67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Ovarian cancer is a significant global health challenge, with cytoreductive surgery and platinum-based chemotherapy serving as established primary treatments. Unfortunately, most patients relapse and ultimately become platinum-resistant, at which point there are limited effective treatment options. Given the success of immunotherapy in inducing durable treatment responses in several other cancers, its potential in platinum-resistant ovarian cancer (PROC) is currently being investigated. However, in unselected advanced ovarian cancer populations, researchers have reported low response rates to immune checkpoint inhibition, and thus far, no validated biomarkers are predictive of response. Understanding the intricate interplay between platinum resistance, immune recognition, and the tumour microenvironment (TME) is crucial. In this review, we examine the research challenges encountered thus far, the biological rationale for immunotherapy, the underlying mechanisms of immune resistance, and new strategies to overcome resistance.
Collapse
Affiliation(s)
| | - Michael Flynn
- Medical Oncology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK
| |
Collapse
|
3
|
Monjé N, Dragomir MP, Sinn BV, Hoffmann I, Makhmut A, Simon T, Kunze CA, Ihlow J, Schmitt WD, Pohl J, Piwonski I, Marchenko S, Keunecke C, Calina TG, Tiso F, Kulbe H, Kreuzinger C, Cacsire Castillo-Tong D, Sehouli J, Braicu EI, Denkert C, Darb-Esfahani S, Kübler K, Capper D, Coscia F, Morkel M, Horst D, Sers C, Taube ET. AHRR and SFRP2 in primary versus recurrent high-grade serous ovarian carcinoma and their prognostic implication. Br J Cancer 2024; 130:1249-1260. [PMID: 38361045 PMCID: PMC11014847 DOI: 10.1038/s41416-023-02550-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND The aim of this study was to analyse transcriptomic differences between primary and recurrent high-grade serous ovarian carcinoma (HGSOC) to identify prognostic biomarkers. METHODS We analysed 19 paired primary and recurrent HGSOC samples using targeted RNA sequencing. We selected the best candidates using in silico survival and pathway analysis and validated the biomarkers using immunohistochemistry on a cohort of 44 paired samples, an additional cohort of 504 primary HGSOCs and explored their function. RESULTS We identified 233 differential expressed genes. Twenty-three showed a significant prognostic value for PFS and OS in silico. Seven markers (AHRR, COL5A2, FABP4, HMGCS2, ITGA5, SFRP2 and WNT9B) were chosen for validation at the protein level. AHRR expression was higher in primary tumours (p < 0.0001) and correlated with better patient survival (p < 0.05). Stromal SFRP2 expression was higher in recurrent samples (p = 0.009) and protein expression in primary tumours was associated with worse patient survival (p = 0.022). In multivariate analysis, tumour AHRR and SFRP2 remained independent prognostic markers. In vitro studies supported the anti-tumorigenic role of AHRR and the oncogenic function of SFRP2. CONCLUSIONS Our results underline the relevance of AHRR and SFRP2 proteins in aryl-hydrocarbon receptor and Wnt-signalling, respectively, and might lead to establishing them as biomarkers in HGSOC.
Collapse
Affiliation(s)
- Nanna Monjé
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Mihnea P Dragomir
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Bruno V Sinn
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Inga Hoffmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Anuar Makhmut
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Tincy Simon
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Catarina A Kunze
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Jana Ihlow
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Wolfgang D Schmitt
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Jonathan Pohl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Iris Piwonski
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Sofya Marchenko
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Carlotta Keunecke
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for Gynecology with the Center for Oncologic Surgery Charité Campus Virchow-Klinikum, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Francesca Tiso
- Center of Functional Genomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Hagen Kulbe
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for Gynecology with the Center for Oncologic Surgery Charité Campus Virchow-Klinikum, Charitéplatz 1, 10117, Berlin, Germany
| | - Caroline Kreuzinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jalid Sehouli
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for Gynecology with the Center for Oncologic Surgery Charité Campus Virchow-Klinikum, Charitéplatz 1, 10117, Berlin, Germany
| | - Elena I Braicu
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for Gynecology with the Center for Oncologic Surgery Charité Campus Virchow-Klinikum, Charitéplatz 1, 10117, Berlin, Germany
| | - Carsten Denkert
- Institute of Pathology, University Hospital Gießen and Marburg, Marburg, Germany
| | - Silvia Darb-Esfahani
- Institute of Pathology, Berlin-Spandau, Stadtrandstraße 555, 13589, Berlin, Germany
| | - Kirsten Kübler
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center of Functional Genomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School Teaching Hospital, Charlestown, MA, USA
| | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Markus Morkel
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - David Horst
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Christine Sers
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Eliane T Taube
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
4
|
Xu AM, Haro M, Walts AE, Hu Y, John J, Karlan BY, Merchant A, Orsulic S. Spatiotemporal architecture of immune cells and cancer-associated fibroblasts in high-grade serous ovarian carcinoma. SCIENCE ADVANCES 2024; 10:eadk8805. [PMID: 38630822 PMCID: PMC11023532 DOI: 10.1126/sciadv.adk8805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
High-grade serous ovarian carcinoma (HGSOC), the deadliest form of ovarian cancer, is typically diagnosed after it has metastasized and often relapses after standard-of-care platinum-based chemotherapy, likely due to advanced tumor stage, heterogeneity, and immune evasion and tumor-promoting signaling from the tumor microenvironment. To understand how spatial heterogeneity contributes to HGSOC progression and early relapse, we profiled an HGSOC tissue microarray of patient-matched longitudinal samples from 42 patients. We found spatial patterns associated with early relapse, including changes in T cell localization, malformed tertiary lymphoid structure (TLS)-like aggregates, and increased podoplanin-positive cancer-associated fibroblasts (CAFs). Using spatial features to compartmentalize the tissue, we found that plasma cells distribute in two different compartments associated with TLS-like aggregates and CAFs, and these distinct microenvironments may account for the conflicting reports about the role of plasma cells in HGSOC prognosis.
Collapse
Affiliation(s)
- Alexander M. Xu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marcela Haro
- Department of Obstetrics and Gynecology and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ye Hu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Joshi John
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Department of Medicine, Division of Geriatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Akil Merchant
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Yao Y, Wang D, Zheng L, Zhao J, Tan M. Advances in prognostic models for osteosarcoma risk. Heliyon 2024; 10:e28493. [PMID: 38586328 PMCID: PMC10998144 DOI: 10.1016/j.heliyon.2024.e28493] [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: 09/30/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
The risk prognosis model is a statistical model that uses a set of features to predict whether an individual will develop a specific disease or clinical outcome. It can be used in clinical practice to stratify disease severity and assess risk or prognosis. With the advancement of large-scale second-generation sequencing technology, along Prognosis models for osteosarcoma are increasingly being developed as large-scale second-generation sequencing technology advances and clinical and biological data becomes more abundant. This expansion greatly increases the number of prognostic models and candidate genes suitable for clinical use. This article will present the predictive effects and reliability of various prognosis models, serving as a reference for their evaluation and application.
Collapse
Affiliation(s)
- Yi Yao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Dapeng Wang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Manli Tan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
6
|
Smith P, Bradley T, Gavarró LM, Goranova T, Ennis DP, Mirza HB, De Silva D, Piskorz AM, Sauer CM, Al-Khalidi S, Funingana IG, Reinius MAV, Giannone G, Lewsley LA, Stobo J, McQueen J, Bryson G, Eldridge M, Macintyre G, Markowetz F, Brenton JD, McNeish IA. The copy number and mutational landscape of recurrent ovarian high-grade serous carcinoma. Nat Commun 2023; 14:4387. [PMID: 37474499 PMCID: PMC10359414 DOI: 10.1038/s41467-023-39867-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 06/28/2023] [Indexed: 07/22/2023] Open
Abstract
The drivers of recurrence and resistance in ovarian high grade serous carcinoma remain unclear. We investigate the acquisition of resistance by collecting tumour biopsies from a cohort of 276 women with relapsed ovarian high grade serous carcinoma in the BriTROC-1 study. Panel sequencing shows close concordance between diagnosis and relapse, with only four discordant cases. There is also very strong concordance in copy number between diagnosis and relapse, with no significant difference in purity, ploidy or focal somatic copy number alterations, even when stratified by platinum sensitivity or prior chemotherapy lines. Copy number signatures are strongly correlated with immune cell infiltration, whilst diagnosis samples from patients with primary platinum resistance have increased rates of CCNE1 and KRAS amplification and copy number signature 1 exposure. Our data show that the ovarian high grade serous carcinoma genome is remarkably stable between diagnosis and relapse and acquired chemotherapy resistance does not select for common copy number drivers.
Collapse
Affiliation(s)
- Philip Smith
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Thomas Bradley
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Teodora Goranova
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Darren P Ennis
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Hasan B Mirza
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Dilrini De Silva
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Anna M Piskorz
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Carolin M Sauer
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Ionut-Gabriel Funingana
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Marika A V Reinius
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Gaia Giannone
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Liz-Anne Lewsley
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jamie Stobo
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - John McQueen
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Gareth Bryson
- Department of Histopathology, Queen Elizabeth University Hospital, Glasgow, UK
| | - Matthew Eldridge
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Geoff Macintyre
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | | | - James D Brenton
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Iain A McNeish
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK.
| |
Collapse
|
7
|
Piki E, Dini A, Raivola J, Salokas K, Zhang K, Varjosalo M, Pellinen T, Välimäki K, Veskimäe KT, Staff S, Hautaniemi S, Murumägi A, Ungureanu D. ROR1-STAT3 signaling contributes to ovarian cancer intra-tumor heterogeneity. Cell Death Discov 2023; 9:222. [PMID: 37400436 DOI: 10.1038/s41420-023-01527-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
Wnt pathway dysregulation through genetic and non-genetic alterations occurs in multiple cancers, including ovarian cancer (OC). The aberrant expression of the non-canonical Wnt signaling receptor ROR1 is thought to contribute to OC progression and drug resistance. However, the key molecular events mediated by ROR1 that are involved in OC tumorigenesis are not fully understood. Here, we show that ROR1 expression is enhanced by neoadjuvant chemotherapy, and Wnt5a binding to ROR1 can induce oncogenic signaling via AKT/ERK/STAT3 activation in OC cells. Proteomics analysis of isogenic ROR1-knockdown OC cells identified STAT3 as a downstream effector of ROR1 signaling. Transcriptomics analysis of clinical samples (n = 125) revealed that ROR1 and STAT3 are expressed at higher levels in stromal cells than in epithelial cancer cells of OC tumors, and these findings were corroborated by multiplex immunohistochemistry (mIHC) analysis of an independent OC cohort (n = 11). Our results show that ROR1 and its downstream STAT3 are co-expressed in epithelial as well as stromal cells of OC tumors, including cancer-associated fibroblasts or CAFs. Our data provides the framework to expand the clinical utility of ROR1 as a therapeutic target to overcome OC progression.
Collapse
Affiliation(s)
- Emilia Piki
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014, Oulu, Finland
| | - Alice Dini
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014, Oulu, Finland
| | - Juuli Raivola
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Kari Salokas
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Kaiyang Zhang
- Research Program in Systems Oncology, Research Program Unit, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland, FIMM, Helsinki Institute of Life Science (HiLIFE) University of Helsinki, 00014, Helsinki, Finland
| | - Katja Välimäki
- Institute for Molecular Medicine Finland, FIMM, Helsinki Institute of Life Science (HiLIFE) University of Helsinki, 00014, Helsinki, Finland
| | - Kristina Tabor Veskimäe
- Department of Obstetrics and Gynecology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland
| | - Synnöve Staff
- Department of Obstetrics and Gynecology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland
| | - Sampsa Hautaniemi
- Research Program in Systems Oncology, Research Program Unit, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Astrid Murumägi
- Institute for Molecular Medicine Finland, FIMM, Helsinki Institute of Life Science (HiLIFE) University of Helsinki, 00014, Helsinki, Finland
| | - Daniela Ungureanu
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014, Oulu, Finland.
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
8
|
Pan L, She H, Wang K, Xia W, Tang H, Fan Y, Ye J. Characterization of the m 6A regulator-mediated methylation modification patterns in oral squamous cell carcinoma. Sci Rep 2023; 13:6617. [PMID: 37095314 PMCID: PMC10126108 DOI: 10.1038/s41598-023-33891-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/20/2023] [Indexed: 04/26/2023] Open
Abstract
N6-methyladenosine (m6A) is a form of posttranscriptional modification that plays important roles in cancer including oral squamous cell carcinoma (OSCC). Most studies to date have focused on a limited number of regulators and oncogenic pathways, thus failing to provide comprehensive insight into the dynamic effects of m6A modification. In addition, the role of m6A modification in shaping immune cell infiltration in OSCC has yet to be clarified. This study was designed to assess m6A modification dynamics in OSCC and to understand how such modifications influence clinical immunotherapeutic treatment outcomes. m6A modification patterns linked with 23 m6A regulators were analyzed in 437 OSCC patients from TCGA and GEO cohorts. These patterns were then quantified through m6A score based on algorithms derived from a principal component analysis (PCA) approach. The m6A modification patterns of OSCC samples were grouped into two clusters based on the m6A regulators expression, and immune cell infiltration was linked with the 5-year survival outcomes of patients in these clusters. 1575 genes associated with OSCC patient prognosis were identified and used to re-cluster these samples into two groups. Patients in clusters exhibiting higher levels of m6A regulator expression exhibited poorer overall survival (OS), whereas patients with high m6A scores survived for longer (p < 0.001). The overall mortality rates in the groups of patients with low and high m6A scores were 55% and 40%, respectively, and the m6A score distributions in clusters of patients grouped by m6A modification patterns and gene expression further supported the link between a high m6A score and better prognostic outcomes. Immunophenoscore (IPS) values for patients in different m6A score groups suggested that the use of PD-1-specific antibodies or CTLA-4 inhibitors alone or in combination would yield superior treatment outcomes in patients in the high-m6A score group relative to the low-m6A score group. m6A modification patterns are relevant to heterogeneity in OSCC. Detailed analyses of m6A modification patterns may thus offer novel insight regarding immune cell infiltration within the OSCC tumor microenvironment, guiding novel efforts to provide patients with more effective immunotherapeutic interventions.
Collapse
Affiliation(s)
- Lu Pan
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - He She
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Keyi Wang
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Wenhui Xia
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Haonan Tang
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China
| | - Yuan Fan
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China.
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China.
| | - Jinhai Ye
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, 136# Hanzhong Road, Nanjing, 210000, Jiangsu, China.
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Jiangsu, China.
| |
Collapse
|
9
|
Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes. Cancers (Basel) 2023; 15:cancers15041282. [PMID: 36831623 PMCID: PMC9953912 DOI: 10.3390/cancers15041282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Collapse
|
10
|
Gertych A, Walts AE, Cheng K, Liu M, John J, Lester J, Karlan BY, Orsulic S. Dynamic Changes in the Extracellular Matrix in Primary, Metastatic, and Recurrent Ovarian Cancers. Cells 2022; 11:3769. [PMID: 36497028 PMCID: PMC9736731 DOI: 10.3390/cells11233769] [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: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) and their extracellular matrix are active participants in cancer progression. While it is known that functionally different subpopulations of CAFs co-exist in ovarian cancer, it is unclear whether certain CAF subsets are enriched during metastatic progression and/or chemotherapy. Using computational image analyses of patient-matched primary high-grade serous ovarian carcinomas, synchronous pre-chemotherapy metastases, and metachronous post-chemotherapy metastases from 42 patients, we documented the dynamic spatiotemporal changes in the extracellular matrix, fibroblasts, epithelial cells, immune cells, and CAF subsets expressing different extracellular matrix components. Among the different CAF subsets, COL11A1+ CAFs were associated with linearized collagen fibers and exhibited the greatest enrichment in pre- and post-chemotherapy metastases compared to matched primary tumors. Although pre- and post-chemotherapy metastases were associated with increased CD8+ T cell infiltration, the infiltrate was not always evenly distributed between the stroma and cancer cells, leading to an increased frequency of the immune-excluded phenotype where the majority of CD8+ T cells are present in the tumor stroma but absent from the tumor parenchyma. Overall, most of the differences in the tumor microenvironment were observed between primary tumors and metastases, while fewer differences were observed between pre- and post-treatment metastases. These data suggest that the tumor microenvironment is largely determined by the primary vs. metastatic location of the tumor while chemotherapy does not have a significant impact on the host microenvironment.
Collapse
Affiliation(s)
- Arkadiusz Gertych
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Faculty of Biomedical Engineering, Silesian University of Technology, 44-100 Zabrze, Poland
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Keyi Cheng
- Department of Mathematics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Manyun Liu
- Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro, GA 30458, USA
| | - Joshi John
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| | - Jenny Lester
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Saha C, Bojdo J, Dunne NJ, Duary RK, Buckley N, McCarthy HO. Nucleic acid vaccination strategies for ovarian cancer. Front Bioeng Biotechnol 2022; 10:953887. [PMID: 36420446 PMCID: PMC9677957 DOI: 10.3389/fbioe.2022.953887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/26/2022] [Indexed: 09/19/2023] Open
Abstract
High grade serous carcinoma (HGSC) is one of the most lethal ovarian cancers that is characterised by asymptomatic tumour growth, insufficient knowledge of malignant cell origin and sub-optimal detection. HGSC has been recently shown to originate in the fallopian tube and not in the ovaries. Conventional treatments such as chemotherapy and surgery depend upon the stage of the disease and have resulted in higher rates of relapse. Hence, there is a need for alternative treatments. Differential antigen expression levels have been utilised for early detection of the cancer and could be employed in vaccination strategies using nucleic acids. In this review the different vaccination strategies in Ovarian cancer are discussed and reviewed. Nucleic acid vaccination strategies have been proven to produce a higher CD8+ CTL response alongside CD4+ T-cell response when compared to other vaccination strategies and thus provide a good arena for antitumour immune therapy. DNA and mRNA need to be delivered into the intracellular matrix. To overcome ineffective naked delivery of the nucleic acid cargo, a suitable delivery system is required. This review also considers the suitability of cell penetrating peptides as a tool for nucleic acid vaccine delivery in ovarian cancer.
Collapse
Affiliation(s)
- Chayanika Saha
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - James Bojdo
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - Nicholas J. Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Raj Kumar Duary
- Department of Food Engineering and Technology, Tezpur University, Tezpur, India
| | - Niamh Buckley
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
- School of Chemical Sciences, Dublin City University, Dublin, Ireland
| |
Collapse
|
12
|
Hudry D, Le Guellec S, Meignan S, Bécourt S, Pasquesoone C, El Hajj H, Martínez-Gómez C, Leblanc É, Narducci F, Ladoire S. Tumor-Infiltrating Lymphocytes (TILs) in Epithelial Ovarian Cancer: Heterogeneity, Prognostic Impact, and Relationship with Immune Checkpoints. Cancers (Basel) 2022; 14:5332. [PMID: 36358750 PMCID: PMC9656626 DOI: 10.3390/cancers14215332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Epithelial ovarian cancers (EOC) are often diagnosed at an advanced stage with carcinomatosis and a poor prognosis. First-line treatment is based on a chemotherapy regimen combining a platinum-based drug and a taxane-based drug along with surgery. More than half of the patients will have concern about a recurrence. To improve the outcomes, new therapeutics are needed, and diverse strategies, such as immunotherapy, are currently being tested in EOC. To better understand the global immune contexture in EOC, several studies have been performed to decipher the landscape of tumor-infiltrating lymphocytes (TILs). CD8+ TILs are usually considered effective antitumor immune effectors that immune checkpoint inhibitors can potentially activate to reject tumor cells. To synthesize the knowledge of TILs in EOC, we conducted a review of studies published in MEDLINE or EMBASE in the last 10 years according to the PRISMA guidelines. The description and role of TILs in EOC prognosis are reviewed from the published data. The links between TILs, DNA repair deficiency, and ICs have been studied. Finally, this review describes the role of TILs in future immunotherapy for EOC.
Collapse
Affiliation(s)
- Delphine Hudry
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Solenn Le Guellec
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Samuel Meignan
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille University, F-59000 Lille, France
| | - Stéphanie Bécourt
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Camille Pasquesoone
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Houssein El Hajj
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | | | - Éric Leblanc
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Fabrice Narducci
- Inserm, U1192–Protéomique Réponse Inflammatoire Spectrométrie de Masse–PRISM, Lille University, F-59000 Lille, France
- Department of Gynecologic Oncology, Oscar Lambret Center, F-59000 Lille, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, F-21000 Dijon, France
- INSERM, CRI-866 Faculty of Medicine, F-21000 Dijon, France
| |
Collapse
|
13
|
Geng R, Chen T, Zhong Z, Ni S, Bai J, Liu J. The m6A-Related Long Noncoding RNA Signature Predicts Prognosis and Indicates Tumor Immune Infiltration in Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14164056. [PMID: 36011053 PMCID: PMC9406778 DOI: 10.3390/cancers14164056] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Background: OV is the most lethal gynecological malignancy. M6A and lncRNAs have a great impact on OV development and patient immunotherapy response. In this paper, we decided to establish a reliable signature of mRLs. Method: The lncRNAs associated with m6A in OV were analyzed and obtained by co-expression analysis of the TCGA-OV database. Univariate, LASSO and multivariate Cox regression analyses were employed to establish the model of mRLs. K-M analysis, PCA, GSEA and nomogram based on the TCGA-OV and GEO database were conducted to prove the predictive value and independence of the model. The underlying relationship between the model and TME and cancer stemness properties were further investigated through immune feature comparison, consensus clustering analysis and pan-cancer analysis. Results: A prognostic signature comprising four mRLs, WAC-AS1, LINC00997, DNM3OS and FOXN3-AS1, was constructed and verified for OV according to the TCGA and GEO database. The expressions of the four mRLs were confirmed by qRT-PCR in clinical samples. Applying this signature, one can identify patients more effectively. The samples were divided into two clusters, and the clusters had different overall survival rates, clinical features and tumor microenvironments. Finally, pan-cancer analysis further demonstrated that the four mRLs were significantly related to immune infiltration, TME and cancer stemness properties in various cancer types. Conclusions: This study provided an accurate prognostic signature for patients with OV and elucidated the potential mechanism of the mRLs in immune modulation and treatment response, giving new insights into identifying new therapeutic targets.
Collapse
Affiliation(s)
- Rui Geng
- Department of Biostatistics, School of Public Heath, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
| | - Tian Chen
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zihang Zhong
- Department of Biostatistics, School of Public Heath, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
| | - Senmiao Ni
- Department of Biostatistics, School of Public Heath, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
| | - Jianling Bai
- Department of Biostatistics, School of Public Heath, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
- Correspondence: (J.B.); (J.L.)
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Correspondence: (J.B.); (J.L.)
| |
Collapse
|
14
|
du Manoir S, Delpech H, Orsetti B, Jacot W, Pirot N, Noel J, Colombo PE, Sardet C, Theillet C. In high grade ovarian carcinoma, platinum-sensitive tumor recurrence and acquired-resistance derive from quiescent residual cancer cells that overexpress CRYAB, CEACAM6 and SOX2. J Pathol 2022; 257:367-378. [PMID: 35302657 DOI: 10.1002/path.5896] [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: 11/09/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/08/2022]
Abstract
Most High-Grade Ovarian Carcinomas (HGOCs) are sensitive to carboplatin (CBP)-based chemotherapy but frequently recur within 24 months. Recurrent tumors remain CBP-sensitive and acquire resistance only after several treatment rounds. Recurrences arise from a small number of residual tumor cells hardly amenable to investigation in patients. We developed Patient-Derived Xenografts (PDXs) that allow the study of these different stages of CBP-sensitive recurrence and acquisition of resistance. We generated PDX models from CBP-sensitive and intrinsically resistant HGOC. PDXs were CBP- or mock-treated and tumors were sampled, after treatment and at recurrence. We also isolated models with acquired-resistance from CBP-sensitive PDXs. All tumors were characterized at the histological and transcriptome levels. PDX models reproduced treatment response seen in the patients. CBP-sensitive residual tumors contained non-proliferating tumor cells clusters embedded in a fibrotic mesh. In non-treated PDX tumors and treated CBP-resistant tumors fibrotic tissue was not prevalent. Residual tumors had marked differences in gene expression when compared to naïve and recurrent tumors, indicating downregulation of cell cycle and proliferation and upregulation of interferon response and epithelial-mesenchymal transition. This gene expression pattern resembled that described in embryonal diapause and 'drug-tolerant persister' states. Residual and acquired-resistance tumors share the overexpression of three genes: CEACAM6, CRYAB, and SOX2.Immunostaining analysis showed strong CEACAM6, CRYAB, and SOX2 protein expression in CBP-sensitive residual and acquired resistance PDX, thus, confirming RNA profiling results. In HGOC PDX, CBP-sensitive recurrences arise from a small population of quiescent, drug-tolerant, residual cells embedded in a fibrotic mesh. These cells overexpress CEACAM6, CRYAB and SOX2, whose overexpression is also associated with acquired resistance and poor patient prognosis. CEACAM6, CRYAB and SOX2 may, thus, serve as a biomarker to predict recurrence and emergence of resistant disease in CBP-treated HGOC patients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Hélène Delpech
- IRCM U1194, INSERM, University of Montpellier, Montpellier, France
| | - Béatrice Orsetti
- IRCM U1194, INSERM, University of Montpellier, Montpellier, France
| | - William Jacot
- IRCM U1194, INSERM, University of Montpellier, Montpellier, France
| | - Nelly Pirot
- IRCM U1194, INSERM, University of Montpellier, Montpellier, France
| | - Jean Noel
- BCM, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Claude Sardet
- IRCM U1194, INSERM, Univ Montpellier, ICM, CNRS, Montpellier, France
| | - Charles Theillet
- IRCM U1194, INSERM, University of Montpellier, Montpellier, France
| |
Collapse
|
15
|
Huo X, Sun H, Liu S, Liang B, Bai H, Wang S, Li S. Identification of a Prognostic Signature for Ovarian Cancer Based on the Microenvironment Genes. Front Genet 2021; 12:680413. [PMID: 34054929 PMCID: PMC8155613 DOI: 10.3389/fgene.2021.680413] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/15/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Ovarian cancer is highly malignant and has a poor prognosis in the advanced stage. Studies have shown that infiltration of tumor microenvironment cells, immune cells and stromal cells has an important impact on the prognosis of cancers. However, the relationship between tumor microenvironment genes and the prognosis of ovarian cancer has not been studied. Methods: Gene expression profiles and SNP data of ovarian cancer were downloaded from the TCGA database. Cluster analysis, WGCNA analysis and univariate survival analysis were used to identify immune microenvironment genes as prognostic signatures for predicting the survival of ovarian cancer patients. External data were used to evaluate the signature. Moreover, the top five significantly correlated genes were evaluated by immunohistochemical staining of ovarian cancer tissues. Results: We systematically analyzed the relationship between ovarian cancer and immune metagenes. Immune metagenes expression were associated with prognosis. In total, we identified 10 genes related to both immunity and prognosis in ovarian cancer according to the expression of immune metagenes. These data reveal that high expression of ETV7 (OS, HR = 1.540, 95% CI 1.023–2.390, p = 0.041), GBP4 (OS, HR = 1.834, 95% CI 1.242–3.055, p = 0.004), CXCL9 (OS, HR = 1.613, 95% CI 1.080 –2.471, p = 0.021), CD3E (OS, HR = 1.590, 95% CI 1.049 –2.459, p = 0.031), and TAP1 (OS, HR = 1.766, 95% CI 1.163 –2.723, p = 0.009) are associated with better prognosis in patients with ovarian cancer. Conclusion: Our study identified 10 immune microenvironment genes related to the prognosis of ovarian cancer. The list of tumor microenvironment-related genes provides new insights into the underlying biological mechanisms driving the tumorigenesis of ovarian cancer.
Collapse
Affiliation(s)
- Xiao Huo
- Peking University Third Hospital Institute of Medical Innovation and Research, Beijing, China
| | - Hengzi Sun
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuangwu Liu
- School of Medicine, ShanDong University, Jinan, China
| | - Bing Liang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuhong Li
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Barger CJ, Chee L, Albahrani M, Munoz-Trujillo C, Boghean L, Branick C, Odunsi K, Drapkin R, Zou L, Karpf AR. Co-regulation and function of FOXM1/ RHNO1 bidirectional genes in cancer. eLife 2021; 10:e55070. [PMID: 33890574 PMCID: PMC8104967 DOI: 10.7554/elife.55070] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
The FOXM1 transcription factor is an oncoprotein and a top biomarker of poor prognosis in human cancer. Overexpression and activation of FOXM1 is frequent in high-grade serous carcinoma (HGSC), the most common and lethal form of human ovarian cancer, and is linked to copy number gains at chromosome 12p13.33. We show that FOXM1 is co-amplified and co-expressed with RHNO1, a gene involved in the ATR-Chk1 signaling pathway that functions in the DNA replication stress response. We demonstrate that FOXM1 and RHNO1 are head-to-head (i.e., bidirectional) genes (BDG) regulated by a bidirectional promoter (BDP) (named F/R-BDP). FOXM1 and RHNO1 each promote oncogenic phenotypes in HGSC cells, including clonogenic growth, DNA homologous recombination repair, and poly-ADP ribosylase inhibitor resistance. FOXM1 and RHNO1 are one of the first examples of oncogenic BDG, and therapeutic targeting of FOXM1/RHNO1 BDG is a potential therapeutic approach for ovarian and other cancers.
Collapse
MESH Headings
- Ataxia Telangiectasia Mutated Proteins/genetics
- Ataxia Telangiectasia Mutated Proteins/metabolism
- Carboplatin/pharmacology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Checkpoint Kinase 1/genetics
- Checkpoint Kinase 1/metabolism
- Databases, Genetic
- Drug Resistance, Neoplasm
- Female
- Forkhead Box Protein M1/genetics
- Forkhead Box Protein M1/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasms, Cystic, Mucinous, and Serous/drug therapy
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Promoter Regions, Genetic
- Recombinational DNA Repair
- Signal Transduction
Collapse
Affiliation(s)
- Carter J Barger
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Linda Chee
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Mustafa Albahrani
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Catalina Munoz-Trujillo
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Lidia Boghean
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Connor Branick
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Kunle Odunsi
- Departments of Gynecologic Oncology, Immunology, and Center for Immunotherapy, Roswell Park Comprehensive Cancer CenterBuffaloUnited States
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Harvard Medical SchoolCharlestownUnited States
| | - Adam R Karpf
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| |
Collapse
|
17
|
Phenotypic Characterization by Mass Cytometry of the Microenvironment in Ovarian Cancer and Impact of Tumor Dissociation Methods. Cancers (Basel) 2021; 13:cancers13040755. [PMID: 33670410 PMCID: PMC7918057 DOI: 10.3390/cancers13040755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC) is the deadliest gynecological malignancy. Despite increasing research on HGSOC, biomarkers for individualized selection of therapy are scarce. In this study, we develop a multiparametric mass cytometry antibody panel to identify differences in the cellular composition of the microenvironment of tumor tissues dissociated to single-cell suspensions. We also investigate how dissociation methods impact results. Application of our antibody panel to HGSOC tissues showed its ability to identify established main cell subsets and subpopulations of these cells. Comparisons between dissociation methods revealed differences in cell fractions for one immune, two stromal, and three tumor cell subpopulations, while functional marker expression was not affected by the dissociation method. The interpatient disparities identified in the tumor microenvironment were more significant than those identified between differently dissociated tissues from one patient, indicating that the panel facilitates the mapping of individual tumor microenvironments in HGSOC patients. Abstract Improved molecular dissection of the tumor microenvironment (TME) holds promise for treating high-grade serous ovarian cancer (HGSOC), a gynecological malignancy with high mortality. Reliable disease-related biomarkers are scarce, but single-cell mapping of the TME could identify patient-specific prognostic differences. To avoid technical variation effects, however, tissue dissociation effects on single cells must be considered. We present a novel Cytometry by Time-of-Flight antibody panel for single-cell suspensions to identify individual TME profiles of HGSOC patients and evaluate the effects of dissociation methods on results. The panel was developed utilizing cell lines, healthy donor blood, and stem cells and was applied to HGSOC tissues dissociated by six methods. Data were analyzed using Cytobank and X-shift and illustrated by t-distributed stochastic neighbor embedding plots, heatmaps, and stacked bar and error plots. The panel distinguishes the main cellular subsets and subpopulations, enabling characterization of individual TME profiles. The dissociation method affected some immune (n = 1), stromal (n = 2), and tumor (n = 3) subsets, while functional marker expressions remained comparable. In conclusion, the panel can identify subsets of the HGSOC TME and can be used for in-depth profiling. This panel represents a promising profiling tool for HGSOC when tissue handling is considered.
Collapse
|
18
|
Cremer SE, Catalfamo JL, Goggs R, Seemann SE, Kristensen AT, Szklanna PB, Maguire PB, Brooks MB. The canine activated platelet secretome (CAPS): A translational model of thrombin-evoked platelet activation response. Res Pract Thromb Haemost 2021; 5:55-68. [PMID: 33537530 PMCID: PMC7845059 DOI: 10.1002/rth2.12450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/20/2020] [Accepted: 10/07/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Domestic dogs represent a translational animal model to study naturally occurring human disease. Proteomics has emerged as a promising tool for characterizing human platelet pathophysiology; thus a detailed characterization of the core canine activated platelet secretome (CAPS) will enhance utilization of the canine model. The objectives of this study were development of a robust, high throughput, label-free approach for proteomic identification and quantification of the canine platelet (i) thrombin releasate proteins, and (ii) the protein subgroup that constitutes CAPS. METHODS Platelets were isolated from 10 healthy dogs and stimulated with 50 nmol/L of γ-thrombin or saline. Proteins were in-solution trypsin-digested and analyzed by nano-liquid chromatography-tandem spectrometry. Core releasate proteins were defined as those present in 10 of 10 dogs, and CAPS defined as releasate proteins with a significantly higher abundance in stimulated versus saline controls (corrected P < .05). RESULTS A total of 2865 proteins were identified; 1126 releasate proteins were present in all dogs, 650 were defined as CAPS. Among the differences from human platelets were a canine lack of platelet factor 4 and vascular endothelial growth factor C, and a 10- to 20-fold lower concentration of proteins such as haptoglobin, alpha-2 macroglobulin, von Willebrand factor, and amyloid-beta A4. Twenty-eight CAPS proteins, including cytokines, adhesion molecules, granule proteins, and calcium regulatory proteins have not previously been attributed to human platelets. CONCLUSIONS CAPS proteins represent a robust characterization of a large animal platelet secretome and a novel tool to model platelet physiology, pathophysiology, and to identify translational biomarkers of platelet-mediated disease.
Collapse
Affiliation(s)
- Signe E. Cremer
- Department of Veterinary Clinical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Population Medicine and Diagnostic SciencesCornell UniversityIthacaNYUSA
| | - James L. Catalfamo
- Department of Population Medicine and Diagnostic SciencesCornell UniversityIthacaNYUSA
| | - Robert Goggs
- Department of Clinical SciencesCornell UniversityIthacaNYUSA
| | - Stefan E. Seemann
- Department of Veterinary and Animal SciencesCenter for Non‐coding RNA in Technology and HealthUniversity of CopenhagenCopenhagenDenmark
| | | | - Paulina B. Szklanna
- School of Biomolecular and Biomedical ScienceUniversity College DublinDublinIreland
| | - Patricia B. Maguire
- School of Biomolecular and Biomedical ScienceUniversity College DublinDublinIreland
| | - Marjory B. Brooks
- Department of Population Medicine and Diagnostic SciencesCornell UniversityIthacaNYUSA
| |
Collapse
|
19
|
Changes in the Tumor Immune Microenvironment during Disease Progression in Patients with Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12123828. [PMID: 33352957 PMCID: PMC7767114 DOI: 10.3390/cancers12123828] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Immunotherapy has been a successful treatment for many cancers. However, no immunotherapy treatment has been approved for ovarian cancer due to low efficacy in this patient group. This study investigated the cellular and molecular changes from primary ovarian tumors, at the time of diagnosis, to recurrence, where the disease returns after surgery and chemotherapies. Here we examined the immune contexture to better understand subdued responses to immunotherapy and identify additional, potentially complimentary, therapeutic targets. Indications of the development of adaptive immune resistance during disease progression were observed, with increases in immune and stromal cell infiltration accompanied by the expression of immune suppressive markers. We observed high gene expression of the immune checkpoint genes LAG3 and HAVCR2 (TIM3) in most tumors and increased expression of the immune checkpoint genes TIGIT and CTLA4 in recurrent tumors, compared to the primaries. These markers may be potential candidates for immunotherapy targeting in ovarian cancer. Abstract Anti-PD1/PDL1 therapy has proven efficacious against many cancers but only reached modest objective response rates against recurrent ovarian cancer. A deeper understanding of the tumor microenvironment (TME) may reveal other immunosuppressive mechanisms that warrant investigation as immunotherapeutic targets for this challenging disease. Matched primary and recurrent tumors from patients with high-grade serous ovarian carcinoma (HGSC) were analyzed by multicolor immunohistochemistry/immunofluorescence for the presence of T cells, B cells, macrophages, and for the expression of immunosuppressive and HLA molecules. Cancer- and immune-related gene expression was assessed by NanoString analysis. Recurrent tumors showed increased infiltration by immune cells, displayed higher expression of PDL1, IDO, and HLA molecules, and contained more stromal tissue. NanoString analysis demonstrated increased expression of gene signatures related to chemokines and T cell functions in recurrent tumors. The ovarian tumors showed high gene expression of LAG3 and HAVCR2 (TIM3) and enhanced levels of TIGIT and CTLA4 in recurrent tumors compared to primary tumors. The majority of HGSC developed into a more inflamed phenotype during progression from primary to recurrent disease, including indications of adaptive immune resistance. This suggests that recurrent tumors may be particularly sensitive to inhibition of adaptive immune resistance mechanisms.
Collapse
|
20
|
Heredia-Soto V, López-Guerrero J, Redondo A, Mendiola M. The hallmarks of ovarian cancer: Focus on angiogenesis and micro-environment and new models for their characterisation. EJC Suppl 2020; 15:49-55. [PMID: 33240442 PMCID: PMC7573462 DOI: 10.1016/j.ejcsup.2019.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/28/2019] [Accepted: 11/16/2019] [Indexed: 12/12/2022] Open
Abstract
Cancers develop by sustained growth, migration and invasion properties of tumour cells, supported by complex interactions with stromal cells within the tumour micro-environment. This review is focused on the latest discoveries regarding the highlighted role of angiogenesis and tumour micro-environment in ovarian cancer. This cancer milieu encompasses non-cancerous cells present in the tumour or nearby, including vessel-forming cells, fibroblasts and immune cells amongst others that work in a cooperative way with cancer cells, impacting tumour behaviour. Angiogenesis, migration and invasion, and more recently immune evasion, are cancer hallmarks clearly dependent on these supporting cells. Moreover, these stromal cells are more genetically stable than tumour cells and thus represent an attractive therapeutic target. A better understanding of the stromal cells function, and their complex interplay with cancer cells, will open additional areas to target, as the tumour-host interface.
Collapse
Affiliation(s)
- V. Heredia-Soto
- Translational Oncology Research Laboratory, La Paz University Hospital Biomedical Research Institute, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Monforte de Lemos 5, Madrid, 28029, Spain
| | - J.A. López-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, Carrer Del Professor Beltrán Báguena, 8, 46009, Valencia, Spain
| | - A. Redondo
- Translational Oncology Research Laboratory, La Paz University Hospital Biomedical Research Institute, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Medical Oncology Department, La Paz University Hospital, Paseo de La Castellana 261, 28046, Madrid, Spain
- Faculty of Medicine, Cátedra UAM-Amgen, Universidad Autónoma de Madrid, Madrid, Spain
| | - M. Mendiola
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Monforte de Lemos 5, Madrid, 28029, Spain
- Molecular Pathology and Therapeutic Targets Research Laboratory, La Paz University Hospital Biomedical Research Institute, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Molecular Pathology Diagnostic Section, Medical and Molecular Medicine Institute, INGEMM, Paseo de La Castellana 261, 28046, Madrid, Spain
| |
Collapse
|
21
|
He X, Lei S, Zhang Q, Ma L, Li N, Wang J. Deregulation of cell adhesion molecules is associated with progression and poor outcomes in endometrial cancer: Analysis of The Cancer Genome Atlas data. Oncol Lett 2020; 19:1906-1914. [PMID: 32194686 PMCID: PMC7039152 DOI: 10.3892/ol.2020.11295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/15/2019] [Indexed: 01/14/2023] Open
Abstract
Cell adhesion molecules (CAMs) determine the behavior of cancer cells during metastasis. Although some CAMs are dysregulated in certain types of cancer and are associated with cancer progression, to the best of our knowledge, a comprehensive study of CAMs has not been undertaken, particularly in endometrial cancer (EC). In the present study the expression of 225 CAMs in EC patients with various clinicopathological phenotypes were evaluated by statistical analysis using publicly available data from The Cancer Genome Atlas database. The Kaplan-Meier method, and univariate and multivariate Cox proportional hazards regression models were used for survival analyses. Among the differentially expressed CAMs that were associated with aggressive clinicopathological phenotypes, 10 CAM genes were independent prognostic factors compared with other clinicopathological prognostic factors, including stage, grade, age, lymph node status, peritoneal cytology and histological subtype. A total of six genes (L1 cell adhesion molecule, mucin 15, cell surface associated, cell adhesion associated, oncogene regulated, immunoglobulin superfamily member 9B, protocadherin 9 and protocadherin β1) were selected for integrative analysis. The six-gene signature was demonstrated to be an independent prognostic factor and could effectively stratify patients with different risks. Patients with more high-expression CAMs had a higher risk of poor overall survival (OS) rate. The mortality risk for patients with elevation of >4 CAMs was 11 times of that in those without elevation of these 6 CAMs. Similar results were obtained when relapse-free survival (RFS) time was used during the analysis. Prognostic reliability of the six-gene model was validated using data of an independent cohort from the International Cancer Genome Consortium. In conclusion, a combination of CAM alterations contributed to progression and aggressiveness of EC. The six-gene signature was effective for predicting worse OS and RFS in patients with EC and could be complementary to the present clinical prognostic criteria.
Collapse
Affiliation(s)
- Xiangjun He
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Shu Lei
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China.,Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Qi Zhang
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Liping Ma
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Na Li
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
22
|
The Many Microenvironments of Ovarian Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1296:199-213. [PMID: 34185294 DOI: 10.1007/978-3-030-59038-3_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most common and deadly subtype of ovarian cancer as it is commonly diagnosed after substantial metastasis has already occurred. The past two decades have been an active era in HGSOC research, with new information on the origin and genomic signature of the tumor cell. Additionally, studies have begun to characterize changes in the HGSOC microenvironment and examine the impact of these changes on tumor progression and response to therapies. While this knowledge may provide valuable insight into better prognosis and treatments for HGSOCs, its collection, synthesis, and application are complicated by the number of unique microenvironments in the disease-the initiating site (fallopian tube), first metastasis (ovary), distal metastases (peritoneum), and recurrent/platinum-resistant setting. Here, we review the state of our understanding of these diverse sites and highlight remaining questions.
Collapse
|
23
|
Kaipio K, Chen P, Roering P, Huhtinen K, Mikkonen P, Östling P, Lehtinen L, Mansuri N, Korpela T, Potdar S, Hynninen J, Auranen A, Grénman S, Wennerberg K, Hautaniemi S, Carpén O. ALDH1A1-related stemness in high-grade serous ovarian cancer is a negative prognostic indicator but potentially targetable by EGFR/mTOR-PI3K/aurora kinase inhibitors. J Pathol 2019; 250:159-169. [PMID: 31595974 DOI: 10.1002/path.5356] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 09/05/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022]
Abstract
Poor chemotherapy response remains a major treatment challenge for high-grade serous ovarian cancer (HGSC). Cancer stem cells are the major contributors to relapse and treatment failure as they can survive conventional therapy. Our objectives were to characterise stemness features in primary patient-derived cell lines, correlate stemness markers with clinical outcome and test the response of our cells to both conventional and exploratory drugs. Tissue and ascites samples, treatment-naive and/or after neoadjuvant chemotherapy, were prospectively collected. Primary cancer cells, cultured under conditions favouring either adherent or spheroid growth, were tested for stemness markers; the same markers were analysed in tissue and correlated with chemotherapy response and survival. Drug sensitivity and resistance testing was performed with 306 oncology compounds. Spheroid growth condition HGSC cells showed increased stemness marker expression (including aldehyde dehydrogenase isoform I; ALDH1A1) as compared with adherent growth condition cells, and increased resistance to platinum and taxane. A set of eight stemness markers separated treatment-naive tumours into two clusters and identified a distinct subgroup of HGSC with enriched stemness features. Expression of ALDH1A1, but not most other stemness markers, was increased after neoadjuvant chemotherapy and its expression in treatment-naive tumours correlated with chemoresistance and reduced survival. In drug sensitivity and resistance testing, five compounds, including two PI3K-mTOR inhibitors, demonstrated significant activity in both cell culture conditions. Thirteen compounds, including EGFR, PI3K-mTOR and aurora kinase inhibitors, were more toxic to spheroid cells than adherent cells. Our results identify stemness markers in HGSC that are associated with a decreased response to conventional chemotherapy and reduced survival if expressed by treatment-naive tumours. EGFR, mTOR-PI3K and aurora kinase inhibitors are candidates for targeting this cell population. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Katja Kaipio
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ping Chen
- Integrated Cardio Metabolic Centre (ICMC), Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Pia Roering
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kaisa Huhtinen
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Piia Mikkonen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Päivi Östling
- Science for Life Laboratory Department of Oncology & Pathology, Karolinska Institutet, Huddinge, Sweden.,Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Laura Lehtinen
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Naziha Mansuri
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Taina Korpela
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland, High Throughput Biomedicine Unit (HTB), University of Helsinki, Helsinki, Finland
| | - Johanna Hynninen
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Annika Auranen
- Department of Obstetrics and Gynaecology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Seija Grénman
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, High Throughput Biomedicine Unit (HTB), University of Helsinki, Helsinki, Finland.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Sampsa Hautaniemi
- Research Programs Unit, Genome-Scale Biology and Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Carpén
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,Research Programs Unit, Genome-Scale Biology and Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Kondratova M, Czerwinska U, Sompairac N, Amigorena SD, Soumelis V, Barillot E, Zinovyev A, Kuperstein I. A multiscale signalling network map of innate immune response in cancer reveals cell heterogeneity signatures. Nat Commun 2019; 10:4808. [PMID: 31641119 PMCID: PMC6805895 DOI: 10.1038/s41467-019-12270-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/02/2019] [Indexed: 12/14/2022] Open
Abstract
The lack of integrated resources depicting the complexity of the innate immune response in cancer represents a bottleneck for high-throughput data interpretation. To address this challenge, we perform a systematic manual literature mining of molecular mechanisms governing the innate immune response in cancer and represent it as a signalling network map. The cell-type specific signalling maps of macrophages, dendritic cells, myeloid-derived suppressor cells and natural killers are constructed and integrated into a comprehensive meta map of the innate immune response in cancer. The meta-map contains 1466 chemical species as nodes connected by 1084 biochemical reactions, and it is supported by information from 820 articles. The resource helps to interpret single cell RNA-Seq data from macrophages and natural killer cells in metastatic melanoma that reveal different anti- or pro-tumor sub-populations within each cell type. Here, we report a new open source analytic platform that supports data visualisation and interpretation of tumour microenvironment activity in cancer. The complexity of the innate immune response to cancer makes interpretation of large data sets challenging. Here, the authors provide an integrated multi-scale map of signalling networks representing the different immune cells and their interactions and show its utility for data interpretation.
Collapse
Affiliation(s)
- Maria Kondratova
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France
| | - Urszula Czerwinska
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France.,Université Paris Descartes, Centre de Recherches Interdisciplinaires, Paris, France
| | - Nicolas Sompairac
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France.,Université Paris Descartes, Centre de Recherches Interdisciplinaires, Paris, France
| | | | - Vassili Soumelis
- Institut Curie, PSL Research University, Inserm, U932, 75005, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France
| | - Inna Kuperstein
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France.
| |
Collapse
|
25
|
Hartl CA, Bertschi A, Puerto RB, Andresen C, Cheney EM, Mittendorf EA, Guerriero JL, Goldberg MS. Combination therapy targeting both innate and adaptive immunity improves survival in a pre-clinical model of ovarian cancer. J Immunother Cancer 2019; 7:199. [PMID: 31362778 PMCID: PMC6668091 DOI: 10.1186/s40425-019-0654-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Despite major advancements in immunotherapy among a number of solid tumors, response rates among ovarian cancer patients remain modest. Standard treatment for ovarian cancer is still surgery followed by taxane- and platinum-based chemotherapy. Thus, there is an urgent need to develop novel treatment options for clinical translation. Methods Our approach was to analyze the effects of standard chemotherapy in the tumor microenvironment of mice harboring orthotopic, syngeneic ID8-Vegf-Defb29 ovarian tumors in order to mechanistically determine a complementary immunotherapy combination. Specifically, we interrogated the molecular and cellular consequences of chemotherapy by analyzing gene expression and flow cytometry data. Results These data show that there is an immunosuppressive shift in the myeloid compartment, with increased expression of IL-10 and ARG1, but no activation of CD3+ T cells shortly after chemotherapy treatment. We therefore selected immunotherapies that target both the innate and adaptive arms of the immune system. Survival studies revealed that standard chemotherapy was complemented most effectively by a combination of anti-IL-10, 2′3’-cGAMP, and anti-PD-L1. Immunotherapy dramatically decreased the immunosuppressive myeloid population while chemotherapy effectively activated dendritic cells. Together, combination treatment increased the number of activated T and dendritic cells as well as expression of cytotoxic factors. It was also determined that the immunotherapy had to be administered concurrently with the chemotherapy to reverse the acute immunosuppression caused by chemotherapy. Mechanistic studies revealed that antitumor immunity in this context was driven by CD4+ T cells, which acquired a highly activated phenotype. Our data suggest that these CD4+ T cells can kill cancer cells directly via granzyme B-mediated cytotoxicity. Finally, we showed that this combination therapy is also effective at delaying tumor growth substantially in an aggressive model of lung cancer, which is also treated clinically with taxane- and platinum-based chemotherapy. Conclusions This work highlights the importance of CD4+ T cells in tumor immunology. Furthermore, the data support the initiation of clinical trials in ovarian cancer that target both innate and adaptive immunity, with a focus on optimizing dosing schedules. Electronic supplementary material The online version of this article (10.1186/s40425-019-0654-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christina A Hartl
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Adrian Bertschi
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Regina Bou Puerto
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Carolin Andresen
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Emily M Cheney
- Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Elizabeth A Mittendorf
- Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02215, USA.,Breast Oncology Program, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Jennifer L Guerriero
- Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Michael S Goldberg
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
26
|
Dzobo K, Adotey S, Thomford NE, Dzobo W. Integrating Artificial and Human Intelligence: A Partnership for Responsible Innovation in Biomedical Engineering and Medicine. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 24:247-263. [PMID: 31313972 DOI: 10.1089/omi.2019.0038] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Historically, the term "artificial intelligence" dates to 1956 when it was first used in a conference at Dartmouth College in the US. Since then, the development of artificial intelligence has in part been shaped by the field of neuroscience. By understanding the human brain, scientists have attempted to build new intelligent machines capable of performing complex tasks akin to humans. Indeed, future research into artificial intelligence will continue to benefit from the study of the human brain. While the development of artificial intelligence algorithms has been fast paced, the actual use of most artificial intelligence (AI) algorithms in biomedical engineering and clinical practice is still markedly below its conceivably broader potentials. This is partly because for any algorithm to be incorporated into existing workflows it has to stand the test of scientific validation, clinical and personal utility, application context, and is equitable as well. In this context, there is much to be gained by combining AI and human intelligence (HI). Harnessing Big Data, computing power and storage capacities, and addressing societal issues emergent from algorithm applications, demand deploying HI in tandem with AI. Very few countries, even economically developed states, lack adequate and critical governance frames to best understand and steer the AI innovation trajectories in health care. Drug discovery and translational pharmaceutical research stand to gain from AI technology provided they are also informed by HI. In this expert review, we analyze the ways in which AI applications are likely to traverse the continuum of life from birth to death, and encompassing not only humans but also all animal, plant, and other living organisms that are increasingly touched by AI. Examples of AI applications include digital health, diagnosis of diseases in newborns, remote monitoring of health by smart devices, real-time Big Data analytics for prompt diagnosis of heart attacks, and facial analysis software with consequences on civil liberties. While we underscore the need for integration of AI and HI, we note that AI technology does not have to replace medical specialists or scientists and rather, is in need of such expert HI. Altogether, AI and HI offer synergy for responsible innovation and veritable prospects for improving health care from prevention to diagnosis to therapeutics while unintended consequences of automation emergent from AI and algorithms should be borne in mind on scientific cultures, work force, and society at large.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Medical Campus, Anzio Road, Observatory 7925, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sampson Adotey
- International Development Innovation Network, D-Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Nicholas E Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Witness Dzobo
- Pathology and Immunology Department, University Hospital Southampton, Mail Point B, Tremona Road, Southampton, UK.,University of Portsmouth, Faculty of Science, St Michael's Building, White Swan Road, Portsmouth, UK
| |
Collapse
|
27
|
Kreuzinger C, von der Decken I, Wolf A, Gamperl M, Koller J, Karacs J, Pfaffinger S, Bartl T, Reinthaller A, Grimm C, Singer CF, Braicu EI, Cunnea P, Gourley C, Smeets D, Boeckx B, Lambrechts D, Perco P, Horvat R, Berns EMJJ, Cacsire Castillo-Tong D. Patient-derived cell line models revealed therapeutic targets and molecular mechanisms underlying disease progression of high grade serous ovarian cancer. Cancer Lett 2019; 459:1-12. [PMID: 31150822 DOI: 10.1016/j.canlet.2019.05.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 01/07/2023]
Abstract
High grade serous ovarian cancer (HGSOC) is the most frequent type of ovarian cancer. Most patients have primary response to platinum-based chemotherapy but frequently relapse, which leads to patient death. A lack of well documented and characterized patient-derived HGSOC cell lines is so far a major barrier to define tumor specific therapeutic targets and to study the molecular mechanisms underlying disease progression. We established 34 patient-derived HGSOC cell lines and characterized them at cellular and molecular level. Particularly, we demonstrated that a cancer-testis antigen PRAME and Estrogen Receptor could serve as therapeutic targets. Notably, data from the cell lines did not demonstrate acquired resistance due to tumor recurrence that matched with clinical observations. Finally, we presented that all HGSOC had no or very low CDKN1A (p21) expression due to loss of wild-type TP53, suggesting that loss of cell cycle control is the determinant for tumorigenesis and progression. In conclusion, patient-derived cell lines reveal that PRAME is a potential tumor specific therapeutic target in HGSOC and counteracting the down-regulation of p21 caused by loss of wild-type TP53 might be the key to impede disease progression.
Collapse
Affiliation(s)
- Caroline Kreuzinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Isabel von der Decken
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Andrea Wolf
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Magdalena Gamperl
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Julia Koller
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Jasmine Karacs
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Stephanie Pfaffinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Bartl
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Alexander Reinthaller
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Christoph Grimm
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Christian F Singer
- Department of Gynecology and Gynecologic Oncology, Gynecologic Cancer Unit, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Elena Ioana Braicu
- Tumor Bank Ovarian Cancer Network, Department of Gynecology, Charité Universitätsmedizin Berlin, 13353, Berlin, Germany; Department of Gynecology, Charité Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Paula Cunnea
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, W12 0HS, United Kingdom
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XR, United Kingdom
| | - Dominiek Smeets
- KU Leuven, Department of Human Genetics, Laboratory for Translational Genetics, 3000, Leuven, Belgium; VIB, VIB Center for Cancer Biology, Laboratory for Translational Genetics, 3000, Leuven, Belgium
| | - Bram Boeckx
- KU Leuven, Department of Human Genetics, Laboratory for Translational Genetics, 3000, Leuven, Belgium; VIB, VIB Center for Cancer Biology, Laboratory for Translational Genetics, 3000, Leuven, Belgium
| | - Diether Lambrechts
- KU Leuven, Department of Human Genetics, Laboratory for Translational Genetics, 3000, Leuven, Belgium; VIB, VIB Center for Cancer Biology, Laboratory for Translational Genetics, 3000, Leuven, Belgium
| | - Paul Perco
- Emergentec Biodevelopment GmbH, 1180, Vienna, Austria
| | - Reinhard Horvat
- Department of Clinical Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - Els M J J Berns
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3000 CA, Rotterdam, the Netherlands
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
28
|
Meshcheryakova A, Svoboda M, Jaritz M, Mungenast F, Salzmann M, Pils D, Cacsire Castillo-Tong D, Hager G, Wolf A, Braicu EI, Sehouli J, Lambrechts S, Vergote I, Mahner S, Birner P, Zimmermann P, Brindley DN, Heinze G, Zeillinger R, Mechtcheriakova D. Interrelations of Sphingolipid and Lysophosphatidate Signaling with Immune System in Ovarian Cancer. Comput Struct Biotechnol J 2019; 17:537-560. [PMID: 31049165 PMCID: PMC6479272 DOI: 10.1016/j.csbj.2019.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/03/2019] [Accepted: 04/05/2019] [Indexed: 12/16/2022] Open
Abstract
The sphingolipid and lysophosphatidate regulatory networks impact diverse mechanisms attributed to cancer cells and the tumor immune microenvironment. Deciphering the complexity demands implementation of a holistic approach combined with higher-resolution techniques. We implemented a multi-modular integrative approach consolidating the latest accomplishments in gene expression profiling, prognostic/predictive modeling, next generation digital pathology, and systems biology for epithelial ovarian cancer. We assessed patient-specific transcriptional profiles using the sphingolipid/lysophosphatidate/immune-associated signature. This revealed novel sphingolipid/lysophosphatidate-immune gene-gene associations and distinguished tumor subtypes with immune high/low context. These were characterized by robust differences in sphingolipid-/lysophosphatidate-related checkpoints and the drug response. The analysis also nominates novel survival models for stratification of patients with CD68, LPAR3, SMPD1, PPAP2B, and SMPD2 emerging as the most prognostically important genes. Alignment of proprietary data with curated transcriptomic data from public databases across a variety of malignancies (over 600 categories; over 21,000 arrays) showed specificity for ovarian carcinoma. Our systems approach identified novel sphingolipid-lysophosphatidate-immune checkpoints and networks underlying tumor immune heterogeneity and disease outcomes. This holds great promise for delivering novel stratifying and targeting strategies.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- Molecular Systems Biology and Pathophysiology Research Group, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Martin Svoboda
- Molecular Systems Biology and Pathophysiology Research Group, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Markus Jaritz
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Felicitas Mungenast
- Molecular Systems Biology and Pathophysiology Research Group, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Martina Salzmann
- Molecular Systems Biology and Pathophysiology Research Group, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Sectionfor Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gudrun Hager
- Molecular Oncology Group, Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Gynecologic Cancer Unit, Medical University of Vienna, Vienna, Austria
| | - Andrea Wolf
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Elena Ioana Braicu
- Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Gynecology, Berlin, Germany
| | - Jalid Sehouli
- Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Gynecology, Berlin, Germany
| | - Sandrina Lambrechts
- Division of Gynecologic Oncology, University Hospital Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Ignace Vergote
- Division of Gynecologic Oncology, University Hospital Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sven Mahner
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Birner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - David N. Brindley
- Cancer Research Institute of Northern Alberta, Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Georg Heinze
- Sectionfor Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Robert Zeillinger
- Molecular Oncology Group, Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Gynecologic Cancer Unit, Medical University of Vienna, Vienna, Austria
| | - Diana Mechtcheriakova
- Molecular Systems Biology and Pathophysiology Research Group, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
Barger CJ, Zhang W, Sharma A, Chee L, James SR, Kufel CN, Miller A, Meza J, Drapkin R, Odunsi K, Klinkebiel D, Karpf AR. Expression of the POTE gene family in human ovarian cancer. Sci Rep 2018; 8:17136. [PMID: 30459449 PMCID: PMC6244393 DOI: 10.1038/s41598-018-35567-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/07/2018] [Indexed: 12/23/2022] Open
Abstract
The POTE family includes 14 genes in three phylogenetic groups. We determined POTE mRNA expression in normal tissues, epithelial ovarian and high-grade serous ovarian cancer (EOC, HGSC), and pan-cancer, and determined the relationship of POTE expression to ovarian cancer clinicopathology. Groups 1 & 2 POTEs showed testis-specific expression in normal tissues, consistent with assignment as cancer-testis antigens (CTAs), while Group 3 POTEs were expressed in several normal tissues, indicating they are not CTAs. Pan-POTE and individual POTEs showed significantly elevated expression in EOC and HGSC compared to normal controls. Pan-POTE correlated with increased stage, grade, and the HGSC subtype. Select individual POTEs showed increased expression in recurrent HGSC, and POTEE specifically associated with reduced HGSC OS. Consistent with tumors, EOC cell lines had significantly elevated Pan-POTE compared to OSE and FTE cells. Notably, Group 1 & 2 POTEs (POTEs A/B/B2/C/D), Group 3 POTE-actin genes (POTEs E/F/I/J/KP), and other Group 3 POTEs (POTEs G/H/M) show within-group correlated expression, and pan-cancer analyses of tumors and cell lines confirmed this relationship. Based on their restricted expression in normal tissues and increased expression and association with poor prognosis in ovarian cancer, POTEs are potential oncogenes and therapeutic targets in this malignancy.
Collapse
Affiliation(s)
- Carter J Barger
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Wa Zhang
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Ashok Sharma
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Linda Chee
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Smitha R James
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Christina N Kufel
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Austin Miller
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jane Meza
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, 68198-4375, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kunle Odunsi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - David Klinkebiel
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
- Department of Biochemistry, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Adam R Karpf
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA.
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
30
|
Binju M, Padilla MA, Singomat T, Kaur P, Suryo Rahmanto Y, Cohen PA, Yu Y. Mechanisms underlying acquired platinum resistance in high grade serous ovarian cancer - a mini review. Biochim Biophys Acta Gen Subj 2018; 1863:371-378. [PMID: 30423357 DOI: 10.1016/j.bbagen.2018.11.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Advanced epithelial ovarian cancer is one of the hardest human malignancies to treat. Standard treatment involves cytoreductive surgery and platinum-based chemotherapy, however, median progression-free survival for patients diagnosed with advanced stage disease (FIGO stages III and IV) is approximately 18 months. There has been little improvement in overall survival over the past decade and less than half of women with advanced stage disease will be living 5 years after diagnosis. A majority of patients initially have a favourable response to platinum-based chemotherapy, but most will eventually relapse and their disease will become platinum resistant. SCOPE OF REVIEW Here, we review our current understanding of mechanisms that promote recurrence and acquired resistance in epithelial ovarian cancer with particular focus on studies that describe differences observed between untreated primary tumors and recurrent tumors, post-first-line chemotherapy. Multiple molecular mechanisms contribute to recurrence in patients following initial treatment for advanced epithelial ovarian cancer including those involving the tumor microenvironment, tumor immune status, cancer stem cells, DNA repair/cell survival pathways and extracellular matrix. MAJOR CONCLUSIONS Due to the adaptive nature of recurrent tumors, the major contributing and specific resistance pattern may largely depend on the nature of the primary tumor itself. GENERAL SIGNIFICANCE Future work that aims to elucidate the complex pattern of acquired resistance will be useful for predicting chemotherapy response/recurrence following primary diagnosis and to develop novel treatment strategies to improve the survival of patients with advanced epithelial ovarian cancer, especially in tumors not harbouring homologous DNA recombination repair deficiencies.
Collapse
Affiliation(s)
- Mudra Binju
- School of Pharmacy and Biomedical Sciences, Curtin University, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Western Australia, Australia
| | - Monica Amaya Padilla
- School of Pharmacy and Biomedical Sciences, Curtin University, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Western Australia, Australia
| | - Terence Singomat
- School of Pharmacy and Biomedical Sciences, Curtin University, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Western Australia, Australia
| | - Pritinder Kaur
- School of Pharmacy and Biomedical Sciences, Curtin University, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Western Australia, Australia
| | - Yohan Suryo Rahmanto
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, United States
| | - Paul A Cohen
- Division of Obstetrics and Gynaecology, Faculty of Health and Medicine, University of Western Australia, Crawley, Western Australia, Australia; Department of Gynaecologic Oncology, Bendat Family Comprehensive Cancer Centre, St John of God Subiaco Hospital, Subiaco, Western Australia, Australia
| | - Yu Yu
- School of Pharmacy and Biomedical Sciences, Curtin University, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Western Australia, Australia.
| |
Collapse
|
31
|
Loessner D, Rockstroh A, Shokoohmand A, Holzapfel BM, Wagner F, Baldwin J, Boxberg M, Schmalfeldt B, Lengyel E, Clements JA, Hutmacher DW. A 3D tumor microenvironment regulates cell proliferation, peritoneal growth and expression patterns. Biomaterials 2018; 190-191:63-75. [PMID: 30396040 DOI: 10.1016/j.biomaterials.2018.10.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/09/2018] [Accepted: 10/14/2018] [Indexed: 02/07/2023]
Abstract
Peritoneal invasion through the mesothelial cell layer is a hallmark of ovarian cancer metastasis. Using tissue engineering technologies, we recreated an ovarian tumor microenvironment replicating this aspect of disease progression. Ovarian cancer cell-laden hydrogels were combined with mesothelial cell-layered melt electrospun written scaffolds and characterized with proliferation and transcriptomic analyses and used as intraperitoneal xenografts. Here we show increased cancer cell proliferation in these 3D co-cultures, which we validated using patient-derived cells and linked to peritoneal tumor growth in vivo. Transcriptome-wide expression analysis identified IGFBP7, PTGS2, VEGFC and FGF2 as bidirectional factors deregulated in 3D co-cultures compared to 3D mono-cultures, which we confirmed by immunohistochemistry of xenograft and patient-derived tumor tissues and correlated with overall and progression-free survival. These factors were further increased upon expression of kallikrein-related proteases. This clinically predictive model allows us to mimic the complexity and processes of the metastatic disease that may lead to therapies that protect from peritoneal invasion or delay the development of metastasis.
Collapse
Affiliation(s)
- Daniela Loessner
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Anja Rockstroh
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Ali Shokoohmand
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Boris M Holzapfel
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany
| | - Ferdinand Wagner
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstr. 4, 80337 Munich, Germany
| | - Jeremy Baldwin
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Melanie Boxberg
- Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany
| | - Barbara Schmalfeldt
- Gynecologic Department, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, 5841 South Maryland Avenue, MC2050, Chicago, IL 60637, USA
| | - Judith A Clements
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Dietmar W Hutmacher
- Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD 4059, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, QLD 4102, Australia; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332-0405, USA; Institute for Advanced Study, Technical University Munich, Lichtenbergstr. 2a, 85748 Garching, Germany.
| |
Collapse
|
32
|
Stanske M, Wienert S, Castillo-Tong DC, Kreuzinger C, Vergote I, Lambrechts S, Gabra H, Gourley C, Ganapathi RN, Kolaschinski I, Budczies J, Sehouli J, Ruscito I, Denkert C, Kulbe H, Schmitt W, Jöhrens K, Braicu I, Darb-Esfahani S. Dynamics of the Intratumoral Immune Response during Progression of High-Grade Serous Ovarian Cancer. Neoplasia 2018; 20:280-288. [PMID: 29466768 PMCID: PMC5852388 DOI: 10.1016/j.neo.2018.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TILs) have an established impact on the prognosis of high-grade serous ovarian carcinoma (HGSOC), however, their role in recurrent ovarian cancer is largely unknown. We therefore systematically investigated TIL densities and MHC class I and II (MHC1, 2) expression in the progression of HGSOC. EXPERIMENTAL DESIGN CD3+, CD4+, CD8+ TILs and MHC1, 2 expression were evaluated by immunohistochemistry on tissue microarrays in 113 paired primary and recurrent HGSOC. TILs were quantified by image analysis. All patients had been included to the EU-funded OCTIPS FP7 project. RESULTS CD3+, CD4+, CD8+ TILs and MHC1 and MHC2 expression showed significant correlations between primary and recurrent tumor levels (Spearman rho 0.427, 0.533, 0.361, 0.456, 0.526 respectively; P<.0001 each). Paired testing revealed higher CD4+ densities and MHC1 expression in recurrent tumors (Wilcoxon P=.034 and P=.018). There was also a shift towards higher CD3+ TILs levels in recurrent carcinomas when analyzing platinum-sensitive tumors only (Wilcoxon P=.026) and in pairs with recurrent tumor tissue from first relapse only (Wilcoxon P=.031). High MHC2 expression was the only parameter to be significantly linked to prolonged progression-free survival after first relapse (PFS2, log-rank P=.012). CONCLUSIONS This is the first study that analyzed the development of TILs density and MHC expression in paired primary and recurrent HGSOC. The level of the antitumoral immune response in recurrent tumors was clearly dependent on the one in the primary tumor. Our data contribute to the understanding of temporal heterogeneity of HGSOC immune microenvironment and have implications for selection of samples for biomarker testing in the setting of immune-targeting therapeutics.
Collapse
Affiliation(s)
- Mandy Stanske
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Stephan Wienert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany; VM Scope GmbH, Charitéplatz 1, 10117 Berlin, Germany.
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | - Caroline Kreuzinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | - Ignace Vergote
- Department of Gynecology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | | | - Hani Gabra
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, MRC IGMM, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK.
| | - Ram N Ganapathi
- Department of Cancer Pharmacology, Levine Cancer Institute, Carolinas Health Care System, 1021 Morehead Medical Drive, Charlotte, NC 28204-2839, USA.
| | - Ivonne Kolaschinski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jan Budczies
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jalid Sehouli
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Ilary Ruscito
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany; UP Cell Therapy and Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy.
| | - Carsten Denkert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Hagen Kulbe
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Wolfgang Schmitt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Korinna Jöhrens
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Ioana Braicu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Silvia Darb-Esfahani
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|